WO2011074621A1 - Mesothelin (msln) antibody and use thereof - Google Patents

Mesothelin (msln) antibody and use thereof Download PDF

Info

Publication number
WO2011074621A1
WO2011074621A1 PCT/JP2010/072608 JP2010072608W WO2011074621A1 WO 2011074621 A1 WO2011074621 A1 WO 2011074621A1 JP 2010072608 W JP2010072608 W JP 2010072608W WO 2011074621 A1 WO2011074621 A1 WO 2011074621A1
Authority
WO
WIPO (PCT)
Prior art keywords
antibody
seq
amino acid
acid sequence
cells
Prior art date
Application number
PCT/JP2010/072608
Other languages
French (fr)
Japanese (ja)
Inventor
栄次 松浦
裕巳 公文
義朗 岸
慶宏 藤井
Original Assignee
株式会社医学生物学研究所
国立大学法人岡山大学
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 株式会社医学生物学研究所, 国立大学法人岡山大学 filed Critical 株式会社医学生物学研究所
Priority to JP2011546157A priority Critical patent/JPWO2011074621A1/en
Publication of WO2011074621A1 publication Critical patent/WO2011074621A1/en

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K51/00Preparations containing radioactive substances for use in therapy or testing in vivo
    • A61K51/02Preparations containing radioactive substances for use in therapy or testing in vivo characterised by the carrier, i.e. characterised by the agent or material covalently linked or complexing the radioactive nucleus
    • A61K51/04Organic compounds
    • A61K51/08Peptides, e.g. proteins, carriers being peptides, polyamino acids, proteins
    • A61K51/10Antibodies or immunoglobulins; Fragments thereof, the carrier being an antibody, an immunoglobulin or a fragment thereof, e.g. a camelised human single domain antibody or the Fc fragment of an antibody
    • A61K51/1045Antibodies or immunoglobulins; Fragments thereof, the carrier being an antibody, an immunoglobulin or a fragment thereof, e.g. a camelised human single domain antibody or the Fc fragment of an antibody against animal or human tumor cells or tumor cell determinants
    • A61K51/1057Antibodies or immunoglobulins; Fragments thereof, the carrier being an antibody, an immunoglobulin or a fragment thereof, e.g. a camelised human single domain antibody or the Fc fragment of an antibody against animal or human tumor cells or tumor cell determinants the tumor cell being from liver or pancreas
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K51/00Preparations containing radioactive substances for use in therapy or testing in vivo
    • A61K51/02Preparations containing radioactive substances for use in therapy or testing in vivo characterised by the carrier, i.e. characterised by the agent or material covalently linked or complexing the radioactive nucleus
    • A61K51/04Organic compounds
    • A61K51/08Peptides, e.g. proteins, carriers being peptides, polyamino acids, proteins
    • A61K51/10Antibodies or immunoglobulins; Fragments thereof, the carrier being an antibody, an immunoglobulin or a fragment thereof, e.g. a camelised human single domain antibody or the Fc fragment of an antibody
    • A61K51/1045Antibodies or immunoglobulins; Fragments thereof, the carrier being an antibody, an immunoglobulin or a fragment thereof, e.g. a camelised human single domain antibody or the Fc fragment of an antibody against animal or human tumor cells or tumor cell determinants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K51/00Preparations containing radioactive substances for use in therapy or testing in vivo
    • A61K51/02Preparations containing radioactive substances for use in therapy or testing in vivo characterised by the carrier, i.e. characterised by the agent or material covalently linked or complexing the radioactive nucleus
    • A61K51/04Organic compounds
    • A61K51/08Peptides, e.g. proteins, carriers being peptides, polyamino acids, proteins
    • A61K51/10Antibodies or immunoglobulins; Fragments thereof, the carrier being an antibody, an immunoglobulin or a fragment thereof, e.g. a camelised human single domain antibody or the Fc fragment of an antibody
    • A61K51/1045Antibodies or immunoglobulins; Fragments thereof, the carrier being an antibody, an immunoglobulin or a fragment thereof, e.g. a camelised human single domain antibody or the Fc fragment of an antibody against animal or human tumor cells or tumor cell determinants
    • A61K51/1051Antibodies or immunoglobulins; Fragments thereof, the carrier being an antibody, an immunoglobulin or a fragment thereof, e.g. a camelised human single domain antibody or the Fc fragment of an antibody against animal or human tumor cells or tumor cell determinants the tumor cell being from breast, e.g. the antibody being herceptin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K51/00Preparations containing radioactive substances for use in therapy or testing in vivo
    • A61K51/02Preparations containing radioactive substances for use in therapy or testing in vivo characterised by the carrier, i.e. characterised by the agent or material covalently linked or complexing the radioactive nucleus
    • A61K51/04Organic compounds
    • A61K51/08Peptides, e.g. proteins, carriers being peptides, polyamino acids, proteins
    • A61K51/10Antibodies or immunoglobulins; Fragments thereof, the carrier being an antibody, an immunoglobulin or a fragment thereof, e.g. a camelised human single domain antibody or the Fc fragment of an antibody
    • A61K51/1045Antibodies or immunoglobulins; Fragments thereof, the carrier being an antibody, an immunoglobulin or a fragment thereof, e.g. a camelised human single domain antibody or the Fc fragment of an antibody against animal or human tumor cells or tumor cell determinants
    • A61K51/1054Antibodies or immunoglobulins; Fragments thereof, the carrier being an antibody, an immunoglobulin or a fragment thereof, e.g. a camelised human single domain antibody or the Fc fragment of an antibody against animal or human tumor cells or tumor cell determinants the tumor cell being from lung
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K51/00Preparations containing radioactive substances for use in therapy or testing in vivo
    • A61K51/02Preparations containing radioactive substances for use in therapy or testing in vivo characterised by the carrier, i.e. characterised by the agent or material covalently linked or complexing the radioactive nucleus
    • A61K51/04Organic compounds
    • A61K51/08Peptides, e.g. proteins, carriers being peptides, polyamino acids, proteins
    • A61K51/10Antibodies or immunoglobulins; Fragments thereof, the carrier being an antibody, an immunoglobulin or a fragment thereof, e.g. a camelised human single domain antibody or the Fc fragment of an antibody
    • A61K51/1045Antibodies or immunoglobulins; Fragments thereof, the carrier being an antibody, an immunoglobulin or a fragment thereof, e.g. a camelised human single domain antibody or the Fc fragment of an antibody against animal or human tumor cells or tumor cell determinants
    • A61K51/1072Antibodies or immunoglobulins; Fragments thereof, the carrier being an antibody, an immunoglobulin or a fragment thereof, e.g. a camelised human single domain antibody or the Fc fragment of an antibody against animal or human tumor cells or tumor cell determinants the tumor cell being from the reproductive system, e.g. ovaria, uterus, testes or prostate
    • 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
    • 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

Definitions

  • the present invention relates to a tumor cell imaging agent comprising an anti-mesothelin antibody.
  • any cancer is desirably diagnosed at an early stage, but early cancers with poor physical findings and subjective symptoms are only detected accidentally in medical examinations.
  • mesothelioma that spreads rapidly along the pleura and peritoneum, or pancreatic cancer that is morphologically unclear is particularly difficult to detect and diagnose and is often too late.
  • diagnostic imaging such as MRI and CT has become widely used in Japan, but it has the merit of visualizing the inside of the body, but it is necessary to read the state of the blood vessel wall from the density and shape of the image. Detection of early mesothelioma and pancreatic cancer is difficult. It also depends on the skills of the doctors and laboratory technicians who perform the tests.
  • paramagnetic gadolinium and iron used in MRI and iodine contrast media used in CT have side effects and nephrotoxicity, and CT is accompanied by radiation exposure. Therefore, development of a new diagnostic method with less patient burden is desired.
  • mesothelin a kind of cell membrane glycoprotein, is expressed exclusively in the mesothelial cells that make up the pleura, peritoneum, pericardium and the like that wrap around organs such as the lung, heart, gastrointestinal tract, and liver.
  • mesothelial cells epidermal mesothelioma
  • mesothelin is high in various cancer tissues such as pancreatic cancer, ovarian cancer, lung cancer, head and neck cancer, colon cancer, and breast cancer. It is known to be expressed (Eur. J. Cancer, 44, 46-53, 2008, Clin. Cancer Res. 10, 3937-3942, 2004).
  • Mesothelin is first synthesized as a precursor protein having a total length of 71 kDa, and then a polypeptide of about 31 kDa called megakaryocyte potentiating factor (MPF) corresponding to positions 34-286 of the extracellular domain by a proteolytic enzyme such as furin,
  • MPF megakaryocyte potentiating factor
  • the GPI anchor breaks down into a 40 kDa polypeptide (mature mesothelin) left on the cell membrane.
  • part of the GPI-anchored membrane mesothelin is separated from the cell membrane and released (soluble mesothelin).
  • MPF and soluble mesothelin have been recognized as tumor markers, and serum measurement systems using the respective antibodies have been developed.
  • An object of the present invention is to provide an imaging agent for tumor cells containing an anti-mesothelin antibody.
  • a tumor cell imaging kit comprising an anti-mesothelin antibody.
  • the antibody is the antibody according to any of (a) to (e) below: (A) an antibody comprising a heavy chain having the amino acid sequence of SEQ ID NO: 4 as CDR1, the amino acid sequence of SEQ ID NO: 6 as CDR2, and the amino acid sequence of SEQ ID NO: 8 as CDR3; (B) an antibody comprising a heavy chain having the amino acid sequence set forth in SEQ ID NO: 2 as the heavy chain variable region; (C) an antibody comprising a light chain having the amino acid sequence of SEQ ID NO: 12 as CDR1, the amino acid sequence of SEQ ID NO: 14 as CDR2, and the amino acid sequence of SEQ ID NO: 16 as CDR3; (D) an antibody comprising a light chain having the amino acid sequence set forth in SEQ ID NO: 10 as the light chain variable region
  • the antibody is the antibody according to any one of the following (a) to (e); (A) an antibody comprising a heavy chain having the amino acid sequence of SEQ ID NO: 4 as CDR1, the amino acid sequence of SEQ ID NO: 6 as CDR2, and the amino acid sequence of SEQ ID NO: 8 as CDR3; (B) an antibody comprising a heavy chain having the amino acid sequence set forth in SEQ ID NO: 2 as the heavy chain variable region; (C) an antibody comprising a light chain having the amino acid sequence of SEQ ID NO: 12 as CDR1, the amino acid sequence of SEQ ID NO: 14 as CDR2, and the amino acid sequence of SEQ ID NO: 16 as CDR3; (D) an antibody comprising a light chain having the amino acid sequence set forth in SEQ ID NO: 10 as the light chain variable region, (E) An antibody having the heavy chain pair described in (a) or (A) an antibody comprising a heavy chain
  • the present invention provides a tumor cell imaging agent comprising an anti-mesothelin antibody.
  • the present invention also provides a method for imaging tumor cells, comprising the step of administering an anti-mesothelin antibody to a subject.
  • MSLN Mesothelin
  • Mesothelin has a molecular weight of about 40 kDa and (i) at least 10 consecutive amino acids of SEQ ID NO: 17 or the known variants listed in Swiss-plot accession number Q13421, or (ii) A polypeptide having an amino acid sequence comprising any of a portion of at least 20 contiguous amino acid residues, wherein the portion of the amino acid residue is the 20 contiguous residues of SEQ ID NO: 17, or the Swiss -Match at least 90% (preferably at least 95% or 100%) with the known variants listed in the accession number Q13421 of the plot.
  • the mesothelin variant is a detected 8-amino acid variant, PQAPRRPL (SEQ ID NO: 18), and the C-terminal VQGGRGGGQARAGGRAGGVEVGALSHPSLCRGPLGDALPPRTWTCSHRRPTAPPSLHPGLRAPLPC (SEQ ID NO: 19), MQEILS (SEQ ID NO: 19) It is known that it includes, but is not limited to, a variant substituted with (GTPCLLGPGPVVLTVALLALLLASTA) (SEQ ID NO: 21).
  • the mesothelin may contain glycophosphatidylinositol (GPI) anchors as post-translational modifications and many N-linked sugar chains.
  • the GPI anchor binds mesothelin to the cell membrane, but may be cleaved by a phospholipase to provide a soluble form of mesothelin.
  • the anti-mesothelin antibody included in the present invention is characterized by binding to the mesothelin.
  • antibodies included in the present invention include, but are not limited to, the following antibodies.
  • A an antibody comprising a heavy chain having the amino acid sequence of SEQ ID NO: 4 as CDR1, the amino acid sequence of SEQ ID NO: 6 as CDR2, and the amino acid sequence of SEQ ID NO: 8 as CDR3;
  • B an antibody comprising a heavy chain having CDR1, CDR2, CDR3 derived from a heavy chain variable region having the amino acid sequence of SEQ ID NO: 2;
  • C an antibody comprising a heavy chain having the amino acid sequence set forth in SEQ ID NO: 2 as the heavy chain variable region,
  • D an antibody comprising a light chain having the amino acid sequence of SEQ ID NO: 12 as CDR1, the amino acid sequence of SEQ ID NO: 14 as CDR2, and the amino acid sequence of SEQ ID NO: 16 as CDR3;
  • E an antibody comprising a light chain having CDR1, CDR2, CDR3 derived from a light chain variable region having the amino acid sequence of SEQ ID NO:
  • the antibody of the present invention includes both polyclonal and monoclonal antibodies. Methods for preparing and purifying monoclonal and polyclonal antibodies are known in the art and are described, for example, in Harlow and Lane, Antibodies: A Laboratory Manual (New York: Cold Spring Harbor Laboratory Press, 1988).
  • the antibodies of the present invention also include recombinant antibodies such as humanized antibodies or chimeric antibodies.
  • “Humanized antibody” refers to an antibody whose structure is similar to that of a human antibody.
  • Such humanized antibodies or chimeric antibodies include human chimeric antibodies (for example, antibodies in which part of the antibody is humanized, antibodies in which the CH2 region is humanized, antibodies in which the Fc region is humanized, constant regions) Humanized antibodies), and human CDR-grafted antibodies (PTJohons et al., Nature 321,522 () other than the CDRs (complementarity determining regions) present in the constant and variable regions) 1986)), fully humanized antibodies and the like.
  • a human chimeric antibody can be prepared, for example, by substituting the constant region of an antibody having the structure of the H chain variable region and / or the structure of the L chain variable region with a constant region of a human antibody.
  • Known constant regions of human antibodies can be employed.
  • the CDR is a CDR derived from an anti-mesothelin antibody
  • the FR is a FR derived from a human immunoglobulin
  • the constant region is a constant region derived from a human immunoglobulin.
  • humanized antibodies Specifically, a heavy chain variable region-derived CDR having the amino acid sequence set forth in SEQ ID NO: 2, a heavy chain variable region FR derived from a human immunoglobulin, and a heavy chain having a heavy chain constant region derived from a human immunoglobulin.
  • a chain, and a CDR derived from a light chain variable region having the amino acid sequence of SEQ ID NO: 10, and an FR derived from a light chain variable region of a human immunoglobulin, and a light chain constant region derived from a human immunoglobulin Mention may be made of humanized antibodies having a pair of light chains.
  • Examples of the chimeric antibody of the present invention include a chimeric antibody characterized in that the variable region is a variable region derived from an anti-mesothelin antibody and the constant region is a constant region derived from human immunoglobulin. .
  • a heavy chain variable region having the amino acid sequence set forth in SEQ ID NO: 2
  • a heavy chain having a heavy chain constant region derived from human immunoglobulin and the amino acid sequence set forth in SEQ ID NO: 10.
  • a chimeric antibody having a pair of a light chain variable region and a light chain having a light chain constant region derived from human immunoglobulin can be mentioned.
  • mRNA is extracted from a hybridoma producing a mouse antibody against a specific target antigen, and cDNA is synthesized according to a conventional method.
  • the synthesized cDNA is incorporated into a vector to construct a cDNA library.
  • a vector containing the H chain gene and the L chain gene is selected by using the H chain gene fragment and the L chain gene fragment as probes.
  • the sequences of the H chain variable region and L chain variable region genes are determined.
  • DNA encoding the H chain variable region is prepared by chemical synthesis, biochemical cleavage / recombination, or the like.
  • the resulting DNA encoding the H chain variable region is ligated with DNA encoding the human H chain constant region and incorporated into an expression vector to prepare an H chain expression vector.
  • expression vectors include, but are not limited to, SV40 virus based vectors, EB virus based vectors, BPV (papilloma virus) based vectors, and the like.
  • an L chain expression vector is prepared by the same method. Host cells are cotransformed with these H chain expression vector and L chain expression vector.
  • CHO cells Choinese hamster ovary
  • the lipofectin method (RWMalone et al., Proc.Natl.Acad.Sci.USA 86,6077 (1989), PLFelgner et al., Proc.Natl.Acad.Sci.USA 84,7413 (1987) ), Electroporation method, calcium phosphate method (FL Graham & AJvan der Eb, Virology 52, 456-467 (1973)), DEAE-Dextran method and the like are preferably used.
  • the human chimeric antibody After culturing the transformant, the human chimeric antibody is separated from the cells of the transformant or from the culture solution.
  • methods such as centrifugation, ammonium sulfate fractionation, salting out, ultrafiltration, affinity chromatography, ion exchange chromatography, gel filtration chromatography and the like can be used in appropriate combination.
  • a human CDR-grafted antibody can be prepared, for example, by the following method. First, the amino acid sequences of the H chain variable region and the L chain variable region of an antibody against a specific antigen and the base sequence encoding the same are determined by the method described in the column of the method for producing a chimeric antibody. In addition, the amino acid sequence and base sequence of each CDR region are determined.
  • the first method is a method using a human antibody frame, such as NEWM and REI, whose three-dimensional structure has already been clarified (Riechmann L. et al., Nature 332, 323-3Z7 (1988); Tempst, PR. Et al., Protein Engineering) 7, 1501-1507 (1994); Ellis JH. et al., J. Immunol 155, 925-937 (1995)).
  • the second method is to select a human antibody variable region having the highest homology with the mouse antibody variable region of interest from the database and use the FR (Queen C.
  • the third method is to select the most commonly used amino acid in human antibody FRs (Sato K. et al., Mol Immunol 31, 371-381 (1994); Kobinger F. et al., Protein Engineering 6, 971-980 (1993); ttleKettleborough CA. et al., Protein Engineering 4, 773-783 (1991)). Any of these methods can be used in the present invention.
  • the amino acid sequence of the selected human FR is modified, it should be used as the FR amino acid sequence as long as the finally obtained human CDR-grafted antibody has a specific binding property to the target antigen. Can do.
  • the number of amino acids to be modified is preferably 30% or less of the entire FR, more preferably 20% or less of the entire FR, and further preferably 10% or less of the entire FR.
  • a DNA encoding the H chain variable region and the L chain variable region is designed by combining the FR selected by any of these methods and the CDR. Based on this design, DNA encoding the H chain variable region and DNA encoding the L chain variable region are respectively prepared by chemical synthesis, biochemical cleavage / recombination, and the like. Then, the DNA encoding the H chain variable region is incorporated into an expression vector together with the DNA encoding the human immunoglobulin H chain constant region to construct an H chain expression vector. Similarly, an L chain expression vector is constructed by incorporating DNA encoding the L chain variable region into an expression vector together with DNA encoding the human immunoglobulin L chain constant region. Examples of expression vectors include, but are not limited to, SV40 virus-based vectors, EB virus-based vectors, BPV (papilloma virus) based vectors, and the like.
  • Host cells are cotransformed with the H chain expression vector and L chain expression vector prepared by the above method.
  • CHO cells Choinese hamster ovary
  • SP2 / 0 cells mouse myeloma
  • the like are preferably used as host cells.
  • the lipofectin method (RWMalone et al., Proc.Natl.Acad.Sci.USA 86,6077 (1989), PLFelgner et al., Proc.Natl.Acad.Sci.USA 84,7413 ( 1987)), electroporation method, calcium phosphate method (FL GrahamGra & AJvan der Eb, Virology 52,456-467 (1973)), DEAE-Dextran method and the like are preferably used.
  • the human CDR-grafted antibody is isolated from the cells of the transformant or the culture solution. Separation and purification of the antibody can be performed by appropriately combining methods such as centrifugation, ammonium sulfate fractionation, salting out, ultrafiltration, affinity chromatography, ion exchange chromatography, and gel filtration chromatography.
  • a method for obtaining a human antibody is also known.
  • human lymphocytes are sensitized with a desired antigen or cells expressing the desired antigen in vitro, the sensitized lymphocytes are fused with human myeloma cells such as U266, and the desired human antibody having binding activity to the antigen is obtained. It can also be obtained (see Japanese Patent Publication No. 1-59878).
  • a desired human antibody can be obtained by immunizing a transgenic animal having all repertoires of human antibody genes with a desired antigen (International Patent Application Publication Nos. WO 93/12227, WO 92/03918, WO 94/02602, WO 94/25585, WO 96/34096, WO 96/33735).
  • antibodies or antibody fragments can be separated from antibody phage libraries produced using the techniques described in McCafferty et al. (Nature, 348: 552-554 (1990)). Clackson et al. (Nature, 352: 624-628 (1991)) and Marks et al. (J. Mol. Biol., 222: 581-597 (1991)) describe the separation of mouse and human antibodies using phage libraries. is doing. Subsequent publications generate high affinity (nM range) human antibodies by chain shuffling (Marks et al., Bio / Technology, 10: 779-783 [1992]), as well as to construct very large phage libraries.
  • bacteriophage (phage) display is a well-known technique that allows large oligopeptide libraries to be searched to identify members of those libraries capable of specifically binding to a polypeptide target.
  • Phage display is a technique by which various polypeptides are presented as fusion proteins to coat proteins on the surface of bacteriophage particles (Scott, J.K. and SmithSG. P. (1990) Science 249: 386).
  • the usefulness of phage display can quickly and effectively classify large libraries of selectively randomized protein variants (or random clone cDNAs) for those sequences that bind to target molecules with high affinity.
  • Peptides on phage (Cwirla, SE et al. (1990) Proc.
  • a phage display library is contacted with a first solution in which a ligand can bind to a target molecule and a second solution in which an affinity ligand does not bind to the target molecule to selectively isolate the bound ligand.
  • a method for isolating affinity ligands is described.
  • WO 97/46251 describes a method of biopanning a random phage display library with affinity purified antibodies, then isolating bound phage, followed by micropanning in the wells of a microplate to isolate high affinity binding phage. To do.
  • WO 97/47314 describes the use of a substrate subtraction library to identify enzyme specificity using a combinatorial library that may be a phage display library.
  • a method for selecting enzymes suitable for use in detergents used for phage display is described in WO 97/09446. Additional methods for selecting proteins that specifically bind are described in US Pat. Nos. 5,498,538, 5,432,018, and WO 98/15833. Methods for preparing peptide libraries and screening these libraries are described in US Pat. Nos. 5,723,286, 5,432,018, 5,580,717, 5,427,908, 5,498,530, 5,770,434 and 5,734,018. Nos. 5,698,426, 5762192, and 5723323.
  • variable region of a human antibody is expressed as a single chain antibody (scFv) on the surface of the phage by the phage display method, and a phage that binds to the antigen can be selected.
  • scFv single chain antibody
  • the DNA sequence encoding the variable region of the human antibody that binds to the antigen can be determined. If the DNA sequence of scFv that binds to the antigen is clarified, a human antibody can be obtained by preparing an appropriate expression vector having the sequence and introducing it into an appropriate host for expression.
  • human antibodies and antibody fragments can be generated in vitro from the immunoglobulin variable (V) domain gene repertoire of non-immunized donors using phage display technology (McCafferty et al., Nature 348: 552-553 [1990]). Can be produced.
  • V domain genes are cloned in frame units with either filamentous bacteriophage, eg, M13 or fd coat protein genes, and displayed as functional antibody fragments on the surface of phage particles. Since the filamentous particle contains a single-stranded DNA copy of the phage genome, the selection of a gene encoding an antibody exhibiting these properties results as a result, even based on selection based on the functional properties of the antibody.
  • phage display can be performed in a variety of formats; see, for example, Johnson, Kevin S. and Chiswell, David J., Current Opinion in Structural Biology 3: 564-571 (1993).
  • V-gene segments can be used for phage display. Clackson et al., Nature, 352: 624-628 (1991) isolated a large number of diverse anti-oxazolone antibodies from a small random combinatorial library of V genes derived from the spleens of immunized mice.
  • V gene repertoire of non-immunized human donors is configurable, and antibodies against a wide variety of antigens (including self-antigens) can be found in Marks et al., J. Mol. Biol. 222: 581-597 (1991), or Griffith. Etc., EMBO J. 12: 725-734 (1993). See also U.S. Pat. Nos. 5,565,332 and 5,573,905.
  • the antibody of the present invention includes functional fragments of antibodies such as Fab, Fab ′, F (ab ′) 2 , Fv, scFv, dsFv, Diabody, sc (Fv) 2, and minibody.
  • multimers for example, dimers, trimers, tetramers, polymers
  • Fab has a molecular weight of about 50,000 composed of L chain and H chain variable region obtained by digesting IgG with papain in the presence of cysteine, and H chain fragment consisting of C H 1 domain and part of hinge part. It is a fragment.
  • the antibody can be obtained by digesting with papain.
  • Fab can be prepared from a transformant obtained by incorporating a DNA encoding part of the H chain and L chain of the above antibody into an appropriate vector and transforming using the vector.
  • Fab ′ is a fragment having a molecular weight of about 50,000 obtained by cleaving a disulfide bond between the H chains of F (ab ′) 2 described later.
  • the antibody is obtained by digesting with pepsin and cleaving disulfide bonds using a reducing agent.
  • it can also be prepared by genetic engineering using DNA encoding Fab ′.
  • F (ab ′) 2 is a fragment (Fab) composed of an L chain and an H chain variable region obtained by digesting IgG with pepsin, and an H chain fragment consisting of a C H 1 domain and a part of the hinge region.
  • Fab fragment having a molecular weight of about 100,000 bonded by a disulfide bond.
  • the antibody is obtained by pepsin digestion.
  • Fv can be expressed in an appropriate host cell after the antibody is treated with an enzyme such as papain or pepsin to generate antibody fragments, or a gene encoding these antibody fragments is constructed and introduced into an expression vector.
  • scFv is an antibody fragment in which an Fv comprising an H chain variable region and an L chain variable region is made into a single chain by linking the C terminus of one chain and the other N terminus with an appropriate peptide linker.
  • the peptide linker for example, (GGGGS) 3 having high flexibility can be used.
  • a DNA encoding an scFv antibody is constructed using DNA encoding the H chain variable region and L chain variable region of the above antibody and DNA encoding a peptide linker, and this is incorporated into an appropriate vector, and the vector is used.
  • ScFv can be prepared from the transformant transformed as described above.
  • dsFv is an Fv fragment in which a Cys residue is introduced at an appropriate position in the H chain variable region and the L chain variable region, and the H chain variable region and the L chain variable region are stabilized by disulfide bonds.
  • the position of Cys residue introduction in each chain can be determined based on the three-dimensional structure predicted by molecular modeling.
  • a three-dimensional structure is predicted from the amino acid sequences of the H chain variable region and the L chain variable region of the above-described antibody, and DNAs encoding the H chain variable region and L chain variable region into which mutations are introduced are constructed based on such prediction Then, this is incorporated into an appropriate vector, and dsFv can be prepared from a transformant transformed with the vector.
  • a fusion antibody or a labeled antibody can be constructed by fusing or binding a low molecular compound, protein, labeling substance or the like to the antibody (including antibody fragment) of the present invention.
  • a radioactive substance such as 125 I can be used as the labeling substance.
  • Diabody refers to a bivalent antibody fragment constructed by gene fusion (Holliger P et al., Proc. Natl. Acad. Sci. USA 90: 6444-6448 (1993), EP 404,097, WO 93 / 11161 etc.). Diabody is a dimer composed of two polypeptide chains. Normally, each polypeptide chain is short to a position where VL and VH cannot bind to each other in the same chain, for example, by a linker of about 5 residues. Are combined. Diabody has two antigen-binding sites because VL and VH encoded on the same polypeptide chain cannot form a single-chain variable region fragment due to the short linker between them, and form a dimer. It will be.
  • Diabody treats an antibody with an enzyme such as papain or pepsin to generate antibody fragments or constructs DNA encoding these antibody fragments and introduces them into an expression vector, and then in an appropriate host cell.
  • an enzyme such as papain or pepsin
  • Diabody treats an antibody with an enzyme such as papain or pepsin to generate antibody fragments or constructs DNA encoding these antibody fragments and introduces them into an expression vector, and then in an appropriate host cell.
  • sc (Fv) 2 is a low molecular weight antibody in which two VHs and two VLs are combined with a linker or the like to form a single chain (Hudson et al, J Immunol. Methods 1999; 231: 177-189). sc (Fv) 2 can be prepared, for example, by linking scFv with a linker.
  • a minibody (low molecular weight antibody) is a full-length antibody (whole antibody, such as whole IgG), which is a parent antibody and includes an antibody fragment in which a part of the full-length antibody is missing, and has a binding ability to an antigen. If it does not specifically limit.
  • the antibody of the present invention includes a fusion protein in which the antibody of the present invention is fused with another peptide or protein.
  • a polynucleotide encoding an antibody of the present invention and a polynucleotide encoding another peptide or polypeptide are linked so that the frames coincide with each other, introduced into an expression vector, and expressed in a host. Any technique known to those skilled in the art can be used.
  • Other peptides or polypeptides to be subjected to fusion with the antibody of the present invention include, for example, FLAG (Hopp, T. P.
  • examples of other polypeptides subjected to fusion with the antibody of the present invention include GST (glutathione-S-transferase), HA (influenza agglutinin), ⁇ -galactosidase, MBP (maltose binding protein) and the like. Can be mentioned.
  • the antibody of the present invention includes an antibody bound with a labeling substance.
  • the labeling substance include, but are not limited to, luminescence by enzyme luminescence (luciferase), a substance using a luminescent low molecule, a substance using a fluorescent protein or a fluorescent small molecule, and a radionuclide.
  • Radionuclides include gamma ray emitting nuclides such as 51 Cr, 59 Fe, 57 Co, 67 Ga, 75 Se, 81 m Kr, 99 m Tc, 111 In, 125 I, 131 I, 133 Xe, 201 Tl, 11 C, Positron emitting nuclides such as 13 N, 15 O, 18 F, 35 m Cl, 76 Br, 45 Ti, 48 V, 60 Cu, 61 Cu, 62 Cu, 66 Ga, 89 Zr, 94 m Tc, 124 I
  • m represents a nuclear isomer.
  • fluorescent labels and luminescent labels those using light emission by enzyme luminescence (luciferase) and fluorescence (fluorescent proteins such as GFP, DsRed, and wedge orange, and small fluorescent molecules such as FITC, Cy5.5, Alexa Fluor 750) Can be used.
  • luminescence by enzyme luminescence (luciferase) administration of a substrate is required separately.
  • those that reduce the influence of the animal's natural autofluorescence are preferred, and labels that emit signals with high skin permeability are preferred.
  • the present invention also provides DNA encoding the antibody of the present invention, a vector into which the DNA has been inserted, and a transformed cell into which the vector has been introduced.
  • vectors include M13 vectors, pUC vectors, pBR322, pBluescript, pCR-Script, and the like.
  • pGEM-T, pDIRECT, pT7 and the like can be mentioned.
  • a DNA encoding the antibody of the present invention, a vector into which the DNA has been inserted, and a transformed cell into which the vector has been introduced are prepared using known techniques.
  • Examples of the DNA encoding the anti-mesothelin antibody of the present invention include the following DNAs.
  • A DNA encoding the heavy chain having the amino acid sequence described in SEQ ID NO: 4 as CDR1, the amino acid sequence described in SEQ ID NO: 6 as CDR2, and the amino acid sequence described in SEQ ID NO: 8 as CDR3;
  • B DNA encoding a heavy chain having the amino acid sequence set forth in SEQ ID NO: 2 as the heavy chain variable region,
  • C DNA encoding a light chain having the amino acid sequence set forth in SEQ ID NO: 12 as CDR1, the amino acid sequence set forth in SEQ ID NO: 14 as CDR2, and the amino acid sequence set forth in SEQ ID NO: 16 as CDR3;
  • D DNA encoding a light chain having the amino acid sequence set forth in SEQ ID NO: 10 as the light chain variable region.
  • the host is E. coli such as JM109, DH5 ⁇ , HB101, XL1-Blue, etc.
  • promoters that can be efficiently expressed in E. coli such as the lacZ promoter (Ward et al., Nature (1989) 341, 544-546; FASEB J. (1992) 6, 2422-2427), araB promoter (Better et al. , (Science (1988) (240), (1041-1043)), or having a T7 promoter or the like.
  • such vectors include pGEX-5X-1 (Pharmacia), “QIAexpress® system” (Qiagen), pEGFP, or pET (in this case, the host expresses T7 RNA polymerase).
  • pGEX-5X-1 Pulacia
  • QIAexpress® system Qiagen
  • pEGFP pEGFP
  • pET in this case, the host expresses T7 RNA polymerase.
  • BL21 is preferred).
  • the vector may also contain a signal sequence for polypeptide secretion.
  • a signal sequence for protein secretion the pelB signal sequence (Lei, S. P. et al J. Bacteriol. (1987) 169, 4379) may be used when the periplasm of E. coli is used for production.
  • Introduction of a vector into a host cell can be performed using, for example, a calcium chloride method or an electroporation method.
  • mammalian-derived expression vectors for example, pcDNA3 (manufactured by Invitrogen), pEGF-BOS (Nucleic Acids. Res.1990, 18 (17), p5322), pEF, pCDM8), insects Expression vectors derived from cells (eg, “Bac-to-BAC baculovairus expression system” (Gibco BRL), pBacPAK8), plant-derived expression vectors (eg, pMH1, pMH2), animal virus-derived expression vectors (eg, pHSV, pMV, pAdexLcw), retrovirus-derived expression vectors (for example, pZIPneo), yeast-derived expression vectors (for example, “Pichia Expression Kit” (manufactured by Invitrogen), pNV11, SP-Q01), Bacillus subtilis-derived expression vectors (For example, pPL608, pKTH50).
  • mammalian-derived expression vectors for
  • promoters necessary for expression in cells such as the SV40 promoter (Mulligan et al., Nature (1979) 277, 108), It is essential to have MMTV-LTR promoter, EF1 ⁇ promoter (Mizushima et al., Nucleic Acids Res. (1990) 18, 5322), CMV promoter, etc., and genes for selecting transformation into cells (for example, More preferably, it has a drug resistance gene that can be discriminated by a drug (neomycin, G418, etc.). Examples of such a vector include pMAM, pDR2, pBK-RSV, pBK-CMV, pOPRSV, and pOP13.
  • a vector having a DHFR gene complementary to the CHO cell lacking the nucleic acid synthesis pathway for example, , PCHOI, etc.
  • amplifying with methotrexate (MTX) for example, COS with a gene expressing SV40 T antigen on the chromosome
  • COS with a gene expressing SV40 T antigen on the chromosome An example is a method of transforming with a vector (such as pcD) having an SV40 replication origin using cells.
  • a vector such as pcD
  • the replication origin those derived from polyoma virus, adenovirus, bovine papilloma virus (BPV) and the like can also be used.
  • the expression vectors are selectable markers: aminoglycoside transferase (APH) gene, thymidine kinase (TK) gene, E. coli xanthine guanine phosphoribosyltransferase (Ecogpt) gene, dihydrofolate reductase ( dhfr) gene and the like.
  • APH aminoglycoside transferase
  • TK thymidine kinase
  • Ecogpt E. coli xanthine guanine phosphoribosyltransferase
  • dhfr dihydrofolate reductase
  • the host cell into which the vector is introduced is not particularly limited, and for example, E. coli or various animal cells can be used.
  • the host cell can be used, for example, as a production system for production and expression of the antibody of the present invention.
  • Production systems for polypeptide production include in vitro and in vivo production systems. Examples of in vitro production systems include production systems using eukaryotic cells and production systems using prokaryotic cells.
  • animal cells for example, animal cells, plant cells, and fungal cells can be used as the host.
  • Animal cells include mammalian cells such as CHO (J. Exp. Med. (1995) 108, 945), COS, 3T3, myeloma, BHK (baby hamster kidney), HeLa, Vero, amphibian cells such as Xenopus eggs Mother cells (Valle, et al., Nature (1981) 291, 358-340) or insect cells such as Sf9, Sf21, and Tn5 are known.
  • CHO-DG44, CHO-DXB11, COS7 cells, and BHK cells are preferably used.
  • CHO cells are particularly preferred for mass expression purposes.
  • Introduction of a vector into a host cell can be performed by, for example, a calcium phosphate method, a DEAE dextran method, a method using a cationic liposome DOTAP (Boehringer Mannheim), an electroporation method, a lipofection method, or the like.
  • yeasts such as the genus Saccharomyces, such as Saccharomyces cerevisiae, Saccharomyces pombe, filamentous fungi such as the genus Aspergillus, such as Aspergillus. Aspergillus niger) is known.
  • Bacterial cells include E. coli (eg, JM109, DH5 ⁇ , HB101, etc.), and Bacillus subtilis is also known.
  • the antibody of the present invention can be obtained by culturing cells transformed with the DNA of the present invention in vitro and purifying the cells by a method commonly used by those skilled in the art.
  • the present invention also provides a host organism having a vector containing a nucleic acid encoding the antibody of the present invention.
  • the host organisms of the present invention are useful for the production of recombinant antibodies.
  • Examples of host organisms in the present invention include goats.
  • the transgenic goat of the present invention can be produced as follows. That is, a fusion gene is constructed in which an antibody gene is inserted in-frame into a gene encoding a protein (such as goat ⁇ -casein) that is uniquely produced in milk.
  • a DNA fragment containing a fusion gene into which an antibody gene has been inserted is injected into a goat embryo, the injected embryo is introduced into a female goat.
  • the antibody of the present invention can be obtained from the milk produced by a transgenic goat born from a goat that has received the embryo or its offspring.
  • hormones can also be used in the transgenic goat as appropriate (Ebert, KM et al., Bio / Technology (1994) 12 , 699-702).
  • the imaging agent of the present invention is administered to a mammal in order to image tumor cells.
  • imaging refers to measuring and visualizing an object by adding a labeling substance to an antibody.
  • a method of observing with a CCD camera using a fluorescent / radioisotope label, PET, SPECT, or CT using MRI is also included.
  • mammals include human and non-human mammals (eg, mouse, rat, hamster, rabbit, pig, monkey, etc.).
  • the imaging agent of the present invention is useful in the diagnosis of the presence or absence of tumor cells.
  • the imaging agent of the present invention can be used for both in vivo use and in vitro use.
  • the tumor cells imaged by the imaging agent of the present invention are cells expressing mesothelin.
  • Cells expressing mesothelin that is, cells imaged by the imaging agent of the present invention include lung cancer, mesothelioma, ovarian cancer, colon cancer, squamous cell carcinoma, pancreatic cancer, uterine cancer, head and neck cancer, breast cancer, etc. However, it is not limited to these.
  • the imaging agent of the present invention is obtained by binding an imaging label or probe that can be directly or indirectly tracked to an anti-mesothelin antibody.
  • the probe is administered to a living body in vivo (for example, intravenously administered)
  • the amount and distribution of tumor cells can be measured using an image measuring device such as PET, SPECT, or CCD camera.
  • CT computer computed tomography
  • PET positron tomography
  • SPECT single photon emission tomography
  • MRI nuclear magnetic resonance imaging
  • contrast agent such as tissue edema, abnormal bone morphology, morphology
  • Contrast-enhanced CT refers to imaging performed after injecting a contrast agent or the like having a high X-ray absorption rate into a blood vessel, and the shape of a tissue rich in blood vessels and blood flow can be observed.
  • next-generation CT for example, helical CT in which the radiation source moves in a spiral
  • multi-row detector CT also called multi-slice CT (MSCT)
  • MSCT multi-slice CT
  • labeling substance examples include, but are not limited to, light emission by enzyme luminescence (luciferase), a substance using a light emitting small molecule, a substance using a fluorescent protein or a fluorescent small molecule, and a radionuclide.
  • luminescence enzyme luminescence
  • Radionuclides include gamma ray emitting nuclides such as 51 Cr, 59 Fe, 57 Co, 67 Ga, 75 Se, 81 m Kr, 99 m Tc, 111 In, 125 I, 131 I, 133 Xe, 201 Tl, 11 C, Positron emitting nuclides such as 13 N, 15 O, 18 F, 35 m Cl, 76 Br, 45 Ti, 48 V, 60 Cu, 61 Cu, 62 Cu, 66 Ga, 89 Zr, 94 m Tc, 124 I
  • m represents a nuclear isomer.
  • indium-111, technetium-99m or iodine-131 can be used particularly preferably for plane scanning or single photon tomography (SPECT).
  • a positron emission label such as fluorine-18, which can be particularly preferably used in positron emission tomography.
  • Paramagnetic ions such as gadolinium (III) or manganese (II) are particularly preferably used in magnetic resonance imaging (MRI).
  • fluorescent labels and luminescent labels those using a luminescence system with an enzyme (luciferase), fluorescent proteins such as GFP, DsRed, and wedge orange, and fluorescent small molecules such as FITC, Cy5.5, Alexa Fluor 750, etc. ) Can be used.
  • luminescence by enzyme luminescence luciferase
  • administration of a substrate is required separately.
  • those that reduce the influence of the animal's natural autofluorescence are preferred, and labels that emit signals with high skin permeability are preferred.
  • the imaging detector magnetic resonance imaging (MRI) PET or SPECT is used.
  • the measuring instrument is preferably a CCD camera. Therefore, labels that emit light at a wavelength that can be captured by a CCD camera, for example, approximately 350-900 nm, are preferred.
  • a machine capable of measuring the intensity of the light source from the measurement value on the body surface of the animal to be measured by the CCD camera is preferable.
  • a fluorescent label either a reflected fluorescent image or a transmitted fluorescent image may be used, but it is preferable to capture both images.
  • CT alone can be sufficiently used (for example, PET or SPECT), the position of the tumor cell, Accumulation amount and distribution can be measured.
  • CT observation of the labeled probe or combined observation with CCD is also possible.
  • a simple CT and / or superimposition with an image of contrast CT
  • CCD image of a fluorescently labeled probe can be used for a CCD image of a fluorescently labeled probe.
  • CT images obtained by extraction of images of organs such as bones and lungs (and / or blood vessel and tissue images of contrast-enhanced CT) by simple CT and fluorescence probe images by a CCD camera are integrated, and tumor cells With regard to position, accumulation amount and distribution, it is possible to more accurately grasp the three-dimensional tissue, the relative positional relationship with blood vessels, and the three-dimensional (localized) image of the tumor cell.
  • the imaging agent of the present invention can be formulated by a known method by introducing a pharmaceutically acceptable carrier in addition to the antibody.
  • a pharmaceutically acceptable carrier for example, it can be used parenterally in the form of a sterile solution with water or other pharmaceutically acceptable liquid, or an injection of suspension.
  • a pharmacologically acceptable carrier or medium specifically, sterile water or physiological saline, vegetable oil, emulsifier, suspension, surfactant, stabilizer, flavoring agent, excipient, vehicle, preservative
  • a pharmaceutical preparation by combining with a binder or the like as appropriate and mixing in a unit dosage form generally required for pharmaceutical practice. The amount of active ingredient in these preparations is such that an appropriate volume within the indicated range can be obtained.
  • Sterile compositions for injection can be formulated according to normal pharmaceutical practice using a vehicle such as distilled water for injection.
  • Aqueous solutions for injection include, for example, isotonic solutions containing physiological saline, glucose and other adjuvants such as D-sorbitol, D-mannose, D-mannitol and sodium chloride, and suitable solubilizers such as You may use together with alcohol, specifically ethanol, polyalcohol, for example, propylene glycol, polyethylene glycol, nonionic surfactant, for example, polysorbate 80 (TM), HCO-50.
  • alcohol specifically ethanol, polyalcohol, for example, propylene glycol, polyethylene glycol, nonionic surfactant, for example, polysorbate 80 (TM), HCO-50.
  • TM polysorbate 80
  • oily liquid examples include sesame oil and soybean oil, which may be used in combination with benzyl benzoate or benzyl alcohol as a solubilizing agent.
  • oily liquid examples include sesame oil and soybean oil, which may be used in combination with benzyl benzoate or benzyl alcohol as a solubilizing agent.
  • buffer for example, phosphate buffer, sodium acetate buffer, a soothing agent, for example, procaine hydrochloride, stabilizer, for example, benzyl alcohol, phenol, antioxidant.
  • the prepared injection solution is usually filled into a suitable ampoule.
  • Administration is preferably parenteral administration, and specific examples include injection, nasal administration, pulmonary administration, and transdermal administration.
  • injection form it can be administered systemically or locally by, for example, intravenous injection, intramuscular injection, intraperitoneal injection, subcutaneous injection, or the like.
  • the administration method can be appropriately selected depending on the age and symptoms of the patient.
  • the dosage of the imaging agent of the present invention can be selected, for example, in the range of 0.0001 mg to 1000 mg per kg of body weight at a time. Alternatively, for example, the dose can be selected in the range of 0.001 to 100,000 mg / body per subject, but is not necessarily limited to these values.
  • the dose and administration method vary depending on the weight, age, symptom, symptom of the subject, the intensity of the fluorescent label per mg antibody, the detection sensitivity of the detection machine, etc., but can be appropriately selected by those skilled in the art.
  • the present invention also provides a tumor cell imaging kit comprising an anti-mesothelin antibody.
  • the kit of the present invention is characterized by imaging tumor cells by being administered to a subject.
  • the kit includes, for example, a labeling substance, an injection (infusion) device for administration of an imaging agent, an auxiliary agent that suppresses non-adsorption (such as albumin), and the like, but is not limited thereto. It is not something.
  • the kit may include instructions included in a normal kit, such as instructions for use in imaging, appropriate containers, and control reagents.
  • composition of the present invention described above can also be expressed as (1) to (3) below.
  • the base sequences and amino acid sequences of the antibodies disclosed herein are described in the sequence listing according to the following SEQ ID NOs.
  • the 11-25 antibody can also be referred to as # 11-25 antibody or clone 11-25.
  • SEQ ID NO: 1 base sequence of heavy chain variable region
  • SEQ ID NO: 2 amino acid sequence of heavy chain variable region
  • SEQ ID NO: 3 base sequence of heavy chain CDR1
  • SEQ ID NO: 4 amino acid sequence of heavy chain CDR1
  • 5 heavy chain CDR2
  • SEQ ID NO: 7 base sequence of heavy chain CDR3
  • SEQ ID NO: 9 base acid sequence of light chain variable region
  • SEQ ID NO: 10 light chain Variable region amino acid sequence
  • SEQ ID NO: 11 light chain CDR1 base sequence
  • SEQ ID NO: 12 light chain CDR1 amino acid sequence
  • SEQ ID NO: 13 light chain C
  • Example 1 The present inventors performed immunostaining on a plurality of cancer cells.
  • the antibody anti-mesothelin antibody used five types of antibodies, namely, 11-25 antibody, IC14-30, IC7-4, IC17-35, 2-9 antibody were used. As a control, one without added antibody was used. The method for obtaining and producing each antibody is as follows. 11-25 antibody: See Reference Examples 1 and 2, JP 2010-014691 IC14-30: Refer to JP2010-014691 IC7-4: See Reference Example 2 IC17-35: See Reference Example 2. 2-9 antibody: see Reference Example 2
  • the cell lines used are NCI-H226 (lung cancer), NCI-H520 (lung cancer), 211H (mesothelioma), PC3 (prostate cancer), PANC-1 (pancreatic cancer), BxPC-3 (pancreas) Cancer), CFPAC-1 (pancreatic cancer).
  • the method for obtaining each cell line is as follows. NCI-H226: Purchased from ATCC (search for CRL-5826). NCI-H520: Purchased from ATCC (search for HTB-182). 211H: Yuji Kashiwakura, et al. Cancer Res 2008; 68: (20). October 15: 8333-8341, 2008, Yuji Kashiwakura, et al.
  • PC3 Obtained from Caliper Life Sciences (The name is PC-3M-luc-C6).
  • PANC-1 Purchased from ATCC (http://www.atcc.org/ATCCAdvancedCatalogSearch/tabid/112/Default.aspx and entered CRL-1469 in ATCC numbers category search) (Sumisho Pharma International Co., Ltd.), See Lieber M, et al. Establishment of a continuous tumor-cell line (panc-1) from a human carcinoma of the exocrine pancreas. Int. J. Cancer 15: 741-747, 1975.
  • BxPC-3 Purchased from ATCC (searched with CRL-1687), see Loor R, et al.
  • pancreas-specific antigen in immunodiagnosis of pancreatic cancer. Clin. Lab. Med. 2: 567-578, 1982.
  • CFPAC-1 Purchased from ATCC (searched by CRL-1918), see Schoumacher RA, et al. A cystic fibrosis pancreatic adenocarcinoma cell line. Proc. Natl. Acad. Sci. USA 87: 4012-4016, 1990.
  • BxPC-3 pancreatic cancer cells were cultured in an 8-well chamber, immunostained with five anti-mesothelin antibodies, and observed with a confocal laser microscope (FIG. 1). Green indicates binding of anti-mesothelin antibody, red indicates actin filaments, and blue indicates nuclei. The 11-25 antibody showed significant anti-mesothelin antibody binding near the cell surface. Binding to the cells was not detected with the other four anti-mesothelin antibodies.
  • CFPAC-1 pancreatic cancer cells were cultured in a 2-well chamber, immunostained with five anti-mesothelin antibodies, and observed with a confocal laser microscope (FIG. 2).
  • PANC-1 pancreatic cancer cells were cultured in an 8-well chamber, immunostained with five anti-mesothelin antibodies, and observed with a confocal laser microscope (FIG. 3). Green indicates binding of anti-mesothelin antibody, red indicates actin filaments, and blue indicates nuclei. Binding of 11-25 anti-mesothelin antibody was observed in the cytoplasm.
  • PC3 cells showed binding of anti-mesothelin antibody to the cell membrane.
  • binding of anti-mesothelin antibody was observed in the cell membrane and cytoplasm.
  • H520 cells showed weak antibody binding to the cytoplasm.
  • anti-mesothelin antibody binding was observed in the membrane and cytoplasm.
  • Example 2 The cancer cells used in Example 1 were transplanted subcutaneously in the back of the back of nude mice, and after 3 weeks, 90 ⁇ g of Alexa 750 (Invitrogen) -labeled or Cy5.5 (GE Healthcare) -labeled 11-25 anti-mesothelin antibody was injected from the tail vein of the mice. After administration, the reflected fluorescence was measured with a biological fluorescence imaging apparatus IVIS200 (Xenogen) over time. When using Alexa 750-labeled antibody, excitation was performed at 745 nm and fluorescence at 800 nm, and when using Cy5.5-labeled antibody, measurement was performed at excitation at 675 nm and fluorescence at 740 nm.
  • Alexa 750 Invitrogen
  • Cy5.5 GE Healthcare
  • mice transplanted subcutaneously with PC3 prostate cancer cells and 211H mesothelioma cells were administered 90 ⁇ g of Alexa750-labeled 11-25 anti-mesothelin antibody via the tail vein, and fluorescence was measured 48 hours later.
  • the PC3 tumor was somewhat weak and the 211H tumor was strongly fluorescent (FIG. 6 (A)).
  • Nude mice transplanted subcutaneously with NCI-H520 and NCI-H226 lung cancer cells were administered 90 ⁇ g of Cy5.5-labeled 11-25 antibody via the tail vein, and the reflected fluorescence was measured 24 hours later.
  • the H520 tumor part did not show specific fluorescence, and the H226 tumor part emitted strong fluorescence (FIG. 6 (B)).
  • Nude mice transplanted subcutaneously with BxPC-3 and PANC-1 pancreatic cancer cells were administered 90 ⁇ g of Alexa750-labeled 11-25 anti-mesothelin antibody via the tail vein, and the reflected fluorescence was measured 24 hours later.
  • the BxPC-3 tumor part showed strong fluorescence, but no fluorescence was observed in the PANC-1 tumor part (FIG. 6 (C)).
  • Nude mice transplanted with NCI-520 lung cancer cells and CFPAC-1 pancreatic cancer cells were administered with tail vein of 90 ⁇ g of Alexa750-labeled 11-25 anti-mesothelin antibody, and the reflected fluorescence was measured 24 hours later.
  • FIG. 6 (D) The CFPAC-1 tumor part showed very strong fluorescence, but no specific fluorescence was observed in the H520 tumor part (FIG. 6 (D)).
  • FIG. 6 (E) is a photograph of the mouse tumor of FIG. 6 (B) taken out. H226 tumor showed strong fluorescence, while H520 tumor showed no specific fluorescence.
  • FIG. 6 (F) is a photograph of the mouse tumor of FIG. 6 (C) taken out. BxPC-3 tumors showed strong fluorescence while PANC-1 tumors did not show specific fluorescence.
  • FIG. 6 (G) is a photograph of the mouse tumor of (D) taken out. CFPAC-1 tumors showed strong fluorescence while H520 tumors did not show specific fluorescence.
  • Recombinant mesothelin protein is composed of 5′-AAATTTCCAAGCTTGTGGAGAAGACAGCCTGTCCTTCAGGCAAG-3 ′ (SEQ ID NO: 22) and 5′-AAGGAAAAAAGCGGCCCCGCCCTGTAGCCCCAGCCAGCGGTCCAG-3 ′ (SEQ ID NO: 23) using a primer called G It was created by amplifying the coding part of amino acids 297-580 derived from cDNA encoding transcription variant 1 of session number NM_005823). The amplified DNA was inserted into the HindIII / NotI sites of the expression vector pSecTag2B (Invitrogen).
  • Amino acids 297-580 of the protein encoded by clone NM005823 are common to human mesothelin isoforms 1 and 3 described by Scholler et al. (Proc Natl Acad Sci US A. 1999; 96 (20): 11531-6). Note that this is an array of The mesothelin expression plasmid was transfected into HEK293T cells. Mesothelin protein was purified from the culture supernatant of the transfected cells using TALON resin. The purified mesothelin protein thus obtained was dialyzed twice with PBS and stored at ⁇ 80 ° C. until use.
  • Culture supernatants were collected after 10 days of culture at 37 ° C., 5% CO 2 , saturated water vapor and screened for the ability to bind to the immunogen by indirect ELISA using recombinant mesothelin protein. Selected positive hybridoma colonies were grown and subcloned by limiting dilution. Subcloned hybridomas were cultured and the subcloned antibody isotype was determined using Roche's isostrip kit. Antibody purification was performed by protein A affinity chromatography. After an immunogen competition assay between the resulting clones, the 11-25 antibody (IgG2b kappa) was selected to construct an ELISA for ELISA detection of MPF. The 11-25 antibody was biotinylated using the GE Healthcare ECL protein biotination module. Similarly to the 11-25 antibody, IC7-4, IC17-35, 2-9 antibodies were prepared.
  • RNA was collected from hybridomas producing each of IC11-25 produced according to Reference Example 2.
  • Messenger RNA in the total RNA collected was reverse transcribed into complementary DNA using oligo (dT) 12-18 primer (purchased from Invitrogen). From this DNA, the nucleotide sequence of the immunoglobulin variable region sequence is 5'-ATGGCTGTCTTGGGGCTGCTCTTCTGC-3 '(SEQ ID NO: 24) and 5'-CAGTGGATAGACTGATGGGGG-3' (SEQ ID NO: 25) for the heavy chain of 11-25.
  • SEQ ID NO: 1 base sequence of heavy chain variable region
  • SEQ ID NO: 2 amino acid sequence of heavy chain variable region
  • SEQ ID NO: 3 base sequence of heavy chain CDR1
  • SEQ ID NO: 4 amino acid sequence of heavy chain CDR1
  • SEQ ID NO: 5 heavy chain CDR2
  • Base sequence SEQ ID NO: 6: amino acid sequence of heavy chain CDR2
  • SEQ ID NO: 7 base sequence of heavy chain CDR3
  • SEQ ID NO: 8 amino acid sequence of heavy chain CDR3
  • SEQ ID NO: 9 base acid sequence of light chain variable region
  • SEQ ID NO: 10 light chain Variable region amino acid sequence
  • SEQ ID NO: 11 light chain CDR1 base sequence
  • SEQ ID NO: 12 light chain CDR1 amino acid sequence
  • SEQ ID NO: 13 light chain CDR2 base sequence
  • SEQ ID NO: 14 light chain CDR2 amino acid sequence
  • SEQ ID NO: 15 light Nucleotide sequence of chain CDR3
  • SEQ ID NO: 16 Amino acid
  • the present inventors have developed a reagent that can directly image cancer derived from an actual patient that highly expresses mesothelin membrane.
  • the present invention is useful for imaging cancers (lung cancer, mesothelioma, ovarian cancer, colon cancer, squamous cell cancer, pancreatic cancer, uterine cancer, etc.) expressing mesothelin. .

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Chemical & Material Sciences (AREA)
  • Oncology (AREA)
  • Immunology (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Epidemiology (AREA)
  • Physics & Mathematics (AREA)
  • Optics & Photonics (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Cell Biology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Organic Chemistry (AREA)
  • Molecular Biology (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Reproductive Health (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Apparatus For Radiation Diagnosis (AREA)
  • Magnetic Resonance Imaging Apparatus (AREA)

Abstract

Disclosed is a tumor cell imaging agent containing anti-mesothelin antibody. As a result of the research of the inventors focusing on the aforementioned mesothelin, cultured cells derived from cancer cells inoculated subcutaneously into mice were imaged using labeled mesothelin antibodies.

Description

メソセリン(MSLN)に対する抗体及びその用途Antibodies against mesothelin (MSLN) and uses thereof
 本発明は、抗メソセリン抗体を含む、腫瘍細胞のイメージング剤に関する。 The present invention relates to a tumor cell imaging agent comprising an anti-mesothelin antibody.
 いずれの癌も早期のうちに診断されることが望ましいが、身体的所見や自覚症状の乏しい早期癌は、健康診断などで偶発的に発見される程度である。特に胸膜や腹膜に沿って急速に広がる中皮腫、あるいは形態的に明確でない膵臓癌は検出ならびに診断が特に難しく、手遅れとなるケースが多い。
 昨今、MRI、CTなどの画像診断が本邦では多用されるようになったが、体の内部を視覚化できるメリットはあるものの、画像の濃淡や形から血管壁の状況を読みとる必要があるため、早期の中皮腫や膵臓癌の検出は困難である。検査を行う医師や検査技師の技量にも左右される。また、MRIで使用する常磁性のガドリニウムや鉄及びCTで使用するヨード造影剤とも、副作用の発現、腎毒性があることや、CTでは放射線被曝を伴うなど、患者への直接的苦痛が強いられることから、患者負担のより少ない新規の診断方法の開発が望まれている。
Any cancer is desirably diagnosed at an early stage, but early cancers with poor physical findings and subjective symptoms are only detected accidentally in medical examinations. In particular, mesothelioma that spreads rapidly along the pleura and peritoneum, or pancreatic cancer that is morphologically unclear is particularly difficult to detect and diagnose and is often too late.
In recent years, diagnostic imaging such as MRI and CT has become widely used in Japan, but it has the merit of visualizing the inside of the body, but it is necessary to read the state of the blood vessel wall from the density and shape of the image. Detection of early mesothelioma and pancreatic cancer is difficult. It also depends on the skills of the doctors and laboratory technicians who perform the tests. In addition, paramagnetic gadolinium and iron used in MRI and iodine contrast media used in CT have side effects and nephrotoxicity, and CT is accompanied by radiation exposure. Therefore, development of a new diagnostic method with less patient burden is desired.
 細胞膜の糖タンパク質の一種であるメソセリンは、肺、心臓、胃腸、肝臓などの臓器を包む胸膜、腹膜、そして心膜などを構成する中皮細胞に限局して発現している。しかしながら、癌化した組織においては、中皮細胞(上皮型中皮腫)だけでなく、膵臓癌、卵巣癌、肺癌、頭頸部癌、大腸癌、乳癌など、多岐の癌組織において、メソセリンが高発現することが分かっている(Eur. J. Cancer, 44, 46-53, 2008, Clin. Cancer Res. 10, 3937-3942, 2004)。
 メソセリンは、まず全長71kDaの前駆体タンパク質として合成された後、フューリンなどのタンパク質分解酵素によって、細胞外ドメインの34-286番目に相当するmegakaryocyte potentiating factor(MPF)と呼ばれる約31kDaのポリペプチドと、GPIアンカーによって細胞膜上に残された40kDaのポリペプチド(成熟型メソセリン)に分解される。さらにGPIアンカー型の膜型メソセリンの一部は細胞膜から切り離され、遊離していることが知られている(可溶型メソセリン)。
 これらの分子的特徴を背景に、MPFと可溶型メソセリンは腫瘍マーカーとして認識され、それぞれの抗体を利用した血清測定系が開発されてきた。しかしながら、血清中の腫瘍マーカーを用いた検査は操作が簡便ではあるが、原発巣の特定、転移の有無は確認できない。また、癌が早期のうちには分泌されるMPFやメソセリンの量は少なく、血清を検体とした早期発見は原理的に困難と言わざるを得ない。また、インジウム111で標識されたメソセリンに対するモノクローナル抗体を用いた癌画像診断の方法論が報告されているが、用いられている癌細胞は、人工的にメソセリン遺伝子を強制的に導入された細胞であり、実際の癌患者における生理的条件を満たしている保証はない(Int J Cancer.80, 559-63, 1999)。
Mesothelin, a kind of cell membrane glycoprotein, is expressed exclusively in the mesothelial cells that make up the pleura, peritoneum, pericardium and the like that wrap around organs such as the lung, heart, gastrointestinal tract, and liver. However, in cancerous tissues, not only mesothelial cells (epithelial mesothelioma) but also mesothelin is high in various cancer tissues such as pancreatic cancer, ovarian cancer, lung cancer, head and neck cancer, colon cancer, and breast cancer. It is known to be expressed (Eur. J. Cancer, 44, 46-53, 2008, Clin. Cancer Res. 10, 3937-3942, 2004).
Mesothelin is first synthesized as a precursor protein having a total length of 71 kDa, and then a polypeptide of about 31 kDa called megakaryocyte potentiating factor (MPF) corresponding to positions 34-286 of the extracellular domain by a proteolytic enzyme such as furin, The GPI anchor breaks down into a 40 kDa polypeptide (mature mesothelin) left on the cell membrane. Furthermore, it is known that part of the GPI-anchored membrane mesothelin is separated from the cell membrane and released (soluble mesothelin).
Against the backdrop of these molecular features, MPF and soluble mesothelin have been recognized as tumor markers, and serum measurement systems using the respective antibodies have been developed. However, tests using serum tumor markers are easy to operate, but the identification of the primary lesion and the presence or absence of metastasis cannot be confirmed. In addition, the amount of MPF and mesothelin secreted in the early stages of cancer is small, and early detection using serum as a specimen must be difficult in principle. Although a methodology for cancer imaging diagnosis using a monoclonal antibody against mesothelin labeled with indium 111 has been reported, the cancer cells used are cells in which the mesothelin gene has been artificially introduced. There is no guarantee that physiological conditions in actual cancer patients will be met (Int J Cancer. 80, 559-63, 1999).
特開2007-71849JP2007-71849A 特許3490125号Patent 3490125
 本発明の課題は、抗メソセリン抗体を含む腫瘍細胞のイメージング剤の提供である。 An object of the present invention is to provide an imaging agent for tumor cells containing an anti-mesothelin antibody.
 本発明者らは、上記課題を解決するために、上記メソセリンに注目し、鋭意研究した。その結果、マウスの皮下に移植したがん細胞由来の培養細胞を、標識メソセリン抗体を用いて画像化することに成功した。
 本願は、この知見に基づき、以下の発明を提供するものである。
〔1〕抗メソセリン抗体を含む、腫瘍細胞のイメージング剤。
〔2〕抗体が下記(a)~(e)のいずれかに記載の抗体である、〔1〕記載のイメージング剤;
(a)CDR1として配列番号:4に記載のアミノ酸配列、CDR2として配列番号:6に記載のアミノ酸配列、及びCDR3として配列番号:8に記載のアミノ酸配列を有する重鎖を含む抗体、
(b)重鎖可変領域として配列番号:2に記載のアミノ酸配列を有する重鎖を含む抗体、
(c)CDR1として配列番号:12に記載のアミノ酸配列、CDR2として配列番号:14に記載のアミノ酸配列、及びCDR3として配列番号:16に記載のアミノ酸配列を有する軽鎖を含む抗体、
(d)軽鎖可変領域として配列番号:10に記載のアミノ酸配列を有する軽鎖を含む抗体、
(e)上記(a)または(b)に記載の重鎖、および、上記(c)または(d)に記載の軽鎖の対を有する抗体。
〔3〕インビボで用いられる、〔1〕または〔2〕記載のイメージング剤。
〔4〕抗メソセリン抗体を含む、腫瘍細胞のイメージング用キット。
〔5〕抗体が下記(a)~(e)のいずれかに記載の抗体である、〔4〕記載のイメージング用キット;
(a)CDR1として配列番号:4に記載のアミノ酸配列、CDR2として配列番号:6に記載のアミノ酸配列、及びCDR3として配列番号:8に記載のアミノ酸配列を有する重鎖を含む抗体、
(b)重鎖可変領域として配列番号:2に記載のアミノ酸配列を有する重鎖を含む抗体、
(c)CDR1として配列番号:12に記載のアミノ酸配列、CDR2として配列番号:14に記載のアミノ酸配列、及びCDR3として配列番号:16に記載のアミノ酸配列を有する軽鎖を含む抗体、
(d)軽鎖可変領域として配列番号:10に記載のアミノ酸配列を有する軽鎖を含む抗体、
(e)上記(a)または(b)に記載の重鎖、および、上記(c)または(d)に記載の軽鎖の対を有する抗体。
〔6〕インビボで用いられる、〔4〕または〔5〕記載のイメージング用キット。
〔7〕抗メソセリン抗体を用いる、腫瘍細胞のイメージング方法。
〔8〕抗体が下記(a)~(e)のいずれかに記載の抗体である、〔7〕記載のイメージング方法;
(a)CDR1として配列番号:4に記載のアミノ酸配列、CDR2として配列番号:6に記載のアミノ酸配列、及びCDR3として配列番号:8に記載のアミノ酸配列を有する重鎖を含む抗体、
(b)重鎖可変領域として配列番号:2に記載のアミノ酸配列を有する重鎖を含む抗体、
(c)CDR1として配列番号:12に記載のアミノ酸配列、CDR2として配列番号:14に記載のアミノ酸配列、及びCDR3として配列番号:16に記載のアミノ酸配列を有する軽鎖を含む抗体、
(d)軽鎖可変領域として配列番号:10に記載のアミノ酸配列を有する軽鎖を含む抗体、
(e)上記(a)または(b)に記載の重鎖、および、上記(c)または(d)に記載の軽鎖の対を有する抗体。
〔9〕腫瘍細胞のイメージング剤の製造における、抗メソセリン抗体の使用。
〔10〕抗体が下記(a)~(e)のいずれかに記載の抗体である、〔9〕記載の使用;
(a)CDR1として配列番号:4に記載のアミノ酸配列、CDR2として配列番号:6に記載のアミノ酸配列、及びCDR3として配列番号:8に記載のアミノ酸配列を有する重鎖を含む抗体、
(b)重鎖可変領域として配列番号:2に記載のアミノ酸配列を有する重鎖を含む抗体、
(c)CDR1として配列番号:12に記載のアミノ酸配列、CDR2として配列番号:14に記載のアミノ酸配列、及びCDR3として配列番号:16に記載のアミノ酸配列を有する軽鎖を含む抗体、
(d)軽鎖可変領域として配列番号:10に記載のアミノ酸配列を有する軽鎖を含む抗体、
(e)上記(a)または(b)に記載の重鎖、および、上記(c)または(d)に記載の軽鎖の対を有する抗体。
〔11〕腫瘍細胞のイメージング方法に使用するための、抗メソセリン抗体。
〔12〕抗体が下記(a)~(e)のいずれかに記載の抗体である、〔11〕記載の抗体;
(a)CDR1として配列番号:4に記載のアミノ酸配列、CDR2として配列番号:6に記載のアミノ酸配列、及びCDR3として配列番号:8に記載のアミノ酸配列を有する重鎖を含む抗体、
(b)重鎖可変領域として配列番号:2に記載のアミノ酸配列を有する重鎖を含む抗体、
(c)CDR1として配列番号:12に記載のアミノ酸配列、CDR2として配列番号:14に記載のアミノ酸配列、及びCDR3として配列番号:16に記載のアミノ酸配列を有する軽鎖を含む抗体、
(d)軽鎖可変領域として配列番号:10に記載のアミノ酸配列を有する軽鎖を含む抗体、
(e)上記(a)または(b)に記載の重鎖、および、上記(c)または(d)に記載の軽鎖の対を有する抗体。
In order to solve the above-mentioned problems, the present inventors paid attention to the above-mentioned mesothelin and conducted intensive research. As a result, we successfully imaged cultured cells derived from cancer cells transplanted under the skin of mice using labeled mesothelin antibodies.
This application provides the following invention based on this knowledge.
[1] An imaging agent for tumor cells, comprising an anti-mesothelin antibody.
[2] The imaging agent according to [1], wherein the antibody is the antibody according to any of (a) to (e) below:
(A) an antibody comprising a heavy chain having the amino acid sequence of SEQ ID NO: 4 as CDR1, the amino acid sequence of SEQ ID NO: 6 as CDR2, and the amino acid sequence of SEQ ID NO: 8 as CDR3;
(B) an antibody comprising a heavy chain having the amino acid sequence set forth in SEQ ID NO: 2 as the heavy chain variable region;
(C) an antibody comprising a light chain having the amino acid sequence of SEQ ID NO: 12 as CDR1, the amino acid sequence of SEQ ID NO: 14 as CDR2, and the amino acid sequence of SEQ ID NO: 16 as CDR3;
(D) an antibody comprising a light chain having the amino acid sequence set forth in SEQ ID NO: 10 as the light chain variable region,
(E) An antibody having the heavy chain pair described in (a) or (b) above and the light chain pair described in (c) or (d) above.
[3] The imaging agent according to [1] or [2], which is used in vivo.
[4] A tumor cell imaging kit comprising an anti-mesothelin antibody.
[5] The imaging kit according to [4], wherein the antibody is the antibody according to any of (a) to (e) below:
(A) an antibody comprising a heavy chain having the amino acid sequence of SEQ ID NO: 4 as CDR1, the amino acid sequence of SEQ ID NO: 6 as CDR2, and the amino acid sequence of SEQ ID NO: 8 as CDR3;
(B) an antibody comprising a heavy chain having the amino acid sequence set forth in SEQ ID NO: 2 as the heavy chain variable region;
(C) an antibody comprising a light chain having the amino acid sequence of SEQ ID NO: 12 as CDR1, the amino acid sequence of SEQ ID NO: 14 as CDR2, and the amino acid sequence of SEQ ID NO: 16 as CDR3;
(D) an antibody comprising a light chain having the amino acid sequence set forth in SEQ ID NO: 10 as the light chain variable region,
(E) An antibody having the heavy chain pair described in (a) or (b) above and the light chain pair described in (c) or (d) above.
[6] The imaging kit according to [4] or [5], which is used in vivo.
[7] A method for imaging tumor cells using an anti-mesothelin antibody.
[8] The imaging method according to [7], wherein the antibody is the antibody according to any of (a) to (e) below:
(A) an antibody comprising a heavy chain having the amino acid sequence of SEQ ID NO: 4 as CDR1, the amino acid sequence of SEQ ID NO: 6 as CDR2, and the amino acid sequence of SEQ ID NO: 8 as CDR3;
(B) an antibody comprising a heavy chain having the amino acid sequence set forth in SEQ ID NO: 2 as the heavy chain variable region;
(C) an antibody comprising a light chain having the amino acid sequence of SEQ ID NO: 12 as CDR1, the amino acid sequence of SEQ ID NO: 14 as CDR2, and the amino acid sequence of SEQ ID NO: 16 as CDR3;
(D) an antibody comprising a light chain having the amino acid sequence set forth in SEQ ID NO: 10 as the light chain variable region,
(E) An antibody having the heavy chain pair described in (a) or (b) above and the light chain pair described in (c) or (d) above.
[9] Use of an anti-mesothelin antibody in the production of an imaging agent for tumor cells.
[10] The use according to [9], wherein the antibody is the antibody according to any one of the following (a) to (e);
(A) an antibody comprising a heavy chain having the amino acid sequence of SEQ ID NO: 4 as CDR1, the amino acid sequence of SEQ ID NO: 6 as CDR2, and the amino acid sequence of SEQ ID NO: 8 as CDR3;
(B) an antibody comprising a heavy chain having the amino acid sequence set forth in SEQ ID NO: 2 as the heavy chain variable region;
(C) an antibody comprising a light chain having the amino acid sequence of SEQ ID NO: 12 as CDR1, the amino acid sequence of SEQ ID NO: 14 as CDR2, and the amino acid sequence of SEQ ID NO: 16 as CDR3;
(D) an antibody comprising a light chain having the amino acid sequence set forth in SEQ ID NO: 10 as the light chain variable region,
(E) An antibody having the heavy chain pair described in (a) or (b) above and the light chain pair described in (c) or (d) above.
[11] An anti-mesothelin antibody for use in a method for imaging tumor cells.
[12] The antibody according to [11], wherein the antibody is the antibody according to any one of the following (a) to (e);
(A) an antibody comprising a heavy chain having the amino acid sequence of SEQ ID NO: 4 as CDR1, the amino acid sequence of SEQ ID NO: 6 as CDR2, and the amino acid sequence of SEQ ID NO: 8 as CDR3;
(B) an antibody comprising a heavy chain having the amino acid sequence set forth in SEQ ID NO: 2 as the heavy chain variable region;
(C) an antibody comprising a light chain having the amino acid sequence of SEQ ID NO: 12 as CDR1, the amino acid sequence of SEQ ID NO: 14 as CDR2, and the amino acid sequence of SEQ ID NO: 16 as CDR3;
(D) an antibody comprising a light chain having the amino acid sequence set forth in SEQ ID NO: 10 as the light chain variable region,
(E) An antibody having the heavy chain pair described in (a) or (b) above and the light chain pair described in (c) or (d) above.
8-wellチャンバーで培養されたBxPC-3膵臓がん細胞の免疫染色写真である。共焦点レーザー走査顕微鏡で観察した。It is an immuno-staining photograph of BxPC-3 pancreatic cancer cells cultured in an 8-well chamber. Observed with a confocal laser scanning microscope. 2-wellチャンバーで培養されたCFPAC-1膵臓がん細胞の免疫染色写真である。共焦点レーザー走査顕微鏡で観察した。It is an immuno-staining photograph of CFPAC-1 pancreatic cancer cells cultured in a 2-well chamber. Observed with a confocal laser scanning microscope. 8-wellチャンバーで培養されたPANC-1膵臓がん細胞の免疫染色写真である。共焦点レーザー走査顕微鏡で観察した。It is an immuno-staining photograph of PANC-1 pancreatic cancer cells cultured in an 8-well chamber. Observed with a confocal laser scanning microscope. 中皮腫細胞(211H)の免疫染色写真である。共焦点レーザー走査顕微鏡で観察した。It is an immuno-staining photograph of a mesothelioma cell (211H). Observed with a confocal laser scanning microscope. PC3前立腺がん細胞、211H中皮腫細胞、H520およびH226肺がん細胞の、11-25抗メソセリン抗体での免疫染色写真である。共焦点レーザー走査顕微鏡で観察した。It is an immuno-staining photograph with 11-25 anti-mesothelin antibody of PC3 prostate cancer cells, 211H mesothelioma cells, H520 and H226 lung cancer cells. Observed with a confocal laser scanning microscope. ヌードマウス皮下に移植したがんのインビボおよびインビトロイメージング写真である。It is the in-vivo and in-vitro imaging photograph of the cancer transplanted to the nude mouse subcutaneously.
 本発明は、抗メソセリン抗体を含む、腫瘍細胞のイメージング剤を提供する。また、本発明は、抗メソセリン抗体を対象に投与する工程を含む、腫瘍細胞のイメージング方法を提供する。 The present invention provides a tumor cell imaging agent comprising an anti-mesothelin antibody. The present invention also provides a method for imaging tumor cells, comprising the step of administering an anti-mesothelin antibody to a subject.
 メソセリン(「MSLN」)は、中皮細胞と、中皮腫と、ある扁平上皮癌と、卵巣癌と、膵臓癌と、肺癌との細胞表面上で観察され(Changら、1996, Proc. Natl. Acad. Sci. USA 93: 136-140;米国特許第6,083,502号公報明細書)、前記癌に対する治療用抗体のための標的として調査中である(例えば、Morphotek社による米国臨床試験第NCT00325494号)。メソセリンは、約40kDaの分子量を有し、かつ、(i)配列番号:17の少なくとも10個の連続するアミノ酸又はスイス-プロットのアクセッション番号Q13421に列挙される既知の前記バリアントか、(ii)少なくとも20個の連続するアミノ酸残基の一部分かのいずれかを含むアミノ酸配列を有する、ポリペプチドをいい、該アミノ酸残基の一部分は、配列番号:17の20個の連続する残基か、スイス-プロットのアクセッション番号Q13421に列挙される既知の前記バリアントに少なくとも90%(好ましくは、少なくとも95%又は100%)一致する。メソセリンのバリアントは、検出される8個のアミノ酸、PQAPRRPL(配列番号:18)のバリアントと、C末端側のVQGGRGGQARAGGRAGGVEVGALSHPSLCRGPLGDALPPRTWTCSHRPGTAPSLHPGLRAPLPC(配列番号:19)が、MQEALS(配列番号:20)及び疎水性GPIリンカーモチーフ(GTPCLLGPGPVLTVLALLLASTLA)(配列番号:21)に置換されているバリアントとを含むが、これらに限られないことが知られる。前記メソセリンは、翻訳後修飾としてのグリコホスファチジルイノシトール(GPI)アンカー及び多くのN-結合型糖鎖を含む場合がある。前記GPIアンカーはメソセリンを細胞膜に結合させるが、可溶型のメソセリンを提供するためのホスホリパーゼによって開裂される場合がある。
 本願発明に包含される抗メソセリン抗体は、上記メソセリンに結合することを特徴とする。
Mesothelin (“MSLN”) is observed on the cell surface of mesothelial cells, mesothelioma, certain squamous cell carcinomas, ovarian cancers, pancreatic cancers, and lung cancers (Chang et al., 1996, Proc. Natl). Acid. Sci. USA 93: 136-140; US Pat. No. 6,083,502), under investigation as a target for therapeutic antibodies against the cancer (eg, US clinical trials by Morphotek) NCT00325494). Mesothelin has a molecular weight of about 40 kDa and (i) at least 10 consecutive amino acids of SEQ ID NO: 17 or the known variants listed in Swiss-plot accession number Q13421, or (ii) A polypeptide having an amino acid sequence comprising any of a portion of at least 20 contiguous amino acid residues, wherein the portion of the amino acid residue is the 20 contiguous residues of SEQ ID NO: 17, or the Swiss -Match at least 90% (preferably at least 95% or 100%) with the known variants listed in the accession number Q13421 of the plot. The mesothelin variant is a detected 8-amino acid variant, PQAPRRPL (SEQ ID NO: 18), and the C-terminal VQGGRGGGQARAGGRAGGVEVGALSHPSLCRGPLGDALPPRTWTCSHRRPTAPPSLHPGLRAPLPC (SEQ ID NO: 19), MQEILS (SEQ ID NO: 19) It is known that it includes, but is not limited to, a variant substituted with (GTPCLLGPGPVVLTVALLALLLASTA) (SEQ ID NO: 21). The mesothelin may contain glycophosphatidylinositol (GPI) anchors as post-translational modifications and many N-linked sugar chains. The GPI anchor binds mesothelin to the cell membrane, but may be cleaved by a phospholipase to provide a soluble form of mesothelin.
The anti-mesothelin antibody included in the present invention is characterized by binding to the mesothelin.
 本願発明に包含される抗体としては、具体的には以下の抗体が挙げられるが、これらに限定されるものではない。
(a)CDR1として配列番号:4に記載のアミノ酸配列、CDR2として配列番号:6に記載のアミノ酸配列、及びCDR3として配列番号:8に記載のアミノ酸配列を有する重鎖を含む抗体、
(b)配列番号:2に記載のアミノ酸配列を有する重鎖可変領域由来のCDR1、CDR2、CDR3を有する重鎖を含む抗体、
(c)重鎖可変領域として配列番号:2に記載のアミノ酸配列を有する重鎖を含む抗体、
(d)CDR1として配列番号:12に記載のアミノ酸配列、CDR2として配列番号:14に記載のアミノ酸配列、及びCDR3として配列番号:16に記載のアミノ酸配列を有する軽鎖を含む抗体、
(e)配列番号:10に記載のアミノ酸配列を有する軽鎖可変領域由来のCDR1、CDR2、CDR3を有する軽鎖を含む抗体、
(f)軽鎖可変領域として配列番号:10に記載のアミノ酸配列を有する軽鎖を含む抗体、
(g)上記(a)~(c)のいずれかに記載の重鎖、および、上記(d)~(f)のいずれかに記載の軽鎖の対を有する抗体。
Specific examples of antibodies included in the present invention include, but are not limited to, the following antibodies.
(A) an antibody comprising a heavy chain having the amino acid sequence of SEQ ID NO: 4 as CDR1, the amino acid sequence of SEQ ID NO: 6 as CDR2, and the amino acid sequence of SEQ ID NO: 8 as CDR3;
(B) an antibody comprising a heavy chain having CDR1, CDR2, CDR3 derived from a heavy chain variable region having the amino acid sequence of SEQ ID NO: 2;
(C) an antibody comprising a heavy chain having the amino acid sequence set forth in SEQ ID NO: 2 as the heavy chain variable region,
(D) an antibody comprising a light chain having the amino acid sequence of SEQ ID NO: 12 as CDR1, the amino acid sequence of SEQ ID NO: 14 as CDR2, and the amino acid sequence of SEQ ID NO: 16 as CDR3;
(E) an antibody comprising a light chain having CDR1, CDR2, CDR3 derived from a light chain variable region having the amino acid sequence of SEQ ID NO: 10;
(F) an antibody comprising a light chain having the amino acid sequence set forth in SEQ ID NO: 10 as the light chain variable region,
(G) An antibody having the heavy chain according to any of (a) to (c) above and the light chain pair according to any of (d) to (f) above.
 本発明の抗体には、ポリクローナル抗体およびモノクローナル抗体の両方が含まれる。モノクローナル抗体およびポリクローナル抗体の調製および精製方法は、当分野で知られており、例えば Harlow and Lane, Antibodies: A Laboratory Manual (New York: Cold Spring Harbor Laboratory Press, 1988)に記載されている。 The antibody of the present invention includes both polyclonal and monoclonal antibodies. Methods for preparing and purifying monoclonal and polyclonal antibodies are known in the art and are described, for example, in Harlow and Lane, Antibodies: A Laboratory Manual (New York: Cold Spring Harbor Laboratory Press, 1988).
 本発明の抗体には、ヒト化(humanized)抗体またはキメラ(chimeric)抗体などの組換え抗体も含まれる。「ヒト化抗体」とは、ヒトの抗体に構造を類似させた抗体のことをいう。このようなヒト化抗体またはキメラ抗体には、ヒト型キメラ抗体(例えば抗体の一部がヒト化された抗体、CH2領域がヒト化された抗体、Fc領域がヒト化された抗体、定常領域がヒト化された抗体)、及び定常領域及び可変領域に存在するCDR(相補性決定領域)以外の部分がヒト化されたヒト型CDR移植(CDR-grafted)抗体(P.T.Johons et al., Nature 321,522(1986))、完全ヒト化抗体などが含まれる。ヒト型CDR移植抗体の抗原結合活性を高めるため、マウス抗体と相同性の高いヒト抗体FRを選択する方法、相同性の高いヒト化抗体を作製する方法、ヒト抗体にマウスCDRを移植した後さらにFRのアミノ酸を置換する方法の改良技術もすでに開発され(米国特許第5585089号、米国特許第5693761号、米国特許第5693762号、米国特許第6180370号、欧州特許第451216号、欧州特許第682040号、特許第2828340号などを参照)、本発明のヒト型CDR移植抗体の作製に利用することもできる。
 ヒト型キメラ抗体は例えば、上記のH鎖可変領域の構造及び/又はL鎖可変領域の構造を有する抗体の定常領域をヒト抗体の定常領域に置換することにより作製することができる。ヒト抗体の定常領域としては公知のものを採用することができる。
The antibodies of the present invention also include recombinant antibodies such as humanized antibodies or chimeric antibodies. “Humanized antibody” refers to an antibody whose structure is similar to that of a human antibody. Such humanized antibodies or chimeric antibodies include human chimeric antibodies (for example, antibodies in which part of the antibody is humanized, antibodies in which the CH2 region is humanized, antibodies in which the Fc region is humanized, constant regions) Humanized antibodies), and human CDR-grafted antibodies (PTJohons et al., Nature 321,522 () other than the CDRs (complementarity determining regions) present in the constant and variable regions) 1986)), fully humanized antibodies and the like. In order to enhance the antigen binding activity of human CDR-grafted antibody, a method of selecting human antibody FR having high homology with mouse antibody, a method of producing humanized antibody having high homology, and further after transplanting mouse CDR to human antibody Techniques for improving the method of substituting amino acids in FR have already been developed (US Patent No. 5585089, US Patent No. 5693761, US Patent No. 5693762, US Patent No. 6180370, European Patent No. 451216, European Patent No. 682040) , Refer to Japanese Patent No. 2828340, etc.) and can also be used to prepare the human CDR-grafted antibody of the present invention.
A human chimeric antibody can be prepared, for example, by substituting the constant region of an antibody having the structure of the H chain variable region and / or the structure of the L chain variable region with a constant region of a human antibody. Known constant regions of human antibodies can be employed.
 本発明のヒト化抗体としては、例えば、CDRが抗メソセリン抗体由来のCDRであり、FRがヒトイムノグロブリン由来のFRであり、かつ定常領域がヒトイムノグロブリン由来の定常領域であることを特徴とするヒト化抗体を挙げることができる。具体的には、配列番号:2に記載のアミノ酸配列を有する重鎖可変領域由来のCDR、ヒトイムノグロブリンの重鎖可変領域由来のFR、および、ヒトイムノグロブリン由来の重鎖定常領域を有する重鎖、ならびに、配列番号:10に記載のアミノ酸配列を有する軽鎖可変領域由来のCDR、および、ヒトイムノグロブリンの軽鎖可変領域由来のFR、および、ヒトイムノグロブリン由来の軽鎖定常領域を有する軽鎖、の対を有するヒト化抗体を挙げることができる。
 また、本発明のキメラ抗体としては、例えば、可変領域が抗メソセリン抗体由来の可変領域であり、かつ定常領域がヒトイムノグロブリン由来の定常領域であることを特徴とするキメラ抗体を挙げることができる。具体的には、配列番号:2に記載のアミノ酸配列を有する重鎖可変領域、および、ヒトイムノグロブリン由来の重鎖定常領域を有する重鎖、ならびに、配列番号:10に記載のアミノ酸配列を有する軽鎖可変領域、および、ヒトイムノグロブリン由来の軽鎖定常領域を有する軽鎖、の対を有するキメラ抗体を挙げることができる。
As a humanized antibody of the present invention, for example, the CDR is a CDR derived from an anti-mesothelin antibody, the FR is a FR derived from a human immunoglobulin, and the constant region is a constant region derived from a human immunoglobulin. And humanized antibodies. Specifically, a heavy chain variable region-derived CDR having the amino acid sequence set forth in SEQ ID NO: 2, a heavy chain variable region FR derived from a human immunoglobulin, and a heavy chain having a heavy chain constant region derived from a human immunoglobulin. A chain, and a CDR derived from a light chain variable region having the amino acid sequence of SEQ ID NO: 10, and an FR derived from a light chain variable region of a human immunoglobulin, and a light chain constant region derived from a human immunoglobulin Mention may be made of humanized antibodies having a pair of light chains.
Examples of the chimeric antibody of the present invention include a chimeric antibody characterized in that the variable region is a variable region derived from an anti-mesothelin antibody and the constant region is a constant region derived from human immunoglobulin. . Specifically, it has a heavy chain variable region having the amino acid sequence set forth in SEQ ID NO: 2, a heavy chain having a heavy chain constant region derived from human immunoglobulin, and the amino acid sequence set forth in SEQ ID NO: 10. A chimeric antibody having a pair of a light chain variable region and a light chain having a light chain constant region derived from human immunoglobulin can be mentioned.
 以下に、ヒト型キメラ抗体の作製方法の一例を示す。
 まず、特定の対象抗原に対するマウス抗体を産生するハイブリドーマよりmRNAを抽出し、常法に従ってcDNAを合成する。合成したcDNAをベクターに組み込みcDNAライブラリーを構築する。このcDNAライブラリーから、H鎖遺伝子フラグメント及びL鎖遺伝子フラグメントをプローブとして用いることにより、H鎖遺伝子及びL鎖遺伝子を含有するベクターを選択する。選択されたベクターの挿入配列のシークエンシングを行うことにより、H鎖可変領域及びL鎖可変領域の遺伝子の配列が決定される。このようにして得られた配列データを基にH鎖可変領域をコードするDNAを化学合成、生化学的切断/再結合等により作製する。得られたH鎖可変領域をコードするDNAを、ヒトH鎖定常領域をコードするDNAとライゲーションして発現用ベクターに組込むことによりH鎖発現ベクターを作製する。発現ベクターとしては例えばSV40 virus basedベクター、EB virus basedベクター、BPV(パピローマウイルス)basedベクターなどを用いることができるが、これらに限定されるものではない。一方、同様の方法によりL鎖発現ベクターを作製する。これらH鎖発現ベクター及びL鎖発現ベクターにより宿主細胞を共形質転換する。宿主細胞としてはCHO細胞(チャイニーズハムスター卵巣)(A.Wright & S.L.Morrison, J.Immunol.160, 3393-3402 (1998))、SP2/0細胞(マウスミエローマ)(K.Motmans et al., Eur.J.Cancer Prev.5,512-519 (1996),R.P.Junghans et al., Cancer Res.50,1495-1502 (1990))などが好適に用いられる。また、形質転換にはリポフェクチン法(R.W.Malone et al., Proc.Natl.Acad.Sci.USA 86,6077 (1989), P.L.Felgner et al., Proc.Natl.Acad.Sci.USA 84,7413 (1987))、エレクトロポレーション法、リン酸カルシウム法(F.L.Graham & A.J.van der Eb,Virology 52,456-467(1973))、DEAE-Dextran法等が好適に用いられる。
An example of a method for producing a human chimeric antibody is shown below.
First, mRNA is extracted from a hybridoma producing a mouse antibody against a specific target antigen, and cDNA is synthesized according to a conventional method. The synthesized cDNA is incorporated into a vector to construct a cDNA library. From this cDNA library, a vector containing the H chain gene and the L chain gene is selected by using the H chain gene fragment and the L chain gene fragment as probes. By sequencing the insertion sequence of the selected vector, the sequences of the H chain variable region and L chain variable region genes are determined. Based on the sequence data thus obtained, DNA encoding the H chain variable region is prepared by chemical synthesis, biochemical cleavage / recombination, or the like. The resulting DNA encoding the H chain variable region is ligated with DNA encoding the human H chain constant region and incorporated into an expression vector to prepare an H chain expression vector. Examples of expression vectors include, but are not limited to, SV40 virus based vectors, EB virus based vectors, BPV (papilloma virus) based vectors, and the like. On the other hand, an L chain expression vector is prepared by the same method. Host cells are cotransformed with these H chain expression vector and L chain expression vector. As host cells, CHO cells (Chinese hamster ovary) (A. Wright & SL Morrison, J. Immunol. 160, 3393-3402 (1998)), SP2 / 0 cells (mouse myeloma) (K. Motmans et al., Eur. J. Cancer Prev. 5, 512-519 (1996), RP Junghans et al., Cancer Res. 50, 1495-1502 (1990)) and the like are preferably used. For transformation, the lipofectin method (RWMalone et al., Proc.Natl.Acad.Sci.USA 86,6077 (1989), PLFelgner et al., Proc.Natl.Acad.Sci.USA 84,7413 (1987) ), Electroporation method, calcium phosphate method (FL Graham & AJvan der Eb, Virology 52, 456-467 (1973)), DEAE-Dextran method and the like are preferably used.
 形質転換体を培養した後、形質転換体の細胞内又は培養液よりヒト型キメラ抗体を分離する。抗体の分離、精製には、遠心分離、硫安分画、塩析、限外濾過、アフィニティークロマトグラフィー、イオン交換クロマトグラフィー、ゲル濾過クロマトグラフィーなどの方法を適宜組み合わせて利用することができる。 After culturing the transformant, the human chimeric antibody is separated from the cells of the transformant or from the culture solution. For separation and purification of antibodies, methods such as centrifugation, ammonium sulfate fractionation, salting out, ultrafiltration, affinity chromatography, ion exchange chromatography, gel filtration chromatography and the like can be used in appropriate combination.
 一方、ヒト型CDR移植抗体は例えば以下の方法により作製することができる。まず、上記キメラ抗体の製造方法の欄で述べた方法により、特定の抗原に対する抗体のH鎖可変領域及びL鎖可変領域のアミノ酸配列及びそれをコードする塩基配列を決定する。併せて各CDR領域のアミノ酸配列及び塩基配列を決定する。 On the other hand, a human CDR-grafted antibody can be prepared, for example, by the following method. First, the amino acid sequences of the H chain variable region and the L chain variable region of an antibody against a specific antigen and the base sequence encoding the same are determined by the method described in the column of the method for producing a chimeric antibody. In addition, the amino acid sequence and base sequence of each CDR region are determined.
 次に、CDR領域を挟んで存在するFR(フレームワーク領域)を選択する。FRの選択には、およそ三つの方法が採用できる。1つめの方法は、NEWM、REIなど既に三次元構造の明らかとなったヒト抗体フレームを用いる方法である(Riechmann L. et al., Nature 332, 323-3Z7 (1988); Tempst, PR. et al., Protein Engineering 7, 1501-1507 (1994); Ellis JH. et al., J. Immunol 155, 925-937 (1995))。2つめの方法は、目的のマウス抗体可変領域と最も高いホモロジーを持つヒト抗体可変領域をデータベースより選択し、そのFRを用いる方法である(Queen C. et al., Proc Natl Acad SciUSA 86, 10029-10033 (1989); Rozak MJ. et al., J Biol Chem 271, 22611-22618 (1996); Shearman CW. et al., J.Immunol 147, 4366-4373 (1991))。3つめの方法は、ヒト抗体のFRで最も共通に用いられるアミノ酸を選択する方法である(Sato K. et al., Mol Immunol 31, 371-381 (1994); Kobinger F. et al., Protein Engineering 6, 971-980 (1993); Kettleborough CA. et al., Protein Engineering 4, 773-783 (1991))。本発明ではこれらいずれの方法を用いることもできる。 Next, select the FR (framework area) that exists across the CDR area. There are approximately three methods for selecting FR. The first method is a method using a human antibody frame, such as NEWM and REI, whose three-dimensional structure has already been clarified (Riechmann L. et al., Nature 332, 323-3Z7 (1988); Tempst, PR. Et al., Protein Engineering) 7, 1501-1507 (1994); Ellis JH. et al., J. Immunol 155, 925-937 (1995)). The second method is to select a human antibody variable region having the highest homology with the mouse antibody variable region of interest from the database and use the FR (Queen C. et al., Proc Natl Acad SciUSA 86, 10029). -10033 (1989); Rozak MJ. Et al., J Biol Chem 271, 22611-22618 (1996); Shearman CW. Et al., J.Immunol 147, 4366-4373 (1991)). The third method is to select the most commonly used amino acid in human antibody FRs (Sato K. et al., Mol Immunol 31, 371-381 (1994); Kobinger F. et al., Protein Engineering 6, 971-980 (1993); ttleKettleborough CA. et al., Protein Engineering 4, 773-783 (1991)). Any of these methods can be used in the present invention.
 尚、選択されたヒトFRのアミノ酸配列を改変したアミノ酸配列であっても、最終的に得られるヒト型CDR移植抗体が対象抗原に対する特異的結合性を有する限り、FRのアミノ酸配列として利用することができる。特に、選択されたヒトFRのアミノ酸の一部をCDRの由来となった抗体のFRのアミノ酸に変更した場合、抗体の特性が維持される可能性が高い。改変されるアミノ酸の数は好ましくはFR全体の30%以下であり、更に好ましくはFR全体の20%以下であり、更に好ましくはFR全体の10%以下である。 Even if the amino acid sequence of the selected human FR is modified, it should be used as the FR amino acid sequence as long as the finally obtained human CDR-grafted antibody has a specific binding property to the target antigen. Can do. In particular, when a part of the amino acid of the selected human FR is changed to the FR amino acid of the antibody derived from CDR, it is highly possible that the characteristics of the antibody are maintained. The number of amino acids to be modified is preferably 30% or less of the entire FR, more preferably 20% or less of the entire FR, and further preferably 10% or less of the entire FR.
 次に、これらいずれかの方法により選択したFRと上記CDRとを組み合わせることによりH鎖可変領域及びL鎖可変領域をコードするDNAを設計する。この設計を基にH鎖可変領域をコードするDNAとL鎖可変領域をコードするDNAを化学合成、生化学的切断/再結合等によりそれぞれ作製する。そしてH鎖可変領域をコードするDNAを、ヒト免疫グロブリンH鎖定常領域をコードするDNAとともに発現ベクターに組み込みH鎖発現ベクターを構築する。同様に、L鎖可変領域をコードするDNAを、ヒト免疫グロブリンL鎖定常領域をコードするDNAとともに発現ベクターに組み込みL鎖発現ベクターを構築する。発現ベクターとしては例えばSV40 virus basedベクター、EB virus basedベクター、BPV(パピローマウイルス)basedベクターなどを用いることができるが、これらに限定されるものではない。 Next, a DNA encoding the H chain variable region and the L chain variable region is designed by combining the FR selected by any of these methods and the CDR. Based on this design, DNA encoding the H chain variable region and DNA encoding the L chain variable region are respectively prepared by chemical synthesis, biochemical cleavage / recombination, and the like. Then, the DNA encoding the H chain variable region is incorporated into an expression vector together with the DNA encoding the human immunoglobulin H chain constant region to construct an H chain expression vector. Similarly, an L chain expression vector is constructed by incorporating DNA encoding the L chain variable region into an expression vector together with DNA encoding the human immunoglobulin L chain constant region. Examples of expression vectors include, but are not limited to, SV40 virus-based vectors, EB virus-based vectors, BPV (papilloma virus) based vectors, and the like.
 以上の方法で作製されたH鎖発現ベクター及びL鎖発現ベクターにより宿主細胞を共形質転換する。宿主細胞としてはCHO細胞(チャイニーズハムスター卵巣)(A.Wright& S.L.Morrison, J.Immunol.160, 3393-3402 (1998))、SP2/0細胞(マウスミエローマ)(K.Motmans et al., Eur.J.Cancer Prev.5,512-519 (1996),R.P.Junghans et al., Cancer Res.50,1495-1502 (1990))などが好適に用いられる。また、形質転換にはリポフェクチン法(R.W.Malone et al., Proc.Natl.Acad.Sci.USA 86,6077 (1989), P.L.Felgner et al., Proc.Natl.Acad.Sci.USA 84,7413 (1987))、エレクトロポレーション法、リン酸カルシウム法(F.L.Graham & A.J.van der Eb,Virology 52,456-467(1973))、DEAE-Dextran法等が好適に用いられる。 Host cells are cotransformed with the H chain expression vector and L chain expression vector prepared by the above method. As host cells, CHO cells (Chinese hamster ovary) (A. Wright & SL Morrison, J. Immunol. 160, 3393-3402 (1998)), SP2 / 0 cells (mouse myeloma) (K. Motmans et al., Eur. J. Cancer Prev. 5, 512-519 1996 (1996), RP Junghans et al., Cancer. Res. 50, 1495-1502 (1990)) and the like are preferably used. For transformation, the lipofectin method (RWMalone et al., Proc.Natl.Acad.Sci.USA 86,6077 (1989), PLFelgner et al., Proc.Natl.Acad.Sci.USA 84,7413 ( 1987)), electroporation method, calcium phosphate method (FL GrahamGra & AJvan der Eb, Virology 52,456-467 (1973)), DEAE-Dextran method and the like are preferably used.
 形質転換体を培養した後、形質転換体の細胞内又は培養液よりヒト型CDR移植抗体を分離する。抗体の分離、精製は、遠心分離、硫安分画、塩析、限外濾過、アフィニティークロマトグラフィー、イオン交換クロマトグラフィー、ゲル濾過クロマトグラフィーなどの方法を適宜組み合わせて行うことができる。 After culturing the transformant, the human CDR-grafted antibody is isolated from the cells of the transformant or the culture solution. Separation and purification of the antibody can be performed by appropriately combining methods such as centrifugation, ammonium sulfate fractionation, salting out, ultrafiltration, affinity chromatography, ion exchange chromatography, and gel filtration chromatography.
 また、ヒト抗体の取得方法も知られている。例えば、ヒトリンパ球をインビトロで所望の抗原または所望の抗原を発現する細胞で感作し、感作リンパ球をヒトミエローマ細胞、例えばU266と融合させ、抗原への結合活性を有する所望のヒト抗体を得ることもできる(特公平1-59878参照)。また、ヒト抗体遺伝子の全てのレパートリーを有するトランスジェニック動物を所望の抗原で免疫することで所望のヒト抗体を取得することができる(国際特許出願公開番号WO 93/12227, WO 92/03918,WO 94/02602, WO 94/25585,WO 96/34096, WO 96/33735参照)。 In addition, a method for obtaining a human antibody is also known. For example, human lymphocytes are sensitized with a desired antigen or cells expressing the desired antigen in vitro, the sensitized lymphocytes are fused with human myeloma cells such as U266, and the desired human antibody having binding activity to the antigen is obtained. It can also be obtained (see Japanese Patent Publication No. 1-59878). Further, a desired human antibody can be obtained by immunizing a transgenic animal having all repertoires of human antibody genes with a desired antigen (International Patent Application Publication Nos. WO 93/12227, WO 92/03918, WO 94/02602, WO 94/25585, WO 96/34096, WO 96/33735).
 更なる実施態様では、抗体又は抗体断片は、McCafferty等(Nature, 348:552-554 (1990))に記載された技術を使用して産生される抗体ファージライブラリーから分離することができる。Clackson等(Nature, 352:624-628 (1991))及び Marks等(J. Mol. Biol., 222:581-597 (1991))は、ファージライブラリーを使用したマウス及びヒト抗体の分離を記述している。続く刊行物は、鎖シャフリングによる高親和性(nM範囲)のヒト抗体の生成(Marks等, Bio/Technology, 10:779-783[1992])、並びに非常に大きなファージライブラリーを構築するための方策としてコンビナトリアル感染とインビボ組換え(Waterhouse等, Nuc. Acids. Res., 21:2265-2266[1993])を記述している。従って、これらの技術はモノクローナル抗体の分離に対する伝統的なモノクローナル抗体ハイブリドーマ法に対する実行可能な別法である。 In a further embodiment, antibodies or antibody fragments can be separated from antibody phage libraries produced using the techniques described in McCafferty et al. (Nature, 348: 552-554 (1990)). Clackson et al. (Nature, 352: 624-628 (1991)) and Marks et al. (J. Mol. Biol., 222: 581-597 (1991)) describe the separation of mouse and human antibodies using phage libraries. is doing. Subsequent publications generate high affinity (nM range) human antibodies by chain shuffling (Marks et al., Bio / Technology, 10: 779-783 [1992]), as well as to construct very large phage libraries. Combinatorial infection and in vivo recombination (Waterhouse et al., Nuc. Acids. Res., 21: 2265-2266 [1993]) have been described as measures for this. These techniques are therefore viable alternatives to traditional monoclonal antibody hybridoma methods for the separation of monoclonal antibodies.
 この点において、バクテリオファージ(ファージ)ディスプレイは、大きなオリゴペプチドライブラリーを検索して、ポリペプチド標的に特異的に結合する能力のあるこれらライブラリーのメンバーの同定を可能にするよく知られた技術の一つである。ファージディスプレイは、様々なポリペプチドがバクテリオファージ粒子の表面上のコートタンパク質に融合タンパク質として提示されることによる技術である(Scott,J.K.及びSmith G. P. (1990) Science 249:386)。ファージディスプレイの有用性は、選択的にランダム化されたタンパク質変異体(又はランダムクローンcDNA)の大きなライブラリーを標的分子に高い親和性で結合するこれらの配列について素早く効果的に分類することができる点にある。ファージでのペプチド(Cwirla,S.E.等 (1990) Proc. Natl. Acad. Sci. USA, 87:6378)又はタンパク質(Lowman,H.B.ら (1991) Biochemistry, 30:10832; Clackson,T.ら (1991) Nature, 352: 624; Marks,J.D.等 (1991), J. Mol. Biol., 222:581; Kang,A.S.等 (1991) Proc. Natl. Acad. Sci. USA, 88:8363)ライブラリーのディスプレイは、特異的に結合する特性を有するものについて無数のポリペプチド又はオリゴペプチドをスクリーニングするために使用されている(Smith, G.P. (1991) Current Opin. Biotechnol., 2:668)。ランダム突然変異体のファージライブラリーの分類は、多数の変異体を構築して増殖させる方法、標的レセプターを用いた親和性精製の方法、及び結合増強の結果を評価する手段を必要とする(米国特許第5223409号、同第5403484号、同第5571689号、及び同第5663143号を参照)。 In this regard, bacteriophage (phage) display is a well-known technique that allows large oligopeptide libraries to be searched to identify members of those libraries capable of specifically binding to a polypeptide target. one of. Phage display is a technique by which various polypeptides are presented as fusion proteins to coat proteins on the surface of bacteriophage particles (Scott, J.K. and SmithSG. P. (1990) Science 249: 386). The usefulness of phage display can quickly and effectively classify large libraries of selectively randomized protein variants (or random clone cDNAs) for those sequences that bind to target molecules with high affinity. In the point. Peptides on phage (Cwirla, SE et al. (1990) Proc. Tl Natl. Acad. Sci. USA, 87: 6378) or proteins (Lowman, HB et al (1991) Biochemistry, 30: 10832; Clackson, T. et al (1991) Nature, 352: 624; Marks, JD et al. (1991), J. Mol. Biol., 222: 581; Kang, AS et al. (1991) Proc. Natl. Acad. Sci. USA, 88: 8363) Library display Has been used to screen a myriad of polypeptides or oligopeptides for specific binding properties (Smith, GP (1991) Current Opin. Biotechnol., 2: 668). Classification of phage libraries of random mutants requires methods to construct and propagate a large number of variants, methods of affinity purification using target receptors, and means to evaluate the results of enhanced binding (US) Nos. 5223409, 5,403,484, 5,571,689, and 5,663,143).
 ほとんどのファージディスプレイ法は繊維状ファージを使用していたが、λファージディスプレイシステム(WO95/34683;米国特許第5627024号)、T4ファージディスプレイシステム(RenJ.らGene 215:439 (1998); Zhuら Cancer Researc, 58(15):3209-3214 (1998); Jiang等 Infection & Immunity, 65(11): 4770-4777 (1997); Ren等, Gene, 195(2):303-311 (1997); Ren, Protein Sci. 5:1833 (1996); Efimov等 Virus Genes 10:173(1995))及びT7ファージディスプレイシステム(Smith及びScott Methods in Enzymology,217, 228-257(1993); 米国特許第5766905号)も知られている。 Although most phage display methods used filamentous phage, the λ phage display system (WO95 / 34683; US Pat. No. 5,627,024), the T4 phage display system (RenJ. Et al. Gene 215: 439 (1998); Zhu et al. Cancer Researc, 58 (15): 3209-3214 (1998); Jiang et al.Infection & Immunity, 65 (11): 4770-4777 (1997); Ren et al., Gene, 195 (2): 303-311 (1997); Ren, Protein Sci. 5: 1833 (1996); Efimov et al. Virus Genes 10: 173 (1995)) and T7 phage display system (Smith and Scott Methods Enzymology, 217, 228-257 (1993); US Pat. No. 5,766,905 ) Is also known.
 現在、基礎的なファージディスプレイ法は多くの改良及び変形が開発されている。これらの改良により、選択された標的分子との結合性など、ペプチドライブラリーやタンパク質ライブラリーから特性、能力などに基づいてスクリーニングする方法が改善された。ファージディスプレイ法のための組み換え反応手段については、WO98/14277に記載がある。ファージディスプレイライブラリーは、二分子相互作用(WO98/20169;WO98/20159)及び拘束性へリックスペプチドの特性(WO98/20036)を分析及び制御するために使用されている。WO97/35196には、リガンドが標的分子に結合しうる第一の溶液、及び親和性リガンドが標的分子に結合しない第二の溶液とファージディスプレイライブラリーを接触させて結合リガンドを選択的に単離する、親和性リガンドの単離方法が記載されている。WO97/46251は、親和性精製抗体でランダムファージディスプレイライブラリーをバイオパニングし、次いで結合ファージを単離し、続いてマイクロプレートのウェルでマイクロパニングして高親和性結合ファージを単離する方法を記載する。黄色ブドウ球菌(Staphlylococcus aureus)タンパク質Aの親和性タグとしての使用も報告されている(Li等, (1998) Mol Biotech., 9:187)。WO97/47314は、ファージディスプレイライブラリーでもよいコンビナトリアルライブラリーを用いて酵素特異性を識別するための基質サブトラクションライブラリーの使用を記載している。ファージディスプレイに用いる洗浄剤での使用に適した酵素を選択する方法はWO97/09446に記載される。特異的に結合するタンパク質を選択する更なる方法は、米国特許第5498538号、同第5432018号、及びWO98/15833に記載されている。ペプチドライブラリーの作製及びこれらのライブラリーのスクリーニングの方法は、米国特許第5723286号、同第5432018号、同第5580717号、同第5427908号、同第5498530号、同第5770434号、同第5734018号、同第5698426号、同第5763192号、及び同第5723323号に記載される。 Currently, many improvements and variations of the basic phage display method have been developed. These improvements have improved the method of screening from peptide libraries and protein libraries based on properties, abilities, etc., such as binding to selected target molecules. Recombination reaction means for the phage display method is described in WO98 / 14277. Phage display libraries have been used to analyze and control bimolecular interactions (WO 98/20169; WO 98/20159) and restricted helix peptide properties (WO 98/20036). In WO97 / 35196, a phage display library is contacted with a first solution in which a ligand can bind to a target molecule and a second solution in which an affinity ligand does not bind to the target molecule to selectively isolate the bound ligand. A method for isolating affinity ligands is described. WO 97/46251 describes a method of biopanning a random phage display library with affinity purified antibodies, then isolating bound phage, followed by micropanning in the wells of a microplate to isolate high affinity binding phage. To do. The use of Staphlylococcus aureus protein A as an affinity tag has also been reported (Li et al., (1998) Mol Biotech., 9: 187). WO 97/47314 describes the use of a substrate subtraction library to identify enzyme specificity using a combinatorial library that may be a phage display library. A method for selecting enzymes suitable for use in detergents used for phage display is described in WO 97/09446. Additional methods for selecting proteins that specifically bind are described in US Pat. Nos. 5,498,538, 5,432,018, and WO 98/15833. Methods for preparing peptide libraries and screening these libraries are described in US Pat. Nos. 5,723,286, 5,432,018, 5,580,717, 5,427,908, 5,498,530, 5,770,434 and 5,734,018. Nos. 5,698,426, 5762192, and 5723323.
 さらに、ヒト抗体ライブラリーを用いて、パニングによりヒト抗体を取得する技術も知られている。例えば、ヒト抗体の可変領域を一本鎖抗体(scFv)としてファージディスプレイ法によりファージの表面に発現させ、抗原に結合するファージを選択することができる。選択されたファージの遺伝子を解析すれば、抗原に結合するヒト抗体の可変領域をコードするDNA配列を決定することができる。抗原に結合するscFvのDNA配列が明らかになれば、当該配列を有する適当な発現ベクターを作製し、適当な宿主に導入して発現させることによりヒト抗体を取得することができる。これらの方法は既に周知であり、WO 92/01047、WO 92/20791、WO 93/06213、WO 93/11236、WO 93/19172、WO 95/01438、WO 95/15388を参考に実施することができる。 Furthermore, a technique for obtaining a human antibody by panning using a human antibody library is also known. For example, the variable region of a human antibody is expressed as a single chain antibody (scFv) on the surface of the phage by the phage display method, and a phage that binds to the antigen can be selected. By analyzing the gene of the selected phage, the DNA sequence encoding the variable region of the human antibody that binds to the antigen can be determined. If the DNA sequence of scFv that binds to the antigen is clarified, a human antibody can be obtained by preparing an appropriate expression vector having the sequence and introducing it into an appropriate host for expression. These methods are already well known and can be carried out with reference to WO 92/01047, WO 92/20791, WO 93/06213, WO 93/11236, WO 93/19172, WO 95/01438, and WO 95/15388. it can.
 別法として、ファージディスプレイ技術(McCafferty等, Nature 348:552-553[1990])を使用して、非免疫化ドナーの免疫グロブリン可変(V)ドメイン遺伝子レパートリーから、インビトロでヒト抗体及び抗体断片を産出させることができる。この技術によれば、抗体Vドメイン遺伝子を、フレーム単位で、繊維状バクテリオファージ、例えばM13又はfdのコートタンパク質遺伝子のどちらかでクローニングし、ファージ粒子の表面で機能的抗体断片として提示させる。繊維状粒子がファージゲノムの一本鎖DNAコピーを含むので、抗体の機能特性に基づいた選択に基づいても、結果としてこれらの特性を示す抗体をコードする遺伝子の選択が成される。よって、このファージはB細胞のいくつかの特性を模倣している。ファージディスプレイは多様な形式で行うことができる;例えばJohnson, Kevin S. 及びChiswell, David J., Current Opinion in Structural Biology 3:564-571(1993)を参照。V-遺伝子セグメントのいくつかの供給源を、ファージディスプレイのために使用できる。Clackson等, Nature, 352:624-628(1991)は、免疫化したマウス脾臓由来のV遺伝子の小さいランダムなコンビナトリアルライブラリーから、多様で多くの抗-オキサゾロン抗体を単離した。非免疫化ヒトドナーのV遺伝子のレパートリーが構成可能であり、多様で多くの抗原(自己抗原を含む)に対する抗体は、Marks等, J. Mol. Biol. 222:581-597(1991)、又はGriffith等, EMBO J. 12:725-734(1993)に記載の技術にそのまま従うことで単離することができる。また、米国特許第5565332号及び同5573905号を参照のこと。 Alternatively, human antibodies and antibody fragments can be generated in vitro from the immunoglobulin variable (V) domain gene repertoire of non-immunized donors using phage display technology (McCafferty et al., Nature 348: 552-553 [1990]). Can be produced. According to this technique, antibody V domain genes are cloned in frame units with either filamentous bacteriophage, eg, M13 or fd coat protein genes, and displayed as functional antibody fragments on the surface of phage particles. Since the filamentous particle contains a single-stranded DNA copy of the phage genome, the selection of a gene encoding an antibody exhibiting these properties results as a result, even based on selection based on the functional properties of the antibody. Thus, this phage mimics some properties of B cells. Phage display can be performed in a variety of formats; see, for example, Johnson, Kevin S. and Chiswell, David J., Current Opinion in Structural Biology 3: 564-571 (1993). Several sources of V-gene segments can be used for phage display. Clackson et al., Nature, 352: 624-628 (1991) isolated a large number of diverse anti-oxazolone antibodies from a small random combinatorial library of V genes derived from the spleens of immunized mice. The V gene repertoire of non-immunized human donors is configurable, and antibodies against a wide variety of antigens (including self-antigens) can be found in Marks et al., J. Mol. Biol. 222: 581-597 (1991), or Griffith. Etc., EMBO J. 12: 725-734 (1993). See also U.S. Pat. Nos. 5,565,332 and 5,573,905.
 本発明の抗体には、Fab、Fab'、F(ab')2、Fv、scFv、dsFv、Diabody、sc(Fv)2、及びminibody等の、抗体の機能的断片も含まれる。また、これら機能的断片の多量体(例えば、ダイマー、トリマー、テトラマー、ポリマー)も、本発明の抗体に含まれる。
 Fabは、IgGをシステイン存在下パパイン消化することにより得られる、L鎖とH鎖可変領域、並びにCH1ドメイン及びヒンジ部の一部からなるH鎖フラグメントとから構成される分子量約5万の断片である。本発明では、上記抗体をパパイン消化することにより得ることができる。また、上記抗体のH鎖の一部及びL鎖をコードするDNAを適当なベクターに組み込み、当該ベクターを用いて形質転換した形質転換体よりFabを調製することもできる。
 Fab'は、後述のF(ab')2のH鎖間のジスルフィド結合を切断することにより得られる分子量が約5万の断片である。本発明では、上記抗体をペプシン消化し、還元剤を用いてジスルフィド結合を切断することにより得られる。また、Fab同様に、Fab'をコードするDNAを用いて遺伝子工学的に調製することもできる。
 F(ab')2は、IgGをペプシン消化することにより得られる、L鎖とH鎖可変領域、並びにCH1ドメイン及びヒンジ部の一部からなるH鎖フラグメントとから構成される断片(Fab')がジスルフィド結合で結合した分子量約10万の断片である。本発明では、上記抗体をペプシン消化することにより得られる。また、Fab同様に、F(ab')2をコードするDNAを用いて遺伝子工学的に調製することもできる。
 Fvは、抗体を酵素、例えばパパイン、ペプシンで処理し抗体断片を生成させるか、または、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できる(例えば、Co, M.S. et al., J. Immunol.(1994)152, 2968-2976、Better, M. &; Horwitz, A. H. Methods in Enzymology(1989)178, 476-496、Plueckthun, A. ; Skerra, A. Methods in Enzymology(1989)178, 476-496、Lamoyi, E., Methods in Enzymology(1989)121, 652-663、Rousseaux, J. et al., Methods in Enzymology(1989)121, 663-669、Bird, R. E. et al., TIBTECH(1991)9, 132-137参照)。
 scFvは、H鎖可変領域とL鎖可変領域とからなるFvを片方の鎖のC末端と他方のN末端とを適当なペプチドリンカーで連結し、一本鎖化した抗体断片である。ペプチドリンカーとしては例えば柔軟性の高い(GGGGS)3などを用いることができる。例えば、上記抗体のH鎖可変領域及びL鎖可変領域をコードするDNAとペプチドリンカーをコードするDNAを用いてscFv抗体をコードするDNAを構築し、これを適当なベクターに組み込み、当該ベクターを用いて形質転換した形質転換体よりscFvを調製することができる。
 dsFvは、H鎖可変領域及びL鎖可変領域の適切な位置にCys残基を導入し、H鎖可変領域とL鎖可変領域とをジスルフィド結合により安定化させたFv断片である。各鎖におけるCys残基の導入位置は分子モデリングにより予測される立体構造に基づき決定することができる。本発明では例えば上記抗体のH鎖可変領域及びL鎖可変領域のアミノ酸配列から立体構造を予測し、かかる予測に基づき変異を導入したH鎖可変領域及びL鎖可変領域をそれぞれコードするDNAを構築し、これを適当なベクターに組み込み、そして当該ベクターを用いて形質転換した形質転換体よりdsFvを調製することができる。
 尚、適当なリンカーを用いてscFv抗体、dsFv抗体などを連結させたり、ストレプトアビジンを融合させたりして抗体断片を多量体化することもできる。本発明の抗体(抗体断片を含む)に低分子化合物、タンパク質、標識物質などを融合又は結合させることにより、融合抗体又は標識化抗体を構成することができる。標識物質としては125I等の放射性物質、などを用いることができる。
The antibody of the present invention includes functional fragments of antibodies such as Fab, Fab ′, F (ab ′) 2 , Fv, scFv, dsFv, Diabody, sc (Fv) 2, and minibody. In addition, multimers (for example, dimers, trimers, tetramers, polymers) of these functional fragments are also included in the antibody of the present invention.
Fab has a molecular weight of about 50,000 composed of L chain and H chain variable region obtained by digesting IgG with papain in the presence of cysteine, and H chain fragment consisting of C H 1 domain and part of hinge part. It is a fragment. In the present invention, the antibody can be obtained by digesting with papain. Alternatively, Fab can be prepared from a transformant obtained by incorporating a DNA encoding part of the H chain and L chain of the above antibody into an appropriate vector and transforming using the vector.
Fab ′ is a fragment having a molecular weight of about 50,000 obtained by cleaving a disulfide bond between the H chains of F (ab ′) 2 described later. In the present invention, the antibody is obtained by digesting with pepsin and cleaving disulfide bonds using a reducing agent. Similarly to Fab, it can also be prepared by genetic engineering using DNA encoding Fab ′.
F (ab ′) 2 is a fragment (Fab) composed of an L chain and an H chain variable region obtained by digesting IgG with pepsin, and an H chain fragment consisting of a C H 1 domain and a part of the hinge region. ') Is a fragment having a molecular weight of about 100,000 bonded by a disulfide bond. In the present invention, the antibody is obtained by pepsin digestion. Similarly to Fab, it can be prepared by genetic engineering using DNA encoding F (ab ′) 2 .
Fv can be expressed in an appropriate host cell after the antibody is treated with an enzyme such as papain or pepsin to generate antibody fragments, or a gene encoding these antibody fragments is constructed and introduced into an expression vector. (For example, Co, MS et al., J. Immunol. (1994) 152, 2968-2976, Better, M. &; Horwitz, AH Methods in Enzymology (1989) 178, 476-496, Plueckthun, A .; Skerra , A. Methods in Enzymology (1989) 178, 476-496, Lamoyi, E., Methods in Enzymology (1989) 121, 652-663, Rousseaux, J. et al., Methods in Enzymology (1989) 121, 663- 669, Bird, RE et al., TIBTECH (1991) 9, 132-137).
scFv is an antibody fragment in which an Fv comprising an H chain variable region and an L chain variable region is made into a single chain by linking the C terminus of one chain and the other N terminus with an appropriate peptide linker. As the peptide linker, for example, (GGGGS) 3 having high flexibility can be used. For example, a DNA encoding an scFv antibody is constructed using DNA encoding the H chain variable region and L chain variable region of the above antibody and DNA encoding a peptide linker, and this is incorporated into an appropriate vector, and the vector is used. ScFv can be prepared from the transformant transformed as described above.
dsFv is an Fv fragment in which a Cys residue is introduced at an appropriate position in the H chain variable region and the L chain variable region, and the H chain variable region and the L chain variable region are stabilized by disulfide bonds. The position of Cys residue introduction in each chain can be determined based on the three-dimensional structure predicted by molecular modeling. In the present invention, for example, a three-dimensional structure is predicted from the amino acid sequences of the H chain variable region and the L chain variable region of the above-described antibody, and DNAs encoding the H chain variable region and L chain variable region into which mutations are introduced are constructed based on such prediction Then, this is incorporated into an appropriate vector, and dsFv can be prepared from a transformant transformed with the vector.
It is also possible to multimerize antibody fragments by linking scFv antibody, dsFv antibody or the like using an appropriate linker, or by fusing streptavidin. A fusion antibody or a labeled antibody can be constructed by fusing or binding a low molecular compound, protein, labeling substance or the like to the antibody (including antibody fragment) of the present invention. As the labeling substance, a radioactive substance such as 125 I can be used.
 Diabodyは、遺伝子融合により構築された二価(bivalent)の抗体断片を指す(Holliger P et al., Proc.Natl.Acad.Sci.USA 90: 6444-6448 (1993)、EP404,097号、WO93/11161号等)。Diabodyは、2本のポリペプチド鎖から構成されるダイマーであり、通常、ポリペプチド鎖は各々、同じ鎖中でVL及びVHが、互いに結合できない位に短い、例えば、5残基程度のリンカーにより結合されている。同一ポリペプチド鎖上にコードされるVLとVHとは、その間のリンカーが短いため単鎖可変領域フラグメントを形成することが出来ず二量体を形成するため、Diabodyは2つの抗原結合部位を有することとなる。Diabodyは、抗体を酵素、例えば、パパイン、ペプシンなどで処理し、抗体断片を生成させるか、又はこれら抗体断片をコードするDNAを構築し、これを発現ベクターに導入した後、適当な宿主細胞で発現させればよい(例えば、Co, M. S. et al., J. Immunol. (1994) 152, 2968-2976; Better, M. and Horwitz, A. H., Methods Enzymol. (1989) 178, 476-496; Pluckthun, A. and Skerra, A., Methods Enzymol. (1989) 178, 497-515; Lamoyi, E., Methods Enzymol. (1986) 121, 652-663 ; Rousseaux, J. et al., Methods Enzymol. (1986) 121, 663-669; Bird, R. E. and Walker, B. W., Trends Biotechnol. (1991) 9, 132-137参照)。
 sc(Fv)2は、2つのVH及び2つのVLをリンカー等で結合して一本鎖にした低分子化抗体である(Hudson et al、J Immunol. Methods 1999;231:177-189)。sc(Fv)2は、例えば、scFvをリンカーで結ぶことによって作製できる。
 minibody(低分子化抗体)とは、全長抗体(whole antibody、例えばwhole IgG等)を親抗体とし、全長抗体の一部分が欠損している抗体断片を含み、抗原への結合能を有していれば特に限定されない。
Diabody refers to a bivalent antibody fragment constructed by gene fusion (Holliger P et al., Proc. Natl. Acad. Sci. USA 90: 6444-6448 (1993), EP 404,097, WO 93 / 11161 etc.). Diabody is a dimer composed of two polypeptide chains. Normally, each polypeptide chain is short to a position where VL and VH cannot bind to each other in the same chain, for example, by a linker of about 5 residues. Are combined. Diabody has two antigen-binding sites because VL and VH encoded on the same polypeptide chain cannot form a single-chain variable region fragment due to the short linker between them, and form a dimer. It will be. Diabody treats an antibody with an enzyme such as papain or pepsin to generate antibody fragments or constructs DNA encoding these antibody fragments and introduces them into an expression vector, and then in an appropriate host cell. (Eg, Co, MS et al., J. Immunol. (1994) 152, 2968-2976; Better, M. and Horwitz, AH, Methods Enzymol. (1989) 178, 476-496; Pluckthun , A. and Skerra, A., Methods Enzymol. (1989) 178, 497-515; Lamoyi, E., Methods Enzymol. (1986) 121, 652-663; Rousseaux, J. et al., Methods Enzymol. 1986) 121, 663-669; Bird, RE and Walker, BW, Trends Biotechnol. (1991) 9, 132-137).
sc (Fv) 2 is a low molecular weight antibody in which two VHs and two VLs are combined with a linker or the like to form a single chain (Hudson et al, J Immunol. Methods 1999; 231: 177-189). sc (Fv) 2 can be prepared, for example, by linking scFv with a linker.
A minibody (low molecular weight antibody) is a full-length antibody (whole antibody, such as whole IgG), which is a parent antibody and includes an antibody fragment in which a part of the full-length antibody is missing, and has a binding ability to an antigen. If it does not specifically limit.
 本発明の抗体には、本発明の抗体と他のペプチド又はタンパク質とが融合した融合タンパク質も含まれる。融合タンパク質を作製する方法は、本発明の抗体をコードするポリヌクレオチドと他のペプチド又はポリペプチドをコードするポリヌクレオチドをフレームが一致するように連結してこれを発現ベクターに導入し、宿主で発現させればよく、当業者に公知の手法を用いることができる。本発明の抗体との融合に付される他のペプチド又はポリペプチドとしては、例えば、FLAG(Hopp, T. P. et al., BioTechnology (1988) 6, 1204-1210 )、6個のHis(ヒスチジン)残基からなる6×His、10×His、インフルエンザ凝集素(HA)、ヒトc-mycの断片、VSV-GPの断片、p18HIVの断片、T7-tag、HSV-tag、E-tag、SV40T抗原の断片、lck tag、α-tubulinの断片、B-tag、Protein C の断片等の公知のペプチドを使用することができる。また、本発明の抗体との融合に付される他のポリペプチドとしては、例えば、GST(グルタチオン-S-トランスフェラーゼ)、HA(インフルエンザ凝集素)、β-ガラクトシダーゼ、MBP(マルトース結合タンパク質)等が挙げられる。 The antibody of the present invention includes a fusion protein in which the antibody of the present invention is fused with another peptide or protein. In the method for producing a fusion protein, a polynucleotide encoding an antibody of the present invention and a polynucleotide encoding another peptide or polypeptide are linked so that the frames coincide with each other, introduced into an expression vector, and expressed in a host. Any technique known to those skilled in the art can be used. Other peptides or polypeptides to be subjected to fusion with the antibody of the present invention include, for example, FLAG (Hopp, T. P. et al., BioTechnology (1988) 6, 1204-1210), 6 His ( (Histidine) 6xHis, 10xHis, influenza agglutinin (HA), human c-myc fragment, VSV-GP fragment, p18HIV fragment, T7-tag, HSV-tag, E-tag, Known peptides such as the SV40T antigen fragment, lck tag, α-tubulin fragment, B-tag, Protein C fragment and the like can be used. Further, examples of other polypeptides subjected to fusion with the antibody of the present invention include GST (glutathione-S-transferase), HA (influenza agglutinin), β-galactosidase, MBP (maltose binding protein) and the like. Can be mentioned.
 本発明の抗体には、標識物質が結合した抗体も含まれる。
 標識物質としては、酵素発光(ルシフェラーゼ)による発光、発光低分子を利用するものと、蛍光タンパクや蛍光低分子)を用いるもの、放射性核種等が挙げられるがこれらには限定されない。放射性核種としては51Cr、59Fe、57Co、67Ga、75Se、81mKr、99mTc、111In、125I、131I、133Xe、201Tlなどのγ線放出核種や、11C、13N、15O、18F、35mCl、76Br、45Ti、48V、60Cu、61Cu、62Cu、66Ga、89Zr、94mTc、124Iなどの陽電子放出核種などが挙げられるがこれらには限定されない。なお当業者には自明であるが"m"とは核異性体を示す。
 蛍光標識や発光標識としては酵素発光(ルシフェラーゼ)による発光を利用するものと、蛍光(GFP、DsRed、クサビラオレンジ等の蛍光タンパク質やFITCやCy5.5、Alexa Fluor 750などの蛍光性低分子)を用いるものとのが使用できる。
 酵素発光(ルシフェラーゼ)による発光の場合は基質投与が別途必要である。
 特に、動物本来の自家蛍光による影響を軽減したものが好ましく、さらには皮膚透過性の高いシグナルを発する標識が好ましい。
The antibody of the present invention includes an antibody bound with a labeling substance.
Examples of the labeling substance include, but are not limited to, luminescence by enzyme luminescence (luciferase), a substance using a luminescent low molecule, a substance using a fluorescent protein or a fluorescent small molecule, and a radionuclide. Radionuclides include gamma ray emitting nuclides such as 51 Cr, 59 Fe, 57 Co, 67 Ga, 75 Se, 81 m Kr, 99 m Tc, 111 In, 125 I, 131 I, 133 Xe, 201 Tl, 11 C, Positron emitting nuclides such as 13 N, 15 O, 18 F, 35 m Cl, 76 Br, 45 Ti, 48 V, 60 Cu, 61 Cu, 62 Cu, 66 Ga, 89 Zr, 94 m Tc, 124 I However, it is not limited to these. As is obvious to those skilled in the art, “m” represents a nuclear isomer.
As fluorescent labels and luminescent labels, those using light emission by enzyme luminescence (luciferase) and fluorescence (fluorescent proteins such as GFP, DsRed, and wedge orange, and small fluorescent molecules such as FITC, Cy5.5, Alexa Fluor 750) Can be used.
In the case of luminescence by enzyme luminescence (luciferase), administration of a substrate is required separately.
In particular, those that reduce the influence of the animal's natural autofluorescence are preferred, and labels that emit signals with high skin permeability are preferred.
 また本発明は、本発明の抗体をコードするDNA、該DNAが挿入されたベクター、及び該ベクターが導入された形質転換細胞を提供する。ベクターの例としては、M13系ベクター、pUC系ベクター、pBR322、pBluescript、pCR-Scriptなどが挙げられる。また、cDNAのサブクローニング、切り出しを目的とした場合、上記ベクターの他に、例えば、pGEM-T、pDIRECT、pT7などが挙げられる。本発明の抗体をコードするDNA、該DNAが挿入されたベクター、及び該ベクターが導入された形質転換細胞は公知の技術を用いて作成される。 The present invention also provides DNA encoding the antibody of the present invention, a vector into which the DNA has been inserted, and a transformed cell into which the vector has been introduced. Examples of vectors include M13 vectors, pUC vectors, pBR322, pBluescript, pCR-Script, and the like. In addition, for the purpose of subcloning and excision of cDNA, in addition to the above vector, for example, pGEM-T, pDIRECT, pT7 and the like can be mentioned. A DNA encoding the antibody of the present invention, a vector into which the DNA has been inserted, and a transformed cell into which the vector has been introduced are prepared using known techniques.
 本発明の抗メソセリン抗体をコードするDNAとしては、以下のDNAが挙げられる。
(a)CDR1として配列番号:4に記載のアミノ酸配列、CDR2として配列番号:6に記載のアミノ酸配列、及びCDR3として配列番号:8に記載のアミノ酸配列を有する重鎖をコードするDNA、
(b)重鎖可変領域として配列番号:2に記載のアミノ酸配列を有する重鎖をコードするDNA、
(c)CDR1として配列番号:12に記載のアミノ酸配列、CDR2として配列番号:14に記載のアミノ酸配列、及びCDR3として配列番号:16に記載のアミノ酸配列を有する軽鎖をコードするDNA、
(d)軽鎖可変領域として配列番号:10に記載のアミノ酸配列を有する軽鎖をコードするDNA。
Examples of the DNA encoding the anti-mesothelin antibody of the present invention include the following DNAs.
(A) DNA encoding the heavy chain having the amino acid sequence described in SEQ ID NO: 4 as CDR1, the amino acid sequence described in SEQ ID NO: 6 as CDR2, and the amino acid sequence described in SEQ ID NO: 8 as CDR3;
(B) DNA encoding a heavy chain having the amino acid sequence set forth in SEQ ID NO: 2 as the heavy chain variable region,
(C) DNA encoding a light chain having the amino acid sequence set forth in SEQ ID NO: 12 as CDR1, the amino acid sequence set forth in SEQ ID NO: 14 as CDR2, and the amino acid sequence set forth in SEQ ID NO: 16 as CDR3;
(D) DNA encoding a light chain having the amino acid sequence set forth in SEQ ID NO: 10 as the light chain variable region.
 発現ベクターとしては、例えば、大腸菌での発現を目的とした場合は、ベクターが大腸菌で増幅されるような上記特徴を持つほかに、宿主をJM109、DH5α、HB101、XL1-Blueなどの大腸菌とした場合においては、大腸菌で効率よく発現できるようなプロモーター、例えば、lacZプロモーター(Wardら, Nature (1989) 341, 544-546;FASEB J. (1992) 6, 2422-2427)、araBプロモーター(Betterら, Science (1988) 240, 1041-1043 )、またはT7プロモーターなどを持っていることが不可欠である。このようなベクターとしては、上記ベクターの他にpGEX-5X-1(ファルマシア社製)、「QIAexpress system」(キアゲン社製)、pEGFP、またはpET(この場合、宿主はT7 RNAポリメラーゼを発現しているBL21が好ましい)などが挙げられる。 As an expression vector, for example, for the purpose of expression in E. coli, in addition to the above characteristics that the vector is amplified in E. coli, the host is E. coli such as JM109, DH5α, HB101, XL1-Blue, etc. In some cases, promoters that can be efficiently expressed in E. coli, such as the lacZ promoter (Ward et al., Nature (1989) 341, 544-546; FASEB J. (1992) 6, 2422-2427), araB promoter (Better et al. , (Science (1988) (240), (1041-1043)), or having a T7 promoter or the like. In addition to the above vectors, such vectors include pGEX-5X-1 (Pharmacia), “QIAexpress® system” (Qiagen), pEGFP, or pET (in this case, the host expresses T7 RNA polymerase). BL21 is preferred).
 また、ベクターには、ポリペプチド分泌のためのシグナル配列が含まれていてもよい。蛋白質分泌のためのシグナル配列としては、大腸菌のペリプラズムに産生させる場合、pelBシグナル配列(Lei, S. P. et al J. Bacteriol. (1987) 169, 4379)を使用すればよい。宿主細胞へのベクターの導入は、例えば塩化カルシウム法、エレクトロポレーション法を用いて行うことができる。 The vector may also contain a signal sequence for polypeptide secretion. As a signal sequence for protein secretion, the pelB signal sequence (Lei, S. P. et al J. Bacteriol. (1987) 169, 4379) may be used when the periplasm of E. coli is used for production. Introduction of a vector into a host cell can be performed using, for example, a calcium chloride method or an electroporation method.
 大腸菌以外にも、例えば、哺乳動物由来の発現ベクター(例えば、pcDNA3 (インビトロゲン社製)や、pEGF-BOS (Nucleic Acids. Res.1990, 18(17),p5322)、pEF 、pCDM8)、昆虫細胞由来の発現ベクター(例えば「Bac-to-BAC baculovairus expression system」(ギブコBRL社製)、pBacPAK8)、植物由来の発現ベクター(例えばpMH1、pMH2)、動物ウィルス由来の発現ベクター(例えば、pHSV、pMV、pAdexLcw)、レトロウィルス由来の発現ベクター(例えば、pZIPneo)、酵母由来の発現ベクター(例えば、「Pichia Expression Kit」(インビトロゲン社製)、pNV11、SP-Q01)、枯草菌由来の発現ベクター(例えば、pPL608、pKTH50)が挙げられる。 In addition to E. coli, for example, mammalian-derived expression vectors (for example, pcDNA3 (manufactured by Invitrogen), pEGF-BOS (Nucleic Acids. Res.1990, 18 (17), p5322), pEF, pCDM8), insects Expression vectors derived from cells (eg, “Bac-to-BAC baculovairus expression system” (Gibco BRL), pBacPAK8), plant-derived expression vectors (eg, pMH1, pMH2), animal virus-derived expression vectors (eg, pHSV, pMV, pAdexLcw), retrovirus-derived expression vectors (for example, pZIPneo), yeast-derived expression vectors (for example, “Pichia Expression Kit” (manufactured by Invitrogen), pNV11, SP-Q01), Bacillus subtilis-derived expression vectors (For example, pPL608, pKTH50).
 CHO細胞、COS細胞、NIH3T3細胞等の動物細胞での発現を目的とした場合には、細胞内で発現させるために必要なプロモーター、例えばSV40プロモーター(Mulliganら, Nature (1979) 277, 108)、MMTV-LTRプロモーター、EF1αプロモーター(Mizushimaら, Nucleic Acids Res. (1990) 18, 5322)、CMVプロモーターなどを持っていることが不可欠であり、細胞への形質転換を選抜するための遺伝子(例えば、薬剤(ネオマイシン、G418など)により判別できるような薬剤耐性遺伝子)を有すればさらに好ましい。このような特性を有するベクターとしては、例えば、pMAM、pDR2、pBK-RSV、pBK-CMV、pOPRSV、pOP13などが挙げられる。 For the purpose of expression in animal cells such as CHO cells, COS cells, NIH3T3 cells, etc., promoters necessary for expression in cells, such as the SV40 promoter (Mulligan et al., Nature (1979) 277, 108), It is essential to have MMTV-LTR promoter, EF1α promoter (Mizushima et al., Nucleic Acids Res. (1990) 18, 5322), CMV promoter, etc., and genes for selecting transformation into cells (for example, More preferably, it has a drug resistance gene that can be discriminated by a drug (neomycin, G418, etc.). Examples of such a vector include pMAM, pDR2, pBK-RSV, pBK-CMV, pOPRSV, and pOP13.
 さらに、遺伝子を安定的に発現させ、かつ、細胞内での遺伝子のコピー数の増幅を目的とする場合には、核酸合成経路を欠損したCHO細胞にそれを相補するDHFR遺伝子を有するベクター(例えば、pCHOIなど)を導入し、メトトレキセート(MTX)により増幅させる方法が挙げられ、また、遺伝子の一過性の発現を目的とする場合には、SV40 T抗原を発現する遺伝子を染色体上に持つCOS細胞を用いてSV40の複製起点を持つベクター(pcDなど)で形質転換する方法が挙げられる。複製開始点としては、また、ポリオーマウィルス、アデノウィルス、ウシパピローマウィルス(BPV)等の由来のものを用いることもできる。さらに、宿主細胞系で遺伝子コピー数増幅のため、発現ベクターは選択マーカーとして、アミノグリコシドトランスフェラーゼ(APH)遺伝子、チミジンキナーゼ(TK)遺伝子、大腸菌キサンチングアニンホスホリボシルトランスフェラーゼ(Ecogpt)遺伝子、ジヒドロ葉酸還元酵素(dhfr)遺伝子等を含むことができる。 Furthermore, when the gene is stably expressed and the purpose is to amplify the copy number of the gene in the cell, a vector having a DHFR gene complementary to the CHO cell lacking the nucleic acid synthesis pathway (for example, , PCHOI, etc.), and amplifying with methotrexate (MTX). For the purpose of transient expression of the gene, COS with a gene expressing SV40 T antigen on the chromosome An example is a method of transforming with a vector (such as pcD) having an SV40 replication origin using cells. As the replication origin, those derived from polyoma virus, adenovirus, bovine papilloma virus (BPV) and the like can also be used. Furthermore, for gene copy number amplification in host cell systems, the expression vectors are selectable markers: aminoglycoside transferase (APH) gene, thymidine kinase (TK) gene, E. coli xanthine guanine phosphoribosyltransferase (Ecogpt) gene, dihydrofolate reductase ( dhfr) gene and the like.
 ベクターが導入される宿主細胞としては特に制限はなく、例えば、大腸菌や種々の動物細胞などを用いることが可能である。宿主細胞は、例えば、本発明の抗体の製造や発現のための産生系として使用することができる。ポリペプチド製造のための産生系は、インビトロおよびインビボの産生系がある。インビトロの産生系としては、真核細胞を使用する産生系や原核細胞を使用する産生系が挙げられる。 The host cell into which the vector is introduced is not particularly limited, and for example, E. coli or various animal cells can be used. The host cell can be used, for example, as a production system for production and expression of the antibody of the present invention. Production systems for polypeptide production include in vitro and in vivo production systems. Examples of in vitro production systems include production systems using eukaryotic cells and production systems using prokaryotic cells.
 真核細胞を使用する場合、例えば、動物細胞、植物細胞、真菌細胞を宿主に用いることができる。動物細胞としては、哺乳類細胞、例えば、CHO(J. Exp. Med. (1995) 108, 945)、COS、3T3、ミエローマ、BHK(baby hamster kidney)、HeLa、Vero、両生類細胞、例えばアフリカツメガエル卵母細胞(Valle, et al., Nature (1981) 291, 358-340)、あるいは昆虫細胞、例えば、 Sf9、 Sf21、 Tn5が知られている。本発明においては、CHO-DG44、CHO-DXB11、COS7細胞、BHK細胞が好適に用いられる。動物細胞において、大量発現を目的とする場合には特にCHO細胞が好ましい。宿主細胞へのベクターの導入は、例えば、リン酸カルシウム法、DEAEデキストラン法、カチオニックリポソームDOTAP(ベーリンガーマンハイム社製)を用いた方法、エレクトロポレーション法、リポフェクションなどの方法で行うことが可能である。 When eukaryotic cells are used, for example, animal cells, plant cells, and fungal cells can be used as the host. Animal cells include mammalian cells such as CHO (J. Exp. Med. (1995) 108, 945), COS, 3T3, myeloma, BHK (baby hamster kidney), HeLa, Vero, amphibian cells such as Xenopus eggs Mother cells (Valle, et al., Nature (1981) 291, 358-340) or insect cells such as Sf9, Sf21, and Tn5 are known. In the present invention, CHO-DG44, CHO-DXB11, COS7 cells, and BHK cells are preferably used. In animal cells, CHO cells are particularly preferred for mass expression purposes. Introduction of a vector into a host cell can be performed by, for example, a calcium phosphate method, a DEAE dextran method, a method using a cationic liposome DOTAP (Boehringer Mannheim), an electroporation method, a lipofection method, or the like.
 植物細胞としては、例えば、ニコチアナ・タバカム(Nicotiana tabacum)由来の細胞が蛋白質生産系として知られており、これをカルス培養すればよい。真菌細胞としては、酵母、例えば、サッカロミセス(Saccharomyces)属、例えば、サッカロミセス・セレビシエ(Saccharomyces cerevisiae)、サッカロミセス・ポンベ(Saccharomyces pombe)、糸状菌、例えば、アスペルギルス(Aspergillus)属、例えば、アスペルギルス・ニガー(Aspergillus niger)が知られている。 As plant cells, for example, cells derived from Nicotiana tabacum are known as protein production systems, and these may be cultured in callus. Fungal cells include yeasts such as the genus Saccharomyces, such as Saccharomyces cerevisiae, Saccharomyces pombe, filamentous fungi such as the genus Aspergillus, such as Aspergillus. Aspergillus niger) is known.
 原核細胞を使用する場合、細菌細胞を用いる産生系がある。細菌細胞としては、大腸菌(E. coli)、例えば、JM109、DH5α、HB101等が挙げられ、その他、枯草菌が知られている。本発明のDNAにより形質転換された細胞をインビトロで培養し、当業者が通常行う方法によって精製することによって、本発明の抗体を得ることが可能である。 When using prokaryotic cells, there is a production system using bacterial cells. Bacterial cells include E. coli (eg, JM109, DH5α, HB101, etc.), and Bacillus subtilis is also known. The antibody of the present invention can be obtained by culturing cells transformed with the DNA of the present invention in vitro and purifying the cells by a method commonly used by those skilled in the art.
 また本発明は、本発明の抗体をコードする核酸を含むベクターを有する宿主生物を提供する。本発明の宿主生物は、組換え型抗体の産生に有用である。本発明における宿主生物としては、ヤギなどが挙げられる。例えば、本発明のトランスジェニックヤギの作成は次のようにして行うことが可能である。即ち、抗体遺伝子が乳汁中に固有に産生されるタンパク質(ヤギβカゼインなど)をコードする遺伝子の内部にインフレームで挿入された融合遺伝子を構築する。抗体遺伝子が挿入された融合遺伝子を含むDNA断片をヤギの胚へ注入すれば、該注入胚が雌のヤギへ導入される。胚を受容したヤギから生まれるトランスジェニックヤギ又はその子孫が産生する乳汁から、本発明の抗体を取得することができる。トランスジェニックヤギから産生される本発明の抗体を含む乳汁量を増加させるために、ホルモンをトランスジェニックヤギに適宜使用することも可能である(Ebert, K.M. et al., Bio/Technology(1994)12, 699-702)。 The present invention also provides a host organism having a vector containing a nucleic acid encoding the antibody of the present invention. The host organisms of the present invention are useful for the production of recombinant antibodies. Examples of host organisms in the present invention include goats. For example, the transgenic goat of the present invention can be produced as follows. That is, a fusion gene is constructed in which an antibody gene is inserted in-frame into a gene encoding a protein (such as goat β-casein) that is uniquely produced in milk. When a DNA fragment containing a fusion gene into which an antibody gene has been inserted is injected into a goat embryo, the injected embryo is introduced into a female goat. The antibody of the present invention can be obtained from the milk produced by a transgenic goat born from a goat that has received the embryo or its offspring. In order to increase the amount of milk containing the antibody of the present invention produced from a transgenic goat, hormones can also be used in the transgenic goat as appropriate (Ebert, KM et al., Bio / Technology (1994) 12 , 699-702).
 本発明のイメージング剤は、腫瘍細胞をイメージングするために哺乳動物に投与される。
 本明細書において、「イメージング」とは、抗体へ標識物質を付加することによって対象物を測定し、可視化することである。蛍光・ラジオアイソトープ標識を用いてCCDカメラにより観察する方法、MRIを用いたPET、SPECT、またはCTなども含む。
 哺乳動物としては、ヒト、非ヒト哺乳動物(例えばマウス、ラット、ハムスター、ウサギ、ブタ、サルなど)が挙げられる。本発明のイメージング剤は、腫瘍細胞の有無の診断において有用である。本発明のイメージング剤はインビボ用、インビトロ用のどちらとしても利用できる。
The imaging agent of the present invention is administered to a mammal in order to image tumor cells.
In this specification, “imaging” refers to measuring and visualizing an object by adding a labeling substance to an antibody. A method of observing with a CCD camera using a fluorescent / radioisotope label, PET, SPECT, or CT using MRI is also included.
Examples of mammals include human and non-human mammals (eg, mouse, rat, hamster, rabbit, pig, monkey, etc.). The imaging agent of the present invention is useful in the diagnosis of the presence or absence of tumor cells. The imaging agent of the present invention can be used for both in vivo use and in vitro use.
 本発明のイメージング剤によってイメージングされる腫瘍細胞は、メソセリンが発現している細胞である。メソセリンが発現している細胞、すなわち、本発明のイメージング剤によってイメージングされる細胞は、肺癌、中皮腫、卵巣癌、大腸癌、扁平上皮癌、膵臓癌、子宮癌、頭頸部癌、乳癌等が挙げられるが、これらに限定されるものではない。 The tumor cells imaged by the imaging agent of the present invention are cells expressing mesothelin. Cells expressing mesothelin, that is, cells imaged by the imaging agent of the present invention include lung cancer, mesothelioma, ovarian cancer, colon cancer, squamous cell carcinoma, pancreatic cancer, uterine cancer, head and neck cancer, breast cancer, etc. However, it is not limited to these.
 本発明のイメージング剤は、抗メソセリン抗体に、直接、あるいは間接的に追尾できる、イメージング用標識、プローブを結合させたものである。
 前記プローブを生体に生体内投与(例えば静脈内投与)した後、PETやSPECT、CCDカメラなどの画像計測装置を用いて腫瘍細胞の蓄積量や分布を測定することができる。
The imaging agent of the present invention is obtained by binding an imaging label or probe that can be directly or indirectly tracked to an anti-mesothelin antibody.
After the probe is administered to a living body in vivo (for example, intravenously administered), the amount and distribution of tumor cells can be measured using an image measuring device such as PET, SPECT, or CCD camera.
 また、近年、透過性線源により、物体を走査し、そのデータをコンピュータを用いて処理することで、物体の内部画像を構成する技術コンピュータ断層撮影(Computed Tomography:以下、「CT」との記載もComputed Tomographyを意味する)の病状診断、臨床への応用がされている。
 そのようなCT技術は、ポジトロン断層法 (PET)や単一光子放射断層撮影 (SPECT)、や核磁気共鳴画像法 (MRI) などの、断面像を得て、物体の(輪切りなどの)2次元断面画像を得る技術であるが、これらの検査技術は単に断面画像として用いられるだけでなく、コンピュータ画像処理技術向上によってその2次元画像を統合し3次元グラフィックスとして表示されることも多く、3次元的な病変の位置の特定、診断、手術方針決定等に有力な技術となっている。
 例えば、単純CTは、造影剤を使用せずにX線照射等の撮影を行うものであり、組織の浮腫、骨の形態異常、形態など造影剤を用いなくても充分に観察でき、一方、造影CTとは、X線吸収率の高い造影剤等を血管内に注射してから撮影を行うものをいい、血管や血流が豊富な組織の形状を観察することができる。さらに、いわゆる次世代CT、例えば、線源がらせん状に動くヘリカルCT、検出器自体を体軸方向に分割した多列検出器CT (MDCT)(マルチスライスCT(MSCT)とも呼ぶ)なども開発され、それらはいずれも特に制限なく、単独又は併用で本発明のイメージング剤の検出に適用可能である。
 イメージング用標識プローブ(本発明のイメージング剤)がX線吸収率の高い放射線核種である場合は、検出器としてのCTの単独使用も可能である。
In recent years, a computer computed tomography (hereinafter referred to as “CT”) that forms an internal image of an object by scanning the object with a transmissive radiation source and processing the data using a computer. (Also means Computed Tomography) and its clinical application.
Such CT techniques can be used to obtain cross-sectional images such as positron tomography (PET), single photon emission tomography (SPECT), and nuclear magnetic resonance imaging (MRI). Although it is a technology to obtain a three-dimensional cross-sectional image, these inspection techniques are not only used as a cross-sectional image, but are often displayed as three-dimensional graphics by integrating the two-dimensional image by improving computer image processing technology. It is a promising technique for specifying the location of three-dimensional lesions, making diagnoses, and determining surgical strategies.
For example, simple CT is to perform imaging such as X-ray irradiation without using a contrast agent, and can be sufficiently observed without using a contrast agent such as tissue edema, abnormal bone morphology, morphology, Contrast-enhanced CT refers to imaging performed after injecting a contrast agent or the like having a high X-ray absorption rate into a blood vessel, and the shape of a tissue rich in blood vessels and blood flow can be observed. In addition, so-called next-generation CT, for example, helical CT in which the radiation source moves in a spiral, multi-row detector CT (MDCT) (also called multi-slice CT (MSCT)) in which the detector itself is divided in the body axis direction are also developed. Any of them is not particularly limited and can be used alone or in combination for detection of the imaging agent of the present invention.
When the labeled probe for imaging (imaging agent of the present invention) is a radionuclide having a high X-ray absorption rate, it is possible to use CT alone as a detector.
 標識物質としては酵素発光(ルシフェラーゼ)による発光、発光低分子を利用するもの、蛍光タンパクや蛍光低分子を用いるもの、放射性核種等が挙げられるがこれらには限定されない。放射性核種としては51Cr、59Fe、57Co、67Ga、75Se、81mKr、99mTc、111In、125I、131I、133Xe、201Tlなどのγ線放出核種や、11C、13N、15O、18F、35mCl、76Br、 45Ti、48V、60Cu、61Cu、62Cu、66Ga、89Zr、94mTc、124Iなどの陽電子放出核種などが挙げられるがこれらには限定されない。なお当業者には自明であるが"m"とは核異性体を示す。特に、インジウム-111、テクネチウム-99mまたはヨウ素-131は、平面走査またはシングルフォトン断層撮影(SPECT)のために特に好ましく使用できる。陽電子放射標識の例えばフッ素-18、陽電子放射断層法において特に好ましく使用できる。常磁性イオンの例えばガドリニウム(III)またはマンガン(II)は特に好ましく磁気共鳴映像法(MRI)において使用できる。 Examples of the labeling substance include, but are not limited to, light emission by enzyme luminescence (luciferase), a substance using a light emitting small molecule, a substance using a fluorescent protein or a fluorescent small molecule, and a radionuclide. Radionuclides include gamma ray emitting nuclides such as 51 Cr, 59 Fe, 57 Co, 67 Ga, 75 Se, 81 m Kr, 99 m Tc, 111 In, 125 I, 131 I, 133 Xe, 201 Tl, 11 C, Positron emitting nuclides such as 13 N, 15 O, 18 F, 35 m Cl, 76 Br, 45 Ti, 48 V, 60 Cu, 61 Cu, 62 Cu, 66 Ga, 89 Zr, 94 m Tc, 124 I However, it is not limited to these. As is obvious to those skilled in the art, “m” represents a nuclear isomer. In particular, indium-111, technetium-99m or iodine-131 can be used particularly preferably for plane scanning or single photon tomography (SPECT). A positron emission label such as fluorine-18, which can be particularly preferably used in positron emission tomography. Paramagnetic ions such as gadolinium (III) or manganese (II) are particularly preferably used in magnetic resonance imaging (MRI).
 蛍光標識や発光標識としては、酵素(ルシフェラーゼ)による発光系を利用するものと、蛍光(GFP、DsRed、クサビラオレンジ等の蛍光タンパク質やFITCやCy5.5、Alexa Fluor 750などの蛍光性低分子)を用いるものとが使用できる。
 酵素発光(ルシフェラーゼ)による発光の場合は基質投与が別途必要である。
 特に、動物本来の自家蛍光による影響を軽減したものが好ましく、さらには皮膚透過性の高いシグナルを発する標識が好ましい。
As fluorescent labels and luminescent labels, those using a luminescence system with an enzyme (luciferase), fluorescent proteins such as GFP, DsRed, and wedge orange, and fluorescent small molecules such as FITC, Cy5.5, Alexa Fluor 750, etc. ) Can be used.
In the case of luminescence by enzyme luminescence (luciferase), administration of a substrate is required separately.
In particular, those that reduce the influence of the animal's natural autofluorescence are preferred, and labels that emit signals with high skin permeability are preferred.
 イメージング検出器としては磁気共鳴映像法(MRI)PETやSPECTが用いられるが、特に、蛍光標識プローブの観察において、低侵しゅう性の観点で、好ましくは、測定機器はCCDカメラである。
 それゆえ、CCDカメラで補足可能な波長、例えばおよそ350~900 nmの光を発する標識が好ましい。さらに生体内の光源の強さの測定には、CCDカメラによる測定動物の体表面における測定値から、該光源の強さを測定できる機械が好ましい。蛍光標識の場合、反射蛍光画像、透過蛍光画像のいずれでも良いが、両方の画像を捉えることが好ましい。また、蛍光画像を多方面(反射、透過を問わず)重ね合わせ、それに線源情報を統合して処理し、立体画像(3次元)として捉えることが、腫瘍細胞の3次元的な位置、分布を正確に構成でき、好ましい。こうして得た3次元画像は、CT画像とさらに統合することもできる。
As the imaging detector, magnetic resonance imaging (MRI) PET or SPECT is used. In particular, in observation of a fluorescently labeled probe, from the viewpoint of low invasiveness, the measuring instrument is preferably a CCD camera.
Therefore, labels that emit light at a wavelength that can be captured by a CCD camera, for example, approximately 350-900 nm, are preferred. Furthermore, for measuring the intensity of the light source in the living body, a machine capable of measuring the intensity of the light source from the measurement value on the body surface of the animal to be measured by the CCD camera is preferable. In the case of a fluorescent label, either a reflected fluorescent image or a transmitted fluorescent image may be used, but it is preferable to capture both images. In addition, it is possible to superimpose fluorescent images in various directions (regardless of reflection and transmission), integrate the source information and process them, and capture them as a three-dimensional image (three-dimensional). Can be configured accurately, which is preferable. The three-dimensional image thus obtained can be further integrated with the CT image.
 イメージング用標識プローブがX線吸収率の高い放射線核種を結合したものである場合、前述したようにイメージング検出器として、CT単独でも十分使用可能であり(例えばPETやSPECT)、腫瘍細胞の位置、蓄積量や分布を測定することができる。 When the labeled probe for imaging binds a radionuclide having a high X-ray absorption rate, as described above, CT alone can be sufficiently used (for example, PET or SPECT), the position of the tumor cell, Accumulation amount and distribution can be measured.
 一方、前記イメージング用標識プローブを生体に生体内投与(例えば静脈内投与)した後、標識された前記プローブのCT観察、又はCCDとの併用観察も可能である。併用観察の例として、蛍光標識したプローブのCCD像を、単純CT(及び/又は造影CTの像と重ね合わせる)を用いることができる。即ち、単純CTにより骨、肺などの臓器像の抽出(及び/又は造影CTの血管、組織像抽出)を行って得られたCT画像と、CCDカメラによる蛍光プローブ画像を統合し、腫瘍細胞の位置、蓄積量や分布について、3次元的な組織、血管との相対的な位置関係、腫瘍細胞の3次元(局在)イメージをより正確に把握することができる。 On the other hand, after the labeled probe for imaging is administered to a living body in vivo (for example, intravenous administration), CT observation of the labeled probe or combined observation with CCD is also possible. As an example of combined observation, a simple CT (and / or superimposition with an image of contrast CT) can be used for a CCD image of a fluorescently labeled probe. In other words, CT images obtained by extraction of images of organs such as bones and lungs (and / or blood vessel and tissue images of contrast-enhanced CT) by simple CT and fluorescence probe images by a CCD camera are integrated, and tumor cells With regard to position, accumulation amount and distribution, it is possible to more accurately grasp the three-dimensional tissue, the relative positional relationship with blood vessels, and the three-dimensional (localized) image of the tumor cell.
 本発明のイメージング剤は、抗体に加えて医薬的に許容し得る担体を導入し、公知の方法で製剤化することが可能である。例えば、水もしくはそれ以外の薬学的に許容し得る液との無菌性溶液、又は懸濁液剤の注射剤の形で非経口的に使用できる。例えば、薬理学上許容される担体もしくは媒体、具体的には、滅菌水や生理食塩水、植物油、乳化剤、懸濁剤、界面活性剤、安定剤、香味剤、賦形剤、ベヒクル、防腐剤、結合剤などと適宜組み合わせて、一般に認められた製薬実施に要求される単位用量形態で混和することによって製剤化することが考えられる。これら製剤における有効成分量は指示された範囲の適当な容量が得られるようにするものである。 The imaging agent of the present invention can be formulated by a known method by introducing a pharmaceutically acceptable carrier in addition to the antibody. For example, it can be used parenterally in the form of a sterile solution with water or other pharmaceutically acceptable liquid, or an injection of suspension. For example, a pharmacologically acceptable carrier or medium, specifically, sterile water or physiological saline, vegetable oil, emulsifier, suspension, surfactant, stabilizer, flavoring agent, excipient, vehicle, preservative It is conceivable to prepare a pharmaceutical preparation by combining with a binder or the like as appropriate and mixing in a unit dosage form generally required for pharmaceutical practice. The amount of active ingredient in these preparations is such that an appropriate volume within the indicated range can be obtained.
 注射のための無菌組成物は注射用蒸留水のようなベヒクルを用いて通常の製剤実施に従って処方することができる。
 注射用の水溶液としては、例えば生理食塩水、ブドウ糖やその他の補助薬を含む等張液、例えばD-ソルビトール、D-マンノース、D-マンニトール、塩化ナトリウムが挙げられ、適当な溶解補助剤、例えばアルコール、具体的にはエタノール、ポリアルコール、例えばプロピレングリコール、ポリエチレングリコール、非イオン性界面活性剤、例えばポリソルベート80(TM)、HCO-50と併用してもよい。
Sterile compositions for injection can be formulated according to normal pharmaceutical practice using a vehicle such as distilled water for injection.
Aqueous solutions for injection include, for example, isotonic solutions containing physiological saline, glucose and other adjuvants such as D-sorbitol, D-mannose, D-mannitol and sodium chloride, and suitable solubilizers such as You may use together with alcohol, specifically ethanol, polyalcohol, for example, propylene glycol, polyethylene glycol, nonionic surfactant, for example, polysorbate 80 (TM), HCO-50.
 油性液としてはゴマ油、大豆油があげられ、溶解補助剤として安息香酸ベンジル、ベンジルアルコールと併用してもよい。また、緩衝剤、例えばリン酸塩緩衝液、酢酸ナトリウム緩衝液、無痛化剤、例えば、塩酸プロカイン、安定剤、例えばベンジルアルコール、フェノール、酸化防止剤と配合してもよい。調製された注射液は通常、適当なアンプルに充填させる。 Examples of the oily liquid include sesame oil and soybean oil, which may be used in combination with benzyl benzoate or benzyl alcohol as a solubilizing agent. Moreover, you may mix | blend with buffer, for example, phosphate buffer, sodium acetate buffer, a soothing agent, for example, procaine hydrochloride, stabilizer, for example, benzyl alcohol, phenol, antioxidant. The prepared injection solution is usually filled into a suitable ampoule.
 投与は好ましくは非経口投与であり、具体的には、注射剤型、経鼻投与剤型、経肺投与剤型、経皮投与型などが挙げられる。注射剤型の例としては、例えば、静脈内注射、筋肉内注射、腹腔内注射、皮下注射などにより全身または局部的に投与することができる。 Administration is preferably parenteral administration, and specific examples include injection, nasal administration, pulmonary administration, and transdermal administration. As an example of the injection form, it can be administered systemically or locally by, for example, intravenous injection, intramuscular injection, intraperitoneal injection, subcutaneous injection, or the like.
 また、患者の年齢、症状により適宜投与方法を選択することができる。本発明のイメージング剤の投与量としては、例えば、一回につき体重1kgあたり0.0001mgから1000mgの範囲で選ぶことが可能である。あるいは、例えば、対象あたり0.001から100000mg/bodyの範囲で投与量を選ぶことができるが、これらの数値に必ずしも制限されるものではない。
 投与量、投与方法は、対象の体重や年齢、症状、mg抗体あたりの蛍光標識の強度・検出機械の検出感度などにより変動するが、当業者であれば適宜選択することが可能である。
The administration method can be appropriately selected depending on the age and symptoms of the patient. The dosage of the imaging agent of the present invention can be selected, for example, in the range of 0.0001 mg to 1000 mg per kg of body weight at a time. Alternatively, for example, the dose can be selected in the range of 0.001 to 100,000 mg / body per subject, but is not necessarily limited to these values.
The dose and administration method vary depending on the weight, age, symptom, symptom of the subject, the intensity of the fluorescent label per mg antibody, the detection sensitivity of the detection machine, etc., but can be appropriately selected by those skilled in the art.
 また本発明は、抗メソセリン抗体を含む、腫瘍細胞のイメージング用キットを提供する。本発明のキットは、対象に投与されることにより、腫瘍細胞をイメージングすることを特徴とする。上記キットには、本発明の抗体に加えて、例えば、標識物質、イメージング剤投与用の注射(点滴)器具、非吸着を抑える補助剤(例えばアルブミン等)等が含まれるが、これらに制限されるものではない。
 また、上記キットには、イメージングに用いるための指示書、適当な容器、コントロール試薬等、通常のキットに含まれるものを含んでいてもよい。
The present invention also provides a tumor cell imaging kit comprising an anti-mesothelin antibody. The kit of the present invention is characterized by imaging tumor cells by being administered to a subject. In addition to the antibody of the present invention, the kit includes, for example, a labeling substance, an injection (infusion) device for administration of an imaging agent, an auxiliary agent that suppresses non-adsorption (such as albumin), and the like, but is not limited thereto. It is not something.
In addition, the kit may include instructions included in a normal kit, such as instructions for use in imaging, appropriate containers, and control reagents.
 また、上述の本発明の組成物の用途は、以下(1)~(3)のように表現することもできる。
(1)腫瘍細胞のイメージング剤の製造における、抗メソセリン抗体の使用。
(2)腫瘍細胞のイメージング方法に使用するための、抗メソセリン抗体。
(3)抗メソセリン抗体を対象に投与する工程を含む、腫瘍細胞のイメージング方法。
The uses of the composition of the present invention described above can also be expressed as (1) to (3) below.
(1) Use of an anti-mesothelin antibody in the production of an imaging agent for tumor cells.
(2) An anti-mesothelin antibody for use in a tumor cell imaging method.
(3) A method for imaging tumor cells, comprising a step of administering an anti-mesothelin antibody to a subject.
 本明細書で開示した抗体の塩基配列およびアミノ酸配列は、以下の配列番号に従って配列表に記載されている。なお、11-25抗体は、#11-25抗体、クローン11-25と言い換えることもできる。
<11-25抗体>
配列番号1:重鎖可変領域の塩基配列
配列番号2:重鎖可変領域のアミノ酸配列
配列番号3:重鎖CDR1の塩基配列
配列番号4:重鎖CDR1のアミノ酸配列
配列番号5:重鎖CDR2の塩基配列
配列番号6:重鎖CDR2のアミノ酸配列
配列番号7:重鎖CDR3の塩基配列
配列番号8:重鎖CDR3のアミノ酸配列
配列番号9:軽鎖可変領域の塩基酸配列
配列番号10:軽鎖可変領域のアミノ酸配列
配列番号11:軽鎖CDR1の塩基配列
配列番号12:軽鎖CDR1のアミノ酸配列
配列番号13:軽鎖CDR2の塩基配列
配列番号14:軽鎖CDR2のアミノ酸配列
配列番号15:軽鎖CDR3の塩基配列
配列番号16:軽鎖CDR3のアミノ酸配列
The base sequences and amino acid sequences of the antibodies disclosed herein are described in the sequence listing according to the following SEQ ID NOs. The 11-25 antibody can also be referred to as # 11-25 antibody or clone 11-25.
<11-25 antibody>
SEQ ID NO: 1: base sequence of heavy chain variable region SEQ ID NO: 2: amino acid sequence of heavy chain variable region SEQ ID NO: 3: base sequence of heavy chain CDR1 SEQ ID NO: 4: amino acid sequence of heavy chain CDR1 SEQ ID NO: 5: heavy chain CDR2 Base sequence SEQ ID NO: 6: amino acid sequence of heavy chain CDR2 SEQ ID NO: 7: base sequence of heavy chain CDR3 SEQ ID NO: 8: amino acid sequence of heavy chain CDR3 SEQ ID NO: 9: base acid sequence of light chain variable region SEQ ID NO: 10: light chain Variable region amino acid sequence SEQ ID NO: 11: light chain CDR1 base sequence SEQ ID NO: 12: light chain CDR1 amino acid sequence SEQ ID NO: 13: light chain CDR2 base sequence SEQ ID NO: 14: light chain CDR2 amino acid sequence SEQ ID NO: 15: light Nucleotide sequence of chain CDR3 SEQ ID NO: 16: Amino acid sequence of light chain CDR3
 なお本明細書において引用されたすべての先行技術文献は、参照として本明細書に組み入れられる。 Note that all prior art documents cited in the present specification are incorporated herein by reference.
 以下、本発明を実施例によって詳細に説明するが、本発明の範囲はこれらの実施例に記載された態様に限定されるものではない。
〔実施例1〕
 本発明者らは、複数のがん細胞に対して、免疫染色を行った。
EXAMPLES Hereinafter, although an Example demonstrates this invention in detail, the scope of the present invention is not limited to the aspect described in these Examples.
[Example 1]
The present inventors performed immunostaining on a plurality of cancer cells.
使用した抗体
 抗メソセリン抗体としては、5種類の抗体、すなわち、11-25抗体、IC14-30、IC7-4、IC17-35、2-9抗体を利用した。コントロールとしては、抗体を添加していないものを用いた。各抗体の入手、作製方法は以下のとおりである。
11-25抗体:参考例1及び2、特開2010-014691参照
IC14-30:特開2010-014691参照
IC7-4:参考例2参照
IC17-35:参考例2参照
2-9抗体:参考例2参照
As the antibody anti-mesothelin antibody used, five types of antibodies, namely, 11-25 antibody, IC14-30, IC7-4, IC17-35, 2-9 antibody were used. As a control, one without added antibody was used. The method for obtaining and producing each antibody is as follows.
11-25 antibody: See Reference Examples 1 and 2, JP 2010-014691
IC14-30: Refer to JP2010-014691
IC7-4: See Reference Example 2
IC17-35: See Reference Example 2.
2-9 antibody: see Reference Example 2
使用した細胞株
 使用した細胞株は、NCI-H226(肺癌)、NCI-H520(肺癌)、211H(中皮腫)、PC3(前立腺癌)、PANC-1(膵臓癌)、BxPC-3(膵臓癌)、CFPAC-1(膵臓癌)である。各細胞株の入手方法は以下のとおりである。
 NCI-H226:ATCC より購入(CRL-5826で検索)。
 NCI-H520:ATCC より購入(HTB-182で検索)。
 211H:Yuji Kashiwakura, et al. Cancer Res 2008; 68: (20). October 15: 8333-8341, 2008、Yuji Kashiwakura, et al. Circulation April 29: 2078-2081, 2003参照
 PC3:Caliper Life Sciencesより入手(名称はPC-3M-luc-C6)。
 PANC-1:ATCCより購入(http://www.atcc.org/ATCCAdvancedCatalogSearch/tabid/112/Default.aspxでATCC numbersカテゴリー検索でCRL-1469と入力して検索)(住商ファーマインターナショナル株式会社)、Lieber M, et al. Establishment of a continuous tumor-cell line (panc-1) from a human carcinoma of the exocrine pancreas. Int. J. Cancer 15: 741-747, 1975参照。
 BxPC-3:ATCC より購入(CRL-1687で検索)、Loor R, et al. Use of pancreas-specific antigen in immunodiagnosis of pancreatic cancer. Clin. Lab. Med. 2: 567-578, 1982参照。
 CFPAC-1:ATCC より購入(CRL-1918で検索)、Schoumacher RA, et al. A cystic fibrosis pancreatic adenocarcinoma cell line. Proc. Natl. Acad. Sci. USA 87: 4012-4016, 1990参照。
The cell lines used are NCI-H226 (lung cancer), NCI-H520 (lung cancer), 211H (mesothelioma), PC3 (prostate cancer), PANC-1 (pancreatic cancer), BxPC-3 (pancreas) Cancer), CFPAC-1 (pancreatic cancer). The method for obtaining each cell line is as follows.
NCI-H226: Purchased from ATCC (search for CRL-5826).
NCI-H520: Purchased from ATCC (search for HTB-182).
211H: Yuji Kashiwakura, et al. Cancer Res 2008; 68: (20). October 15: 8333-8341, 2008, Yuji Kashiwakura, et al. Circulation April 29: 2078-2081, 2003 PC3: Obtained from Caliper Life Sciences (The name is PC-3M-luc-C6).
PANC-1: Purchased from ATCC (http://www.atcc.org/ATCCAdvancedCatalogSearch/tabid/112/Default.aspx and entered CRL-1469 in ATCC numbers category search) (Sumisho Pharma International Co., Ltd.), See Lieber M, et al. Establishment of a continuous tumor-cell line (panc-1) from a human carcinoma of the exocrine pancreas. Int. J. Cancer 15: 741-747, 1975.
BxPC-3: Purchased from ATCC (searched with CRL-1687), see Loor R, et al. Use of pancreas-specific antigen in immunodiagnosis of pancreatic cancer. Clin. Lab. Med. 2: 567-578, 1982.
CFPAC-1: Purchased from ATCC (searched by CRL-1918), see Schoumacher RA, et al. A cystic fibrosis pancreatic adenocarcinoma cell line. Proc. Natl. Acad. Sci. USA 87: 4012-4016, 1990.
免疫染色
 それぞれの細胞をラブテックII 8well(または2well)チャンバースライドにて3日間培養し、10%ホルマリン・PBSで固定後、10 mM グリシンを含むPBS(PBS-G)で洗浄、3% BSAを含むPBSでブロッキングし、抗メソセリン抗体で1時間処理した。その後0.1% BSAを含むPBSで洗浄し、TRITC標識ロバ抗マウスIgG (Santa Cruz、100倍希釈)、DAPI(4000倍希釈;核染色)、ローダミンファロイジン(molecular probe 社R415、200倍希釈)の混液で1時間処理した。洗浄後、蛍光用封入剤とカバーグラスで封入した。
 オリンパス IX71蛍光顕微鏡または共焦点レーザー走査顕微鏡 ZEISS LSM510型にて観察した。
Immunostaining respective cells were cultured for 3 days at Rabutekku II 8well (or 2Well) chamber slides were fixed with 10% formalin · PBS, washed with PBS (PBS-G) containing 10 mM glycine, containing 3% BSA Blocked with PBS and treated with anti-mesothelin antibody for 1 hour. After washing with PBS containing 0.1% BSA, TRITC-labeled donkey anti-mouse IgG (Santa Cruz, diluted 100-fold), DAPI (4000-fold diluted; nuclear staining), rhodamine phalloidin (molecular probe R415, diluted 200-fold) For 1 hour. After washing, it was sealed with a fluorescent mounting medium and a cover glass.
The images were observed with an Olympus IX71 fluorescence microscope or a confocal laser scanning microscope ZEISS LSM510.
結果
 BxPC-3膵臓がん細胞を8-wellチャンバーで培養し、抗メソセリン抗体5種でそれぞれ免疫染色を行い、共焦点レーザー顕微鏡で観察した(図1)。緑色は抗メソセリン抗体の結合、赤色はアクチンフィラメント、青色は核を示す。11-25抗体により細胞表面付近に著しい抗メソセリン抗体の結合が認められた。それ以外の4種類の抗メソセリン抗体では細胞への結合は検出されなかった。
 CFPAC-1膵臓がん細胞を2-wellチャンバーで培養し、抗メソセリン抗体5種でそれぞれ免疫染色を行い、共焦点レーザー顕微鏡で観察した(図2)。緑色は抗メソセリン抗体の結合、赤色はアクチンフィラメント、青色は核を示す。細胞表面付近に強い11-25抗メソセリン抗体の結合が小さな塊状に分布しているのが認められた。他の4種類の抗メソセリン抗体では細胞への結合は検出されなかった。
 PANC-1膵臓がん細胞を8-wellチャンバーで培養し、抗メソセリン抗体5種でそれぞれ免疫染色を行い、共焦点レーザー顕微鏡で観察した(図3)。緑色は抗メソセリン抗体の結合、赤色はアクチンフィラメント、青色は核を示す。細胞質に11-25抗メソセリン抗体の結合が認められた。それ以外の4種類の抗メソセリン抗体では細胞への結合はほとんど検出されなかった。
 211H中皮腫細胞を8-wellチャンバーで培養し、抗メソセリン抗体2種(11-25抗体及び#IC14-30抗体)でそれぞれ免疫染色を行い、共焦点レーザー顕微鏡で観察した(図4)。細胞表面および細胞質中に11-25抗体の結合が認められたが、#IC14-30抗体の結合は認められなかった。
 PC3前立腺がん細胞、211H中皮腫細胞、H520およびH226肺がん細胞の4種類について、11-25抗メソセリン抗体で染色を行い、共焦点レーザー顕微鏡で観察した(図5)。PC3細胞では細胞膜に抗メソセリン抗体の結合が見られた。211H細胞では、細胞膜および細胞質に抗メソセリン抗体の結合が見られた。H520細胞では細胞質に弱い抗体の結合を認めた。H226細胞では膜および細胞質に抗メソセリン抗体の結合が見られた。
Results BxPC-3 pancreatic cancer cells were cultured in an 8-well chamber, immunostained with five anti-mesothelin antibodies, and observed with a confocal laser microscope (FIG. 1). Green indicates binding of anti-mesothelin antibody, red indicates actin filaments, and blue indicates nuclei. The 11-25 antibody showed significant anti-mesothelin antibody binding near the cell surface. Binding to the cells was not detected with the other four anti-mesothelin antibodies.
CFPAC-1 pancreatic cancer cells were cultured in a 2-well chamber, immunostained with five anti-mesothelin antibodies, and observed with a confocal laser microscope (FIG. 2). Green indicates binding of anti-mesothelin antibody, red indicates actin filaments, and blue indicates nuclei. It was found that the binding of strong 11-25 anti-mesothelin antibody was distributed in small clumps near the cell surface. No binding to cells was detected with the other four types of anti-mesothelin antibodies.
PANC-1 pancreatic cancer cells were cultured in an 8-well chamber, immunostained with five anti-mesothelin antibodies, and observed with a confocal laser microscope (FIG. 3). Green indicates binding of anti-mesothelin antibody, red indicates actin filaments, and blue indicates nuclei. Binding of 11-25 anti-mesothelin antibody was observed in the cytoplasm. With the other four types of anti-mesothelin antibodies, binding to cells was hardly detected.
211H mesothelioma cells were cultured in an 8-well chamber, immunostained with two anti-mesothelin antibodies (11-25 antibody and # IC14-30 antibody), and observed with a confocal laser microscope (FIG. 4). 11-25 antibody binding was observed on the cell surface and cytoplasm, but # IC14-30 antibody binding was not observed.
Four types of PC3 prostate cancer cells, 211H mesothelioma cells, H520 and H226 lung cancer cells were stained with 11-25 anti-mesothelin antibody and observed with a confocal laser microscope (FIG. 5). PC3 cells showed binding of anti-mesothelin antibody to the cell membrane. In 211H cells, binding of anti-mesothelin antibody was observed in the cell membrane and cytoplasm. H520 cells showed weak antibody binding to the cytoplasm. In H226 cells, anti-mesothelin antibody binding was observed in the membrane and cytoplasm.
 抗メソセリン抗体11-25の反応性を表1にまとめる。
[表1]
―――――――――――――――――――――――――
       免疫染色(陽性部位)  インビボ
                   イメージング
―――――――――――――――――――――――――
211H(中皮腫)  + (細胞質&膜)    +
PC3 (前立腺癌)  +/-(細胞膜)    +/-
NCI-H226(肺癌) + (細胞質&膜)    +
NCI-H520(肺癌) +/-(細胞質)    -
PANC-1 (膵臓癌) +/-(細胞質)    -
BxPC-3(膵臓癌)  ++ (細胞膜)    ++
CFPAC-1(膵臓癌) ++ (細胞膜)    +++
―――――――――――――――――――――――――
The reactivity of anti-mesothelin antibody 11-25 is summarized in Table 1.
[Table 1]
―――――――――――――――――――――――――
Immunostaining (positive site) In vivo imaging ―――――――――――――――――――――――――
211H (mesothelioma) + (cytoplasm & membrane) +
PC3 (prostate cancer) +/- (cell membrane) +/-
NCI-H226 (Lung cancer) + (Cytoplasm & membrane) +
NCI-H520 (lung cancer) +/- (cytoplasm)-
PANC-1 (pancreatic cancer) +/- (cytoplasm)-
BxPC-3 (pancreatic cancer) ++ (cell membrane) ++
CFPAC-1 (pancreatic cancer) ++ (cell membrane) ++++
―――――――――――――――――――――――――
 免疫染色に用いることができた11-25抗体について、生細胞の細胞表面に対する反応性を確認するために、フローサイトメトリーを行った。NCI-H226,NCI-H520,BxPC-3,CFPAC-1それぞれの細胞(1.0 x105 Cells/sample)に対して50μlの11-25抗体(1.0 μg/ml)で4℃にて1時間の染色を行い、2次抗体にはPE標識ゴート抗マウスIgG抗体(IM0855ベックマンコールター)を用いた。フローサイトメトリーはベックマンコールターのFC500を使用した。その結果、NCI-H226, BxPC-3,CFPAC-1細胞について、11-25抗体の反応性が確認された。 For the 11-25 antibody that could be used for immunostaining, flow cytometry was performed in order to confirm the reactivity of living cells to the cell surface. NCI-H226, NCI-H520, BxPC-3, CFPAC-1 cells (1.0 x 10 5 Cells / sample) stained with 50 µl of 11-25 antibody (1.0 µg / ml) for 1 hour at 4 ° C PE-labeled goat anti-mouse IgG antibody (IM0855 Beckman Coulter) was used as the secondary antibody. For flow cytometry, Beckman Coulter FC500 was used. As a result, the reactivity of the 11-25 antibody was confirmed in NCI-H226, BxPC-3, and CFPAC-1 cells.
〔実施例2〕
 ヌードマウス背部後方皮下に実施例1で用いた癌細胞を移植し、3週間後にマウスの尾静脈からAlexa 750 (Invitrogen)標識あるいはCy5.5 (GEヘルスケア)標識11-25抗メソセリン抗体90μgを投与し、経時的に生体蛍光発光イメージング装置IVIS200 (Xenogen)にて反射蛍光を測定した。Alexa 750標識抗体を用いた場合は、励起745nm、蛍光800nmで、Cy5.5標識抗体を用いた場合は励起675nm、蛍光740nmで測定した。
[Example 2]
The cancer cells used in Example 1 were transplanted subcutaneously in the back of the back of nude mice, and after 3 weeks, 90 μg of Alexa 750 (Invitrogen) -labeled or Cy5.5 (GE Healthcare) -labeled 11-25 anti-mesothelin antibody was injected from the tail vein of the mice. After administration, the reflected fluorescence was measured with a biological fluorescence imaging apparatus IVIS200 (Xenogen) over time. When using Alexa 750-labeled antibody, excitation was performed at 745 nm and fluorescence at 800 nm, and when using Cy5.5-labeled antibody, measurement was performed at excitation at 675 nm and fluorescence at 740 nm.
結果
 PC3前立腺がん細胞および211H中皮腫細胞を皮下に移植したヌードマウスにAlexa750標識11-25抗メソセリン抗体90μgを尾静脈投与し、48時間後の蛍光を測定した。PC3腫瘍はやや弱く、211H腫瘍は強く蛍光を発していた(図6(A))。
 NCI-H520およびNCI-H226肺がん細胞を皮下に移植したヌードマウスにCy5.5標識11-25抗体90μgを尾静脈投与し、24時間後に反射蛍光を測定した。H520腫瘍部分は特異的蛍光を示さず、H226腫瘍部分は強く蛍光を発した(図6(B))。
 BxPC-3およびPANC-1膵臓がん細胞を皮下に移植したヌードマウスにAlexa750標識11-25抗メソセリン抗体90μgを尾静脈投与し、24時間後に反射蛍光を測定した。BxPC-3腫瘍部分は強い蛍光を示していたが、PANC-1腫瘍部分には蛍光を認めなかった(図6(C))。
 NCI-520肺がん細胞およびCFPAC-1膵臓がん細胞を移植したヌードマウスにAlexa750標識11-25抗メソセリン抗体90μgを尾静脈投与し、24時間後に反射蛍光を測定した。CFPAC-1腫瘍部分は非常に強い蛍光を示したが、H520腫瘍部分には特異的蛍光を認めなかった(図6(D))。
 図6(E)は、図6(B)のマウスの腫瘍を取り出して撮影したものである。H226腫瘍は強い蛍光を示したがH520腫瘍は特異的蛍光を示さなかった。
 図6(F)は、図6(C)のマウスの腫瘍を取り出して撮影したものである。BxPC-3腫瘍は強い蛍光を示したがPANC-1腫瘍は特異的蛍光を示さなかった。
 図6(G)は、(D)のマウスの腫瘍を取り出して撮影したものである。CFPAC-1腫瘍は強い蛍光を示したがH520腫瘍は特異的蛍光を示さなかった。
Results Nude mice transplanted subcutaneously with PC3 prostate cancer cells and 211H mesothelioma cells were administered 90 μg of Alexa750-labeled 11-25 anti-mesothelin antibody via the tail vein, and fluorescence was measured 48 hours later. The PC3 tumor was somewhat weak and the 211H tumor was strongly fluorescent (FIG. 6 (A)).
Nude mice transplanted subcutaneously with NCI-H520 and NCI-H226 lung cancer cells were administered 90 μg of Cy5.5-labeled 11-25 antibody via the tail vein, and the reflected fluorescence was measured 24 hours later. The H520 tumor part did not show specific fluorescence, and the H226 tumor part emitted strong fluorescence (FIG. 6 (B)).
Nude mice transplanted subcutaneously with BxPC-3 and PANC-1 pancreatic cancer cells were administered 90 μg of Alexa750-labeled 11-25 anti-mesothelin antibody via the tail vein, and the reflected fluorescence was measured 24 hours later. The BxPC-3 tumor part showed strong fluorescence, but no fluorescence was observed in the PANC-1 tumor part (FIG. 6 (C)).
Nude mice transplanted with NCI-520 lung cancer cells and CFPAC-1 pancreatic cancer cells were administered with tail vein of 90 μg of Alexa750-labeled 11-25 anti-mesothelin antibody, and the reflected fluorescence was measured 24 hours later. The CFPAC-1 tumor part showed very strong fluorescence, but no specific fluorescence was observed in the H520 tumor part (FIG. 6 (D)).
FIG. 6 (E) is a photograph of the mouse tumor of FIG. 6 (B) taken out. H226 tumor showed strong fluorescence, while H520 tumor showed no specific fluorescence.
FIG. 6 (F) is a photograph of the mouse tumor of FIG. 6 (C) taken out. BxPC-3 tumors showed strong fluorescence while PANC-1 tumors did not show specific fluorescence.
FIG. 6 (G) is a photograph of the mouse tumor of (D) taken out. CFPAC-1 tumors showed strong fluorescence while H520 tumors did not show specific fluorescence.
〔参考例1 組換えメソセリンタンパク質の生産〕
 組換えメソセリンタンパク質は、5’-AAATTTCCCAAGCTTGTGGAGAAGACAGCCTGTCCTTCAGGCAAG-3’(配列番号:22)と、5’-AAGGAAAAAAGCGGCCGCGCCCTGTAGCCCCAGCCCCAGCGTGTCCAG-3’(配列番号:23)というプライマーを用いるポリメラーゼ連鎖反応によってヒトメソセリン前駆体タンパク質(Genbankアクセッション番号NM_005823)の転写バリアント1をエンコードするcDNAに由来するアミノ酸297-580番目のコーディング部分を増幅することによって作成された。増幅DNAは、発現ベクターpSecTag2B(Invitrogen)のHindIII/NotI部位に挿入された。クローンNM005823によってエンコードされるタンパク質のアミノ酸297-580番目は、Schollerら(Proc Natl Acad Sci U S A. 1999;96(20):11531-6)によって説明されるヒトメソセリンのアイソフォーム1及び3に共通の配列である点に留意すべきである。前記メソセリン発現プラスミドは、HEK293T細胞にトランスフェクションされた。メソセリンタンパク質は、トランスフェクションされた細胞の培養上清からTALON樹脂を用いて精製された。このようにして得られる精製メソセリンタンパク質はPBSで2回透析され、使用時まで-80°Cで保存された。
[Reference Example 1 Production of recombinant mesothelin protein]
Recombinant mesothelin protein is composed of 5′-AAATTTCCAAGCTTGTGGAGAAGACAGCCTGTCCTTCAGGCAAG-3 ′ (SEQ ID NO: 22) and 5′-AAGGAAAAAAGCGGCCCCGCCCTGTAGCCCCAGCCCCAGCGGTCCAG-3 ′ (SEQ ID NO: 23) using a primer called G It was created by amplifying the coding part of amino acids 297-580 derived from cDNA encoding transcription variant 1 of session number NM_005823). The amplified DNA was inserted into the HindIII / NotI sites of the expression vector pSecTag2B (Invitrogen). Amino acids 297-580 of the protein encoded by clone NM005823 are common to human mesothelin isoforms 1 and 3 described by Scholler et al. (Proc Natl Acad Sci US A. 1999; 96 (20): 11531-6). Note that this is an array of The mesothelin expression plasmid was transfected into HEK293T cells. Mesothelin protein was purified from the culture supernatant of the transfected cells using TALON resin. The purified mesothelin protein thus obtained was dialyzed twice with PBS and stored at −80 ° C. until use.
〔参考例2 メソセリン抗体の生産〕
 メソセリンに対するモノクローナル抗体(mAbs)を生産するために、4ないし6週齢のBALB/cマウスは、精製組換えメソセリンタンパク質を0日目、7日目、14日目及び16日目に腹腔内注射(10マイクログラム/注射)して免疫された。4回目の免疫後18日目に脾臓のリンパ球は採取され、50%ポリエチレングリコール4000溶液を用いてP3U1メラノーマ細胞と融合された。前記融合細胞は、15% ウシ胎児血清、ペニシリン/ストレプトマイシン及びHAT溶液(Invitrogen)を含む、RPMI-1640培地を用いて96ウェルプレート上に播種された。37°C、5% CO、飽和水蒸気下で10日間の培養後に培養上清は採取され、組換えメソセリンタンパク質を用いる間接ELISAによって免疫原に結合する能力がスクリーニングされた。選択された陽性ハイブリドーマコロニーは増殖され、限界希釈によってサブクローン化された。サブクローン化ハイブリドーマは培養され、サブクローン化抗体のアイソタイプはRocheのアイソストリップキットを用いて決定された。抗体精製はタンパク質A親和性クロマトグラフィーによって実施された。得られたクローン間の免疫原競合アッセイ後に、11-25抗体(IgG2bカッパー)が、MPFのELISA検出のためのELISAを構築するために選択された。11-25抗体は、GE HealthcareのECLタンパク質ビオチネーションモジュールを用いてビオチン化された。
 また、11-25抗体と同様に、IC7-4、IC17-35、2-9抗体を作製した。
[Reference Example 2 Production of Mesothelin Antibody]
To produce monoclonal antibodies (mAbs) against mesothelin, 4-6 week old BALB / c mice were treated with purified recombinant mesothelin protein intraperitoneally on days 0, 7, 14, and 16. Immunized by injection (10 micrograms / injection). On day 18 after the fourth immunization, splenic lymphocytes were collected and fused with P3U1 melanoma cells using a 50% polyethylene glycol 4000 solution. The fused cells were seeded on 96-well plates using RPMI-1640 medium containing 15% fetal bovine serum, penicillin / streptomycin and HAT solution (Invitrogen). Culture supernatants were collected after 10 days of culture at 37 ° C., 5% CO 2 , saturated water vapor and screened for the ability to bind to the immunogen by indirect ELISA using recombinant mesothelin protein. Selected positive hybridoma colonies were grown and subcloned by limiting dilution. Subcloned hybridomas were cultured and the subcloned antibody isotype was determined using Roche's isostrip kit. Antibody purification was performed by protein A affinity chromatography. After an immunogen competition assay between the resulting clones, the 11-25 antibody (IgG2b kappa) was selected to construct an ELISA for ELISA detection of MPF. The 11-25 antibody was biotinylated using the GE Healthcare ECL protein biotination module.
Similarly to the 11-25 antibody, IC7-4, IC17-35, 2-9 antibodies were prepared.
〔参考例3 MSLN抗体の特徴付け〕
 抗MSLNモノクローナル抗体、11-25抗体のCDR領域の核酸配列を決定するために、全量RNAは、参考例2に従って生産されるIC11-25のそれぞれを産生するハイブリドーマから採取された。採取される前記全量RNA中のメッセンジャーRNAは、(Invitrogenから購入される)オリゴ(dT)12-18プライマーを用いて相補性DNAに逆転写された。このDNAから、免疫グロブリン可変領域配列のヌクレオチド配列は、11-25の重鎖のための5’-ATGGCTGTCTTGGGGCTGCTCTTCTGC-3’(配列番号:24)及び5’-CAGTGGATAGACTGATGGGGG-3’(配列番号:25)プライマーと、11-25の軽鎖のための5’-ATGAAGTTGCCTGTTAGGCTGTTGGTGCTG-3’(配列番号:26)及び5’-ACTGGATGGTGGGAAGATGG-3’(配列番号:27)プライマーとを用いるPCR技術によって増幅された。増幅cDNAは、pBluescript II KS(+)プラスミド(Stratagene)のEcoRV部位に挿入された。次に、VH及びVL領域の配列は遺伝子配列決定装置(3130/3100-Avant(登録商標)Genetic Analyzer Capillary Arrays、Applied Biosystems)によって解析された。FR1、2、3及び4と、CDR1、2及び3との領域の決定は、IgG blast(NCBI データベース)及びSEQUENCES OF PROTEINS OF IMMUNOLOGICAL INTEREST、1巻、第5版、1991;NIH出版第91-3242頁(Elvin A. Kabatら)を用いて実施された。11-25抗体の各領域の配列は以下のとおりである。
<11-25抗体>
配列番号1:重鎖可変領域の塩基配列
配列番号2:重鎖可変領域のアミノ酸配列
配列番号3:重鎖CDR1の塩基配列
配列番号4:重鎖CDR1のアミノ酸配列
配列番号5:重鎖CDR2の塩基配列
配列番号6:重鎖CDR2のアミノ酸配列
配列番号7:重鎖CDR3の塩基配列
配列番号8:重鎖CDR3のアミノ酸配列
配列番号9:軽鎖可変領域の塩基酸配列
配列番号10:軽鎖可変領域のアミノ酸配列
配列番号11:軽鎖CDR1の塩基配列
配列番号12:軽鎖CDR1のアミノ酸配列
配列番号13:軽鎖CDR2の塩基配列
配列番号14:軽鎖CDR2のアミノ酸配列
配列番号15:軽鎖CDR3の塩基配列
配列番号16:軽鎖CDR3のアミノ酸配列
[Reference Example 3 Characterization of MSLN Antibody]
To determine the nucleic acid sequence of the CDR regions of the anti-MSLN monoclonal antibody, 11-25 antibody, total RNA was collected from hybridomas producing each of IC11-25 produced according to Reference Example 2. Messenger RNA in the total RNA collected was reverse transcribed into complementary DNA using oligo (dT) 12-18 primer (purchased from Invitrogen). From this DNA, the nucleotide sequence of the immunoglobulin variable region sequence is 5'-ATGGCTGTCTTGGGGCTGCTCTTCTGC-3 '(SEQ ID NO: 24) and 5'-CAGTGGATAGACTGATGGGGG-3' (SEQ ID NO: 25) for the heavy chain of 11-25. Amplified by PCR technique using primers and 5′-ATGAAGTTGCCCGTTAGGCTGTGTGTGCTG-3 ′ (SEQ ID NO: 26) and 5′-ACTGGATGGTGGAGAATGG-3 ′ (SEQ ID NO: 27) primers for 11-25 light chains. The amplified cDNA was inserted into the EcoRV site of the pBluescript II KS (+) plasmid (Stratagene). Next, the sequences of the VH and VL regions were analyzed by a gene sequencing device (3130 / 3100-Avant (R) Genetic Analyzer Capillary Arrays, Applied Biosystems). The determination of the region between FR1, 2, 3 and 4 and CDR1, 2 and 3 is as follows: IgG blast (NCBI database) and SEQUENCES OF PROTEINS OF IMMUNOLOGICAL INTEREST, Volume 1, 5th edition, 1991; NIH Publication 91-3242 P. (Elvin A. Kabat et al.). The sequence of each region of the 11-25 antibody is as follows.
<11-25 antibody>
SEQ ID NO: 1: base sequence of heavy chain variable region SEQ ID NO: 2: amino acid sequence of heavy chain variable region SEQ ID NO: 3: base sequence of heavy chain CDR1 SEQ ID NO: 4: amino acid sequence of heavy chain CDR1 SEQ ID NO: 5: heavy chain CDR2 Base sequence SEQ ID NO: 6: amino acid sequence of heavy chain CDR2 SEQ ID NO: 7: base sequence of heavy chain CDR3 SEQ ID NO: 8: amino acid sequence of heavy chain CDR3 SEQ ID NO: 9: base acid sequence of light chain variable region SEQ ID NO: 10: light chain Variable region amino acid sequence SEQ ID NO: 11: light chain CDR1 base sequence SEQ ID NO: 12: light chain CDR1 amino acid sequence SEQ ID NO: 13: light chain CDR2 base sequence SEQ ID NO: 14: light chain CDR2 amino acid sequence SEQ ID NO: 15: light Nucleotide sequence of chain CDR3 SEQ ID NO: 16: Amino acid sequence of light chain CDR3
 本発明者らは、膜型メソセリンを高発現する実際の患者由来の癌を直接画像化できる試薬を開発した。本発明は、メソセリンを発現しているがん(肺がん、中皮腫、卵巣がん、大腸がん、扁平上皮がん、膵臓がん、子宮がん等)を画像化するために有用である。 The present inventors have developed a reagent that can directly image cancer derived from an actual patient that highly expresses mesothelin membrane. The present invention is useful for imaging cancers (lung cancer, mesothelioma, ovarian cancer, colon cancer, squamous cell cancer, pancreatic cancer, uterine cancer, etc.) expressing mesothelin. .

Claims (6)

  1. 抗メソセリン抗体を含む、腫瘍細胞のイメージング剤。 A tumor cell imaging agent comprising an anti-mesothelin antibody.
  2. 抗体が下記(a)~(e)のいずれかに記載の抗体である、請求項1記載のイメージング剤;
    (a)CDR1として配列番号:4に記載のアミノ酸配列、CDR2として配列番号:6に記載のアミノ酸配列、及びCDR3として配列番号:8に記載のアミノ酸配列を有する重鎖を含む抗体、
    (b)重鎖可変領域として配列番号:2に記載のアミノ酸配列を有する重鎖を含む抗体、
    (c)CDR1として配列番号:12に記載のアミノ酸配列、CDR2として配列番号:14に記載のアミノ酸配列、及びCDR3として配列番号:16に記載のアミノ酸配列を有する軽鎖を含む抗体、
    (d)軽鎖可変領域として配列番号:10に記載のアミノ酸配列を有する軽鎖を含む抗体、
    (e)上記(a)または(b)に記載の重鎖、および、上記(c)または(d)に記載の軽鎖の対を有する抗体。
    The imaging agent according to claim 1, wherein the antibody is an antibody according to any one of the following (a) to (e):
    (A) an antibody comprising a heavy chain having the amino acid sequence of SEQ ID NO: 4 as CDR1, the amino acid sequence of SEQ ID NO: 6 as CDR2, and the amino acid sequence of SEQ ID NO: 8 as CDR3;
    (B) an antibody comprising a heavy chain having the amino acid sequence set forth in SEQ ID NO: 2 as the heavy chain variable region;
    (C) an antibody comprising a light chain having the amino acid sequence of SEQ ID NO: 12 as CDR1, the amino acid sequence of SEQ ID NO: 14 as CDR2, and the amino acid sequence of SEQ ID NO: 16 as CDR3;
    (D) an antibody comprising a light chain having the amino acid sequence set forth in SEQ ID NO: 10 as the light chain variable region,
    (E) An antibody having the heavy chain pair described in (a) or (b) above and the light chain pair described in (c) or (d) above.
  3. インビボで用いられる、請求項1または2記載のイメージング剤。 The imaging agent according to claim 1 or 2, which is used in vivo.
  4. 抗メソセリン抗体を含む、腫瘍細胞のイメージング用キット。 A kit for imaging tumor cells, comprising an anti-mesothelin antibody.
  5. 抗体が下記(a)~(e)のいずれかに記載の抗体である、請求項4記載のイメージング用キット;
    (a)CDR1として配列番号:4に記載のアミノ酸配列、CDR2として配列番号:6に記載のアミノ酸配列、及びCDR3として配列番号:8に記載のアミノ酸配列を有する重鎖を含む抗体、
    (b)重鎖可変領域として配列番号:2に記載のアミノ酸配列を有する重鎖を含む抗体、
    (c)CDR1として配列番号:12に記載のアミノ酸配列、CDR2として配列番号:14に記載のアミノ酸配列、及びCDR3として配列番号:16に記載のアミノ酸配列を有する軽鎖を含む抗体、
    (d)軽鎖可変領域として配列番号:10に記載のアミノ酸配列を有する軽鎖を含む抗体、
    (e)上記(a)または(b)に記載の重鎖、および、上記(c)または(d)に記載の軽鎖の対を有する抗体。
    The imaging kit according to claim 4, wherein the antibody is the antibody according to any of the following (a) to (e):
    (A) an antibody comprising a heavy chain having the amino acid sequence of SEQ ID NO: 4 as CDR1, the amino acid sequence of SEQ ID NO: 6 as CDR2, and the amino acid sequence of SEQ ID NO: 8 as CDR3;
    (B) an antibody comprising a heavy chain having the amino acid sequence set forth in SEQ ID NO: 2 as the heavy chain variable region;
    (C) an antibody comprising a light chain having the amino acid sequence of SEQ ID NO: 12 as CDR1, the amino acid sequence of SEQ ID NO: 14 as CDR2, and the amino acid sequence of SEQ ID NO: 16 as CDR3;
    (D) an antibody comprising a light chain having the amino acid sequence set forth in SEQ ID NO: 10 as the light chain variable region,
    (E) An antibody having the heavy chain pair described in (a) or (b) above and the light chain pair described in (c) or (d) above.
  6. インビボで用いられる、請求項4または5記載のイメージング用キット。 The imaging kit according to claim 4 or 5, which is used in vivo.
PCT/JP2010/072608 2009-12-18 2010-12-16 Mesothelin (msln) antibody and use thereof WO2011074621A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
JP2011546157A JPWO2011074621A1 (en) 2009-12-18 2010-12-16 Antibody to mesothelin (MSLN) and use thereof

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
JP2009-287506 2009-12-18
JP2009287506 2009-12-18

Publications (1)

Publication Number Publication Date
WO2011074621A1 true WO2011074621A1 (en) 2011-06-23

Family

ID=44167372

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2010/072608 WO2011074621A1 (en) 2009-12-18 2010-12-16 Mesothelin (msln) antibody and use thereof

Country Status (2)

Country Link
JP (1) JPWO2011074621A1 (en)
WO (1) WO2011074621A1 (en)

Cited By (32)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2016500655A (en) * 2012-09-27 2016-01-14 ザ・ユナイテッド・ステイツ・オブ・アメリカ・アズ・リ Mesothelin antibodies and methods for eliciting potent anti-tumor activity
WO2016123593A1 (en) * 2015-01-30 2016-08-04 Academia Sinica Compositions and methods relating to universal glycoforms for enhanced antibody efficacy
US9547009B2 (en) 2012-08-21 2017-01-17 Academia Sinica Benzocyclooctyne compounds and uses thereof
US9879042B2 (en) 2014-09-08 2018-01-30 Academia Sinica Human iNKT cell activation using glycolipids
WO2018048975A1 (en) 2016-09-09 2018-03-15 Bristol-Myers Squibb Company Use of an anti-pd-1 antibody in combination with an anti-mesothelin antibody in cancer treatment
US9975965B2 (en) 2015-01-16 2018-05-22 Academia Sinica Compositions and methods for treatment and detection of cancers
US9981030B2 (en) 2013-06-27 2018-05-29 Academia Sinica Glycan conjugates and use thereof
US9982041B2 (en) 2014-01-16 2018-05-29 Academia Sinica Compositions and methods for treatment and detection of cancers
JP6339283B1 (en) * 2017-10-31 2018-06-06 国立大学法人 岡山大学 DNA, polypeptide, anti-mesothelin antibody, tumor imaging agent and complex
US10005847B2 (en) 2014-05-27 2018-06-26 Academia Sinica Anti-HER2 glycoantibodies and uses thereof
US10023892B2 (en) 2014-05-27 2018-07-17 Academia Sinica Compositions and methods relating to universal glycoforms for enhanced antibody efficacy
US10087236B2 (en) 2009-12-02 2018-10-02 Academia Sinica Methods for modifying human antibodies by glycan engineering
US10086054B2 (en) 2013-06-26 2018-10-02 Academia Sinica RM2 antigens and use thereof
US10111951B2 (en) 2013-09-06 2018-10-30 Academia Sinica Human iNKT cell activation using glycolipids with altered glycosyl groups
US10118969B2 (en) 2014-05-27 2018-11-06 Academia Sinica Compositions and methods relating to universal glycoforms for enhanced antibody efficacy
US10119972B2 (en) 2014-03-27 2018-11-06 Academia Sinica Reactive labelling compounds and uses thereof
US10130714B2 (en) 2012-04-14 2018-11-20 Academia Sinica Enhanced anti-influenza agents conjugated with anti-inflammatory activity
US10150818B2 (en) 2014-01-16 2018-12-11 Academia Sinica Compositions and methods for treatment and detection of cancers
US10214765B2 (en) 2012-08-18 2019-02-26 Academia Sinica Cell-permeable probes for identification and imaging of sialidases
US10274488B2 (en) 2008-07-15 2019-04-30 Academia Sinica Glycan arrays on PTFE-like aluminum coated glass slides and related methods
US10317393B2 (en) 2007-03-23 2019-06-11 Academia Sinica Alkynyl sugar analogs for labeling and visualization of glycoconjugates in cells
US10338069B2 (en) 2010-04-12 2019-07-02 Academia Sinica Glycan arrays for high throughput screening of viruses
US10336784B2 (en) 2016-03-08 2019-07-02 Academia Sinica Methods for modular synthesis of N-glycans and arrays thereof
US10342858B2 (en) 2015-01-24 2019-07-09 Academia Sinica Glycan conjugates and methods of use thereof
US10495645B2 (en) 2015-01-16 2019-12-03 Academia Sinica Cancer markers and methods of use thereof
WO2019232449A1 (en) 2018-06-01 2019-12-05 Eisai R&D Management Co., Ltd. Splicing modulator antibody-drug conjugates and methods of use
US10538592B2 (en) 2016-08-22 2020-01-21 Cho Pharma, Inc. Antibodies, binding fragments, and methods of use
WO2020123836A2 (en) 2018-12-13 2020-06-18 Eisai R&D Management Co., Ltd. Herboxidiene splicing modulator antibody-drug conjugates and methods of use
US11332523B2 (en) 2014-05-28 2022-05-17 Academia Sinica Anti-TNF-alpha glycoantibodies and uses thereof
US11377485B2 (en) 2009-12-02 2022-07-05 Academia Sinica Methods for modifying human antibodies by glycan engineering
WO2023115528A1 (en) * 2021-12-24 2023-06-29 Zhejiang Shimai Pharmaceutical Co., Ltd. Antibodies against mesothelin and uses thereof
US11884739B2 (en) 2014-05-27 2024-01-30 Academia Sinica Anti-CD20 glycoantibodies and uses thereof

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2007071849A (en) * 2005-08-09 2007-03-22 Meneki Seibutsu Kenkyusho:Kk Diagnostic agent and diagnostic kit for mesothelioma

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2007071849A (en) * 2005-08-09 2007-03-22 Meneki Seibutsu Kenkyusho:Kk Diagnostic agent and diagnostic kit for mesothelioma

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
ATSUSHI TSUJI ET AL.: "Shuyo Kenkyu Jo no Yuyosei", NIRS-M, March 2009 (2009-03-01), pages 11 - 12 *
HASSAN,R. ET AL.: "lndium-labeled monoclonal antibody Kl: biodistribution study in nude mice bearing a human carcinoma xenograft expressing mesothelin", INT J CANCER, vol. 80, no. 4, 1999, pages 559 - 63 *
HASSAN,R. ET AL.: "Mesothelin targeted cancer immunotherapy", EUR J CANCER, vol. 44, no. 1, 2008, pages 46 - 53 *
IWAHORI,K. ET AL.: "Megakaryocyte potentiating factor as a tumor marker of malignant pleural mesothelioma: evaluation in comparison with mesothelin", LUNG CANCER, vol. 62, no. 1, 2008, pages 45 - 54 *

Cited By (40)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10317393B2 (en) 2007-03-23 2019-06-11 Academia Sinica Alkynyl sugar analogs for labeling and visualization of glycoconjugates in cells
US10274488B2 (en) 2008-07-15 2019-04-30 Academia Sinica Glycan arrays on PTFE-like aluminum coated glass slides and related methods
US10087236B2 (en) 2009-12-02 2018-10-02 Academia Sinica Methods for modifying human antibodies by glycan engineering
US11377485B2 (en) 2009-12-02 2022-07-05 Academia Sinica Methods for modifying human antibodies by glycan engineering
US11267870B2 (en) 2009-12-02 2022-03-08 Academia Sinica Methods for modifying human antibodies by glycan engineering
US10338069B2 (en) 2010-04-12 2019-07-02 Academia Sinica Glycan arrays for high throughput screening of viruses
US10130714B2 (en) 2012-04-14 2018-11-20 Academia Sinica Enhanced anti-influenza agents conjugated with anti-inflammatory activity
US10214765B2 (en) 2012-08-18 2019-02-26 Academia Sinica Cell-permeable probes for identification and imaging of sialidases
US9547009B2 (en) 2012-08-21 2017-01-17 Academia Sinica Benzocyclooctyne compounds and uses thereof
JP2016500655A (en) * 2012-09-27 2016-01-14 ザ・ユナイテッド・ステイツ・オブ・アメリカ・アズ・リ Mesothelin antibodies and methods for eliciting potent anti-tumor activity
US10086054B2 (en) 2013-06-26 2018-10-02 Academia Sinica RM2 antigens and use thereof
US9981030B2 (en) 2013-06-27 2018-05-29 Academia Sinica Glycan conjugates and use thereof
US10918714B2 (en) 2013-09-06 2021-02-16 Academia Sinica Human iNKT cell activation using glycolipids with altered glycosyl groups
US10111951B2 (en) 2013-09-06 2018-10-30 Academia Sinica Human iNKT cell activation using glycolipids with altered glycosyl groups
US9982041B2 (en) 2014-01-16 2018-05-29 Academia Sinica Compositions and methods for treatment and detection of cancers
US10150818B2 (en) 2014-01-16 2018-12-11 Academia Sinica Compositions and methods for treatment and detection of cancers
US10119972B2 (en) 2014-03-27 2018-11-06 Academia Sinica Reactive labelling compounds and uses thereof
US11884739B2 (en) 2014-05-27 2024-01-30 Academia Sinica Anti-CD20 glycoantibodies and uses thereof
US10023892B2 (en) 2014-05-27 2018-07-17 Academia Sinica Compositions and methods relating to universal glycoforms for enhanced antibody efficacy
US10005847B2 (en) 2014-05-27 2018-06-26 Academia Sinica Anti-HER2 glycoantibodies and uses thereof
US10118969B2 (en) 2014-05-27 2018-11-06 Academia Sinica Compositions and methods relating to universal glycoforms for enhanced antibody efficacy
US11319567B2 (en) 2014-05-27 2022-05-03 Academia Sinica Fucosidase from bacteroides and methods using the same
US10618973B2 (en) 2014-05-27 2020-04-14 Academia Sinica Anti-HER2 glycoantibodies and uses thereof
US11332523B2 (en) 2014-05-28 2022-05-17 Academia Sinica Anti-TNF-alpha glycoantibodies and uses thereof
US9879042B2 (en) 2014-09-08 2018-01-30 Academia Sinica Human iNKT cell activation using glycolipids
US10533034B2 (en) 2014-09-08 2020-01-14 Academia Sinica Human iNKT cell activation using glycolipids
US9975965B2 (en) 2015-01-16 2018-05-22 Academia Sinica Compositions and methods for treatment and detection of cancers
US10495645B2 (en) 2015-01-16 2019-12-03 Academia Sinica Cancer markers and methods of use thereof
US10342858B2 (en) 2015-01-24 2019-07-09 Academia Sinica Glycan conjugates and methods of use thereof
WO2016123593A1 (en) * 2015-01-30 2016-08-04 Academia Sinica Compositions and methods relating to universal glycoforms for enhanced antibody efficacy
US10336784B2 (en) 2016-03-08 2019-07-02 Academia Sinica Methods for modular synthesis of N-glycans and arrays thereof
US10538592B2 (en) 2016-08-22 2020-01-21 Cho Pharma, Inc. Antibodies, binding fragments, and methods of use
WO2018048975A1 (en) 2016-09-09 2018-03-15 Bristol-Myers Squibb Company Use of an anti-pd-1 antibody in combination with an anti-mesothelin antibody in cancer treatment
JP2019080531A (en) * 2017-10-31 2019-05-30 国立大学法人 岡山大学 DNA, polypeptide, anti-mesothelin antibody, tumor imaging agent and complex
CN111247249A (en) * 2017-10-31 2020-06-05 国立大学法人冈山大学 DNA, polypeptide, anti-mesothelin antibody, tumor imaging agent and complex
JP6339283B1 (en) * 2017-10-31 2018-06-06 国立大学法人 岡山大学 DNA, polypeptide, anti-mesothelin antibody, tumor imaging agent and complex
US11975080B2 (en) 2017-10-31 2024-05-07 National University Corporation Okayama University Anti-mesothelin polypeptide, and tumor imaging agents and complexes thereof
WO2019232449A1 (en) 2018-06-01 2019-12-05 Eisai R&D Management Co., Ltd. Splicing modulator antibody-drug conjugates and methods of use
WO2020123836A2 (en) 2018-12-13 2020-06-18 Eisai R&D Management Co., Ltd. Herboxidiene splicing modulator antibody-drug conjugates and methods of use
WO2023115528A1 (en) * 2021-12-24 2023-06-29 Zhejiang Shimai Pharmaceutical Co., Ltd. Antibodies against mesothelin and uses thereof

Also Published As

Publication number Publication date
JPWO2011074621A1 (en) 2013-04-25

Similar Documents

Publication Publication Date Title
WO2011074621A1 (en) Mesothelin (msln) antibody and use thereof
JP5616592B2 (en) Antibody against oxidized LDL / β2GPI complex and use thereof
JP5739157B2 (en) Antibodies against calcified globules and uses thereof
EP2044123B1 (en) Tumor-targeting monoclonal antibodies to fzd10 and uses thereof
CA2646329C (en) Engineered anti-prostate stem cell antigen (psca) antibodies for cancer targeting
US9790274B2 (en) Monoclonal antibodies targeting EpCAM for detection of prostate cancer lymph node metastases
KR102512833B1 (en) Novel anti-human MUC1 antibody Fab fragment
CA2800565C (en) Anti-icam-1 single domain antibody and uses thereof
JP2021512071A (en) Nanobody-based imaging and targeting and development of ECM in disease
JP5990523B2 (en) Imaging diagnostic agent containing anti-human transferrin receptor antibody
JP2016534114A (en) CX3CR1 targeted imaging agent and its use in disease diagnosis and treatment
JP2016056165A (en) ANTIBODY FORMULATIONS AND ANTIBODY KITS FOR DIAGNOSIS OR TREATMENT OF CANCER OR TUMOR UTILIZING ANTI-α6β4-INTEGRIN ANTIBODIES
JP2010280568A (en) Diagnostic agent and therapeutic agent for lung cancer, prostate cancer, breast cancer, ovarian cancer and melanoma

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: 10837646

Country of ref document: EP

Kind code of ref document: A1

DPE1 Request for preliminary examination filed after expiration of 19th month from priority date (pct application filed from 20040101)
WWE Wipo information: entry into national phase

Ref document number: 2011546157

Country of ref document: JP

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 10837646

Country of ref document: EP

Kind code of ref document: A1