WO2020158959A1 - Anti-mc16 antibody - Google Patents

Anti-mc16 antibody Download PDF

Info

Publication number
WO2020158959A1
WO2020158959A1 PCT/JP2020/004695 JP2020004695W WO2020158959A1 WO 2020158959 A1 WO2020158959 A1 WO 2020158959A1 JP 2020004695 W JP2020004695 W JP 2020004695W WO 2020158959 A1 WO2020158959 A1 WO 2020158959A1
Authority
WO
WIPO (PCT)
Prior art keywords
annexin
cancer
terminal region
antibody
monoclonal antibody
Prior art date
Application number
PCT/JP2020/004695
Other languages
French (fr)
Japanese (ja)
Inventor
道子 福田
元裕 野中
Original Assignee
国立研究開発法人産業技術総合研究所
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 国立研究開発法人産業技術総合研究所 filed Critical 国立研究開発法人産業技術総合研究所
Publication of WO2020158959A1 publication Critical patent/WO2020158959A1/en

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [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
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/02Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving viable microorganisms
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer

Definitions

  • the present invention relates generally to the field of cancer biology, and more specifically to an antibody that specifically binds to the N-terminal region of Annexin A1 and its use.
  • brain tumors such as meningioma and schwannoma
  • originating from the meninges and the cranial/spinal nerves are benign brain tumors and can be completely cured if they can be removed by surgery.
  • neuroepithelial tumors such as gliomas are basically malignant brain tumors.
  • the prognosis is extremely poor (5-year survival rate is about 10%) even if radiation therapy and chemotherapy are performed after excision by craniotomy.
  • the presence of the blood-brain barrier is a major reason why effective chemotherapy has not been implemented for brain tumors.
  • Non-Patent Document 1 Non-Patent Document 1
  • a target peptide for malignant tumor blood vessels IF7, dTIT7, etc.
  • IF7, dTIT7, etc. a target peptide for malignant tumor blood vessels that accumulates at a relatively high concentration and can actively cross the wall of vascular endothelium
  • a drug delivery system a target peptide for malignant tumor blood vessels (IF7, dTIT7, etc.) that accumulates at a relatively high concentration and can actively cross the wall of vascular endothelium.
  • the peptide intravenously injected into a tumor-bearing mouse model targets malignant tumor blood vessels, it is considered that the N-terminal region of annexin A1 is actually expressed on the surface of malignant tumor vascular endothelium.
  • the N-terminal region of annexin A1 which is a receptor for IF7 and dTIT7, needs to be present on the surface of vascular endothelium in order for the anticancer drug bound by IF7 and dTIT7 developed by the present inventors to exert a therapeutic effect. .. Therefore, when a patient suffering from a brain tumor or the like is treated with an anticancer agent to which the IF7 or dTIT7 is bound, the tissue of the tumor or the like is preliminarily considered from the viewpoint of the physical and economic burden of the patient.
  • a diagnostic agent capable of detecting the presence of the N-terminal region of annexin A1 on the surface of vascular endothelium is strongly desired.
  • An antibody that specifically binds to the N-terminal region of Annexin A1 that can be used for immunostaining is suitable as a diagnostic agent for detecting the presence or absence of the N-terminal region of Annexin A1 in the tumor tissue. To our knowledge, such an antibody is not commercially available and has not been reported in academic papers.
  • An object of the present invention is to provide a monoclonal antibody that specifically binds to the N-terminal region of annexin A1, a hybridoma that produces the antibody, a reagent for detecting the presence of the N-terminal region of annexin A1 containing the antibody, and a surface of a tumor blood vessel. It is intended to provide a method for detecting the presence of the N-terminal region of annexin A1 in E. coli.
  • the present inventors have found a monoclonal antibody that specifically binds to the N-terminal region of annexin A1, and found that the monoclonal antibody can positively stain vascular endothelium in immunostaining in malignant brain tumors. Heading out, the present invention has been completed.
  • the present invention is as follows.
  • a method for detecting the presence of an N-terminal region of annexin A1 on the surface of a tumor blood vessel comprising: (1) Contacting a sample derived from a subject with a monoclonal antibody that specifically binds to the N-terminal region of annexin A1 according to any one of [1] to [3] (2) In the sample , Detecting the N-terminal region of Annexin A1 on the surface of tumor blood vessels by immunostaining.
  • a method for assisting selection of an application target of cancer treatment using a conjugate of a peptide that binds to the N-terminal region of annexin A1 and an anticancer agent (1) Contacting a sample derived from a subject with a monoclonal antibody that specifically binds to the N-terminal region of annexin A1 according to any one of [1] to [3] (2) In the sample , Detecting the N-terminal region of Annexin A1 on the surface of tumor blood vessels by immunostaining.
  • a method for treating cancer which comprises using a conjugate of a peptide that binds to the N-terminal region of annexin A1 and an anticancer agent, (1) Contacting a sample derived from a subject with a monoclonal antibody that specifically binds to the N-terminal region of annexin A1 according to any one of [1] to [3] (2) In the sample Detecting the N-terminal region of annexin A1 on the surface of the tumor blood vessel by immunostaining (3) selecting an application target of the treatment based on the detection result (4) selecting annexin A1 A method comprising administering a conjugate of an anticancer drug and a peptide that binds to the N-terminal region.
  • the subject-derived sample is a tissue section prepared using a biopsy sample containing cancer cells collected by biopsy from the subject. The method described.
  • the cancer is solid cancer or liquid cancer.
  • a monoclonal antibody that specifically binds to the N-terminal region of annexin A1, a hybridoma producing the antibody, a reagent containing the antibody for detecting the presence of the N-terminal region of annexin A1, a surface of a tumor blood vessel It is possible to provide a method for detecting the presence of the N-terminal region of Annexin A1 in the above.
  • FIG. 1 shows the results of measuring the binding of anti-MC16 antibody (clone 31-8D) to human annexin A1 by ELISA.
  • FIG. 2 shows the results of detection of binding of anti-MC16 antibody to human annexin A1 and a mutant of human annexin A1 lacking the N-terminal domain by Western blotting.
  • FIG. 3 shows the results of fluorescent immunostaining images of mouse vascular endothelial F2 cells with anti-MC16 antibody.
  • FIG. 4 shows the results of immunostaining of glioblastoma tissue with anti-MC16 antibody (left: immunostaining, right: hematoxylin-eosin staining).
  • FIG. 1 shows the results of measuring the binding of anti-MC16 antibody (clone 31-8D) to human annexin A1 by ELISA.
  • FIG. 2 shows the results of detection of binding of anti-MC16 antibody to human annexin A1 and a mutant of human annexin A1 lacking the
  • FIG. 5 shows the results of immunostaining of various cancer tumor tissues (breast cancer) with anti-MC16 antibody and anti-annexin A1 antibody commercially available from Invitrogen.
  • tissue specimen a multiple cancer tissue micro array from Biomax was used.
  • FIG. 6 shows the results of immunostaining of various cancer tumor tissues (lung cancer) with anti-MC16 antibody and anti-annexin A1 antibody marketed by Invitrogen.
  • tissue specimen a multiple cancer tissue micro array from Biomax was used.
  • FIG. 7 shows the results of immunostaining of various cancer tumor tissues (hepatocellular carcinoma) with anti-MC16 antibody and anti-annexin A1 antibody marketed by Invitrogen.
  • tissue specimen a multiple cancer tissue micro array from Biomax was used.
  • FIG. 8 shows the results of immunostaining of various cancer tumor tissues (ovarian cancer) with anti-MC16 antibody and anti-annexin A1 antibody marketed by Invitrogen.
  • tissue specimen a multiple cancer tissue micro array from Biomax was used.
  • the present invention provides a monoclonal antibody that specifically binds to the N-terminal region of annexin A1 (RefSeq No. NP — 000691.1) (hereinafter sometimes referred to as “ANXA1” or “Anxa1”). .. Specifically, the antibody is an antibody that specifically binds to an epitope that is present in the N-terminal region of annexin A1 and that contains the amino acid sequence represented by SEQ ID NO: 1 (MAMVSEFLKQAWFIE).
  • the antibody may belong to any immunoglobulin class of IgG, IgA, IgM, IgD or IgE, but is preferably IgG, more preferably IgG2b.
  • Specific examples of the antibody include an antibody (mouse IgG2b) produced by the monoclonal antibody-producing hybridoma (clone 31-8D) of the present invention described below (hereinafter sometimes referred to as "anti-MC16 antibody").
  • the monoclonal antibody of the present invention specifically binds to the N-terminal region of Annexin A1 expressed on the blood side of neovascular endothelial cells formed in (malignant) tumors, the presence or absence of the N-terminal region of Annexin A1 is checked. Suitable for detecting. Therefore, the monoclonal antibody of the present invention is, for example, a cancer treatment comprising a conjugate containing a peptide that binds to the N-terminal region of annexin A1 and an anticancer agent, and the conjugate and a pharmaceutically acceptable carrier. It can be used for companion diagnosis for predicting whether or not a subject is an application target of the treatment before performing cancer treatment using the composition for treatment and the like.
  • a subject is any animal that expresses annexin A1, particularly a mammal.
  • mammals include, for example, mice, rats, hamsters, experimental animals such as rodents and rabbits such as guinea pigs, pigs, cows, goats, horses, sheep, mink and other domestic animals, dogs, pets such as cats, humans, and the like.
  • rodents and rabbits such as guinea pigs, pigs, cows, goats, horses, sheep, mink and other domestic animals, dogs, pets such as cats, humans, and the like.
  • primates such as monkeys, rhesus monkeys, marmosets, orangutans, and chimpanzees.
  • the monoclonal antibody of the present invention and an antibody similar thereto can be prepared by a method known per se (phage display method, hybridoma method).
  • phage display method hybridoma method
  • an immunogenic antigenic peptide is subcutaneously or intramuscularly administered to a mammal such as mouse, rat or hamster. Immunization is carried out by injecting once or several times into a vein, a hood, an abdominal cavity or the like, or by transplanting.
  • antibody-producing cells are isolated from the immunized mammal, the cells are fused with myeloma cells to form a hybridoma, the hybridoma is cloned, and immunogenic used for immunization of the mammal. It is produced by selecting a clone that produces an antibody showing a specific affinity for the antigenic peptide. Further, as the antibody-producing cells, antibody-producing cells produced by reacting previously isolated spleen cells, lymphocytes, etc. with an immunogenic antigen peptide in a culture medium can also be used. In this case, human-derived antibody-producing cells can also be prepared.
  • the immunogenic antigen peptide can be used for immunization by diluting it with PBS (Phosphate-Buffered Saline) or physiological saline at an appropriate dilution ratio.
  • the immunogenic antigenic peptide may be administered together with an adjuvant, for example, it may be mixed with complete or incomplete Freund's adjuvant (FCA or FIA) and emulsified.
  • FCA or FIA complete or incomplete Freund's adjuvant
  • a suitable carrier may be used when immunizing with an immunogenic antigen.
  • a carrier protein such as albumin or keyhole limpet hemocyanin.
  • synthetic peptide in which cysteine is added to the C-terminal of the amino acid sequence from the N-terminal to the 15th amino acid of human annexin A1 (SEQ ID NO: 1 MAMVSEFLKQAWFIE) (hereinafter, “synthetic peptide”).
  • synthetic peptide A mixture of bovine serum albumin-bound substance and complete Freund's adjuvant was used as an immunogenic antigen peptide.
  • Hybridomas that secrete monoclonal antibodies can be prepared according to the method of Kohler and Milstein (Nature, Vol. 256, pp. 495-497, 1975) and its modifications. That is, the monoclonal antibody of the present invention, splenocytes, lymph node cells, peripheral lymphocytes, myeloma cells, tonsil cells, etc.
  • myeloma cells used for cell fusion include mouse-derived myeloma P3/X63-AG8, P3/NSI/1-Ag4-1, P3/X63-Ag8. U1, SP2/0-Ag14, F0 or BW5147, rat-derived myeloma 210RCY3-Ag1.2.3. , Human-derived myeloma U-266AR1, GML500-6TG-A1-2, UC729-6, CEM-AGR, D1R11 or CEM-T15.
  • the screening of the hybridoma clone producing the monoclonal antibody of the present invention is carried out by culturing the hybridoma in, for example, a microtiter plate, and measuring the reactivity of the culture supernatant in the wells in which proliferation is observed against the immunogenic antigenic peptide.
  • the measurement can be performed by ELISA, Western blotting, immunostaining, or the like.
  • a hybridoma that produced an antibody showing high reactivity with the synthetic peptide MC16 was subjected to cell cloning by the limiting dilution method to obtain a monoclonal antibody-producing hybridoma of the present invention (clone 31-8D).
  • Clone 31-8D will be placed on October 24, 2018, under the accession number NITE P-02801, at the Patent Microorganism Depositary Center (2-5-8, Kazusa Kamafoot, Kisarazu City, Chiba Prefecture) under the National Institute for Product Evaluation Technology, National Institute for Product Evaluation Technology. Deposited.
  • the isolation and purification of the monoclonal antibody is carried out by subjecting the antibody-containing culture supernatant or ascites produced by the above-mentioned method to, for example, ion exchange chromatography, anti-immunoglobulin column or affinity column chromatography such as protein A or protein G column. It can be performed by attaching it to a graph.
  • the monoclonal antibody of the present invention may be labeled with a detectable label.
  • the label include fluorescent label, low molecular weight compound label, peptide label, enzyme label and the like.
  • the monoclonal antibody of the present invention may form a conjugate with other drugs.
  • the monoclonal antibody of the present invention is not limited to the above-mentioned production method, and may be obtained by any method.
  • a gene cloned from the hybridoma of the present invention which encodes the monoclonal antibody of the present invention, may be expressed as an antibody by incorporating the gene into a suitable vector and introducing it into a host.
  • Methods for the isolation of antibody-encoding genes, their introduction into vectors, and the transformation of host cells have been established (eg, Vandamme, AM et al. Eur. J. Biochem. (1990). 192, 767-775).
  • RNA is usually extracted from the hybridoma.
  • guanidine ultracentrifugation method Chirgwin, JM, et al., Biochemistry (1979) 18, 5294-5299
  • AGPC method Chemczynski, P., et al., Anal. Biochem. (1987) 162, 156-159
  • the extracted mRNA can be purified using mRNA Purification Kit (manufactured by GE Healthcare Bioscience).
  • kits for directly extracting total mRNA from cells such as QuickPrep mRNA Purification Kit (manufactured by GE Healthcare Bioscience), is commercially available. Such a kit can also be used to obtain total mRNA from hybridomas.
  • CDNA encoding the antibody V region can be synthesized from the obtained mRNA using reverse transcriptase.
  • the cDNA can be synthesized by AMV Reverse Transcriptase First-strand cDNA Synthesis Kit (manufactured by Seikagaku Corporation).
  • the desired cDNA fragment is purified from the resulting PCR product and then operably linked to vector DNA.
  • a desired recombinant vector can be prepared from E. coli forming the colony. Whether or not the recombinant vector has the nucleotide sequence of the desired cDNA can be confirmed by a method known per se, such as the dideoxynucleotide chain termination method.
  • a monoclonal antibody has a sugar chain having a different structure depending on the type of mammal to be immunized, but the monoclonal antibody in the present invention is not limited by the structural difference of the sugar chain, and a monoclonal antibody derived from any mammal It also includes. Furthermore, for example, a recombinant human monoclonal antibody obtained from a transgenic animal into which a human immunoglobulin gene has been incorporated, or a constant region (Fc) of a monoclonal antibody derived from a certain mammal is recombined with the Fc region of a human-derived monoclonal antibody.
  • Fc constant region
  • chimeric monoclonal antibodies are also included in the monoclonal antibody of the present invention.
  • chimeric monoclonal antibodies in which the entire region other than the complementarity determining region (CDR) that can directly bind complementarily with an antigen is recombined with the corresponding region of a human-derived monoclonal antibody. To be done.
  • CDR complementarity determining region
  • the antibodies of the present invention include natural antibodies such as the above-mentioned monoclonal antibody (mAb), chimeric antibodies that can be produced by gene recombination technology, humanized antibodies and single chain antibodies, and these antibodies. Fragment of.
  • the antibody fragment means a partial region of the aforementioned antibody having specific binding activity, and specifically includes Fab, Fab′, F(ab′) 2 , scAb, scFv or scFv-Fc and the like. ..
  • antibody fragments can be obtained by a method known per se, and specifically, the monoclonal antibody of the present invention is treated with an enzyme such as papain or pepsin to produce an antibody fragment, or these antibodies are produced.
  • a gene encoding the fragment may be constructed, introduced into an expression vector, and then expressed in an appropriate host cell. The obtained fragment can be evaluated for reactivity with an antigen, specificity and the like in the same manner as for the monoclonal antibody of the present invention.
  • a fusion antibody of the antibody or fragment of the present invention with another peptide or protein, or a modified antibody having a modifying agent bound thereto.
  • Other peptides or proteins used for fusion are not particularly limited as long as they do not reduce the binding activity of the antibody, and examples thereof include human serum albumin, various tag peptides, artificial helix motif peptides, maltose binding protein, glutathione S transferase, Examples include various toxins and peptides or proteins capable of promoting multimerization.
  • the modifying agent used for modification is not particularly limited as long as it does not reduce the binding activity of the antibody, and examples thereof include polyethylene glycol, sugar chains, phospholipids, liposomes, and low molecular weight compounds.
  • the monoclonal antibody of the present invention specifically binds to the N-terminal region of annexin A1 expressed on the blood side of neovascular endothelial cells formed in (malignant) tumors, it can be targeted to malignant tumors. It is suitable for detecting the presence or absence of the N-terminal region of annexin A1, which is a receptor for therapeutic agents for malignant tumors using active IF7 or dTIT7. Therefore, the present invention also provides a reagent for detecting the presence of the N-terminal region of Annexin A1, which comprises the monoclonal antibody of the present invention.
  • the monoclonal antibody of the present invention can be used for companion diagnosis for predicting whether or not a specific cancer treatment is applied. Therefore, the present invention provides a conjugate comprising a peptide that binds to the N-terminal region of annexin A1 and the anticancer agent, which comprises the monoclonal antibody of the present invention, and a composition comprising the conjugate and a pharmaceutically acceptable carrier. Also provided is a reagent for determining the applicability of cancer treatment using the above.
  • malignant tumor may be any type of cancer, and may be solid cancer or liquid cancer.
  • the solid cancer preferably includes a solid cancer that expresses annexin A1 on the cell surface, and thus an angiogenic solid cancer.
  • Examples of solid cancer include brain/nervous system cancer (for example, brain tumor, spinal cord tumor, etc.), head and neck cancer (for example, laryngeal cancer, oral cancer, salivary gland cancer, sinus cancer, thyroid cancer).
  • gastrointestinal cancer for example, gastric cancer, esophageal cancer, small intestine cancer, colon cancer, rectal cancer, anal cancer, liver cancer, biliary tract cancer, pancreatic cancer, etc.
  • urinary organ or Reproductive organ cancer eg, renal cancer, renal cell cancer, bladder cancer, prostate cancer, renal pelvis and ureteral cancer, gallbladder cancer, bile duct cancer, testicular cancer, penis cancer, uterine cancer
  • Endometrial cancer uterine sarcoma, cervical cancer, vaginal cancer, vulvar cancer, ovarian cancer, fallopian tube cancer, etc.
  • respiratory cancer eg lung cancer (small cell lung cancer, non Small cell lung cancer, including metastatic lung cancer), bronchial cancer, etc.
  • breast cancer eg, skin cancer (eg, malignant melanoma), bone cancer (eg, osteosarcoma), muscle cancer (eg, Rhabdomyosarcoma) and the like.
  • Liquid cancers include leukemia, malignant lymphoma, multiple myeloma, myelodysplastic syndrome, etc.
  • leukemia include acute myelogenous leukemia, acute lymphocytic leukemia, chronic myelogenous leukemia, chronic lymphocytic leukemia and the like.
  • Malignant lymphoma is classified into Hodgkin lymphoma and non-Hodgkin lymphoma.
  • non-Hodgkin lymphoma B-cell lymphoma, adult T-cell lymphoma, lymphoblastic lymphoma, diffuse large cell lymphoma, Burkitt lymphoma, follicular lymphoma , MALT lymphoma, peripheral T cell lymphoma, mantle cell lymphoma and the like.
  • a peptide that binds to the N-terminal region of annexin A1 administered to a subject, a conjugate of an anticancer agent, or the conjugate and a pharmaceutically acceptable carrier are used.
  • the composition containing the compound can efficiently cross the blood-brain tumor barrier, and therefore a brain tumor can be mentioned as a particularly preferable target.
  • the brain tumor may be a primary brain tumor or a metastatic brain tumor.
  • the brain tumor may be benign (eg, meningioma, pituitary adenoma, schwannoma, etc.) or malignant brain tumor, preferably malignant brain tumor.
  • Malignant brain tumors include grade 2 brain tumors such as astrocytoma (astrocytoma) and oligodendrogliomas, anaplastic astrocytoma, anaplastic oligodendroglioma, anaplastic Examples thereof include grade 3 brain tumors such as oligoastrocytoma, and grade 4 brain tumors such as glioblastoma.
  • the reagent of the present invention may consist only of the monoclonal antibody of the present invention or may contain a pharmaceutically acceptable carrier.
  • a pharmaceutically acceptable carrier when the reagent of the present invention is prepared as a liquid agent, various carriers commonly used as a formulation material, for example, diluents, solvents, solubilizers, suspending agents, isotonic agents, etc. Agents, buffers, etc. may be included. Further, a surfactant such as Tween 20 (registered trademark) or Tween 80 (registered trademark) may be added to prevent antibody aggregation.
  • the diluent include water and physiological saline.
  • the solvent include water, physiological saline, ethanol and the like.
  • solubilizing agent examples include cyclodextrins.
  • suspending agent examples include gum arabic and carmellose.
  • isotonicity agent examples include inorganic salts such as sodium chloride and potassium chloride, carbohydrates such as glycerin, mannitol and sorbitol.
  • the buffer examples include phosphate buffer, acetate buffer, borate buffer, carbonate buffer, citrate buffer, Tris buffer and the like. Those skilled in the art can appropriately determine the blending ratio of these carriers.
  • the monoclonal antibody of the present invention specifically binds to the N-terminal region of annexin A1 expressed on the blood side of neovascular endothelial cells formed in (malignant) tumors. It is suitable for detecting the presence or absence of the N-terminal region. Therefore, the present invention provides a method for detecting the presence of the N-terminal region of annexin A1 (annexin A1 on the surface of tumor blood vessels) expressed on the blood side of neovascular endothelial cells formed in a tumor using the monoclonal antibody. Also provide.
  • the method of the present invention comprises (1) contacting a sample derived from a subject with a monoclonal antibody of the present invention that specifically binds to the N-terminal region of annexin A1, and (2) in the sample Detecting the N-terminal region of annexin A1 (annexin A1 on the surface of tumor blood vessels) expressed on the blood side of neovascular endothelial cells formed in the tumor by immunostaining.
  • the monoclonal antibody of the present invention can be used for companion diagnosis for predicting whether or not a specific cancer treatment is applied.
  • the present invention provides a conjugate comprising a peptide that binds to the N-terminal region of annexin A1 and an anticancer agent using the monoclonal antibody, a composition containing the conjugate and a pharmaceutically acceptable carrier, and the like.
  • the present invention also provides a method for selecting an application target of cancer treatment using the method or a method for assisting the selection.
  • the present invention provides a composition comprising the conjugate selected from a peptide that binds to the N-terminal region of annexin A1 and an anticancer agent, and the conjugate and a pharmaceutically acceptable carrier in the subject selected as described above. It may include actually treating cancer by administering a substance or the like.
  • the method of the present invention comprises (1) contacting a sample derived from a subject with a monoclonal antibody of the present invention that specifically binds to the N-terminal region of annexin A1, and (2) in the sample Detecting the N-terminal region of annexin A1 (annexin A1 on the surface of tumor blood vessels) expressed on the blood side of neovascular endothelial cells formed in the tumor by immunostaining.
  • the measuring means for detecting the presence of the N-terminal region of Annexin A1 using the monoclonal antibody of the present invention is not particularly limited, but an immunostaining method is preferably used.
  • Specific means using the immunostaining method include immunohistochemical staining (IHC) method, flow cytometry analysis, mass cytometry analysis and the like.
  • Immunohistochemical staining can be performed according to a method known per se (for example, the revised fourth edition Watanabe-Nakone enzyme antibody method, published by Interdisciplinary Planning Co., Ltd., edited by Hiroshi Nagura, Yoshiyuki Nagamura, Hiroshi Tsutsumi, ISBN4-906514).
  • -73-X C304 preferably the enzyme antibody method (Enzyme labeled antibody method) and the fluorescent antibody method are used.
  • the N-terminal region of annexin A1 is detected with an enzyme-labeled antibody. Labeling may be done directly or indirectly using a secondary antibody.
  • Various improvements have been made to increase the detection sensitivity, and very high sensitivity is achieved with improved methods such as the ABC method using biotin-streptavidin and the tyramide method (TSA method, Tyramide Signal Amplification) using tyramide (tylamide). realizable.
  • HRP horseradish peroxidase
  • AP alkaline phosphatase
  • a cell sample deparaffinized or fixed as necessary is prepared, and if necessary, pretreatment (for example, decolorization or removal treatment of vital dye, blocking treatment) or antigen activation (heat bath, microwave treatment, autoclave) Treatment, protease treatment, etc., and react with primary antibody (for example, 0.5° C. to 24 hours at 4° C. to room temperature).
  • primary antibody for example, 0.5° C. to 24 hours at 4° C. to room temperature.
  • secondary antibody for example, 4° C.
  • the cell sample is washed at room temperature for 5 to 120 minutes).
  • a color-developing substrate such as diaminobenzidine (3,3-diaminobenzidine:DAB) is added to develop the color.
  • counterstaining such as hematoxylin staining may be further performed.
  • the N-terminal region of annexin A1 may be detected as a fluorescent antibody method by using an antibody labeled with a fluorescent dye instead of the enzyme labeling by the above-mentioned enzyme antibody method. Also in the fluorescent antibody method, labeling may be performed directly or indirectly using a secondary antibody.
  • Take an image of the immunostained sample using an optical microscope equipped with an imaging device or a fluorescence microscope (in the case of fluorescence staining), and save it in a computer as a digital image.
  • the digital image may be two-dimensional data or three-dimensional data, but two-dimensional data can be sufficiently analyzed in the present invention. Image processing such as filtering may be performed before the analysis.
  • Image analysis of immunostained samples can be performed using image analysis software installed in the microscope used and other commercially available software.
  • An example of commercially available software is ScanScope (registered trademark, Aperio Inc.).
  • Classification of immunostaining intensity can follow standard criteria by pathologists for immunohistochemical staining, such as score 0 (undetected), score +1 (weak), score +2 (moderate), and score +3 (significant). ) May be classified. Further, for example, when the score is +1 or more, a conjugate of a peptide that binds to the N-terminal region of annexin A1 and an anticancer agent, a composition containing the conjugate and a pharmaceutically acceptable carrier, and the like were used. It may be judged that the cancer treatment is applied.
  • a sample derived from a subject can be contacted with a monoclonal antibody of the present invention that specifically binds to the N-terminal region of Annexin A1 to detect the presence or absence of the N-terminal region of Annexin A1.
  • a monoclonal antibody of the present invention that specifically binds to the N-terminal region of Annexin A1 to detect the presence or absence of the N-terminal region of Annexin A1. It is not particularly limited as long as it is one, but is preferably a tissue section prepared by using a biopsy sample containing cancer cells collected by biopsy from a subject, and more preferably a paraffin-embedded tissue. It is a section.
  • Examples of the peptide that binds to the N-terminal region of annexin A1 used in the method of the present invention include, for example, the following (I) to (III Peptide containing any of the amino acid sequences of: (I) Amino acid sequence of (X1)[D]P[D](X2)[D] (wherein X1 represents W or F, X2 represents S or T), (II) P[D]T[D](X) n F[D] amino acid sequence (wherein (X) n represents n arbitrary amino acids selected independently from each other, and n is Indicates an integer of 0 to 4), (III) An amino acid sequence which is retro-inverso of the amino acid sequence of (I) or (II) above can be mentioned.
  • the peptide used in the method of the present invention is, for example, a peptide containing any of the following amino acid sequences (i) to (vii): (I) the amino acid sequence of T[D]I[D]T[D]W[D]P[D]T[D]M[D], (Ii) the amino acid sequence of L[D]R[D]F[D]P[D]T[D]V[D]L[D], (Iii) L[D]L[D]S[D]W[D]P[D]S[D]A[D] amino acid sequence, (Iv) the amino acid sequence of S[D]P[D]T[D]S[D]L[D]L[D]F[D], (V) the amino acid sequence of M[D]P[D]T[D]L[D]T[D]F[D]R[D], (Vi) an amino acid sequence having one or several amino acid insertions, substitutions or deletions, or a combination thereof in the
  • amino acid sequence of a chain peptide is described with the N-terminal side on the left side and the C-terminal side on the right side according to the convention of peptide notation.
  • each amino acid symbol immediately after the symbol [D] in the amino acid sequence represents the D-form of the amino acid, and each amino acid symbol without immediately following the symbol [D] in the amino acid sequence is uncontextual. Unless otherwise indicated, the L form of the amino acid is shown.
  • amino acid sequence of (I) above is W[D]P[D]S[D], W[D]P[D]T[D], F[D]P[D]S[D], or F It may be any of [D]P[D]T[D].
  • n is 0 to 4, preferably 2 to 3.
  • X may be any amino acid selected independently of each other.
  • the peptide used in the method of the present invention may consist of any of the amino acid sequences of (I) to (III) and (i) to (vii) described above, or the N-terminal side of these sequences. And/or one or more amino acids may be added to the C-terminal side.
  • the peptide used in the method of the present invention refers to a peptide in which two or more amino acids are bound to each other.
  • the length of the peptide of the present invention is not particularly limited, but for example, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 11 , At least 12, at least 13, at least 14, or at least 15 amino acids.
  • the peptides used in the method of the present invention include up to 50, 45, 40, 35, 30, 25, 20, 19, 18, 18 and 17 peptides. , Up to 16, up to 15, up to 14, up to 13, up to 12, up to 11, up to 10, up to 9, up to 8, or up to 7 amino acids Good.
  • the peptide used in the method of the present invention may be composed of a combination of D-type amino acids and L-type amino acids. Further, the peptide used in the method of the present invention is 37 C.I. F. R. It may or may not contain modified or unusual amino acids, such as those mentioned in 1.821-1.822.
  • amino terminus and/or carboxy terminus of the peptide used in the method of the present invention may be modified. Further, the peptide used in the method of the present invention may have various modifications other than at the N-terminus or C-terminus.
  • substitution in the amino acid sequence of (vi) above is preferably a conservative amino acid substitution.
  • conservative amino acid substitutions are well known in the art.
  • a conservative amino acid substitution can be defined as a substitution between amino acids with similar side chain properties.
  • the retro-inverso isomer of a peptide is one in which the chirality of each amino acid residue is opposite to that of the original peptide (“inverso”) and the direction of the amino acid sequence is reversed (“Retro”).
  • the Retro-inverso isomer is known to exhibit a structure and a function similar to that of the original peptide (eg, Acc. Chem. Res., 1993, 26(5), pp 266-273 and PLoS One. 2013 Dec 2;8(12):e80390).
  • the peptide used in the method of the present invention may contain two or more sequences selected from the above (I) to (III) and (i) to (vii).
  • the peptide used in the method of the present invention is a tandem repeat of any one of the sequences (I) to (III) and (i) to (vii) (that is, identical sequence portions are directly linked to each other). Structure) may be included.
  • the peptide used in the method of the present invention may include a structure in which two or more sequences (I) to (III) and (i) to (vii) different from each other are directly linked.
  • the peptide used in the method of the present invention may constitute a multivalent peptide by a dendrimer.
  • the peptide used in the method of the present invention may be a free form or a salt form.
  • Salts of the peptides used in the method of the present invention include pharmaceutically acceptable acid addition salts and base addition salts.
  • the acid addition salt include salts with inorganic acids such as hydrochloric acid, sulfuric acid, nitric acid and phosphoric acid, and salts with organic acids such as acetic acid, malic acid, succinic acid, tartaric acid and citric acid.
  • Base addition salts include salts with alkali metals such as sodium and potassium, salts with alkaline earth metals such as calcium and magnesium, and salts with amines such as ammonium and triethylamine.
  • the peptide used in the method of the present invention is a region consisting of amino acids 1 to 15 shown in SEQ ID NO: 1 in human annexin A1, or 1 to 15 shown in SEQ ID NO: 3 (MAMVSEFLKQARFLE) in mouse annexin A1. Can bind to a region consisting of the th amino acid.
  • the peptide used in the method of the present invention is preferably less than 10 ⁇ 6 M, and more preferably less than 10 ⁇ 6 M, when the intermolecular interaction with, for example, mouse or human annexin A1 is measured using the QCM (Quartz Crystal Microbalance) method.
  • Kd value a dissociation constant (Kd value) of less than 10 ⁇ 7 M, even more preferably less than 5 ⁇ 10 ⁇ 8 M.
  • a human recombinant annexin A1 protein composed of 346 amino acid residues commercially available from ATGen (Seongnam-si, South Korea) can be used.
  • the peptide used in the method of the present invention can be produced according to a known peptide synthesis method.
  • the peptide synthesis method may be, for example, either a solid phase synthesis method or a liquid phase synthesis method.
  • a partial peptide or amino acid that can form the peptide of the present invention is condensed with a residual portion, and when the product has a protecting group, the target peptide is produced by removing the protecting group by a method known per se. You can
  • the peptide thus obtained can be purified and isolated by a known purification method.
  • the purification method include solvent extraction, distillation, column chromatography, liquid chromatography, recrystallization, a combination thereof, and the like.
  • the anticancer drug used in the method of the present invention refers to a drug intended to suppress the growth of malignant tumor (cancer).
  • the action mechanism of the anticancer agent is not particularly limited.
  • the anticancer agent may be an antimetabolite, an alkylating agent, an anticancer antibiotic, a microtubule inhibitor, a platinum preparation, a topoisomerase inhibitor, a molecular targeting drug and the like.
  • the conjugate of the present invention may contain two or more same or different anticancer agents.
  • Antimetabolites include, for example, antifolates, dihydropteroate synthase inhibitors, dihydrofolate reductase inhibitors (DHFR inhibitors), pyrimidine metabolism inhibitors, thymidylate synthase inhibitors, purine metabolism inhibitors, IMPDH inhibitors , Ribonucleotide reductase inhibitors, ribonucleotide reductase inhibitors, nucleotide analogs, L-asparaginase and the like.
  • antimetabolites include enocitabine (san-rabine), capecitabine (xeloda), carmofur (mifurol), cladribine (leustatin), gemcitabine (gemzar), cytarabine (kiloside), cytarabine ocphosphate (staracide), tegafur (tegafur).
  • alkylating agent examples include cyclophosphamide (endoxane), ifosfamide (ifomide), melphalan (alkeran), busulfan, thiotepa (tespamin), and other nitrogen mustard-based alkylating agents, nimustine (nidoran), ranimustine. (Cimerine), dacarbacin (dacarbacin), procarbacin (procarbacin hydrochloride), temozolomide (temodal), carmustine (giliadel), streptozotocin (zanocer), bendamustine (treaxin), and other nitrosourea-based alkylating agents.
  • cyclophosphamide endoxane
  • ifosfamide ifomide
  • melphalan alkeran
  • busulfan thiotepa (tespamin)
  • thiotepa tespamin
  • other nitrogen mustard-based alkylating agents
  • anticancer antibiotics include actinomycin D (cosmegen), aclarubicin (acracinone), amrubicin (calced), idarubicin (idamycin), epirubicin (epirubicin hydrochloride, famorubicin), zinostatin stimalamer (smanx). ), daunorubicin (daunomycin), doxorubicin (adriacin), pirarubicin (pinorbin, teralubicin), bleomycin (bleo), peplomycin (pepleo), mitomycin C (mitomycin), mitoxantrone (novantron), liposomal doxorubicin (doxyl), etc.
  • actinomycin D cosmegen
  • aclarubicin acracinone
  • amrubicin calced
  • idarubicin idamycin
  • epirubicin epirubicin hydrochloride
  • Microtubule inhibitors include, for example, vinca alkaloid-based microtubule polymerization inhibitors such as vinblastine (exar), vincristine (oncobin), and vindesine (fordesin), and taxane-based microtubule depolymerization such as paclitaxel (taxol) and docetaxel (taxotere). Examples include inhibitors.
  • platinum preparations include oxaliplatin (elprat), carboplatin (carboplatin, carbomerk, paraplatin), cisplatin (ie equol, conaburi, cisplatin, etc.), nedaplatin (acpra), etc.
  • topoisomerase inhibitors examples include type I topoisomerase inhibitors such as camptothecin and its derivatives (eg, irinotecan (campto), nogitecan (hycamtin), SN-38); anthracycline drugs such as doxorubicin (adriacin); etoposide.
  • type II topoisomerase inhibitors such as epipodophyllotoxin drugs such as (Lasted and bepside) and quinolone drugs such as levofloxacin (clavit) and ciprofloxacin (ciproxan).
  • molecularly targeted drugs examples include regorafenib (stibagga), cetuximab (erbitux), panitumumab (vetivics), ramucirumab (cyramza), gefitinib (iressa), erlotinib (tarseva), afatinib (diotryf), crizotinib (crizotinib).
  • Alectinib (Alesensor), Ceritinib, Lenvatinib (Lenvima), Trastuzumab (Herceptin), Lapatinib (Tykerb), Pertuzumab (Perjeta), Sunitinib (Sutent), Sorafenib (Nexavar), Axitinib (Inraitabi), Panitab (Inraitab) Opdivo), pembrolizumab, ipilimumab (Yarvoy), vemurafenib (Zelboraf), everolimus (Afinitor), temsirolimus (Torisel), rituximab (Rituxan), bevacizumab (Avastin), geldanamycin and the like.
  • the anti-cancer agent may also be an anti-angiogenic agent.
  • Anti-angiogenic agents can be those that inhibit vascular endothelial growth factor (VEGF) or other angiogenic factors, or their receptors.
  • VEGF vascular endothelial growth factor
  • Specific examples of the anti-angiogenic agent include angiostatin, endostatin, metastatin, anti-VEGF antibody (for example, Avastin), VEGFR-2 inhibitor (for example, SU5416, SU6668) and the like.
  • the mode of binding between the peptide that binds to the N-terminal region of annexin A1 in the conjugate used in the method of the present invention and one or more anticancer agents is not particularly limited.
  • the bond may be direct, or indirect via a linker or the like.
  • the binding may be covalent, non-covalent, or a combination thereof.
  • One or more anti-cancer agents may be attached, directly or indirectly, at the N-terminus, C-terminus, or other position of the peptide that binds to the N-terminal region of Annexin A1.
  • the linkage between the peptide and the anticancer drug is well known in the art, and in the conjugate of the present invention, the binding may be by any known means.
  • composition containing the conjugate used in the method of the present invention and a pharmaceutically acceptable carrier can be provided as a dosage form suitable for oral or parenteral administration.
  • a pharmaceutically acceptable carrier for example, injections, suppositories, etc. are used, and the injections are in the form of intravenous injection, subcutaneous injection, intradermal injection, intramuscular injection, drip injection, etc. Can be included. These can be prepared according to a method known per se.
  • compositions for oral administration include solid or liquid dosage forms, specifically tablets (including sugar-coated tablets, film-coated tablets), pills, granules, powders, capsules (including soft capsules), syrup. Agents, emulsions, suspensions and the like.
  • Such a composition is produced by a known method and may contain a carrier, a diluent or an excipient which is usually used in the field of formulation.
  • composition may contain other active ingredients as long as it does not cause an unfavorable interaction with the above-mentioned conjugate.
  • the above-mentioned parenteral or oral pharmaceutical composition is conveniently prepared in a unit dosage form suitable for the dose of the active ingredient.
  • dosage forms of such dosage units include tablets, pills, capsules, injections (ampoules), and suppositories.
  • the content of the peptide or conjugate is usually 1 to 500 mg per dosage unit dosage form, particularly 1 to 100 mg for injections, and preferably 10 to 250 mg for the other dosage forms. ..
  • the dose of the composition containing the conjugate used in the method of the present invention and a pharmaceutically acceptable carrier varies depending on the purpose of administration, administration subject, target disease, symptom, administration route, etc., and is appropriately set by those skilled in the art. can do.
  • Example 1 production of mouse monoclonal antibody
  • the C-terminal cysteine (Cys) of synthetic peptide MC16 was bound to a carrier protein (BSA) using a bivalent reactive reagent to prepare an immunogen.
  • BSA carrier protein
  • HRP Label for Screening C-terminal Cys of synthetic peptide MC16 was bound to horseradish-derived peroxidase (HRP) using a divalent reactive reagent to prepare an HRP label for screening.
  • mice were selected for cell fusion.
  • Collection of lymphocytes and antiserum Three days after the final immunization, the spleen was collected, lymphocytes were separated, and then frozen and stored at -80°C. At the same time, blood was collected, serum was separated, and then frozen and stored at -40°C. The obtained antiserum was used as a positive control during screening.
  • Example 2 Selection of monoclonal antibody-producing hybridoma (clone 31-8D)
  • Six highly reactive clones were selected from the clones prepared in Example 1, and the clone (#31-8D, mouse IgG2b) that gave the best results in immunostaining of cultured mouse vascular endothelial cells was selected.
  • Example 3 binding assay of antibody (anti-MC16 antibody) produced by clone 31-8D) Plate assay was performed to confirm the binding of anti-MC16 antibody to ANXA1 protein.
  • full-length human ANXA1 protein was produced in insect cells using a baculovirus vector using recombinant DNA, and the produced ANXA1 protein was attached to an ELISA plastic well.
  • a control medium (culture medium for cell culture), a control IgG (mouse IgG2b), and an anti-MC16 antibody purified with a protein A column were reacted, and then HRP-anti-mouse IgG was reacted to determine the enzymatic activity of peroxidase, It was quantified by color development using a substrate for peroxidase. In addition, the specificity of the anti-MC16 antibody was confirmed using Western blotting.
  • Example 4 (Investigation of fluorescent immunostaining) Mouse vascular endothelial cells F2 were cultured in a monolayer, an extract of a tumor formed subcutaneously in mice with mouse melanoma B16 cells was added, and the cells were cultured for 2 days. The surface of F2 cells was reacted with an anti-MC16 antibody and then with a fluorescently labeled secondary antibody. As can be seen from the results of the fluorescent immunostaining image of mouse vascular endothelial F2 cells, it was strongly suggested that the anti-MC16 antibody of the present invention also binds to mouse Anxa1 protein. It was also suggested that the B16 tumor extract may contain an active substance that induces the expression of the N-terminal domain of Annexin A1 on the surface of F2 cells (Fig. 3).
  • Example 5 (antibody peroxidase staining) Paraffin-embedded tissue sections of tumor tissue collected from patients with glioblastoma were immunostained by the peroxidase method using anti-MC16 antibody. As a result, it was confirmed that the anti-MC16 antibody positively stained vascular endothelial cells in the tumor (Fig. 4, left). The right of FIG. 4 is a hematoxylin-eosin stain.
  • Example 6 (antibody peroxidase staining) Paraffin-embedded tissue sections of prostate cancer tumor tissue were immunostained by the peroxidase method using an anti-MC16 antibody and a rabbit anti-ANXA1 antibody commercially available from Invitrogen. As a result, both antibodies stained tumor blood vessels positively, but it was revealed that the staining result of the anti-MC16 antibody was excellent with a low background (FIGS. 5 to 8).
  • the present invention is based on Japanese Patent Application No. 2019-017446 filed in Japan (filing date: February 1, 2019), and all the contents are included in the present specification.
  • the monoclonal antibody of the present invention specifically binds to the N-terminal region of Annexin A1, it is possible to detect the presence or absence of the N-terminal region of Annexin A1 on the vascular endothelial surface of a tissue such as a tumor. It can be used for selecting an application target of cancer treatment using a conjugate of an anticancer agent and a peptide that binds to the N-terminal region of annexin A1.

Abstract

The present invention provides: a monoclonal antibody capable of binding specifically to an N-terminal region of annexin A1; a hybridoma capable of producing the antibody; a reagent for detecting the presence of an N-terminal region of annexin A1, which comprises the antibody; a method for detecting the presence of an N-terminal region of annexin A1 on the surface of a tumor vasculature; and others.

Description

抗MC16抗体Anti-MC16 antibody
 本発明は、概してがん生物学の分野に関し、より詳細にはアネキシンA1のN末端領域に特異的に結合する抗体及びその用途に関する。 The present invention relates generally to the field of cancer biology, and more specifically to an antibody that specifically binds to the N-terminal region of Annexin A1 and its use.
 脳腫瘍の中でも髄膜腫や神経鞘腫など、髄膜や脳・脊髄神経から発生する腫瘍は多くが良性脳腫瘍であり、外科手術によって摘出できれば完全治癒が可能である。それに対して、神経膠腫(グリオーマ)を始めとする神経上皮性腫瘍は基本的に悪性脳腫瘍である。特にグレード4の膠芽腫(グリオブラストーマ)に至っては、頭開手術による摘出後、放射線療法と化学療法を行ったとしても、予後は極めて悪い(5年生存率は10%程度)。脳腫瘍に対して有効な化学療法が実施できていない大きな理由として、血液脳関門の存在が挙げられる。 Most brain tumors, such as meningioma and schwannoma, originating from the meninges and the cranial/spinal nerves are benign brain tumors and can be completely cured if they can be removed by surgery. In contrast, neuroepithelial tumors such as gliomas are basically malignant brain tumors. In particular, for grade 4 glioblastoma, the prognosis is extremely poor (5-year survival rate is about 10%) even if radiation therapy and chemotherapy are performed after excision by craniotomy. The presence of the blood-brain barrier is a major reason why effective chemotherapy has not been implemented for brain tumors.
 これまでに本発明者らは、腫瘍血管特異的マーカー分子の中で最も特異性が高いことが知られているアネキシンA1(非特許文献1)のN末端領域に結合することによって、脳腫瘍に圧倒的高濃度で集積し、かつ積極的に血管内皮の壁を越えることができる悪性腫瘍血管標的ペプチド(IF7、dTIT7等)を開発し、そして該ペプチドを用いた悪性腫瘍血管特異的な抗がん剤デリバリーシステムも開発してきた。また、担癌マウスモデルに静脈注射した該ペプチドは悪性腫瘍血管に標的するので、アネキシンA1のN末端領域が、実際に悪性腫瘍血管内皮表面に発現していると考えられる。 To date, the present inventors have overwhelmed brain tumors by binding to the N-terminal region of annexin A1 (Non-Patent Document 1), which is known to have the highest specificity among tumor blood vessel-specific marker molecules. Of a target peptide for malignant tumor blood vessels (IF7, dTIT7, etc.) that accumulates at a relatively high concentration and can actively cross the wall of vascular endothelium, and uses the peptide to malignant tumor blood vessel-specific anticancer We have also developed a drug delivery system. Further, since the peptide intravenously injected into a tumor-bearing mouse model targets malignant tumor blood vessels, it is considered that the N-terminal region of annexin A1 is actually expressed on the surface of malignant tumor vascular endothelium.
 最近の医療費高騰は患者の経済的負担に留まらず、我が国の国家財政破綻が危ぶまれるほどに緊迫しており、また海外の国民皆保険がない国においては、高価な医薬品は手の届かないものになり、想像を絶する医療格差を招きつつある。このような状況の中、本発明者らの開発したIF7やdTIT7及びこれらを用いた抗がん剤のデリバリーシステムは、効率よく血液−脳腫瘍関門を越えて脳腫瘍等で優れた治療効果を上げることができ、且つ安価であるため、世界規模での医療格差の是正などのために期待されている。 The recent surge in medical expenses is not just an economic burden for patients, it is tense enough to threaten Japan's financial failure, and expensive medicines are out of reach in countries without overseas universal health insurance. It is becoming a reality, leading to unimaginable medical disparities. Under such circumstances, the delivery system of IF7 and dTIT7 developed by the present inventors and an anticancer agent using them can efficiently cross the blood-brain tumor barrier and exert an excellent therapeutic effect on a brain tumor or the like. Because it is possible and cheap, it is expected to correct the medical disparity on a global scale.
 本発明者らの開発したIF7やdTIT7を結合した抗がん剤が治療効果を発揮するには、IF7やdTIT7の受容体であるアネキシンA1のN末端領域が血管内皮表面に存在する必要がある。そのため、脳腫瘍等に罹患する患者において、該IF7やdTIT7を結合した抗がん剤を用いた治療等を実施する際、患者の身体的及び経済的負担の観点などから、事前に該腫瘍等組織の血管内皮表面にアネキシンA1のN末端領域が存在することを検出し得る診断薬が切望されている。 The N-terminal region of annexin A1, which is a receptor for IF7 and dTIT7, needs to be present on the surface of vascular endothelium in order for the anticancer drug bound by IF7 and dTIT7 developed by the present inventors to exert a therapeutic effect. .. Therefore, when a patient suffering from a brain tumor or the like is treated with an anticancer agent to which the IF7 or dTIT7 is bound, the tissue of the tumor or the like is preliminarily considered from the viewpoint of the physical and economic burden of the patient. A diagnostic agent capable of detecting the presence of the N-terminal region of annexin A1 on the surface of vascular endothelium is strongly desired.
 該腫瘍組織におけるアネキシンA1のN末端領域の存在の有無を検出する診断薬としては、免疫染色に使えるアネキシンA1のN末端領域に特異的に結合する抗体が適しているが、本発明者らの知る限り、そのような抗体は市販されておらず、学術論文等において報告もされていない。 An antibody that specifically binds to the N-terminal region of Annexin A1 that can be used for immunostaining is suitable as a diagnostic agent for detecting the presence or absence of the N-terminal region of Annexin A1 in the tumor tissue. To our knowledge, such an antibody is not commercially available and has not been reported in academic papers.
 本発明の課題は、アネキシンA1のN末端領域に特異的に結合するモノクローナル抗体、該抗体を産生するハイブリドーマ、該抗体を含むアネキシンA1のN末端領域の存在を検出するための試薬、腫瘍血管表面におけるアネキシンA1のN末端領域の存在を検出する方法等を提供することにある。 An object of the present invention is to provide a monoclonal antibody that specifically binds to the N-terminal region of annexin A1, a hybridoma that produces the antibody, a reagent for detecting the presence of the N-terminal region of annexin A1 containing the antibody, and a surface of a tumor blood vessel. It is intended to provide a method for detecting the presence of the N-terminal region of annexin A1 in E. coli.
 本発明者らは、鋭意研究を行った結果、アネキシンA1のN末端領域に特異的に結合するモノクローナル抗体を見出し、そして該モノクローナル抗体が悪性脳腫瘍における免疫染色において血管内皮を陽性染色し得ることを見出し、本発明を完成させるに至った。 As a result of intensive studies, the present inventors have found a monoclonal antibody that specifically binds to the N-terminal region of annexin A1, and found that the monoclonal antibody can positively stain vascular endothelium in immunostaining in malignant brain tumors. Heading out, the present invention has been completed.
 すなわち、本発明は以下の通りである。
[1]アネキシンA1のN末端領域に特異的に結合するモノクローナル抗体。
[2]前記N末端領域が、配列番号1で示されるアミノ酸配列を含む領域である、請求項1に記載のモノクローナル抗体。
[3]受託番号NITE P−02801として寄託されたハイブリドーマにより産生されるモノクローナル抗体。
[4]受託番号NITE P−02801として受託されたハイブリドーマ。
[5][1]~[3]のいずれか1つに記載のモノクローナル抗体を含む、アネキシンA1のN末端領域の存在を検出するための試薬。
[6][1]~[3]のいずれか1つに記載のモノクローナル抗体を含む、アネキシンA1のN末端領域に結合するペプチドと抗がん剤とのコンジュゲートを用いたがん治療の適用の可否を判定するための試薬。
[7]がんが固形がん又は液性がんである、[6]に記載の試薬。
[8]腫瘍血管表面におけるアネキシンA1のN末端領域の存在を検出する方法であって、
(1)[1]~[3]のいずれか1つに記載のアネキシンA1のN末端領域に特異的に結合するモノクローナル抗体と、対象由来の試料とを接触させること
(2)前記試料中において、腫瘍血管表面におけるアネキシンA1のN末端領域を、免疫染色によって検出すること
を含む、方法。
[9]アネキシンA1のN末端領域に結合するペプチドと抗がん剤とのコンジュゲートを用いたがん治療の適用対象の選別を補助するための方法であって、
(1)[1]~[3]のいずれか1つに記載のアネキシンA1のN末端領域に特異的に結合するモノクローナル抗体と、対象由来の試料とを接触させること
(2)前記試料中において、腫瘍血管表面におけるアネキシンA1のN末端領域を、免疫染色によって検出すること
を含む、方法。
[10]アネキシンA1のN末端領域に結合するペプチドと抗がん剤とのコンジュゲートを用いたがんの治療方法であって、
(1)[1]~[3]のいずれか1つに記載のアネキシンA1のN末端領域に特異的に結合するモノクローナル抗体と、対象由来の試料とを接触させること
(2)前記試料中において、腫瘍血管表面におけるアネキシンA1のN末端領域を、免疫染色によって検出すること
(3)前記検出結果に基づいて、前記治療の適用対象を選別すること
(4)前記選別した対象に、アネキシンA1のN末端領域に結合するペプチドと抗がん剤とのコンジュゲートを投与すること
を含む、方法。
[11]前記対象由来の試料が、対象から生検により採取されたがん細胞を含む生検サンプルを用いて作製された組織切片である、[8]~[10]のいずれか1つに記載の方法。
[12]前記がんが固形がん又は液性がんである、[9]~[11]のいずれか1つに記載の方法。
That is, the present invention is as follows.
[1] A monoclonal antibody that specifically binds to the N-terminal region of annexin A1.
[2] The monoclonal antibody according to claim 1, wherein the N-terminal region is a region containing the amino acid sequence represented by SEQ ID NO: 1.
[3] A monoclonal antibody produced by the hybridoma deposited under accession number NITE P-02801.
[4] The hybridoma deposited under the deposit number NITE P-02801.
[5] A reagent for detecting the presence of the N-terminal region of annexin A1, which comprises the monoclonal antibody according to any one of [1] to [3].
[6] Application of cancer treatment using a conjugate of a peptide that binds to the N-terminal region of annexin A1 and the anticancer agent, which comprises the monoclonal antibody according to any one of [1] to [3] A reagent for determining the availability of.
[7] The reagent according to [6], wherein the cancer is solid cancer or liquid cancer.
[8] A method for detecting the presence of an N-terminal region of annexin A1 on the surface of a tumor blood vessel, comprising:
(1) Contacting a sample derived from a subject with a monoclonal antibody that specifically binds to the N-terminal region of annexin A1 according to any one of [1] to [3] (2) In the sample , Detecting the N-terminal region of Annexin A1 on the surface of tumor blood vessels by immunostaining.
[9] A method for assisting selection of an application target of cancer treatment using a conjugate of a peptide that binds to the N-terminal region of annexin A1 and an anticancer agent,
(1) Contacting a sample derived from a subject with a monoclonal antibody that specifically binds to the N-terminal region of annexin A1 according to any one of [1] to [3] (2) In the sample , Detecting the N-terminal region of Annexin A1 on the surface of tumor blood vessels by immunostaining.
[10] A method for treating cancer, which comprises using a conjugate of a peptide that binds to the N-terminal region of annexin A1 and an anticancer agent,
(1) Contacting a sample derived from a subject with a monoclonal antibody that specifically binds to the N-terminal region of annexin A1 according to any one of [1] to [3] (2) In the sample Detecting the N-terminal region of annexin A1 on the surface of the tumor blood vessel by immunostaining (3) selecting an application target of the treatment based on the detection result (4) selecting annexin A1 A method comprising administering a conjugate of an anticancer drug and a peptide that binds to the N-terminal region.
[11] In any one of [8] to [10], wherein the subject-derived sample is a tissue section prepared using a biopsy sample containing cancer cells collected by biopsy from the subject. The method described.
[12] The method according to any one of [9] to [11], wherein the cancer is solid cancer or liquid cancer.
 本発明によれば、アネキシンA1のN末端領域に特異的に結合するモノクローナル抗体、該抗体を産生するハイブリドーマ、該抗体を含むアネキシンA1のN末端領域の存在を検出するための試薬、腫瘍血管表面におけるアネキシンA1のN末端領域の存在を検出する方法等を提供することができる。 According to the present invention, a monoclonal antibody that specifically binds to the N-terminal region of annexin A1, a hybridoma producing the antibody, a reagent containing the antibody for detecting the presence of the N-terminal region of annexin A1, a surface of a tumor blood vessel It is possible to provide a method for detecting the presence of the N-terminal region of Annexin A1 in the above.
図1は、ELISAによる抗MC16抗体(クローン31−8D)のヒト アネキシンA1に対する結合を測定した結果である。FIG. 1 shows the results of measuring the binding of anti-MC16 antibody (clone 31-8D) to human annexin A1 by ELISA. 図2は、ウェスタンブロットによる抗MC16抗体のヒト アネキシンA1とN末端ドメインを欠損したヒト アネキシンA1の変異体に対する結合を検出した結果である。FIG. 2 shows the results of detection of binding of anti-MC16 antibody to human annexin A1 and a mutant of human annexin A1 lacking the N-terminal domain by Western blotting. 図3は、抗MC16抗体によるマウス血管内皮F2細胞の蛍光免疫染色イメージの結果である。FIG. 3 shows the results of fluorescent immunostaining images of mouse vascular endothelial F2 cells with anti-MC16 antibody. 図4は、抗MC16抗体による膠芽腫組織の免疫染色の結果である(左:免疫染色、右:ヘマトキシリン‐エオシン染色)。FIG. 4 shows the results of immunostaining of glioblastoma tissue with anti-MC16 antibody (left: immunostaining, right: hematoxylin-eosin staining). 図5は、抗MC16抗体とInvitrogenから市販されている抗アネキシンA1抗体による様々な癌腫瘍組織(乳がん)の免疫染色の結果である。組織標本はBiomax社のmultiple cancer tissue micro arrayを使用した。FIG. 5 shows the results of immunostaining of various cancer tumor tissues (breast cancer) with anti-MC16 antibody and anti-annexin A1 antibody commercially available from Invitrogen. As the tissue specimen, a multiple cancer tissue micro array from Biomax was used. 図6は、抗MC16抗体とInvitrogenから市販されている抗アネキシンA1抗体による様々な癌腫瘍組織(肺がん)の免疫染色の結果である。組織標本はBiomax社のmultiple cancer tissue micro arrayを使用した。FIG. 6 shows the results of immunostaining of various cancer tumor tissues (lung cancer) with anti-MC16 antibody and anti-annexin A1 antibody marketed by Invitrogen. As the tissue specimen, a multiple cancer tissue micro array from Biomax was used. 図7は、抗MC16抗体とInvitrogenから市販されている抗アネキシンA1抗体による様々な癌腫瘍組織(肝細胞がん)の免疫染色の結果である。組織標本はBiomax社のmultiple cancer tissue micro arrayを使用した。FIG. 7 shows the results of immunostaining of various cancer tumor tissues (hepatocellular carcinoma) with anti-MC16 antibody and anti-annexin A1 antibody marketed by Invitrogen. As the tissue specimen, a multiple cancer tissue micro array from Biomax was used. 図8は、抗MC16抗体とInvitrogenから市販されている抗アネキシンA1抗体による様々な癌腫瘍組織(卵巣がん)の免疫染色の結果である。組織標本はBiomax社のmultiple cancer tissue micro arrayを使用した。FIG. 8 shows the results of immunostaining of various cancer tumor tissues (ovarian cancer) with anti-MC16 antibody and anti-annexin A1 antibody marketed by Invitrogen. As the tissue specimen, a multiple cancer tissue micro array from Biomax was used.
1.本発明のモノクローナル抗体
 本発明は、アネキシンA1(RefSeq No.NP_000691.1)(以下、「ANXA1」あるいは「Anxa1」と称する場合がある)のN末端領域に特異的に結合するモノクローナル抗体を提供する。該抗体は、具体的には、配列番号1で示されるアミノ酸配列(MAMVSEFLKQAWFIE)を含む、アネキシンA1のN末端領域に存在するエピトープと特異的に結合する抗体である。該抗体は、IgG、IgA、IgM、IgD又はIgEのいずれの免疫グロブリンクラスに属するものであってもよいが、好ましくはIgGであり、より好ましくはIgG2bである。該抗体の具体例としては、後述する本発明のモノクローナル抗体産生ハイブリドーマ(クローン31−8D)が産生する抗体(マウスIgG2b)(以下、「抗MC16抗体」と称する場合がある)が挙げられる。
1. Monoclonal Antibody of the Present Invention The present invention provides a monoclonal antibody that specifically binds to the N-terminal region of annexin A1 (RefSeq No. NP — 000691.1) (hereinafter sometimes referred to as “ANXA1” or “Anxa1”). .. Specifically, the antibody is an antibody that specifically binds to an epitope that is present in the N-terminal region of annexin A1 and that contains the amino acid sequence represented by SEQ ID NO: 1 (MAMVSEFLKQAWFIE). The antibody may belong to any immunoglobulin class of IgG, IgA, IgM, IgD or IgE, but is preferably IgG, more preferably IgG2b. Specific examples of the antibody include an antibody (mouse IgG2b) produced by the monoclonal antibody-producing hybridoma (clone 31-8D) of the present invention described below (hereinafter sometimes referred to as "anti-MC16 antibody").
 本発明のモノクローナル抗体は、(悪性)腫瘍内にできた新生血管内皮細胞の血液側に発現するアネキシンA1のN末端領域に特異的に結合するため、アネキシンA1のN末端領域の存在の有無を検出するために適している。そのため、本発明のモノクローナル抗体は、例えば、アネキシンA1のN末端領域に結合するペプチドと抗がん剤とを含むコンジュゲート、該コンジュゲートと薬学的に許容される担体とを含むがんの治療用組成物等を用いたがん治療を行う前に、対象が該治療の適用対象であるか否かを予測するためのコンパニオン診断に用いることができる。 Since the monoclonal antibody of the present invention specifically binds to the N-terminal region of Annexin A1 expressed on the blood side of neovascular endothelial cells formed in (malignant) tumors, the presence or absence of the N-terminal region of Annexin A1 is checked. Suitable for detecting. Therefore, the monoclonal antibody of the present invention is, for example, a cancer treatment comprising a conjugate containing a peptide that binds to the N-terminal region of annexin A1 and an anticancer agent, and the conjugate and a pharmaceutically acceptable carrier. It can be used for companion diagnosis for predicting whether or not a subject is an application target of the treatment before performing cancer treatment using the composition for treatment and the like.
 本明細書において、対象とは、アネキシンA1を発現する任意の動物、特には哺乳動物である。哺乳動物としては、例えば、マウス、ラット、ハムスター、モルモットなどのげっ歯類やウサギなどの実験動物、ブタ、ウシ、ヤギ、ウマ、ヒツジ、ミンクなどの家畜、イヌ、ネコなどのペット、ヒト、サル、アカゲザル、マーモセット、オランウータン、チンパンジーなどの霊長類などを挙げることができるが、これらに限定されない。 As used herein, a subject is any animal that expresses annexin A1, particularly a mammal. Examples of mammals include, for example, mice, rats, hamsters, experimental animals such as rodents and rabbits such as guinea pigs, pigs, cows, goats, horses, sheep, mink and other domestic animals, dogs, pets such as cats, humans, and the like. Examples thereof include, but are not limited to, primates such as monkeys, rhesus monkeys, marmosets, orangutans, and chimpanzees.
2.本発明のモノクローナル抗体の作製
 本発明のモノクローナル抗体及びこれに類似する抗体は、自体公知の方法(ファージディスプレイ法、ハイブリドーマ法)により作製することができる。該抗体を細胞融合によって製造されるハイブリドーマ(融合細胞)から取得する場合、具体的には、例えば、まず免疫原性の抗原ペプチドを、マウス、ラット、ハムスターなどの哺乳動物の皮下内、筋肉内、静脈内、フッドパッド内、腹腔内等に1~数回注射するか又は移植することにより免疫を施す。次に、該免疫した哺乳動物から抗体産生細胞を単離し、該細胞と骨髄腫細胞とを融合させてハイブリドーマを形成させ、該ハイブリドーマをクローン化し、該哺乳動物の免疫に用いた免疫原性の抗原ペプチドに対して特異的な親和性を示す抗体を産生するクローンを選択することによって製造される。
 また、抗体産生細胞については、予め単離された脾細胞、リンパ球等に培養液中で免疫原性の抗原ペプチドを作用させて生じる抗体産生細胞も使用することができる。この場合にはヒト由来の抗体産生細胞も調製可能である。
2. Preparation of Monoclonal Antibody of the Present Invention The monoclonal antibody of the present invention and an antibody similar thereto can be prepared by a method known per se (phage display method, hybridoma method). When the antibody is obtained from a hybridoma (fused cell) produced by cell fusion, specifically, for example, first, an immunogenic antigenic peptide is subcutaneously or intramuscularly administered to a mammal such as mouse, rat or hamster. Immunization is carried out by injecting once or several times into a vein, a hood, an abdominal cavity or the like, or by transplanting. Next, antibody-producing cells are isolated from the immunized mammal, the cells are fused with myeloma cells to form a hybridoma, the hybridoma is cloned, and immunogenic used for immunization of the mammal. It is produced by selecting a clone that produces an antibody showing a specific affinity for the antigenic peptide.
Further, as the antibody-producing cells, antibody-producing cells produced by reacting previously isolated spleen cells, lymphocytes, etc. with an immunogenic antigen peptide in a culture medium can also be used. In this case, human-derived antibody-producing cells can also be prepared.
 免疫原性の抗原ペプチドは、PBS(Phosphate−Buffered Saline)や生理食塩水等で適当な希釈倍率で希釈して免疫に使用することができる。また、免疫原性の抗原ペプチドは、アジュバントとともに投与してもよく、例えば、完全又は不完全フロイントアジュバント(FCA又はFIA)と混和し、乳化して用いてもよい。さらに、免疫原性の抗原の免疫時には適当な担体を使用してもよい。特に分子量の小さいペプチドが免疫原性の抗原として用いられる場合には、該免疫原性の抗原ペプチドをアルブミン、キーホールリンペットヘモシアニン等の担体タンパク質と結合させて免疫することが望ましい。
 後述する実施例では、ヒト アネキシンA1のN端から15番目までのアミノ酸配列(配列番号1:MAMVSEFLKQAWFIE)のC端にシステインを付加させた合成ペプチド(配列番号2:MAMVSEFLKQAWFIEC)(以下、「合成ペプチドMC16」又は単に「MC16」と称する場合がある)をウシ血清アルブミンに結合させた物質と、完全フロイントアジュバントを混和したものを免疫原性の抗原ペプチドとして用いた。
The immunogenic antigen peptide can be used for immunization by diluting it with PBS (Phosphate-Buffered Saline) or physiological saline at an appropriate dilution ratio. The immunogenic antigenic peptide may be administered together with an adjuvant, for example, it may be mixed with complete or incomplete Freund's adjuvant (FCA or FIA) and emulsified. Furthermore, a suitable carrier may be used when immunizing with an immunogenic antigen. Particularly when a peptide having a small molecular weight is used as an immunogenic antigen, it is desirable to immunize the immunogenic antigen peptide by binding it to a carrier protein such as albumin or keyhole limpet hemocyanin.
In Examples described later, a synthetic peptide (SEQ ID NO:2: MAMVSEFLKQAWFIEC) in which cysteine is added to the C-terminal of the amino acid sequence from the N-terminal to the 15th amino acid of human annexin A1 (SEQ ID NO: 1 MAMVSEFLKQAWFIE) (hereinafter, “synthetic peptide”). A mixture of bovine serum albumin-bound substance and complete Freund's adjuvant was used as an immunogenic antigen peptide.
 モノクローナル抗体を分泌するハイブリドーマの調製はケーラー及びミルシュタインの方法(Nature,Vol.256,pp.495−497,1975)、及びその変法に従って行うことができる。即ち、本発明のモノクローナル抗体は、前述のごとく免疫された動物から取得される脾細胞、リンパ節細胞、末梢リンパ球、骨髄腫細胞、扁桃細胞等、好ましくは脾細胞に含まれる抗体産生細胞と、好ましくは同種のマウス、ラット、モルモット、ハムスター、ウサギ又はヒト等の哺乳動物、より好ましくはマウス、ラット又はヒトの骨髄腫細胞(ミエローマ)との融合により得られるハイブリドーマを培養することにより調製される。培養は、インビトロ又はマウス、ラット、モルモット、ハムスター、ウサギ等の哺乳動物、好ましくはマウス又はラット、より好ましくはマウスの腹腔内等でのインビボで行うことができ、抗体はそれぞれ培養上清あるいは哺乳動物の腹水から取得することができる。 Hybridomas that secrete monoclonal antibodies can be prepared according to the method of Kohler and Milstein (Nature, Vol. 256, pp. 495-497, 1975) and its modifications. That is, the monoclonal antibody of the present invention, splenocytes, lymph node cells, peripheral lymphocytes, myeloma cells, tonsil cells, etc. obtained from the immunized animal as described above, preferably antibody-producing cells contained in splenocytes and It is preferably prepared by culturing a hybridoma obtained by fusion with a mammal such as mouse, rat, guinea pig, hamster, rabbit or human of the same species, more preferably mouse, rat or human myeloma cells (myeloma). It Culturing can be performed in vitro or in vivo in mammals such as mouse, rat, guinea pig, hamster, rabbit, etc., preferably mouse or rat, more preferably in the abdominal cavity of mouse, and the antibody is the culture supernatant or mammalian, respectively. Can be obtained from the ascites of an animal.
 細胞融合に用いられる骨髄腫細胞としては、例えば、マウス由来ミエローマP3/X63−AG8、P3/NSI/1−Ag4−1、P3/X63−Ag8.U1、SP2/0−Ag14、F0或いはBW5147、ラット由来ミエローマ210RCY3−Ag1.2.3.、ヒト由来ミエローマU−266AR1、GML500−6TG−A1−2、UC729−6、CEM−AGR、D1R11或いはCEM−T15等が挙げられる。 Examples of myeloma cells used for cell fusion include mouse-derived myeloma P3/X63-AG8, P3/NSI/1-Ag4-1, P3/X63-Ag8. U1, SP2/0-Ag14, F0 or BW5147, rat-derived myeloma 210RCY3-Ag1.2.3. , Human-derived myeloma U-266AR1, GML500-6TG-A1-2, UC729-6, CEM-AGR, D1R11 or CEM-T15.
 本発明のモノクローナル抗体を産生するハイブリドーマクローンのスクリーニングは、ハイブリドーマを、例えば、マイクロタイタープレート中で培養し、増殖の見られたウェル中の培養上清の免疫原性の抗原ペプチドに対する反応性を、例えば、ELISA法、ウェスタンブロット法、免疫染色法等によって測定することにより行うことができる。 The screening of the hybridoma clone producing the monoclonal antibody of the present invention is carried out by culturing the hybridoma in, for example, a microtiter plate, and measuring the reactivity of the culture supernatant in the wells in which proliferation is observed against the immunogenic antigenic peptide. For example, the measurement can be performed by ELISA, Western blotting, immunostaining, or the like.
 後述する実施例に記載の通り、合成ペプチドMC16に高い反応性を示す抗体を産生したハイブリドーマを、限界希釈法によって細胞クローニングし、本発明のモノクローナル抗体産生ハイブリドーマ(クローン31−8D)として取得した。クローン31−8Dを平成30年10月24日に、受託番号NITE P−02801として独立行政法人製品評価技術基盤機構 特許微生物寄託センター(千葉県木更津市かずさ鎌足2−5−8 122号室)に寄託した。 As described in Examples below, a hybridoma that produced an antibody showing high reactivity with the synthetic peptide MC16 was subjected to cell cloning by the limiting dilution method to obtain a monoclonal antibody-producing hybridoma of the present invention (clone 31-8D). Clone 31-8D will be placed on October 24, 2018, under the accession number NITE P-02801, at the Patent Microorganism Depositary Center (2-5-8, Kazusa Kamafoot, Kisarazu City, Chiba Prefecture) under the National Institute for Product Evaluation Technology, National Institute for Product Evaluation Technology. Deposited.
 モノクローナル抗体の単離精製は、上述のような方法によって製造される該抗体含有培養上清或いは腹水を、例えば、イオン交換クロマトグラフィー、抗イムノグロブリンカラム又はプロテインAやプロテインGカラム等のアフィニティーカラムクロマトグラフィーに付すことにより行うことができる。 The isolation and purification of the monoclonal antibody is carried out by subjecting the antibody-containing culture supernatant or ascites produced by the above-mentioned method to, for example, ion exchange chromatography, anti-immunoglobulin column or affinity column chromatography such as protein A or protein G column. It can be performed by attaching it to a graph.
 本発明のモノクローナル抗体は、検出可能な標識で標識されていてもよい。標識には蛍光標識、低分子化合物による標識、ペプチドによる標識、酵素による標識等が挙げられる。また、本発明のモノクローナル抗体は、他の薬剤とともにコンジュゲートを形成していてもよい。 The monoclonal antibody of the present invention may be labeled with a detectable label. Examples of the label include fluorescent label, low molecular weight compound label, peptide label, enzyme label and the like. In addition, the monoclonal antibody of the present invention may form a conjugate with other drugs.
 本発明のモノクローナル抗体は、上述の製造方法に限定されることなく、いかなる方法で得られたものであってもよい。例えば、本発明のハイブリドーマからクローニングされた、本発明のモノクローナル抗体をコードする遺伝子を、適当なベクターに組み込んで宿主に導入することによって、抗体として発現させたものを利用してもよい。抗体をコードする遺伝子の単離と、ベクターへの導入、そして宿主細胞の形質転換のための方法は既に確立されている(例えば、Vandamme,A.M.らEur.J.Biochem.(1990)192,767−775を参照)。 The monoclonal antibody of the present invention is not limited to the above-mentioned production method, and may be obtained by any method. For example, a gene cloned from the hybridoma of the present invention, which encodes the monoclonal antibody of the present invention, may be expressed as an antibody by incorporating the gene into a suitable vector and introducing it into a host. Methods for the isolation of antibody-encoding genes, their introduction into vectors, and the transformation of host cells have been established (eg, Vandamme, AM et al. Eur. J. Biochem. (1990). 192, 767-775).
 例えば、本発明のモノクローナル抗体を産生するハイブリドーマから、該抗体の可変領域(V領域)をコードするcDNAを得ることができる。そのためには、通常、まずハイブリドーマから全RNAが抽出される。細胞からmRNAを抽出するための方法として、たとえば、グアニジン超遠心法(Chirgwin,J.M.ら、Biochemistry(1979)18,5294−5299)、AGPC法(Chomczynski,P.ら、Anal.Biochem.(1987)162,156−159)などを用いることができる。抽出されたmRNAは、mRNA Purification Kit(GEヘルスケアバイオサイエンス製)等を使用して精製することができる。あるいは、QuickPrep mRNA Purification Kit(GEヘルスケアバイオサイエンス製)などのように、細胞から直接全mRNAを抽出するためのキットも市販されている。このようなキットを用いて、ハイブリドーマから全mRNAを得ることもできる。得られたmRNAから逆転写酵素を用いて抗体V領域をコードするcDNAを合成することができる。cDNAは、AMV Reverse Transcriptase First−strand cDNA Synthesis Kit(生化学工業社製)等によって合成することができる。また、cDNAの合成および増幅のために、5’−Ampli FINDER RACE Kit(Clontech製)及びPCRを用いた5’−RACE法(Frohman,M.A.ら、Proc.Natl.Acad.Sci.USA(1988)85,8998−9002、Belyavsky,A.らNucleic Acids Res.(1989)17,2919−2932)を利用することができる。さらに、このようなcDNAの合成の過程において、cDNAの両末端に適切な制限酵素サイトを導入してもよい。 For example, from a hybridoma producing the monoclonal antibody of the present invention, cDNA encoding the variable region (V region) of the antibody can be obtained. For that purpose, first, total RNA is usually extracted from the hybridoma. As a method for extracting mRNA from cells, for example, guanidine ultracentrifugation method (Chirgwin, JM, et al., Biochemistry (1979) 18, 5294-5299), AGPC method (Chomczynski, P., et al., Anal. Biochem. (1987) 162, 156-159) and the like can be used. The extracted mRNA can be purified using mRNA Purification Kit (manufactured by GE Healthcare Bioscience). Alternatively, a kit for directly extracting total mRNA from cells, such as QuickPrep mRNA Purification Kit (manufactured by GE Healthcare Bioscience), is commercially available. Such a kit can also be used to obtain total mRNA from hybridomas. CDNA encoding the antibody V region can be synthesized from the obtained mRNA using reverse transcriptase. The cDNA can be synthesized by AMV Reverse Transcriptase First-strand cDNA Synthesis Kit (manufactured by Seikagaku Corporation). For the synthesis and amplification of cDNA, 5'-Ampli FINDER RACE Kit (manufactured by Clontech) and 5'-RACE method using PCR (Frohman, MA, et al., Proc. Natl. Acad. Sci. USA). (1988) 85, 8998-9002, Belyavsky, A. et al. Nucleic Acids Res. (1989) 17, 2919-2932). Furthermore, in the process of synthesizing such a cDNA, appropriate restriction enzyme sites may be introduced at both ends of the cDNA.
 得られたPCR産物から目的とするcDNA断片が精製され、次いで、ベクターDNAと作動可能に連結される。このように組換えベクターが作製され、大腸菌等に導入されコロニーが選択された後に、該コロニーを形成した大腸菌から所望の組換えベクターが調製できる。そして、該組換えベクターが目的とするcDNAの塩基配列を有しているか否かについて、自体公知の方法、例えば、ジデオキシヌクレオチドチェインターミネーション法等により確認できる。 The desired cDNA fragment is purified from the resulting PCR product and then operably linked to vector DNA. After the recombinant vector is prepared in this manner and introduced into Escherichia coli or the like to select a colony, a desired recombinant vector can be prepared from E. coli forming the colony. Whether or not the recombinant vector has the nucleotide sequence of the desired cDNA can be confirmed by a method known per se, such as the dideoxynucleotide chain termination method.
 通常モノクローナル抗体は免疫を施す哺乳動物の種類によりそれぞれ異なる構造の糖鎖を有するが、本発明におけるモノクローナル抗体は、該糖鎖の構造差異により限定されるものではなく、あらゆる哺乳動物由来のモノクローナル抗体をも包含するものである。さらに、例えば、ヒト免疫グロブリン遺伝子を組み込まれたトランスジェニック動物から得られる組換えヒト型モノクローナル抗体、あるいはある哺乳動物由来のモノクローナル抗体の定常領域(Fc)をヒト由来モノクローナル抗体のFc領域と組換えたキメラモノクローナル抗体、さらには抗原と相補的に直接結合し得る相補性決定部位(CDR)以外の全領域をヒト由来モノクローナル抗体の対応領域と組換えたキメラモノクローナル抗体も本発明のモノクローナル抗体に包含される。 Usually, a monoclonal antibody has a sugar chain having a different structure depending on the type of mammal to be immunized, but the monoclonal antibody in the present invention is not limited by the structural difference of the sugar chain, and a monoclonal antibody derived from any mammal It also includes. Furthermore, for example, a recombinant human monoclonal antibody obtained from a transgenic animal into which a human immunoglobulin gene has been incorporated, or a constant region (Fc) of a monoclonal antibody derived from a certain mammal is recombined with the Fc region of a human-derived monoclonal antibody. Also included in the monoclonal antibody of the present invention are chimeric monoclonal antibodies, and further, chimeric monoclonal antibodies in which the entire region other than the complementarity determining region (CDR) that can directly bind complementarily with an antigen is recombined with the corresponding region of a human-derived monoclonal antibody. To be done.
 また、本発明の抗体には、前記のモノクローナル抗体(mAb)等の天然型抗体、遺伝子組換え技術を用いて製造され得るキメラ抗体、ヒト化抗体や一本鎖抗体に加えて、これらの抗体の断片が含まれる。抗体の断片とは、特異的結合活性を有する前述の抗体の一部分の領域を意味し、具体的にはFab、Fab’、F(ab’)、scAb、scFv又はscFv−Fc等を包含する。 The antibodies of the present invention include natural antibodies such as the above-mentioned monoclonal antibody (mAb), chimeric antibodies that can be produced by gene recombination technology, humanized antibodies and single chain antibodies, and these antibodies. Fragment of. The antibody fragment means a partial region of the aforementioned antibody having specific binding activity, and specifically includes Fab, Fab′, F(ab′) 2 , scAb, scFv or scFv-Fc and the like. ..
 これらの抗体断片は、自体公知の方法を用いて得ることができ、具体的には、本発明のモノクローナル抗体を酵素、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遺伝子を構築し、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。得られたフラグメントは、本発明のモノクローナル抗体と同様にして抗原との反応性、特異性等を評価することができる。 These antibody fragments can be obtained by a method known per se, and specifically, the monoclonal antibody of the present invention is treated with an enzyme such as papain or pepsin to produce an antibody fragment, or these antibodies are produced. A gene encoding the fragment may be constructed, introduced into an expression vector, and then expressed in an appropriate host cell. The obtained fragment can be evaluated for reactivity with an antigen, specificity and the like in the same manner as for the monoclonal antibody of the present invention.
 また、当業者であれば、本発明の抗体又は断片と他のペプチドやタンパク質との融合抗体を作製することや、修飾剤を結合させた修飾抗体を作製することも可能である。融合に用いられる他のペプチドやタンパク質は、抗体の結合活性を低下させないものである限り特に限定されず、例えば、ヒト血清アルブミン、各種tagペプチド、人工ヘリックスモチーフペプチド、マルトース結合タンパク質、グルタチオンSトランスフェラーゼ、各種毒素、その他多量体化を促進し得るペプチド又はタンパク質等が挙げられる。修飾に用いられる修飾剤は、抗体の結合活性を低下させないものである限り特に限定されず、例えば、ポリエチレングリコール、糖鎖、リン脂質、リポソーム、低分子化合物等が挙げられる。 Further, those skilled in the art can also prepare a fusion antibody of the antibody or fragment of the present invention with another peptide or protein, or a modified antibody having a modifying agent bound thereto. Other peptides or proteins used for fusion are not particularly limited as long as they do not reduce the binding activity of the antibody, and examples thereof include human serum albumin, various tag peptides, artificial helix motif peptides, maltose binding protein, glutathione S transferase, Examples include various toxins and peptides or proteins capable of promoting multimerization. The modifying agent used for modification is not particularly limited as long as it does not reduce the binding activity of the antibody, and examples thereof include polyethylene glycol, sugar chains, phospholipids, liposomes, and low molecular weight compounds.
3.本発明の試薬
 上述したように、本発明のモノクローナル抗体は(悪性)腫瘍内にできた新生血管内皮細胞の血液側に発現するアネキシンA1のN末端領域に特異的に結合するため、悪性腫瘍標的活性のあるIF7やdTIT7を用いた悪性腫瘍治療薬の受容体であるアネキシンA1のN末端領域の存在の有無を検出するために適している。従って、本発明は、本発明のモノクローナル抗体を含む、アネキシンA1のN末端領域の存在を検出するための試薬も提供する。
3. Reagent of the present invention As described above, since the monoclonal antibody of the present invention specifically binds to the N-terminal region of annexin A1 expressed on the blood side of neovascular endothelial cells formed in (malignant) tumors, it can be targeted to malignant tumors. It is suitable for detecting the presence or absence of the N-terminal region of annexin A1, which is a receptor for therapeutic agents for malignant tumors using active IF7 or dTIT7. Therefore, the present invention also provides a reagent for detecting the presence of the N-terminal region of Annexin A1, which comprises the monoclonal antibody of the present invention.
 また上述したように、本発明のモノクローナル抗体は、特定のがん治療の適用対象であるか否かを予測するためのコンパニオン診断に用いることができる。従って、本発明は、本発明のモノクローナル抗体を含む、アネキシンA1のN末端領域に結合するペプチドと抗がん剤とのコンジュゲート、該コンジュゲートと薬学的に許容される担体とを含む組成物等を用いたがん治療の適用の可否を判定するための試薬も提供する。 Further, as described above, the monoclonal antibody of the present invention can be used for companion diagnosis for predicting whether or not a specific cancer treatment is applied. Therefore, the present invention provides a conjugate comprising a peptide that binds to the N-terminal region of annexin A1 and the anticancer agent, which comprises the monoclonal antibody of the present invention, and a composition comprising the conjugate and a pharmaceutically acceptable carrier. Also provided is a reagent for determining the applicability of cancer treatment using the above.
 本明細書において、悪性腫瘍(がん)は任意の種類のがんであってよく、固形がん又は液性がんであってもよい。固形がんとしては、好ましくはアネキシンA1を細胞表面に発現する固形がん、従って血管新生する固形がんが挙げられる。固形がんとしては、例えば、脳・神経系のがん(例えば、脳腫瘍、脊髄腫瘍など)、頭頸部がん(例えば、喉頭がん、口腔がん、唾液腺がん、副鼻腔がん、甲状腺がんなど)、消化器がん(例えば、胃がん、食道がん、小腸がん、結腸がん、直腸がん、肛門がん、肝臓がん、胆道がん、膵臓がんなど)、泌尿器又は生殖器のがん(例えば、腎がん、腎細胞がん、膀胱がん、前立腺がん、腎盂および尿管がん、胆嚢がん、胆管がん、精巣がん、陰茎がん、子宮がん、子宮内膜がん、子宮肉腫、子宮頚がん、膣がん、外陰がん、卵巣がん、卵管がんなど)、呼吸器系のがん(例えば、肺がん(小細胞肺がん、非小細胞肺がん、転移性肺がんを含む)、気管支がんなど)、乳がん、皮膚がん(例えば、悪性黒色腫など)、骨のがん(例えば、骨肉腫など)、筋肉のがん(例えば、横紋筋肉腫など)などが挙げられる。 In the present specification, malignant tumor (cancer) may be any type of cancer, and may be solid cancer or liquid cancer. The solid cancer preferably includes a solid cancer that expresses annexin A1 on the cell surface, and thus an angiogenic solid cancer. Examples of solid cancer include brain/nervous system cancer (for example, brain tumor, spinal cord tumor, etc.), head and neck cancer (for example, laryngeal cancer, oral cancer, salivary gland cancer, sinus cancer, thyroid cancer). Cancer, etc.), gastrointestinal cancer (for example, gastric cancer, esophageal cancer, small intestine cancer, colon cancer, rectal cancer, anal cancer, liver cancer, biliary tract cancer, pancreatic cancer, etc.), urinary organ or Reproductive organ cancer (eg, renal cancer, renal cell cancer, bladder cancer, prostate cancer, renal pelvis and ureteral cancer, gallbladder cancer, bile duct cancer, testicular cancer, penis cancer, uterine cancer) , Endometrial cancer, uterine sarcoma, cervical cancer, vaginal cancer, vulvar cancer, ovarian cancer, fallopian tube cancer, etc., respiratory cancer (eg lung cancer (small cell lung cancer, non Small cell lung cancer, including metastatic lung cancer), bronchial cancer, etc.), breast cancer, skin cancer (eg, malignant melanoma), bone cancer (eg, osteosarcoma), muscle cancer (eg, Rhabdomyosarcoma) and the like.
 液性がんとしては、白血病、悪性リンパ腫、多発性骨髄腫、骨髄異型性症候群などが挙げられる。白血病としては、急性骨髄性白血病、急性リンパ性白血病、慢性骨髄性白血病、慢性リンパ性白血病などが挙げられる。悪性リンパ腫は、ホジキンリンパ腫と非ホジキンリンパ腫に分類され、非ホジキンリンパ腫としては、B細胞性リンパ腫、成人T細胞リンパ腫、リンパ芽球性リンパ腫、びまん性大細胞型リンパ腫、バーキットリンパ腫、濾胞性リンパ腫、MALTリンパ腫、末梢性T細胞リンパ腫、マントル細胞リンパ腫などが挙げられる。 Liquid cancers include leukemia, malignant lymphoma, multiple myeloma, myelodysplastic syndrome, etc. Examples of leukemia include acute myelogenous leukemia, acute lymphocytic leukemia, chronic myelogenous leukemia, chronic lymphocytic leukemia and the like. Malignant lymphoma is classified into Hodgkin lymphoma and non-Hodgkin lymphoma. As non-Hodgkin lymphoma, B-cell lymphoma, adult T-cell lymphoma, lymphoblastic lymphoma, diffuse large cell lymphoma, Burkitt lymphoma, follicular lymphoma , MALT lymphoma, peripheral T cell lymphoma, mantle cell lymphoma and the like.
 本発明の試薬を用いたコンパニオン診断の結果として、対象に投与されるアネキシンA1のN末端領域に結合するペプチドと抗がん剤のコンジュゲートや該コンジュゲートと薬学的に許容される担体とを含む組成物等は、後述するように、効率よく血液−脳腫瘍関門を越えることができるので、特に好ましい標的として脳腫瘍を挙げることができる。 As a result of the companion diagnosis using the reagent of the present invention, a peptide that binds to the N-terminal region of annexin A1 administered to a subject, a conjugate of an anticancer agent, or the conjugate and a pharmaceutically acceptable carrier are used. As will be described later, the composition containing the compound can efficiently cross the blood-brain tumor barrier, and therefore a brain tumor can be mentioned as a particularly preferable target.
 脳腫瘍は、原発性脳腫瘍又は転移性脳腫瘍であってもよい。また、脳腫瘍は良性(例えば、髄膜腫、下垂体腺腫、神経鞘腫など)又は悪性の脳腫瘍であってもよく、好ましくは悪性の脳腫瘍である。悪性の脳腫瘍としては、星細胞腫(アストロサイトーマ)、乏突起神経膠腫(オリゴデンドログリオーマ)などのグレード2の脳腫瘍、退形成性星細胞腫、退形成性乏突起膠腫、退形成性乏突起星細胞腫などのグレード3の脳腫瘍、膠芽腫(グリオブラストーマ)などのグレード4の脳腫瘍が挙げられる。 The brain tumor may be a primary brain tumor or a metastatic brain tumor. The brain tumor may be benign (eg, meningioma, pituitary adenoma, schwannoma, etc.) or malignant brain tumor, preferably malignant brain tumor. Malignant brain tumors include grade 2 brain tumors such as astrocytoma (astrocytoma) and oligodendrogliomas, anaplastic astrocytoma, anaplastic oligodendroglioma, anaplastic Examples thereof include grade 3 brain tumors such as oligoastrocytoma, and grade 4 brain tumors such as glioblastoma.
 本発明の試薬は、本発明のモノクローナル抗体のみからなるものであってもよいし、医薬的に許容される担体を含んでいてもよい。医薬的に許容される担体としては、本発明の試薬を液剤として調製する場合、製剤素材として慣用されている各種担体、例えば、希釈剤、溶剤、溶解補助剤、懸濁化剤、等張化剤、緩衝剤などを含んでいてもよい。さらに抗体の凝集を防ぐために、Tween20(登録商標)、Tween80(登録商標)などの界面活性剤を添加してもよい。希釈剤としては、水、生理用食塩水などが挙げられる。溶剤としては、水、生理用食塩水、エタノールなどが挙げられる。溶解補助剤としては、シクロデキストリン類などが挙げられる。懸濁化剤としては、アラビアゴム、カルメロースなどが挙げられる。等張化剤としては、塩化ナトリウム、塩化カリウムなどの無機塩類、グリセリン、マンニトール、ソルビトールなどの炭水化物などが挙げられる。緩衝剤としては、リン酸緩衝液、酢酸緩衝液、ホウ酸緩衝液、炭酸緩衝液、クエン酸緩衝液、トリス緩衝液などが挙げられる。これらの担体の配合比は、当業者が適宜決定することができる。 The reagent of the present invention may consist only of the monoclonal antibody of the present invention or may contain a pharmaceutically acceptable carrier. As the pharmaceutically acceptable carrier, when the reagent of the present invention is prepared as a liquid agent, various carriers commonly used as a formulation material, for example, diluents, solvents, solubilizers, suspending agents, isotonic agents, etc. Agents, buffers, etc. may be included. Further, a surfactant such as Tween 20 (registered trademark) or Tween 80 (registered trademark) may be added to prevent antibody aggregation. Examples of the diluent include water and physiological saline. Examples of the solvent include water, physiological saline, ethanol and the like. Examples of the solubilizing agent include cyclodextrins. Examples of the suspending agent include gum arabic and carmellose. Examples of the isotonicity agent include inorganic salts such as sodium chloride and potassium chloride, carbohydrates such as glycerin, mannitol and sorbitol. Examples of the buffer include phosphate buffer, acetate buffer, borate buffer, carbonate buffer, citrate buffer, Tris buffer and the like. Those skilled in the art can appropriately determine the blending ratio of these carriers.
4.本発明の方法
 上述したように、本発明のモノクローナル抗体は(悪性)腫瘍内にできた新生血管内皮細胞の血液側に発現するアネキシンA1のN末端領域に特異的に結合するため、アネキシンA1のN末端領域の存在の有無を検出するために適している。従って、本発明は、該モノクローナル抗体を用いた、腫瘍内にできた新生血管内皮細胞の血液側に発現しているアネキシンA1(腫瘍血管表面におけるアネキシンA1)のN端領域の存在を検出する方法も提供する。
4. Method of the present invention As described above, the monoclonal antibody of the present invention specifically binds to the N-terminal region of annexin A1 expressed on the blood side of neovascular endothelial cells formed in (malignant) tumors. It is suitable for detecting the presence or absence of the N-terminal region. Therefore, the present invention provides a method for detecting the presence of the N-terminal region of annexin A1 (annexin A1 on the surface of tumor blood vessels) expressed on the blood side of neovascular endothelial cells formed in a tumor using the monoclonal antibody. Also provide.
 具体的には、本発明の方法は(1)本発明のアネキシンA1のN端領域に特異的に結合するモノクローナル抗体と、対象由来の試料とを接触させること、及び(2)前記試料中において、腫瘍内にできた新生血管内皮細胞の血液側に発現しているアネキシンA1(腫瘍血管表面におけるアネキシンA1)のN端領域を、免疫染色によって検出することを含む。 Specifically, the method of the present invention comprises (1) contacting a sample derived from a subject with a monoclonal antibody of the present invention that specifically binds to the N-terminal region of annexin A1, and (2) in the sample Detecting the N-terminal region of annexin A1 (annexin A1 on the surface of tumor blood vessels) expressed on the blood side of neovascular endothelial cells formed in the tumor by immunostaining.
 また上述したように、本発明のモノクローナル抗体は、特定のがん治療の適用対象であるか否かを予測するためのコンパニオン診断に用いることができる。従って、本発明は、該モノクローナル抗体を用いた、アネキシンA1のN末端領域に結合するペプチドと抗がん剤とのコンジュゲート、該コンジュゲートと薬学的に許容される担体とを含む組成物等を用いたがん治療の適用対象の選別方法あるいは選別を補助するための方法も提供する。さらに、本発明は、上記のように選別した対象に、アネキシンA1のN末端領域に結合するペプチドと抗がん剤とのコンジュゲート、該コンジュゲートと薬学的に許容される担体とを含む組成物等を投与することにより実際にがん治療を行うことを含んでもよい。 Further, as described above, the monoclonal antibody of the present invention can be used for companion diagnosis for predicting whether or not a specific cancer treatment is applied. Accordingly, the present invention provides a conjugate comprising a peptide that binds to the N-terminal region of annexin A1 and an anticancer agent using the monoclonal antibody, a composition containing the conjugate and a pharmaceutically acceptable carrier, and the like. The present invention also provides a method for selecting an application target of cancer treatment using the method or a method for assisting the selection. Furthermore, the present invention provides a composition comprising the conjugate selected from a peptide that binds to the N-terminal region of annexin A1 and an anticancer agent, and the conjugate and a pharmaceutically acceptable carrier in the subject selected as described above. It may include actually treating cancer by administering a substance or the like.
 具体的には、本発明の方法は(1)本発明のアネキシンA1のN端領域に特異的に結合するモノクローナル抗体と、対象由来の試料とを接触させること、及び(2)前記試料中において、腫瘍内にできた新生血管内皮細胞の血液側に発現しているアネキシンA1(腫瘍血管表面におけるアネキシンA1)のN端領域を、免疫染色によって検出することを含む。 Specifically, the method of the present invention comprises (1) contacting a sample derived from a subject with a monoclonal antibody of the present invention that specifically binds to the N-terminal region of annexin A1, and (2) in the sample Detecting the N-terminal region of annexin A1 (annexin A1 on the surface of tumor blood vessels) expressed on the blood side of neovascular endothelial cells formed in the tumor by immunostaining.
 本発明のモノクローナル抗体を用いてアネキシンA1のN端領域の存在を検出する場合の測定手段は特に限定されないが、好ましくは、免疫染色法が用いられる。免疫染色法を用いた具体的手段としては、免疫組織化学染色(IHC)法、フローサイトメトリー解析、マスサイトメトリー解析などが挙げられる。 The measuring means for detecting the presence of the N-terminal region of Annexin A1 using the monoclonal antibody of the present invention is not particularly limited, but an immunostaining method is preferably used. Specific means using the immunostaining method include immunohistochemical staining (IHC) method, flow cytometry analysis, mass cytometry analysis and the like.
 免疫組織化学染色は、自体公知の方法に従って行うことができ(例えば、改訂四版渡辺・中根酵素抗体法、発行:学際企画株式会社、編集:名倉宏、長村義之、堤寛、ISBN4−906514−73−X C304を参照)、好ましくは、酵素抗体法(Enzyme labeled antibody method)及び蛍光抗体法等が用いられる。 Immunohistochemical staining can be performed according to a method known per se (for example, the revised fourth edition Watanabe-Nakone enzyme antibody method, published by Interdisciplinary Planning Co., Ltd., edited by Hiroshi Nagura, Yoshiyuki Nagamura, Hiroshi Tsutsumi, ISBN4-906514). -73-X C304), preferably the enzyme antibody method (Enzyme labeled antibody method) and the fluorescent antibody method are used.
 例えば、酵素抗体法では、酵素で標識した抗体により、アネキシンA1のN端領域を検出する。標識を直接行う場合と、二次抗体を用いて間接的に行う場合がある。検出感度を上げるために種々の改良がなされ、ビオチン−ストレプトアビジンを用いたABC法、チラミド(タイラマイド)を用いたチラミド法(TSA法、Tyramide Signal Amplification)などの改良法で、非常に高い感度を実現できる。 For example, in the enzyme antibody method, the N-terminal region of annexin A1 is detected with an enzyme-labeled antibody. Labeling may be done directly or indirectly using a secondary antibody. Various improvements have been made to increase the detection sensitivity, and very high sensitivity is achieved with improved methods such as the ABC method using biotin-streptavidin and the tyramide method (TSA method, Tyramide Signal Amplification) using tyramide (tylamide). realizable.
 標識酵素は、ペルオキシダーゼ(HRP、horseradish peroxidase)及びアルカリホスファターゼ(AP、alkaline phosphatase)が汎用されており、本発明においても好適に使用することができる。 As the labeling enzyme, peroxidase (HRP, horseradish peroxidase) and alkaline phosphatase (AP, alkaline phosphatase) are widely used and can be suitably used in the present invention.
 一般的なプロトコールとして、必要により脱パラフィン又は固定処理した細胞標本を準備し、必要により前処理(例えば、生体色素の脱色又は除去処理、ブロッキング処理)又は抗原賦活化(温浴、マイクロウェーブ処理、オートクレーブ処理、プロテアーゼ処理等)し、一次抗体と反応させ(例えば、4℃~室温で0.5~24時間)、反応終了後、細胞標本を洗浄し、二次抗体と反応させ(例えば、4℃~室温で5~120分)、反応終了後、細胞標本を洗浄する。次いで、ジアミノベンジジン(3,3−diaminobenzidine:DAB)などの発色基質を添加し、発色させる。顕微鏡での形態観察のために、さらにヘマトキシリン染色等の対比染色を行ってもよい。 As a general protocol, a cell sample deparaffinized or fixed as necessary is prepared, and if necessary, pretreatment (for example, decolorization or removal treatment of vital dye, blocking treatment) or antigen activation (heat bath, microwave treatment, autoclave) Treatment, protease treatment, etc., and react with primary antibody (for example, 0.5° C. to 24 hours at 4° C. to room temperature). After the reaction is complete, the cell sample is washed and reacted with secondary antibody (for example, 4° C.). After completion of the reaction, the cell sample is washed at room temperature for 5 to 120 minutes). Then, a color-developing substrate such as diaminobenzidine (3,3-diaminobenzidine:DAB) is added to develop the color. For morphological observation with a microscope, counterstaining such as hematoxylin staining may be further performed.
 また、上述の酵素抗体法での酵素標識に換えて、蛍光色素で標識した抗体を用いることで、蛍光抗体法として、アネキシンA1のN端領域を検出してもよい。蛍光抗体法においても、標識を直接行ってもよく、また二次抗体を用いて間接的に行ってもよい。 Alternatively, the N-terminal region of annexin A1 may be detected as a fluorescent antibody method by using an antibody labeled with a fluorescent dye instead of the enzyme labeling by the above-mentioned enzyme antibody method. Also in the fluorescent antibody method, labeling may be performed directly or indirectly using a secondary antibody.
 免疫染色された試料を、撮影装置を搭載した光学顕微鏡又は蛍光顕微鏡(蛍光染色の場合)などを用いて撮像し、デジタル画像としてコンピュータに保存する。デジタル画像は二次元データであっても三次元データであってもよいが、本発明においては二次元データで十分に解析可能である。解析を行う前に、フィルタリングなどの画像処理を行ってもよい。  Take an image of the immunostained sample using an optical microscope equipped with an imaging device or a fluorescence microscope (in the case of fluorescence staining), and save it in a computer as a digital image. The digital image may be two-dimensional data or three-dimensional data, but two-dimensional data can be sufficiently analyzed in the present invention. Image processing such as filtering may be performed before the analysis.
 免疫染色された試料の画像解析は、使用する顕微鏡等に搭載されている画像解析用ソフトウエア、その他の市販のソフトウエアを用いて行うことができる。市販ソフトウエアの一例として、ScanScope(登録商標、Aperio社)が挙げられる。 Image analysis of immunostained samples can be performed using image analysis software installed in the microscope used and other commercially available software. An example of commercially available software is ScanScope (registered trademark, Aperio Inc.).
 免疫染色強度の分類は、免疫組織染色の病理専門医による標準的な判定基準に従うことができ、例えばスコア0(未検出)、スコア+1(弱い)、スコア+2(中程度)、及びスコア+3(顕著)のように分類してもよい。また、例えば、スコア+1以上の場合、アネキシンA1のN末端領域に結合するペプチドと抗がん剤とのコンジュゲート、該コンジュゲートと薬学的に許容される担体とを含む組成物等を用いたがん治療の適用対象と判断してもよい。 Classification of immunostaining intensity can follow standard criteria by pathologists for immunohistochemical staining, such as score 0 (undetected), score +1 (weak), score +2 (moderate), and score +3 (significant). ) May be classified. Further, for example, when the score is +1 or more, a conjugate of a peptide that binds to the N-terminal region of annexin A1 and an anticancer agent, a composition containing the conjugate and a pharmaceutically acceptable carrier, and the like were used. It may be judged that the cancer treatment is applied.
 本明細書において、対象由来の試料とは、本発明のアネキシンA1のN端領域に特異的に結合するモノクローナル抗体と接触させて、アネキシンA1のN末端領域の存在の有無を検出することができるものであれば特に限定されないが、好ましくは、対象から生検等により採取されたがん細胞を含む生検サンプルを用いて作製された組織切片であり、より好ましくは、パラフィン包埋された組織切片である。 In the present specification, a sample derived from a subject can be contacted with a monoclonal antibody of the present invention that specifically binds to the N-terminal region of Annexin A1 to detect the presence or absence of the N-terminal region of Annexin A1. It is not particularly limited as long as it is one, but is preferably a tissue section prepared by using a biopsy sample containing cancer cells collected by biopsy from a subject, and more preferably a paraffin-embedded tissue. It is a section.
 本発明の方法において使用されるアネキシンA1のN末端領域に結合するペプチド(以下、「本発明の方法で使用されるペプチド」と称する場合がある)としては、例えば、下記(I)~(III)のいずれかのアミノ酸配列を含むペプチド:
(I)(X1)[D]P[D](X2)[D]のアミノ酸配列(該配列中、X1はW又はFを示し、X2はS又はTを示す。)、
(II)P[D]T[D](X)F[D]のアミノ酸配列(該配列中、(X)は互いに独立して選択されるn個の任意のアミノ酸を示し、nは0~4の整数を示す。)、
(III)前記(I)又は(II)のいずれかのアミノ酸配列のRetro−inversoであるアミノ酸配列
が挙げられる。
Examples of the peptide that binds to the N-terminal region of annexin A1 used in the method of the present invention (hereinafter sometimes referred to as “peptide used in the method of the present invention”) include, for example, the following (I) to (III Peptide containing any of the amino acid sequences of:
(I) Amino acid sequence of (X1)[D]P[D](X2)[D] (wherein X1 represents W or F, X2 represents S or T),
(II) P[D]T[D](X) n F[D] amino acid sequence (wherein (X) n represents n arbitrary amino acids selected independently from each other, and n is Indicates an integer of 0 to 4),
(III) An amino acid sequence which is retro-inverso of the amino acid sequence of (I) or (II) above can be mentioned.
 また、本発明の方法で使用されるペプチドは、例えば、下記(i)~(vii)のいずれかのアミノ酸配列を含むペプチド:
(i)T[D]I[D]T[D]W[D]P[D]T[D]M[D]のアミノ酸配列、
(ii)L[D]R[D]F[D]P[D]T[D]V[D]L[D]のアミノ酸配列、
(iii)L[D]L[D]S[D]W[D]P[D]S[D]A[D]のアミノ酸配列、
(iv)S[D]P[D]T[D]S[D]L[D]L[D]F[D]のアミノ酸配列、
(v)M[D]P[D]T[D]L[D]T[D]F[D]R[D]のアミノ酸配列、
(vi)前記(i)~(v)のいずれかのアミノ酸配列において1若しくは数個のアミノ酸の挿入、置換若しくは欠失、又はこれらの組み合わせを有するアミノ酸配列、
(vii)前記(i)~(vi)のいずれかのアミノ酸配列のRetro−inversoであるアミノ酸配列
が挙げられる。
The peptide used in the method of the present invention is, for example, a peptide containing any of the following amino acid sequences (i) to (vii):
(I) the amino acid sequence of T[D]I[D]T[D]W[D]P[D]T[D]M[D],
(Ii) the amino acid sequence of L[D]R[D]F[D]P[D]T[D]V[D]L[D],
(Iii) L[D]L[D]S[D]W[D]P[D]S[D]A[D] amino acid sequence,
(Iv) the amino acid sequence of S[D]P[D]T[D]S[D]L[D]L[D]F[D],
(V) the amino acid sequence of M[D]P[D]T[D]L[D]T[D]F[D]R[D],
(Vi) an amino acid sequence having one or several amino acid insertions, substitutions or deletions, or a combination thereof in the amino acid sequence of any of (i) to (v) above,
(Vii) An amino acid sequence which is retro-inverso of the amino acid sequence of any one of (i) to (vi) above can be mentioned.
 本明細書において、鎖状ペプチドのアミノ酸配列は、ペプチド標記の慣例に従って左側がN末端側、右側がC末端側で記載される。また、アミノ酸配列中の直後に記号[D]を付した各アミノ酸記号は該アミノ酸のD体を示し、アミノ酸配列中の直後に記号[D]を付していない各アミノ酸記号は、文脈に反しない限り、該アミノ酸のL体を示す。 In this specification, the amino acid sequence of a chain peptide is described with the N-terminal side on the left side and the C-terminal side on the right side according to the convention of peptide notation. In addition, each amino acid symbol immediately after the symbol [D] in the amino acid sequence represents the D-form of the amino acid, and each amino acid symbol without immediately following the symbol [D] in the amino acid sequence is uncontextual. Unless otherwise indicated, the L form of the amino acid is shown.
 上記(I)のアミノ酸配列は、W[D]P[D]S[D]、W[D]P[D]T[D]、F[D]P[D]S[D]、又はF[D]P[D]T[D]のいずれであってもよい。 The amino acid sequence of (I) above is W[D]P[D]S[D], W[D]P[D]T[D], F[D]P[D]S[D], or F It may be any of [D]P[D]T[D].
 上記(II)のアミノ酸配列において、整数nは0~4であり、好ましくは2~3である。Xは互いに独立して選択される任意のアミノ酸であってもよい。 In the amino acid sequence of (II) above, the integer n is 0 to 4, preferably 2 to 3. X may be any amino acid selected independently of each other.
 本発明の方法で使用されるペプチドは上記の(I)~(III)及び(i)~(vii)のいずれかのアミノ酸配列からなるものであってもよいし、これらの配列のN末端側及び/又はC末端側に1つ以上のアミノ酸が付加されていてもよい。 The peptide used in the method of the present invention may consist of any of the amino acid sequences of (I) to (III) and (i) to (vii) described above, or the N-terminal side of these sequences. And/or one or more amino acids may be added to the C-terminal side.
 本明細書において、本発明の方法で使用されるペプチドとは、2つ以上のアミノ酸がペプチド結合したものをいう。本発明のペプチドの長さは特に限定されないが、例えば、少なくとも3個、少なくとも4個、少なくとも5個、少なくとも6個、少なくとも7個、少なくとも8個、少なくとも9個、少なくとも10個、少なくとも11個、少なくとも12個、少なくとも13個、少なくとも14個、又は少なくとも15個のアミノ酸を含んでもよい。本発明の方法で使用されるペプチドは、50個まで、45個まで、40個まで、35個まで、30個まで、25個まで、20個まで、19個まで、18個まで、17個まで、16個まで、15個まで、14個まで、13個まで、12個まで、11個まで、10個まで、9個まで、8個まで、又は7個までのアミノ酸からなるものであってもよい。 In the present specification, the peptide used in the method of the present invention refers to a peptide in which two or more amino acids are bound to each other. The length of the peptide of the present invention is not particularly limited, but for example, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 11 , At least 12, at least 13, at least 14, or at least 15 amino acids. The peptides used in the method of the present invention include up to 50, 45, 40, 35, 30, 25, 20, 19, 18, 18 and 17 peptides. , Up to 16, up to 15, up to 14, up to 13, up to 12, up to 11, up to 10, up to 9, up to 8, or up to 7 amino acids Good.
 本発明の方法で使用されるペプチドは、D型アミノ酸及びL型アミノ酸の組み合わせにより構成されていてもよい。また、本発明の方法で使用されるペプチドは、37 C.F.R. 1.821−1.822に言及されるものなどの、修飾(modified)又は異常(unusual)アミノ酸を含んでいてもよいし、含まなくてもよい。 The peptide used in the method of the present invention may be composed of a combination of D-type amino acids and L-type amino acids. Further, the peptide used in the method of the present invention is 37 C.I. F. R. It may or may not contain modified or unusual amino acids, such as those mentioned in 1.821-1.822.
 本発明の方法で使用されるペプチドのアミノ末端及び/又はカルボキシ末端は、改変されていてもよい。また、本発明の方法で使用されるペプチドは、N末端又はC末端以外においても種々の修飾を受けていてもよい。 The amino terminus and/or carboxy terminus of the peptide used in the method of the present invention may be modified. Further, the peptide used in the method of the present invention may have various modifications other than at the N-terminus or C-terminus.
 上記(vi)のアミノ酸配列中の置換は、保存的アミノ酸置換であることが好ましい。「保存的アミノ酸置換」は当該技術分野においてよく知られている。例えば、保存的アミノ酸置換は、側鎖の性質が類似するアミノ酸の間の置換として定義することができる。 The substitution in the amino acid sequence of (vi) above is preferably a conservative amino acid substitution. "Conservative amino acid substitutions" are well known in the art. For example, a conservative amino acid substitution can be defined as a substitution between amino acids with similar side chain properties.
 上記(vi)の配列は、上記(I)又は(II)の配列に包含されるものであってもよく、その場合、上記(vii)の配列は上記(III)の配列に包含されるものとなる。 The above (vi) sequence may be included in the above (I) or (II) sequence, in which case the above (vii) sequence is included in the above (III) sequence. Becomes
 ペプチドのRetro−inverso異性体は、元となるペプチドに対して、各アミノ酸残基のキラリティが反対であり(″inverso″)、且つアミノ酸配列の方向が反転している(″Retro″)ものをいう。Retro−inverso異性体は、元となるペプチドと類似の構造及び機能を示すことが知られている(例えば、Acc.Chem.Res.,1993,26(5),pp 266−273及びPLoS One.2013 Dec 2;8(12):e80390)。 The retro-inverso isomer of a peptide is one in which the chirality of each amino acid residue is opposite to that of the original peptide (“inverso”) and the direction of the amino acid sequence is reversed (“Retro”). Say. The Retro-inverso isomer is known to exhibit a structure and a function similar to that of the original peptide (eg, Acc. Chem. Res., 1993, 26(5), pp 266-273 and PLoS One. 2013 Dec 2;8(12):e80390).
 本発明の方法で使用されるペプチドは、上記(I)~(III)及び(i)~(vii)から選択される配列を2つ以上含んでいてもよい。本発明の方法で使用されるペプチドは、上記(I)~(III)及び(i)~(vii)のいずれかの配列のタンデムリピート(即ち、同一の配列部分が互いに直接的に連結された構造)を含んでもよい。また、本発明の方法で使用されるペプチドは、上記(I)~(III)及び(i)~(vii)の異なる2以上の配列が直接的に連結された構造を含んでもよい。具体的なリピート構造として、例えば、T[D]I[D]T[D]W[D]P[D]T[D]M[D]−T[D]I[D]T[D]E[D]P[D]T[D]M[D]、及びT[D]I[D]T[D]−T[D]I[D]T[D]が挙げられる。あるいは、本発明の方法で使用されるペプチドは、デンドリマーにより多価ペプチドを構成していてもよい。 The peptide used in the method of the present invention may contain two or more sequences selected from the above (I) to (III) and (i) to (vii). The peptide used in the method of the present invention is a tandem repeat of any one of the sequences (I) to (III) and (i) to (vii) (that is, identical sequence portions are directly linked to each other). Structure) may be included. In addition, the peptide used in the method of the present invention may include a structure in which two or more sequences (I) to (III) and (i) to (vii) different from each other are directly linked. As a specific repeat structure, for example, T[D]I[D]T[D]W[D]P[D]T[D]M[D]-T[D]I[D]T[D] E[D]P[D]T[D]M[D], and T[D]I[D]T[D]-T[D]I[D]T[D]. Alternatively, the peptide used in the method of the present invention may constitute a multivalent peptide by a dendrimer.
 本発明の方法で使用されるペプチドは遊離体であってもよいし、塩の形態であってもよい。本発明の方法で使用されるペプチドの塩としては、医薬的に許容される酸付加塩及び塩基付加塩が挙げられる。酸付加塩としては、塩酸、硫酸、硝酸及びリン酸などの無機酸との塩、酢酸、リンゴ酸、コハク酸、酒石酸及びクエン酸などの有機酸との塩が挙げられる。塩基付加塩としては、ナトリウム及びカリウムなどのアルカリ金属との塩、カルシウム及びマグネシウムなどのアルカリ土類金属との塩、アンモニウム及びトリエチルアミンなどのアミン類との塩が挙げられる。 The peptide used in the method of the present invention may be a free form or a salt form. Salts of the peptides used in the method of the present invention include pharmaceutically acceptable acid addition salts and base addition salts. Examples of the acid addition salt include salts with inorganic acids such as hydrochloric acid, sulfuric acid, nitric acid and phosphoric acid, and salts with organic acids such as acetic acid, malic acid, succinic acid, tartaric acid and citric acid. Base addition salts include salts with alkali metals such as sodium and potassium, salts with alkaline earth metals such as calcium and magnesium, and salts with amines such as ammonium and triethylamine.
 本発明の方法で使用されるペプチドは、ヒトアネキシンA1中の配列番号1で示される1~15番目のアミノ酸からなる領域、又はマウスアネキシンA1中の配列番号3(MAMVSEFLKQARFLE)で示される1~15番目のアミノ酸からなる領域に結合し得る。本発明の方法で使用されるペプチドは、例えばマウス又はヒトのアネキシンA1との分子間相互作用をQCM(Quartz Crystal Microbalance)法を用いて測定したときに、好ましくは10−6M未満、より好ましくは10−7M未満、更により好ましくは5x10−8M未満の解離定数(Kd値)を有し得る。上記測定のために、ATGen社(Seongnam−si,South Korea)から市販されている346アミノ酸残基から構成されるヒト組換えアネキシンA1タンパク質を用いることができる。 The peptide used in the method of the present invention is a region consisting of amino acids 1 to 15 shown in SEQ ID NO: 1 in human annexin A1, or 1 to 15 shown in SEQ ID NO: 3 (MAMVSEFLKQARFLE) in mouse annexin A1. Can bind to a region consisting of the th amino acid. The peptide used in the method of the present invention is preferably less than 10 −6 M, and more preferably less than 10 −6 M, when the intermolecular interaction with, for example, mouse or human annexin A1 is measured using the QCM (Quartz Crystal Microbalance) method. May have a dissociation constant (Kd value) of less than 10 −7 M, even more preferably less than 5×10 −8 M. For the above measurement, a human recombinant annexin A1 protein composed of 346 amino acid residues commercially available from ATGen (Seongnam-si, South Korea) can be used.
 本発明の方法で使用されるペプチドは、公知のペプチド合成法に従って製造することができる。ペプチド合成法は、例えば、固相合成法、液相合成法のいずれであってもよい。本発明のペプチドを構成し得る部分ペプチド若しくはアミノ酸と残余部分とを縮合し、生成物が保護基を有する場合は自体公知の方法により保護基を脱離することにより目的とするペプチドを製造することができる。 The peptide used in the method of the present invention can be produced according to a known peptide synthesis method. The peptide synthesis method may be, for example, either a solid phase synthesis method or a liquid phase synthesis method. A partial peptide or amino acid that can form the peptide of the present invention is condensed with a residual portion, and when the product has a protecting group, the target peptide is produced by removing the protecting group by a method known per se. You can
 このようにして得られたペプチドは、公知の精製法により精製単離することができる。ここで、精製法としては、例えば、溶媒抽出、蒸留、カラムクロマトグラフィー、液体クロマトグラフィー、再結晶、これらの組み合わせなどが挙げられる。
 上記方法で得られるペプチドが遊離体である場合には、該遊離体を公知の方法あるいはそれに準じる方法によって適当な塩に変換することができるし、逆にペプチドが塩として得られた場合には、該塩を公知の方法あるいはそれに準じる方法によって遊離体又は他の塩に変換することができる。
The peptide thus obtained can be purified and isolated by a known purification method. Here, examples of the purification method include solvent extraction, distillation, column chromatography, liquid chromatography, recrystallization, a combination thereof, and the like.
When the peptide obtained by the above method is a free form, the free form can be converted into an appropriate salt by a known method or a method analogous thereto, and conversely, when the peptide is obtained as a salt, The salt can be converted into a free form or another salt by a known method or a method analogous thereto.
 本発明の方法において使用される抗がん剤とは、悪性腫瘍(がん)の増殖を抑えることを目的とした薬剤をいう。抗がん剤の作用機序は特に限定されない。抗がん剤は、代謝拮抗剤、アルキル化剤、抗がん性抗生物質、微小管阻害剤、白金製剤、トポイソメラーゼ阻害剤、分子標的薬などであってよい。本発明のコンジュゲートは、2つ以上の同一又は異なる抗がん剤を含んでいてもよい。 The anticancer drug used in the method of the present invention refers to a drug intended to suppress the growth of malignant tumor (cancer). The action mechanism of the anticancer agent is not particularly limited. The anticancer agent may be an antimetabolite, an alkylating agent, an anticancer antibiotic, a microtubule inhibitor, a platinum preparation, a topoisomerase inhibitor, a molecular targeting drug and the like. The conjugate of the present invention may contain two or more same or different anticancer agents.
 代謝拮抗剤は、例えば、葉酸代謝拮抗薬、ジヒドロプテロイン酸シンターゼ阻害薬、ジヒドロ葉酸レダクターゼ阻害薬(DHFR阻害薬)、ピリミジン代謝阻害薬、チミジル酸シンターゼ阻害薬、プリン代謝阻害薬、IMPDH阻害薬、リボヌクレオチドレダクターゼ阻害薬、リボヌクレオチドレダクターゼ阻害薬、ヌクレオチドアナログ、L−アスパラギナーゼなどであってもよい。代謝拮抗剤の具体例としては、エノシタビン(サンラビン)、カペシタビン(ゼローダ)、カルモフール(ミフロール)、クラドリビン(ロイスタチン)、ゲムシタビン(ジェムザール)、シタラビン(キロサイド)、シタラビンオクホスファート(スタラシド)、テガフール(アチロン、アフトフール、テフシール、フトラフール、ルナシンほか)、テガフール・ウラシル(ユーエフティ)、テガフール・ギメラシル・オテラシルカリウム(TS−1:ティーエスワン)、ドキシフルリジン(フルツロン)、ネララビン(アラノンジー)、ヒドロキシカルバミド(ハイドレア)、フルオロウラシル(5−FU、カルゾナール、ベンナン、ルナコール、ルナボン)、フルダラビン(フルダラ)、ペメトレキセド(アリムタ)、ペントスタチン(コホリン)、メルカプトプリン(ロイケリン)、メトトレキサート(メソトレキセート)などが挙げられる。 Antimetabolites include, for example, antifolates, dihydropteroate synthase inhibitors, dihydrofolate reductase inhibitors (DHFR inhibitors), pyrimidine metabolism inhibitors, thymidylate synthase inhibitors, purine metabolism inhibitors, IMPDH inhibitors , Ribonucleotide reductase inhibitors, ribonucleotide reductase inhibitors, nucleotide analogs, L-asparaginase and the like. Specific examples of the antimetabolites include enocitabine (san-rabine), capecitabine (xeloda), carmofur (mifurol), cladribine (leustatin), gemcitabine (gemzar), cytarabine (kiloside), cytarabine ocphosphate (staracide), tegafur (tegafur). Athyron, Aftful, Tefseal, Futrafur, Lunacin, etc.), Tegafur Uracil (Yufti), Tegafur Guimeracil Oteracil Potassium (TS-1: TS-1), Doxyfluridine (Furtulon), Nerarabine (Alanony), Hydroxycarbamide (Hydrea) ), fluorouracil (5-FU, carzonal, bennan, lunacol, lunabon), fludarabine (fludara), pemetrexed (alimuta), pentostatin (cophorin), mercaptopurine (leukerin), methotrexate (methotrexate) and the like.
 アルキル化剤の具体例としては、シクロホスファミド(エンドキサン)、イホスファミド(イホマイド)、メルファラン(アルケラン)、ブスルファン、チオテパ(テスパミン)などのナイトロジェンマスタード系アルキル化剤、ニムスチン(ニドラン)、ラニムスチン(サイメリン)、ダカルバシン(ダカルバシン)、プロカルバシン(塩酸プロカルバシン)、テモゾロマイド(テモダール)、カルムスチン(ギリアデル)、ストレプトゾトシン(ザノサー)、ベンダムスチン(トレアキシン)などのニトロソウレア系アルキル化剤などが挙げられる。 Specific examples of the alkylating agent include cyclophosphamide (endoxane), ifosfamide (ifomide), melphalan (alkeran), busulfan, thiotepa (tespamin), and other nitrogen mustard-based alkylating agents, nimustine (nidoran), ranimustine. (Cimerine), dacarbacin (dacarbacin), procarbacin (procarbacin hydrochloride), temozolomide (temodal), carmustine (giliadel), streptozotocin (zanocer), bendamustine (treaxin), and other nitrosourea-based alkylating agents.
 抗がん性抗生物質の具体例としては、アクチノマイシンD(コスメゲン)、アクラルビシン(アクラシノン)、アムルビシン(カルセド)、イダルビシン(イダマイシン)、エピルビシン(エピルビシン塩酸塩、ファモルビシン)、ジノスタチンスチマラマー(スマンクス)、ダウノルビシン(ダウノマイシン)、ドキソルビシン(アドリアシン)、ピラルビシン(ピノルビン、テラルビシン)、ブレオマイシン(ブレオ)、ペプロマイシン(ペプレオ)、マイトマイシンC(マイトマイシン)、ミトキサントロン(ノバントロン)、リポソーマルドキソルビシン(ドキシル)などが挙げられる。 Specific examples of the anticancer antibiotics include actinomycin D (cosmegen), aclarubicin (acracinone), amrubicin (calced), idarubicin (idamycin), epirubicin (epirubicin hydrochloride, famorubicin), zinostatin stimalamer (smanx). ), daunorubicin (daunomycin), doxorubicin (adriacin), pirarubicin (pinorbin, teralubicin), bleomycin (bleo), peplomycin (pepleo), mitomycin C (mitomycin), mitoxantrone (novantron), liposomal doxorubicin (doxyl), etc. Are listed.
 微小管阻害剤は、例えば、ビンブラスチン(エクザール)やビンクリスチン(オンコビン)、ビンデシン(フォルデシン)などのビンカアルカロイド系微小管重合阻害薬、パクリタキセル(タキソール)やドセタキセル(タキソテール)などのタキサン系微小管脱重合阻害薬などが挙げられる。 Microtubule inhibitors include, for example, vinca alkaloid-based microtubule polymerization inhibitors such as vinblastine (exar), vincristine (oncobin), and vindesine (fordesin), and taxane-based microtubule depolymerization such as paclitaxel (taxol) and docetaxel (taxotere). Examples include inhibitors.
 白金製剤としては、例えば、オキサリプラチン(エルプラット)、カルボプラチン(カルボプラチン、カルボメルク、パラプラチン)、シスプラチン(アイエーコール、コナブリ、シスプラチンなど)、ネダプラチン(アクプラ)などが挙げられる。 Examples of platinum preparations include oxaliplatin (elprat), carboplatin (carboplatin, carbomerk, paraplatin), cisplatin (ie equol, conaburi, cisplatin, etc.), nedaplatin (acpra), etc.
 トポイソメラーゼ阻害剤としては、例えば、カンプトテシン及びその誘導体(例えば、イリノテカン(カンプト)、ノギテカン(ハイカムチン)、SN−38など)などのI型トポイソメラーゼ阻害剤;ドキソルビシン(アドリアシン)などのアントラサイクリン系薬物、エトポシド(ラステッド、ベプシド)などのエピポドフィロトキシン系薬物、レボフロキサシン(クラビット)やシプロフロキサシン(シプロキサン)などのキノロン系薬物などのII型トポイソメラーゼ阻害剤が挙げられる。 Examples of topoisomerase inhibitors include type I topoisomerase inhibitors such as camptothecin and its derivatives (eg, irinotecan (campto), nogitecan (hycamtin), SN-38); anthracycline drugs such as doxorubicin (adriacin); etoposide. Examples thereof include type II topoisomerase inhibitors such as epipodophyllotoxin drugs such as (Lasted and bepside) and quinolone drugs such as levofloxacin (clavit) and ciprofloxacin (ciproxan).
 分子標的薬としては、例えば、レゴラフェニブ(スチバーガ)、セツキシマブ(アービタックス)、パニツムマブ(ベクティビックス)、ラムシルマブ(サイラムザ)、ゲフィチニブ(イレッサ)、エルロチニブ(タルセバ)、アファチニブ(ジオトリフ)、クリゾチニブ(ザーコリ)、アレクチニブ(アレセンサ)、セリチニブ、レンバチニブ(レンビマ)、トラスツズマブ(ハーセプチン)、ラパチニブ(タイケルブ)、ペルツズマブ(パージェタ)、スニチニブ(スーテント)、ソラフェニブ(ネクサバール)、アキシチニブ(インライタ)、パゾパニブ(ヴォトリエント)、ニボルマブ(オプジーボ)、ペムブロリズマブ、イピリムマブ(ヤーボイ)、ベムラフェニブ(ゼルボラフ)、エベロリムス(アフィニトール)、テムシロリムス(トーリセル)、リツキシマブ(リツキサン)、ベバシズマブ(アバスチン)、ゲルダナマイシンなどが挙げられる。 Examples of molecularly targeted drugs include regorafenib (stibagga), cetuximab (erbitux), panitumumab (vetivics), ramucirumab (cyramza), gefitinib (iressa), erlotinib (tarseva), afatinib (diotryf), crizotinib (crizotinib). Alectinib (Alesensor), Ceritinib, Lenvatinib (Lenvima), Trastuzumab (Herceptin), Lapatinib (Tykerb), Pertuzumab (Perjeta), Sunitinib (Sutent), Sorafenib (Nexavar), Axitinib (Inraitabi), Panitab (Inraitab) Opdivo), pembrolizumab, ipilimumab (Yarvoy), vemurafenib (Zelboraf), everolimus (Afinitor), temsirolimus (Torisel), rituximab (Rituxan), bevacizumab (Avastin), geldanamycin and the like.
 抗がん剤はまた、抗血管新生剤であってもよい。抗血管新生剤は、血管内皮成長因子(VEGF)若しくは他の血管新生因子、又はこれらの受容体を阻害するものであり得る。抗血管新生剤の具体例としては、アンギオスタチン、エンドスタチン、メタスタチン、抗VEGF抗体(例えば、アバスチン)、VEGFR−2インヒビター(例えば、SU5416、SU6668)などが挙げられる。 The anti-cancer agent may also be an anti-angiogenic agent. Anti-angiogenic agents can be those that inhibit vascular endothelial growth factor (VEGF) or other angiogenic factors, or their receptors. Specific examples of the anti-angiogenic agent include angiostatin, endostatin, metastatin, anti-VEGF antibody (for example, Avastin), VEGFR-2 inhibitor (for example, SU5416, SU6668) and the like.
 本発明の方法で使用されるコンジュゲートにおけるアネキシンA1のN末端領域に結合するペプチドと1つ以上の抗がん剤との間の結合の様式は特に限定されない。結合は、直接的なものであってもよいし、又はリンカーなどを介した間接的なものであってもよい。結合は、共有結合、非共有結合、又はこれらの組み合わせによるものであってもよい。1つ以上の抗がん剤は、直接的又は間接的に、アネキシンA1のN末端領域に結合するペプチドのN末端、C末端、又はそれ以外の位置において結合していてもよい。ペプチドと抗がん剤との連結は当該技術分野において周知であり、本発明のコンジュゲートにおいても、該結合は任意の公知の手段によるものであってよい。 The mode of binding between the peptide that binds to the N-terminal region of annexin A1 in the conjugate used in the method of the present invention and one or more anticancer agents is not particularly limited. The bond may be direct, or indirect via a linker or the like. The binding may be covalent, non-covalent, or a combination thereof. One or more anti-cancer agents may be attached, directly or indirectly, at the N-terminus, C-terminus, or other position of the peptide that binds to the N-terminal region of Annexin A1. The linkage between the peptide and the anticancer drug is well known in the art, and in the conjugate of the present invention, the binding may be by any known means.
 本発明の方法で使用されるコンジュゲート及び薬学的に許容される担体を含む組成物は、経口又は非経口投与に適する剤形として提供され得る。非経口投与のための組成物としては、例えば、注射剤、坐剤などが用いられ、注射剤は静脈注射剤、皮下注射剤、皮内注射剤、筋肉注射剤、点滴注射剤などの剤形を包含し得る。これらは、自体公知の方法に従って調製できる。 The composition containing the conjugate used in the method of the present invention and a pharmaceutically acceptable carrier can be provided as a dosage form suitable for oral or parenteral administration. As the composition for parenteral administration, for example, injections, suppositories, etc. are used, and the injections are in the form of intravenous injection, subcutaneous injection, intradermal injection, intramuscular injection, drip injection, etc. Can be included. These can be prepared according to a method known per se.
 経口投与のための組成物としては、固体又は液体の剤形、具体的には錠剤(糖衣錠、フィルムコーティング錠を含む)、丸剤、顆粒剤、散剤、カプセル剤(ソフトカプセル剤を含む)、シロップ剤、乳剤、懸濁剤などが挙げられる。このような組成物は公知の方法によって製造され、製剤分野において通常用いられる担体、希釈剤若しくは賦形剤を含有していても良い。 Compositions for oral administration include solid or liquid dosage forms, specifically tablets (including sugar-coated tablets, film-coated tablets), pills, granules, powders, capsules (including soft capsules), syrup. Agents, emulsions, suspensions and the like. Such a composition is produced by a known method and may contain a carrier, a diluent or an excipient which is usually used in the field of formulation.
 なお前記した組成物は、上記コンジュゲートとの配合により好ましくない相互作用を生じない限り他の活性成分を含有してもよい。 Note that the above-mentioned composition may contain other active ingredients as long as it does not cause an unfavorable interaction with the above-mentioned conjugate.
 上記の非経口用又は経口用医薬組成物は、活性成分の投与量に適合するような投薬単位の剤形に調製されることが好都合である。このような投薬単位の剤形としては、例えば、錠剤、丸剤、カプセル剤、注射剤(アンプル)、坐剤が挙げられる。ペプチド又はコンジュゲートの含有量としては、投薬単位剤形当たり通常1~500mg、とりわけ注射剤では1~100mg、その他の剤形では10~250mgの上記ペプチド又はコンジュゲートが含有されていることが好ましい。 The above-mentioned parenteral or oral pharmaceutical composition is conveniently prepared in a unit dosage form suitable for the dose of the active ingredient. Examples of dosage forms of such dosage units include tablets, pills, capsules, injections (ampoules), and suppositories. The content of the peptide or conjugate is usually 1 to 500 mg per dosage unit dosage form, particularly 1 to 100 mg for injections, and preferably 10 to 250 mg for the other dosage forms. ..
 本発明の方法で使用されるコンジュゲート及び薬学的に許容される担体を含む組成物の投与量は、投与目的、投与対象、対象疾患、症状、投与ルートなどによっても異なり、当業者により適宜設定することができる。 The dose of the composition containing the conjugate used in the method of the present invention and a pharmaceutically acceptable carrier varies depending on the purpose of administration, administration subject, target disease, symptom, administration route, etc., and is appropriately set by those skilled in the art. can do.
 以下に実施例などを挙げて本発明をより具体的に説明するが、本発明は以下の実施例などにより限定されるものではない。 The present invention will be described in more detail with reference to the following examples, but the present invention is not limited to the following examples.
実施例1(マウスモノクローナル抗体の作製)
(1)免疫原の作成
 合成ペプチドMC16のC末端のシステイン(Cys)を、二価反応性試薬を用いてキャリアタンパク質(BSA)に結合させて免疫原を作製した。
(2)スクリーニング用HRP標識体の作製
 合成ペプチドMC16のC末端のCysを、二価反応性試薬を用いて西洋わさび由来ペルオキシダーゼ(HRP)に結合させてスクリーニング用HRP標識体を作製した。
Example 1 (production of mouse monoclonal antibody)
(1) Preparation of immunogen The C-terminal cysteine (Cys) of synthetic peptide MC16 was bound to a carrier protein (BSA) using a bivalent reactive reagent to prepare an immunogen.
(2) Preparation of HRP Label for Screening C-terminal Cys of synthetic peptide MC16 was bound to horseradish-derived peroxidase (HRP) using a divalent reactive reagent to prepare an HRP label for screening.
(3)免疫
 上記(1)で作製した免疫原性を持つ抗原を、FCAと混和して乳化させ、Balb/cマウス1匹に対して、計算上200μgを尾根部筋肉内に注射(免疫)した(合計3匹)。初回の抗体活性測定の結果を踏まえ、さらに同じ抗原を、FIAと混和して乳化させ、2週間間隔で2回、マウス1匹に対して、計算上50μgを皮下に注射(追加免疫)した。2回の追加免疫後の抗体活性測定の結果を踏まえ、マウスの腹腔に最終免疫(マウス1匹に対して計算上100μg)を実施した。
(3) Immunization The immunogenic antigen prepared in (1) above was mixed with FCA to emulsify, and one Balb/c mouse was calculated and injected with 200 μg intramuscularly in the ridge (immunization). (3 in total). Based on the result of the first antibody activity measurement, the same antigen was further mixed with FIA to emulsify, and 50 μg was subcutaneously injected (boost) to one mouse twice at two-week intervals. Based on the results of antibody activity measurement after two booster immunizations, a final immunization (calculated 100 μg per mouse) was performed on the abdominal cavity of the mice.
(4)試血及び抗体活性測定
 初回及び追加免疫から一定期間経過した後、マウスより試血して、合成MC16ペプチドをプラスチックプレートに結合させたELISAにより、抗体活性の測定を実施した。その結果から、細胞融合を実施するマウスを選択した。
(5)リンパ球及び抗血清の採取
最終免疫より3日後に脾臓を採取し、リンパ球を分離した後、−80℃で凍結保存した。同時に、採血を実施し、血清を分離した後、−40℃で凍結保存した。得られた抗血清を、スクリーニング時の陽性コントロールとして用いた。
(4) Test Blood and Measurement of Antibody Activity After a certain period of time from the initial and booster immunizations, blood was sampled from mice, and antibody activity was measured by ELISA in which a synthetic MC16 peptide was bound to a plastic plate. From the results, mice were selected for cell fusion.
(5) Collection of lymphocytes and antiserum Three days after the final immunization, the spleen was collected, lymphocytes were separated, and then frozen and stored at -80°C. At the same time, blood was collected, serum was separated, and then frozen and stored at -40°C. The obtained antiserum was used as a positive control during screening.
(6)細胞融合~培養
 上記(5)で得られたリンパ球とマウスミエローマを、50%ポリエチレングリコールの存在下で細胞融合を実施した。融合細胞(ハイブリドーマ)を、HAT培地に分散させ、96ウェルマイクロプレート4枚に分注し、37℃炭酸ガスインキュベーターで培養した。細胞融合から6日間にHT培地に交換し、さらに培養を続けた。
(6) Cell fusion-culture The lymphocytes obtained in (5) above and mouse myeloma were subjected to cell fusion in the presence of 50% polyethylene glycol. The fused cells (hybridomas) were dispersed in HAT medium, dispensed into four 96-well microplates, and cultured in a 37° C. carbon dioxide incubator. 6 days after cell fusion, the medium was replaced with HT medium, and the culture was further continued.
(7)一次スクリーニング~培養
 細胞融合から9日後、96ウェルマイクロプレートの各ウェルから培養上清をサンプリングし、培養上清のMC16に対する反応を、合成MC16ペプチドをプラスチックプレートに結合させたELISAで測定し、一次スクリーニングを実施した。
(8)二次スクリーニング及び細胞の凍結保存
 24ウェルマイクロプレートに移して3日間拡大培養させた後、マイクロプレートの各ウェルから培養上清をサンプリングし、上記と同様のELISAにより、二次スクリーニングを実施し、選択されたウェルの細胞の凍結保存を実施した。
(7) Primary screening-culture 9 days after cell fusion, the culture supernatant was sampled from each well of a 96-well microplate, and the reaction of the culture supernatant with MC16 was measured by an ELISA in which a synthetic MC16 peptide was bound to a plastic plate. Then, the primary screening was performed.
(8) Secondary screening and cryopreservation of cells After transferring to a 24-well microplate and expanding and culturing for 3 days, the culture supernatant is sampled from each well of the microplate, and the secondary screening is performed by the same ELISA as above. Performed, cryopreservation of cells in selected wells was performed.
(9)抗体活性測定
 2回目の追加免疫から2週間後、試血で得られた抗血清について、上記と同様のELISAにより、抗体活性を測定した。その結果、免疫したすべてのマウスで目的の抗原に対する抗体活性が確認され、細胞融合の実施に十分であると判断された。
(9) Antibody activity measurement Two weeks after the second booster, the antibody activity of the antiserum obtained by blood test was measured by the same ELISA as above. As a result, antibody activity against the antigen of interest was confirmed in all immunized mice, and it was judged that this was sufficient for carrying out cell fusion.
(10)一次スクリーニング
 上記と同様のELISAを実施し、目的の特異性を示す40個のクローンを選択し、拡大培養した。
(11)二次スクリーニング及び細胞の凍結保存
 上記と同様のELISAを実施した結果、多数の陽性クローンが得られた。該陽性クローンうち、18クローンの細胞を凍結保存した。
(10) Primary screening The same ELISA as described above was carried out, 40 clones showing the desired specificity were selected, and expanded and cultured.
(11) Secondary screening and cryopreservation of cells As a result of performing the same ELISA as above, a large number of positive clones were obtained. Of the positive clones, 18 clones of cells were cryopreserved.
実施例2(モノクローナル抗体産生ハイブリドーマ(クローン31−8D)の選択)
 実施例1にて作製したクローンから反応性の高いクローンを6個選び、培養マウス血管内皮細胞の免疫染色で最も優れた結果を与えたクローン(#31−8D,マウスIgG2b)を選択した。
Example 2 (Selection of monoclonal antibody-producing hybridoma (clone 31-8D))
Six highly reactive clones were selected from the clones prepared in Example 1, and the clone (#31-8D, mouse IgG2b) that gave the best results in immunostaining of cultured mouse vascular endothelial cells was selected.
実施例3(クローン31−8Dが生産する抗体(抗MC16抗体)の結合アッセイ)
 抗MC16抗体のANXA1タンパク質に対する結合性を確認するためにプレートアッセイを行った。具体的には、全長ヒトANXA1タンパク質を組み換えDNAを用いたバキュロウイルスベクターを用いて昆虫細胞で作製し、該作製したANXA1タンパク質をELISAプラスチックウェルに付着させた。コントロール培地(細胞培養用の培養液)、コントロールIgG(mouse IgG2b)、プロテインAカラムで精製した抗MC16抗体をそれぞれ反応させた後、HRP−anti−mouse IgGを反応させパーオキシダーゼの酵素活性を、パーオキシダーゼの基質を用いた発色で定量した。また、抗MC16抗体の特異性について、ウェスタンブロッティングを用いて確認した。
Example 3 (binding assay of antibody (anti-MC16 antibody) produced by clone 31-8D)
Plate assay was performed to confirm the binding of anti-MC16 antibody to ANXA1 protein. Specifically, full-length human ANXA1 protein was produced in insect cells using a baculovirus vector using recombinant DNA, and the produced ANXA1 protein was attached to an ELISA plastic well. A control medium (culture medium for cell culture), a control IgG (mouse IgG2b), and an anti-MC16 antibody purified with a protein A column were reacted, and then HRP-anti-mouse IgG was reacted to determine the enzymatic activity of peroxidase, It was quantified by color development using a substrate for peroxidase. In addition, the specificity of the anti-MC16 antibody was confirmed using Western blotting.
 上記アッセイの結果、クローン31−8Dが生産する抗体(抗MC16抗体)は、全長ヒトアネキシンA1と結合するが、N末端ドメイン(N末端の9アミノ酸)を欠損したアネキシンA1とは結合しないことを、ELISA(図1)及びウェスタンブロット(図2右)により確認した。
 一方、ウサギの抗アネキシンA1抗体(Invitrogen)は、全長ヒトアネキシンA1及びN末端ドメイン(N末端の9アミノ酸)を欠損したアネキシンA1の両方に結合した(図2)。
As a result of the above assay, it was confirmed that the antibody (anti-MC16 antibody) produced by clone 31-8D binds to full-length human annexin A1 but does not bind to annexin A1 lacking the N-terminal domain (N-terminal 9 amino acids). , ELISA (Fig. 1) and Western blot (Fig. 2 right).
On the other hand, the rabbit anti-annexin A1 antibody (Invitrogen) bound to both full-length human annexin A1 and annexin A1 lacking the N-terminal domain (N-terminal 9 amino acids) (FIG. 2).
実施例4(蛍光免疫染色の検討)
 マウス血管内皮細胞F2を単層培養し、マウスメラノーマB16細胞でマウスの皮下に形成した腫瘍の抽出液を加え2日間培養した。F2細胞の表面を抗MC16抗体と反応させた後、蛍光標識した二次抗体で反応させた。
 マウス血管内皮F2細胞の蛍光免疫染色イメージの結果から分かるように、本発明の抗MC16抗体がマウスのAnxa1タンパク質にも結合する事が強く示唆された。また、B16腫瘍抽出液にはF2細胞表面にアネキシンA1のN末端ドメインを発現誘導する活性物質が含まれている可能性があることも示唆された(図3)。
Example 4 (Investigation of fluorescent immunostaining)
Mouse vascular endothelial cells F2 were cultured in a monolayer, an extract of a tumor formed subcutaneously in mice with mouse melanoma B16 cells was added, and the cells were cultured for 2 days. The surface of F2 cells was reacted with an anti-MC16 antibody and then with a fluorescently labeled secondary antibody.
As can be seen from the results of the fluorescent immunostaining image of mouse vascular endothelial F2 cells, it was strongly suggested that the anti-MC16 antibody of the present invention also binds to mouse Anxa1 protein. It was also suggested that the B16 tumor extract may contain an active substance that induces the expression of the N-terminal domain of Annexin A1 on the surface of F2 cells (Fig. 3).
実施例5(抗体パーオキシダーゼ染色)
 膠芽腫を持った患者から採取した腫瘍組織のパラフィン包埋組織切片を、抗MC16抗体を用いたパーオキシダーゼ法で免疫染色した。その結果、抗MC16抗体が、腫瘍内の血管内皮細胞を陽性に染色することを確認した(図4左)。図4右は、ヘマトキシリン−エオシン染色である。
Example 5 (antibody peroxidase staining)
Paraffin-embedded tissue sections of tumor tissue collected from patients with glioblastoma were immunostained by the peroxidase method using anti-MC16 antibody. As a result, it was confirmed that the anti-MC16 antibody positively stained vascular endothelial cells in the tumor (Fig. 4, left). The right of FIG. 4 is a hematoxylin-eosin stain.
実施例6(抗体パーオキシダーゼ染色)
 前立腺がん腫瘍組織のパラフィン包埋組織切片を、抗MC16抗体とInvitrogenから市販のウサギanti−ANXA1抗体を用いたパーオキシダーゼ法で免疫染色した。その結果、両抗体とも腫瘍血管を陽性に染色したが、抗MC16抗体の染色結果はバックグラウンドも低く優れている事が判明した(図5~8)。
Example 6 (antibody peroxidase staining)
Paraffin-embedded tissue sections of prostate cancer tumor tissue were immunostained by the peroxidase method using an anti-MC16 antibody and a rabbit anti-ANXA1 antibody commercially available from Invitrogen. As a result, both antibodies stained tumor blood vessels positively, but it was revealed that the staining result of the anti-MC16 antibody was excellent with a low background (FIGS. 5 to 8).
 本発明は、日本国で出願された特願2019−017446(出願日:2019年2月1日)を基礎としており、その内容はすべて本明細書に包含されるものとする。 The present invention is based on Japanese Patent Application No. 2019-017446 filed in Japan (filing date: February 1, 2019), and all the contents are included in the present specification.
 本発明のモノクローナル抗体は、アネキシンA1のN末端領域に特異的に結合するため、腫瘍等組織の血管内皮表面におけるアネキシンA1のN末端領域の存在の有無を検出することができ、従って、例えば、アネキシンA1のN末端領域に結合するペプチドと抗がん剤とのコンジュゲートを用いたがん治療の適用対象の選別等を行うために利用可能である。
Since the monoclonal antibody of the present invention specifically binds to the N-terminal region of Annexin A1, it is possible to detect the presence or absence of the N-terminal region of Annexin A1 on the vascular endothelial surface of a tissue such as a tumor. It can be used for selecting an application target of cancer treatment using a conjugate of an anticancer agent and a peptide that binds to the N-terminal region of annexin A1.

Claims (11)

  1.  アネキシンA1のN末端領域に特異的に結合するモノクローナル抗体。 A monoclonal antibody that specifically binds to the N-terminal region of Annexin A1.
  2.  前記N末端領域が、配列番号1で示されるアミノ酸配列を含む領域である、請求項1に記載のモノクローナル抗体。 The monoclonal antibody according to claim 1, wherein the N-terminal region is a region containing the amino acid sequence represented by SEQ ID NO: 1.
  3.  受託番号NITE P−02801として寄託されたハイブリドーマにより産生されるモノクローナル抗体。 Monoclonal antibody produced by the hybridoma deposited under accession number NITE P-02801.
  4.  受託番号NITE P−02801として受託されたハイブリドーマ。 Hybridoma that has been entrusted with the consignment number NITE P-02801.
  5.  請求項1~3のいずれか1項に記載のモノクローナル抗体を含む、アネキシンA1のN末端領域の存在を検出するための試薬。 A reagent for detecting the presence of the N-terminal region of annexin A1, which comprises the monoclonal antibody according to any one of claims 1 to 3.
  6.  請求項1~3のいずれか1項に記載のモノクローナル抗体を含む、アネキシンA1のN末端領域に結合するペプチドと抗がん剤とのコンジュゲートを用いたがん治療の適用の可否を判定するための試薬。 Determining the applicability of cancer treatment using the conjugate of the peptide that binds to the N-terminal region of annexin A1 and the anticancer agent, which comprises the monoclonal antibody according to any one of claims 1 to 3. For reagents.
  7.  がんが固形がん又は液性がんである、請求項6に記載の試薬。 The reagent according to claim 6, wherein the cancer is solid cancer or liquid cancer.
  8.  腫瘍血管表面におけるアネキシンA1のN末端領域の存在を検出する方法であって、
    (1)請求項1~3のいずれか1項に記載のアネキシンA1のN末端領域に特異的に結合するモノクローナル抗体と、対象由来の試料とを接触させること
    (2)前記試料中において、腫瘍血管表面におけるアネキシンA1のN末端領域を、免疫染色によって検出すること
    を含む、方法。
    A method for detecting the presence of an N-terminal region of annexin A1 on the surface of a tumor blood vessel, comprising:
    (1) Contacting a monoclonal antibody that specifically binds to the N-terminal region of annexin A1 according to any one of claims 1 to 3 with a sample derived from a subject (2) tumor in the sample A method comprising detecting the N-terminal region of Annexin A1 on the surface of blood vessels by immunostaining.
  9.  アネキシンA1のN末端領域に結合するペプチドと抗がん剤とのコンジュゲートを用いたがん治療の適用対象の選別を補助するための方法であって、
    (1)請求項1~3のいずれか1項に記載のアネキシンA1のN末端領域に特異的に結合するモノクローナル抗体と、対象由来の試料とを接触させること
    (2)前記試料中において、腫瘍血管表面におけるアネキシンA1のN末端領域を、免疫染色によって検出すること
    を含む、方法。
    A method for assisting selection of an application target of cancer treatment using a conjugate of a peptide that binds to the N-terminal region of annexin A1 and an anticancer agent,
    (1) Contacting a monoclonal antibody that specifically binds to the N-terminal region of annexin A1 according to any one of claims 1 to 3 with a sample derived from a subject (2) tumor in the sample A method comprising detecting the N-terminal region of Annexin A1 on the surface of blood vessels by immunostaining.
  10.  前記対象由来の試料が、対象から生検により採取されたがん細胞を含む生検サンプルを用いて作製された組織切片である、請求項8又は9に記載の方法。 The method according to claim 8 or 9, wherein the sample derived from the subject is a tissue section prepared by using a biopsy sample containing cancer cells collected by biopsy from the subject.
  11.  前記がんが固形がん又は液性がんである、請求項9又は10に記載の方法。 The method according to claim 9 or 10, wherein the cancer is solid cancer or liquid cancer.
PCT/JP2020/004695 2019-02-01 2020-01-31 Anti-mc16 antibody WO2020158959A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
JP2019-017446 2019-02-01
JP2019017446A JP7224628B2 (en) 2019-02-01 2019-02-01 Anti-MC16 antibody

Publications (1)

Publication Number Publication Date
WO2020158959A1 true WO2020158959A1 (en) 2020-08-06

Family

ID=71841104

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2020/004695 WO2020158959A1 (en) 2019-02-01 2020-01-31 Anti-mc16 antibody

Country Status (2)

Country Link
JP (1) JP7224628B2 (en)
WO (1) WO2020158959A1 (en)

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2012510507A (en) * 2008-12-02 2012-05-10 クイーン メアリー アンド ウェストフィールド カレッジ Method of treating autoimmune disease by modulating annexin-1 (lipocortin 1)
JP2013515745A (en) * 2009-12-23 2013-05-09 サンフォード−バーナム メディカル リサーチ インスティテュート Methods and compositions relating to annexin 1-binding compounds
JP2013534914A (en) * 2010-06-09 2013-09-09 クイーン マリー アンド ウエストフィールド カレッジ、ユニバーシティ オブ ロンドン Annexin 1 antibody
US20140243507A1 (en) * 2013-02-28 2014-08-28 Samsung Electronics Co., Ltd. Method of screening antibodies with high antigen selectivity
JP2015501829A (en) * 2011-12-14 2015-01-19 クイーン マリー アンド ウエストフィールド カレッジ、ユニバーシティ オブ ロンドン Use of antibodies
US20150148253A1 (en) * 2013-11-28 2015-05-28 Samsung Electronics Co., Ltd. Cell with surface coated with anxa1 and use thereof
WO2018034356A1 (en) * 2016-08-16 2018-02-22 国立研究開発法人産業技術総合研究所 Malignant tumor target peptide

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2012510507A (en) * 2008-12-02 2012-05-10 クイーン メアリー アンド ウェストフィールド カレッジ Method of treating autoimmune disease by modulating annexin-1 (lipocortin 1)
JP2013515745A (en) * 2009-12-23 2013-05-09 サンフォード−バーナム メディカル リサーチ インスティテュート Methods and compositions relating to annexin 1-binding compounds
JP2013534914A (en) * 2010-06-09 2013-09-09 クイーン マリー アンド ウエストフィールド カレッジ、ユニバーシティ オブ ロンドン Annexin 1 antibody
JP2015501829A (en) * 2011-12-14 2015-01-19 クイーン マリー アンド ウエストフィールド カレッジ、ユニバーシティ オブ ロンドン Use of antibodies
US20140243507A1 (en) * 2013-02-28 2014-08-28 Samsung Electronics Co., Ltd. Method of screening antibodies with high antigen selectivity
US20150148253A1 (en) * 2013-11-28 2015-05-28 Samsung Electronics Co., Ltd. Cell with surface coated with anxa1 and use thereof
WO2018034356A1 (en) * 2016-08-16 2018-02-22 国立研究開発法人産業技術総合研究所 Malignant tumor target peptide

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
FUKUDA, MICHIKO,: "D-type peptideovercoming the blood-brain barrier and withtherapeutic activity against brain malignancies", FINAL RESEARCH REPORT OF 2017 'PROJECT FOR DEVELOPMENT OF INNOVATIVE RESEARCH ON CANCER THERAPEUTICS', JAPAN AGENCY FOR MEDICAL RESEARCH AND DEVELOPMENT, 19 March 2018 (2018-03-19) *
FUKUDA, MICHIKO,: "Therapeutic method for malignant tumor using carbohydrate-mimeticpeptide", SYMPOSIUM OF THE 12TH JAPAN CONSORTIUM FOR GLYCOBIOLOGY & GLYCOTECHNOLOGY), 2014, pages 56 - 57 *
HATAKEYAMA, S. ET AL.: "Targeted drug delivery to tumor vasculature by a carbohydrate mimetic peptide", PNAS, vol. 108, no. 49, 6 December 2011 (2011-12-06), pages 19587 - 19592, XP055685086, DOI: 10.1073/pnas.1105057108 *

Also Published As

Publication number Publication date
JP7224628B2 (en) 2023-02-20
JP2020125253A (en) 2020-08-20

Similar Documents

Publication Publication Date Title
US20220340682A1 (en) Anti-ceacam5 antibodies and uses thereof
US8841424B2 (en) Humanized AXL antibodies
JP2022534725A (en) Antibody drug conjugate dosage
CA2510315C (en) Antibodies against gpr64 and uses thereof
KR20190104160A (en) Anti GPR20 Antibody and Anti GPR20 Antibody-Drug Conjugate
JP5816188B2 (en) Anti-clusterin antibodies and antigen-binding fragments and their use to reduce tumor volume
JP2013522167A (en) uPAR binder and method of using the same
JP7440509B2 (en) CDCP1-targeted therapy
WO2016057683A2 (en) Novel anti-nodal antibodies and methods of using same
JP2024516581A (en) Anti-human CD73 antibodies and their applications
WO2023165615A1 (en) Combination therapies comprising a kras inhibitor for the treatment of cancer
WO2020158959A1 (en) Anti-mc16 antibody
WO2017147247A1 (en) Anti-sas1b antibodies and methods of use
WO2021251459A1 (en) Humanized anti-gpc-1 antibody
WO2022075482A1 (en) Medicine for treating cancer
US20220227882A1 (en) Anti-adam8 antibodies and uses of the same
TW202404646A (en) Dosage regimen of an anti-cdh6 antibody-drug conjugate
KR102661801B1 (en) Anti-ceacam5 antibodies and uses thereof
CN116410313A (en) CD30 humanized murine monoclonal antibody development

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: 20749236

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 20749236

Country of ref document: EP

Kind code of ref document: A1