US20020165363A1 - Cachexia remedy - Google Patents

Cachexia remedy Download PDF

Info

Publication number
US20020165363A1
US20020165363A1 US09/423,800 US42380099A US2002165363A1 US 20020165363 A1 US20020165363 A1 US 20020165363A1 US 42380099 A US42380099 A US 42380099A US 2002165363 A1 US2002165363 A1 US 2002165363A1
Authority
US
United States
Prior art keywords
antibody
ser
dna
gly
chain
Prior art date
Legal status (The legal status 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 status listed.)
Abandoned
Application number
US09/423,800
Other languages
English (en)
Inventor
Koh Sato
Toshiaki Tsunenari
Kimie Ishii
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Chugai Pharmaceutical Co Ltd
Original Assignee
Chugai Pharmaceutical Co Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Chugai Pharmaceutical Co Ltd filed Critical Chugai Pharmaceutical Co Ltd
Assigned to CHUGAI SEIYAKU KABUSHIKI KAISHA reassignment CHUGAI SEIYAKU KABUSHIKI KAISHA ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: ISHII, KIMIE, SATO, KOH, TSUNENARI, TOSHIAKI
Publication of US20020165363A1 publication Critical patent/US20020165363A1/en
Priority to US10/337,981 priority Critical patent/US7468184B2/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/22Hormones
    • A61K38/29Parathyroid hormone, i.e. parathormone; Parathyroid hormone-related peptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/575Hormones
    • C07K14/635Parathyroid hormone, i.e. parathormone; Parathyroid hormone-related peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • 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
    • C07K16/26Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against hormones ; against hormone releasing or inhibiting factors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide

Definitions

  • the present invention relates to a therapeutic agent for cachexia comprising a substance capable of inhibiting the binding between parathyroid hormone related protein (PTHrP) and a receptor thereof as an active ingredient.
  • PTHrP parathyroid hormone related protein
  • Cachexia found in terminal cancer patients is one of the common paraneoplastic syndromes of malignancy, and characterized by systemic disorders with anorexia, weight loss, anemia, electrolyte imbalance and compromised immune function as main symptoms.
  • the development of cachexia in cancer patients leads to fatal and terminal symptoms; impairs the Quality-of-life (QOL) of the patients; and gives strong psychological, physical and social impacts on the patients and their families and surrounding people.
  • QOL Quality-of-life
  • cachectin which is believed to be a causative agent of cancer cachexia, is identical to tumor necrosis factor (TNF).
  • TNF tumor necrosis factor
  • cytokines e.g., interleukin(IL)-1, IL-6, LIF, IFN
  • OCC-1 cell line derived from human oral cavity carcinoma produces various types of liquid factors involved in cancer cachexia.
  • a nude mouse implanted with OCC-1 cells comes to develop various syndromes including cachexia (Kajimura N. et al., Cancer Chemother. Pharmacol., 1996, 38 Suppl. pS48-52; Tanaka R. et al., Jpn. J. Clin. Oncology April 1996, 26 (2) p88-94).
  • OCC-1 cell line implanted into the nude mouse produces various cytokines (e.g., G-CSF, IL-6, LIF, IL-11, PTHrP) with the growth of the cells, and these factors act compositely in the nude mouse to cause such symptoms.
  • cytokines e.g., G-CSF, IL-6, LIF, IL-11, PTHrP
  • the object of the present invention is to provide a therapeutic agent for cachexia comprising, as an active ingredient, a substance capable of inhibiting the binding between parathyroid hormone related protein (PTHrP) and a receptor thereof.
  • PTHrP parathyroid hormone related protein
  • the present inventors have made extensive and intensive studies on discovering such therapeutic agent. As a result, they found that development of a substance that can inhibit the binding between PTHrP and a receptor thereof could achieve such object. This finding leads the accomplishment of the invention.
  • the present invention relates to a therapeutic agent for cachexia comprising, as an active ingredient, a substance capable of inhibiting the binding between PTHrP and a receptor thereof.
  • the term “cachexia” encompasses those induced by cancer.
  • the present invention relates to a therapeutic agent for cachexia comprising, as an active ingredient, a substance capable of inhibiting the binding between parathyroid hormone related protein (hereinafter, referred to as “PTHrP”) and a receptor thereof (hereinafter, referred to as “PTHrP receptor”).
  • PTHrP parathyroid hormone related protein
  • PTHrP receptor a receptor thereof
  • PTHrP receptor refers to any receptor which binds to PTHrP (such as those as described in Japanese National Phase Laid-open Publication No. 6-506598), regardless of whether the PTHrP receptor is present on a target organ (e.g., bone, kidney) or not.
  • a substance capable of inhibiting the binding between PTHrP and a receptor thereof refers to any substance that can bind to PTHrP to prevent the binding of the PTHrP to a PTHrP receptor, such as an anti-PTHrP antibody; any substance that can bind to a PTHrP receptor to prevent the binding of the PTHrP receptor to PTHrP, such as an antagonist against a PTHrP receptor (a PTHrP antagonist), specifically a peptide having replacement or deletion of at least one amino acid residue in the PTHrP peptide or a partial sequence of the PTHrP peptide; or a combination thereof.
  • the anti-PTHrP antibody includes those of any known types, such as a humanized antibody, a human antibody (WO 96/33735) or a chimeric antibody (Japanese Patent Application Laid-open No. 4-228089), and the antibody exemplary used in the present invention (#23-57-137-1 antibody).
  • the antibody may be of polyclonal type or monoclonal type, but preferably of monoclonal type.
  • the PTHrP antagonist includes a polypeptide or a low molecular weight substance.
  • the PTHrP antagonist includes a substance that binds to a PTHrP receptor in an antagonistic manner against PTHrP, such as a polypeptide having a PTHrP antagonistic activity as described in Japanese Patent Application Laid-open No. 7-165790; Peptides (UNITED STATES), 1995, 16(6) 1031-1037; Biochemistry (UNITED STATES) Apr. 28 1992, 31(16) 4026-4033; and Japanese National Phase Laid-open No. 5-509098. These polypeptides may have deletion, replacement, addition or insertion of at least one amino acid residue, as long as they can exhibit an equivalent level of PTHrP antagonistic activity, which are also encompassed in the PTHrP antagonists of the present invention.
  • the anti-PTHrP antibody used in the present invention may be any one as long as it can exhibit a therapeutic effect on cachexia, regardless of its source, type (monoclonal or polyclonal) and configuration.
  • the anti-PTHrP antibody used in the present invention can be produced by any known method as a polyclonal or monoclonal antibody.
  • the anti-PTHrP antibody is a monoclonal antibody derived from a mammal.
  • the monoclonal antibody from a mammal includes those produced from a hybridoma and those produced by a genetic engineering technique from a host transformed with a recombinant expression vector carrying a gene for the antibody.
  • the antibody used in the present invention is one that can bind to PTHrP to prevent the binding of the PTHrP to a PTH/PTHrP receptor, thus blocking the signal transduction of the PTHrP and consequently inhibiting the biological activity of PTHrP.
  • a specific example of such antibody is #23-57-137-1 antibody which can be produced with a hybridoma clone #23-57-137-1.
  • hybridoma clone #23-57-137-1 has been designated “mouse-mouse hybrodima #23-57-137-1” and deposited under the terms of the Budapest Treaty on Aug. 15, 1996 at the National Institute of Bioscience and Human-technology, Agency of Industrial Science and Technology, Japan (1-3, Higashi 1-chome, Tsukuba-shi, Ibaraki, Japan) under the accession No. FERM BP-5631.
  • a monoclonal antibody-producing hybridoma can basically be produced by any known technique. That is, PTHrP is used as an antigen for immunization in accordance with a conventional immunization method.
  • the resultant immunocytes are fused to known parent cells by a conventional cell fusion method, and monoclonal antibody-producing cells are screened from the fused cells by a conventional screening method.
  • the monoclonal antibody-producing cell can be prepared as follows.
  • a human PTHrP which is used as an sensitizing antigen for producing the antibody, is prepared by expressing the PTHrP gene/amino acid sequence disclosed in Suva, L. J. et al., Science (1987) 237, 893. That is, a nucleotide sequence encoding the PTHrP is inserted into any known expression vector, and a suitable host cell is transformed with the expression vector. The PTHrP protein is then isolated and purified from the transformed host cell or from a culture supernatant of the transformed host cell by any known method.
  • the purified PTHrP protein is used as a sensitizing antigen.
  • a 34-amino acid peptide of the N-terminal region of the PTHrP may be used as a sensitizing antigen, which can be chemically synthesized.
  • the mammal to be immunized with the sensitizing antigen is not particularly limited. However, the mammal is preferably selected taking into consideration of compatibility with the parent cell used for cell fusion. Generally, a rodent (e.g., mouse, rat, hamster, rabbit) or monkey may be used.
  • a rodent e.g., mouse, rat, hamster, rabbit
  • monkey may be used.
  • the immunization of the mammal with the sensitizing antigen can be performed in accordance with any known method, for example, by injecting the sensitizing antigen to a mammal intraperitoneally or subcutaneously. More specifically, the sensitizing antigen is diluted with and suspended to phosphate-buffered saline (PBS) or normal saline properly, the resultant suspension is then mixed with an appropriate amount of an adjuvant (e.g., Freund's complete adjuvant) to give an emulsion. The emulsion is injected to a mammal several times at intervals of 4 to 21 days. In the immunization, the sensitizing antigen may be attached to a suitable carrier.
  • PBS phosphate-buffered saline
  • an adjuvant e.g., Freund's complete adjuvant
  • the sensitizing antigen may be attached to a suitable carrier.
  • the serum antibody level is checked.
  • immunocytes are isolated from the mammal and then subjected to cell fusion.
  • a preferable immunocyte is a spleen cell.
  • the parent cell used for the cell fusion is a myeloma cell derived from a mammal.
  • the myeloma cell is of any known cell line, and, for example, P3 (P3x63Ag8.653) (J. Immunol. (1979) 123, 1548-1550), P3x63Ag8U.1 (Current Topics in Microbiology and Immunology (1978) 81, 1-7), NS-1 (Kohler, G. and Milstein, C. Eur. J. Immunol. (1976) 6, 511-519), MPC-11 (Margulies, D. H.
  • Cell fusion of the immunocyte to the myeloma cell is basically performed in accordance with any known method such as the method of Milstein et al. (Kohler, G. and Milstein, C., Methods Enzymol. (1981) 73, 3-46) may be preferably used.
  • the cell fusion is performed, for example, in a conventional nutrient culture medium in the presence of a cell fusion promoter.
  • the cell fusion promoter may be polyethylene glycol (PEG) or a Sendai virus (hemagglutinating virus of Japan; HVJ). If desired, for the purpose of improving the fusion efficiency, an additive such as dimethyl sulfoxide may also be incorporated.
  • the ratio between the immunocytes and the myeloma cells for the cell fusion may be any one.
  • the immunocytes are used in the amount 1-10 times larger than the myeloma cells.
  • the culture medium used for the cell fusion is, for example, RPMI 1640 medium or MEM medium suitable for the growth of the myeloma cell line, or other medium conventionally used for the culture of such cells.
  • a serum supplement such as feral calf serum (FCS) may be added to the culture medium.
  • the cell fusion is performed by well mixing the immunocytes and the myeloma cells of given amounts in the culture medium, adding PEG solution (e.g., mean molecular weight: about 1000-6000) (which has been previously warmed to about 37° C.) thereto usually to a concentration of 30-60% (w/v), and then mixing the resultant solution, thereby giving fusion cells (hybridomas). Subsequently, an appropriate culture medium is added to the culture solution, and centrifuged to remove the supernatant. This procedure is repeated several times to remove the cell fusion promoter or the like that are undesirable for the growth of the hybridomas from the culture medium.
  • PEG solution e.g., mean molecular weight: about 1000-6000
  • the obtained hybridomas can be selected by cultivating in a conventional selective medium, such as hypoxanthine-aminopterin-thymidine (HAT) medium.
  • a conventional selective medium such as hypoxanthine-aminopterin-thymidine (HAT) medium.
  • HAT hypoxanthine-aminopterin-thymidine
  • the cultivation of the hybridomas in HAT medium is performed for the time of period enough to cause to death of the cells other than the desired hybridomas (i.e., cells that fail to fuse), usually for several days to several weeks.
  • a conventional limiting dilution method is performed to screen and mono-clone the hybridomas that are secreting the desired antibody.
  • a human antibody having a binding activity against the PTHrP may be prepared by sensitizing a human lymphocyte with PTHrP in vitro, and then subjecting the sensitized lymphocyte to cell fusion to a human-derived myeloma cell capable of infinite growth (Japanese Patent Publication No. 1-59878).
  • a human antibody against PTHrP may be prepared by injecting PTHrP as an antigen to a transgenic animal that has the entire repertories of the human antibody genes to give an anti-PTHrP antibody-producing cell, and immortalizing the cells, thus the human antibody can be produced from the immortalized cell (International Publication Nos. WO 94/25585, WO 93/12227, WO 92/03918 and WO 94/02602).
  • the monoclonal antibody-producing hybridoma prepared as above can be subcultured in a conventional culture medium and stored under liquid nitrogen for a long time of period.
  • a method involving cultivating the hybridoma in accordance with a conventional method and collecting the monoclonal antibody from the culture supernatant, or a method involving injecting the hybridoma to a mammal compatible with the hybridoma to grow the hybridoma in the mammal body and collecting the hybridoma from the ascites of the mammal may be employed.
  • the former method is suitable for producing the antibody in high purity, while the latter method is suitable for producing the antibody in a large amount.
  • a recombinant-type monoclonal antibody may also be used, which can be produced by cloning an antibody gene from the hybridoma, integrating the antibody gene into a suitable vector, introducing the vector into a host, and producing the antibody from the host according to a conventional genetic recombination technique (see, for example, Vandamme, A. M. et al., Eur. J. Biochem. (1990) 192, 767-775) More specifically, mRNA encoding variable (V) region of an anti-PTHrP antibody is isolated from the anti-PTHrP antibody-producing hybridoma.
  • the isolation of the mRNA is performed by preparing a total RNA by any known method, such as guanidium ultracentrifugation method (Chirgwin, J. M. et al., Biochemistry (1979) 18, 5294-5299) and AGPC method (Chomczynski, P. et al., Anal. Biochem. (1987) 162, 156-159), and then producing the desired mRNA from the total RNA using mRNA Purification Kit (Pharmacia) or the like.
  • the mRNA may also be prepared directly using QuickPrep mRNA Purification Kit (Pharmacia).
  • cDNA for the antibody V-region is synthesized from the mRNA with a reverse transcriptase.
  • the synthesis of the cDNA is performed using AMV Reverse Transcriptase First-strand cDNA Synthesis Kit (Seikagaku Corporation) or the like.
  • the cDNA may also be synthesized or amplified by 5′-RACE method (Frohman, M. A. et al., Proc. Natl. Acad. Sci. USA (1988) 85, 8998-9002; Belyavsky, A. et al., Nucleic Acids Res. (1989) 17, 2919-2932) using 5′-Ampli FINDER RACE Kit (Clontech) in combination with a PCR method, or the like.
  • a DNA fragment of interest is isolated and purified from the resultant PCR product and then ligated to a vector DNA to give a recombinant vector.
  • the recombinant vector is introduced into a host such as E. coli, and a colony containing a desired recombinant vector is selected.
  • the nucleotide sequence of the DNA of interest in the recombinant vector is confirmed by, for example, dideoxynucleotide chain termination method.
  • the antibody gene is integrated into an expression vector so that the antibody gene can be expressed under the control of expression control regions (e.g., enhancer, promoter).
  • expression control regions e.g., enhancer, promoter.
  • DNA encoding heavy (H) chain and DNA encoding light (L) chain of the antibody may be integrated into separate expression vectors, and then a host cell is co-transformed with the resultant recombinant expression vectors.
  • both DNA encoding H-chain and DNA encoding L-chain of the antibody may be integrated together into a single expression vector, and then a host cell is transformed with the resultant recombinant expression vector (WO 94/11523).
  • a transgenic animal may also be used as a host.
  • the antibody gene is inserted into a predetermined site of a gene encoding a protein inherently produced in the milk of an animal (e.g., goat ⁇ -casein) to give a fusion gene.
  • a DNA fragment containing the antibody gene-introduced fusion gene is injected into an embryo of a goat, and the embryo is then introduced into a female goat.
  • the female goat having the embryo therein bears a transgenic goat.
  • the antibody of interest is secreted in the milk from the transgenic goat or a progeny thereof.
  • an appropriate hormone may be administered to the transgenic goat (Ebert, K. M. et al., Bio/Technology (1994) 12, 699-702).
  • an artificially modified recombinant antibody for the purpose of reducing the heterogenisity against a human body or the like, an artificially modified recombinant antibody may be used, including a chimeric antibody and a humanized antibody. These modified antibodies can be prepared by any known method.
  • a chimeric antibody usable in the present invention can be prepared by ligating the DNA encoding the antibody V-region prepared as mentioned above to DNA encoding a human antibody C-region, integrating the ligation product into an expression vector, and introducing the resultant recombinant expression vector into a host to produce the chimeric antibody.
  • a humanized antibody is also referred to as “reshaped human antibody”, in which the complementarity determining regions (CDRs) of an antibody of a non-human mammal (e.g., a mouse) are grafted to those of a human antibody.
  • CDRs complementarity determining regions
  • the general genetic recombination procedure for producing such humanized antibody is also known (EP 125023; WO 96/02576).
  • a DNA sequence in which mouse antibody CDRs are ligated through framework regions (FRs) is designed, and synthesized by a PCR method using several oligonucleotides as primers which were designed to have regions overlapping to the terminal regions of the CDRs and the FRs.
  • the resultant DNA is ligated to DNA encoding the human antibody C-region, and the ligation product is integrated into an expression vector.
  • the resultant recombinant expression vector is introduced into a host, thereby producing the humanized antibody (EP 239044, WO 96/02576).
  • the FRs ligated through the CDRs are selected so that the CDRs can form a satisfactory antigen binding site. If necessary, an amino acid(s) in the FRs of the antibody V-region may be replaced so that the CDRs of the reshaped human antibody can form an appropriate antigen binding site (Sato, K. et al., Cancer Res. (1993) 53, 851-856).
  • the C-region of the chimeric or humanized antibody may be any human antibody C-region; such as C ⁇ 1, C ⁇ 2, C ⁇ 3 or C ⁇ 4 for the H-chain, and C ⁇ or C ⁇ for the L-chain.
  • the human antibody C-region may be modified for the purpose of improving the stability of the antibody or ensuring the stable production of the antibody.
  • the chimeric antibody is composed of V-regions derived from a non-human mammal antibody and C-regions derived from a human antibody.
  • the humanized antibody is composed of CDRs derived from a non-human mammal antibody and FRs and C-regions derived from a human antibody.
  • the humanized antibody is especially useful as an active ingredient for the therapeutic agent of the present invention, because the antigenicity of the antibody against a human body is reduced.
  • a specific example of the humanized antibody used in the present invention is humanized #23-57-137-1 antibody; in which the CDRs are derived from mouse-derived #23-57-137-1 antibody; and the L-chain is composed of the CDRs ligated through three FRs (FR1, FR2 and FR3) derived from human antibody HSU 03868 (GEN-BANK, Deftos, M. et al., Scand. J. Immunol., 39, 95-103, 1994) and a FR (FR4) derived from human antibody S25755 (NBRF-PDB); and the H-chain is composed of the CDRs ligated through FRs derived from human antibody S31679 (NBRF-PDB, Cuisinier, A. M. et al., Eur. J. Immunol. 23, 110-118, 1993) in which a portion of the amino acid residues in the FRs is replaced so that the reshaped humanized antibody can exhibit an antigen-binding activity.
  • the E. coli strains containing the plasmids having DNA encoding the H-chain and the L-chain of the humanized #23-57-137-1 antibody, respectively, are designated Escherichia coli JM109 (hMBC1HcDNA/PUC19) (for H-chain) and Escherichia coli JM109 (hMBC1Lq ⁇ /pUC19) (for L-chain), respectively.
  • Escherichia coli JM109 hMBC1HcDNA/PUC19
  • hMBC1Lq ⁇ /pUC19 for L-chain
  • FERM BP-5629 for Escherichia coli JM109 (hMBC1HcDNA/PUC19), and under the accession No. FERM BP-5630 for Escherichia coli JM109 (hMBC1Lq ⁇ /pUC19).
  • the antibody used in the present invention may be any fragment thereof or a modified product of the fragment, as long as it can bind to PTHrP and inhibit the activity of the PTHrP.
  • the fragment of the antibody includes Fab, F(ab′) 2 , Fv, or a single chain Fv (scFv) composed of a H-chain Fv fragment or a L-chain Fv fragment linked together through a suitable linker.
  • such antibody fragments can be produced by cleaving the antibody with an enzyme (e.g., papain, pepsin) into antibody fragments, or by constructing a gene encoding the antibody fragment and inserting the gene into an expression vector and introducing the resultant recombinant expression vector into a suitable host cell, thereby expressing the antibody fragment (see, for example, Co, M. S., et al., J. Immunol. (1994), 152, 2968-2976; Better, M. & Horwitz, A. H., Methods in Enzymology (1989), 178, 476-496, Academic Press, Inc.; Plueckthun, A.
  • an enzyme e.g., papain, pepsin
  • a scFv can be produced by ligating the H-chain V-region to the L-chain V-region through a linker, preferably a peptide linker (Huston, J. S. et al., Proc. Natl. Acad. Sci. USA (1988) 85, 5879-5883).
  • the H-chain V-region and the L-chain V-region in the scFv may be derived from any one of the antibodies described herein.
  • the peptide linker which binds the V-regions may be any single chain peptide, for example, of 12-19 amino acid residues.
  • the DNA encoding the scFv can be prepared by first amplifying the DNA encoding the H-chain V-region and the DNA encoding the L-chain V-region of the antibody separately using a DNA fragment encoding the entire region of the H-chain or a portion thereof that includes the V-region and a DNA fragment encoding the entire region of the L-chain or a portion thereof that includes the V-region as templates and primer pairs that define the terminal ends of the DNA fragments; and then amplifying the DNA encoding the peptide linker using a DNA fragment encoding the peptide linker as a template and a primer pair that define the terminal ends of the DNA fragment so that each terminal end of the peptide linker is ligated to the H-chain V-region and the L-chain V-region, respectively.
  • an expression vector carrying the DNA and a host transformed with the expression vector can be prepared by conventional methods.
  • the scFv can be produced from the transformed host in any conventional method.
  • the antibody fragments used in the present invention may be produced by preparing genes for the fragments and expressing the genes in suitable hosts as described above. These antibody fragments are also encompassed in the “antibody” of the present invention.
  • anti-PTHrP antibody conjugated to any molecule may also be used.
  • modified antibodies are also encompassed in the “antibody” of the present invention.
  • the modified antibodies can be prepared by chemical modifications of the antibodies. The chemical modification techniques suitable for this purpose have already been established in the art.
  • the antibody gene constructed as described above can be produced and expressed by known methods.
  • a conventional useful promoter for the expression in a mammalian cell, a conventional useful promoter, the antibody gene to be expressed and a poly(A) signal (located downstream to the 3′ end of the antibody gene) are operably linked.
  • a useful promoter/enhancer system a human cytomegalovirus immediate early promoter/enhancer system may be used.
  • promoter/enhancer systems for example, those derived from viruses (e.g., retrovirus, polyoma virus, adenovirus and simian virus 40 (Sv40)) and those derived from mammalian cells (e.g., human elongation factor 1 ⁇ (HEF1 ⁇ )), may also be used for the expression of the antibody in the present invention.
  • viruses e.g., retrovirus, polyoma virus, adenovirus and simian virus 40 (Sv40)
  • mammalian cells e.g., human elongation factor 1 ⁇ (HEF1 ⁇ )
  • HEF1 ⁇ human elongation factor 1 ⁇
  • the gene expression may be performed readily by the method of Mulligan et al. (Nature (1979) 277, 108).
  • HEF1 ⁇ promoter/enhancer system the gene expression may be performed readily by the method of Mizushima et al. (Nucleic Acids Res. (1990) 18, 5322).
  • a signal sequence for secreting the antibody of interest and the antibody gene may be operably linked.
  • lacZ promoter or araB promoter may be used.
  • the gene expression may be performed by the method of Ward et al. (Nature (1098) 341, 544-546; FASBE J. (1992) 6, 2422-2427), while when araB promoter is used, the gene expression may be performed by the method of Better et al. (Better et al., Science (1988) 240, 1041-1043).
  • pe1B signal sequence (Lei, S. P. et al., J. Bacteriol. (1987) 169, 4379) may be used.
  • the antibody secreted into the periplasmic space is isolated and then refolded so that the antibody takes an appropriate configuration.
  • the replication origin derived from viruses e.g., SV40, polyoma virus, adenovirus, bovine papilloma virus (BPV)
  • viruses e.g., SV40, polyoma virus, adenovirus, bovine papilloma virus (BPV)
  • the expression vector may further contain a selective marker gene, such as an aminoglycoside phosphotranferase (APH) gene, a thymidine kinase (TK) gene, an E. coli xanthine-guanine phosphoribosyltransferase (Ecogpt) gene and a dihydrofolate reductase (dhfr) gene.
  • APH aminoglycoside phosphotranferase
  • TK thymidine kinase
  • Ecogpt E. coli xanthine-guanine phosphoribosyltransferase
  • any expression system including eukaryotic and prokaryotic cell systems may be used.
  • the eukaryotic cell includes established cell lines of animals (e.g., mammals, insects, molds and fungi, yeast).
  • the prokaryotic cell includes bacterial cells such as E. coli cells.
  • the antibody used in the present invention be expressed in a mammalian cell, such as a CHO, COS, myeloma, BHK, Vero and HeLa cell.
  • a mammalian cell such as a CHO, COS, myeloma, BHK, Vero and HeLa cell.
  • the transformed host cell is cultured in vitro or in vivo to produce the antibody of interest.
  • the cultivation of the host cell may be performed by any known method.
  • the culture medium usable herein may be DMEM, MEM, RPMI 1640 or IMDM medium.
  • the culture medium may contain a serum supplement, such as fetal calf serum (FCS).
  • the antibody expressed and produced as described above may be isolated from the cells or the host animal body and purified to uniformity.
  • the isolation and purification of the antibody used in the present invention may be performed on an affinity column.
  • a protein A column include Hyper D, POROS and Sepharose F.F. (Pharmacia).
  • Other methods conventionally used for the isolation and purification of an antibody may be also be used; thus the method is not particularly limited.
  • various chromatographs using columns including the above-mentioned affinity column, filtration, ultrafiltration, salting out and dialysis may be used singly or in combination to isolate and purify the antibody of interest (Antibodies A Laboratory Manual. Ed. Harlow, David Lane, Cold Spring Harbor Laboratory, 1988).
  • ELISA enzyme-linked immunosorbent assay
  • EIA enzyme immunoassay
  • RIA radioimmunoassay
  • fluorescent antibody technique for example, when enzyme immunoassay is employed, a sample solution containing the anti-PTHrP antibody (e.g., a culture supernatant of anti-PTHrP antibody-producing cells, or the anti-PTHrP antibody per se in a purified form) is added to a plate on which PTHrP (1-34) is previously coated.
  • a secondary antibody labeled with an enzyme e.g., alkaline phosphatease
  • an enzyme e.g., alkaline phosphatease
  • a substrate for the enzyme e.g., p-nitrophenylphosphoric acid
  • the absorbance of the solution in the plate is measured to evaluate the antigen-binding activity of the antibody.
  • a neutralizing activity of the antibody e.g., anti-PTHrP antibody
  • the therapeutic agent of the present invention can be used for treatment or amelioration of cachexia.
  • the cachexia to be treated or ameliorated by the present invention may be of any type, including cancer-induced type.
  • Examples of the cancer-induced cachexia include those as described in J. Urol. (UNITED STATES) March 1995, 153 (3 Pt 1) p.854-857; Langenbecks Arch. Chir. Suppl II Verh Dtsch Ges Chir (GERMANY) 1990, p.261-265; Oncology (SWITZERLAND) 1990, 47 (1) p.87-91; Int. J. Pancreatol. (UNITED STATES) August-November 1990, 7 (1-3) p.141-150; J. Natl. Cancer Inst. (UNITED STATES) Dec. 19, 1990, 82 (24) p.1922-1926.
  • Examples of cachexia other than the cancer-induced cachexia include those as described in JPEN J. Parenter. Enteral Nutr. (UNITED STATES) November-December 1990, 14 (6) p.605-609; Chest (UNITED STATED) November 1990, 98 (5) p.1091-1094; Bone Marrow Transplant. (ENGLAND) July 1990, 6 (1) p.53-57.
  • the therapeutic agent comprising the anti-PTHrP antibody as an active ingredient of the present invention may be administered orally or parenterally, but preferably parenterally.
  • the therapeutic agent may take any dosage form, such as a transpulmonary agent (e.g., an agent administered with the help of a device such as a nebulizer), a nasogastric agent, a transdermic agent (e.g., ointment, cream) or an injection.
  • a transpulmonary agent e.g., an agent administered with the help of a device such as a nebulizer
  • a nasogastric agent e.g., ointment, cream
  • transdermic agent e.g., ointment, cream
  • injection include an intervenous injection (e.g., drops), an intramuscular injection, an intraperitoneal injection and a subcutaneous injection for systemic or topical administration.
  • the route of administration may be properly selected depending on the age of a patient and
  • An effective single dose may be selected from the range of 0.001 to 1,000 mg per kg of body weight.
  • the dose to a patient may be selected from the range of 0.01 to 100,000 mg/body.
  • the dose of the therapeutic agent comprising the anti-PTHrP antibody of the present invention is not particularly limited to the above-mentioned ranges.
  • the therapeutic agent may be administered to a patient at any stage, including before or after the development of cachexia. Alternatively, the therapeutic agent may be administered at the stage where the development of weight loss is predicted in the patient.
  • the therapeutic agent comprising the anti-PTHrP antibody as an active ingredient of the present invention may be formulated by any conventional method (Remington's Pharmaceutical Science, latest edition, Mark Publishing Company, Easton, USA).
  • the formulation may further comprise pharmaceutically acceptable carriers and additives.
  • Examples of such carriers and additives include water, pharmaceutically acceptable organic solvents, collagen, polyvinyl alcohol, polyvinyl pyrrolidone, carboxyvinyl polymer, sodium carboxymethyl cellulose, poly(sodium acrylate), sodium arginate, water soluble dextran, sodium carboxymethyl starch, pectin, methyl cellulose, ethyl cellulose, xanthane gum, gum arabic, casein, agar, polyethylene glycol, diglycerin, glycerin, propylene glycol, vaseline, paraffin, stearyl alcohol, stearic acid, human serum albumin (HSA), mannitol, sorbitol, lactose, and surfactants acceptable as pharmaceutical additives.
  • water pharmaceutically acceptable organic solvents
  • collagen polyvinyl alcohol, polyvinyl pyrrolidone
  • carboxyvinyl polymer sodium carboxymethyl cellulose, poly(sodium acrylate), sodium arginate
  • the additive is properly selected from the above members either singly or in combination depending on the dosage form employed, but not limited thereto.
  • an injection may be used which is prepared by dissolving the anti-PTHrP antibody in a purified form into a solvent (e.g., normal saline, a buffer, a grape sugar solution) and then further adding an adsorption-preventing agent (e.g., Tween 80, Tween 20, a gelatin, human serum albumin) thereto.
  • a solvent e.g., normal saline, a buffer, a grape sugar solution
  • an adsorption-preventing agent e.g., Tween 80, Tween 20, a gelatin, human serum albumin
  • the therapeutic agent of the present invention may also be in a re-constitutive, freeze-dried form, which is dissolved before use.
  • an excipient such as a sugar alcohol (e.g., mannitol, grape sugar) and a sugar may be incorporated.
  • FIG. 1 is a graphical illustration of the therapeutic effect of an anti-PTHrP antibody on cachexia.
  • FIG. 2 is a graphical illustration of the therapeutic effect of an anti-PTHrP antibody on cachexia.
  • FIG. 3 is a graphical illustration of the therapeutic effect of an anti-PTHrP antibody on cachexia.
  • FIG. 4 is a graphical illustration of the therapeutic effect of an anti-PTHrP antibody on cachexia.
  • FIG. 5 is a graphical illustration of the measurement results of the antigen-binding activity.
  • FIG. 6 is a graphical illustration of the measurement results of the antigen-binding activity.
  • FIG. 7 is a graphical illustration of the measurement results of the antigen-binding activity.
  • FIG. 8 is a graphical illustration of the measurement results of the antigen-binding activity.
  • FIG. 9 is a graphical illustration of the measurement results of the antigen-binding activity.
  • FIG. 10 is a graphical illustration of the measurement results of the antigen-binding activity.
  • FIG. 11 is a graphical illustration of the measurement results of the antigen-binding activity.
  • FIG. 12 is a graphical illustration of the measurement results of the antigen-binding activity.
  • FIG. 13 is a graphical illustration of the neutralizing activity of a humanized antibody.
  • FIG. 14 is a graphical illustration of the neutralizing activity of a humanized antibody.
  • FIG. 15 is a graphical illustration of the neutralizing activity of a humanized antibody.
  • FIG. 16 is a graphical illustration of the therapeutic effect of a humanized antibody on cachexia.
  • FIG. 17 is a graphical illustration of the therapeutic effect of a humanized antibody on cachexia.
  • FIG. 18 is a graphical illustration of the therapeutic effect of a humanized antibody on cachexia.
  • FIG. 19 is a graphical illustration of the therapeutic effect of a humanized antibody on cachexia.
  • a nude mouse implanted with human oral cavity carcinoma OCC-1 [purchased from the Central Institute for Experimental Animals] was used. It has been known that a nude mouse implanted with human oral cavity carcinoma OCC-1 exhibits an increased blood calcium level as increasing the tumor volume and develops cachexia symptoms such as weight loss and decrease in movements. In this test, amelioration of such human oral cavity carcinoma OCC-1-induced cachexia symptoms by the murine monoclonal antibody was evaluated with respect to blood calcium level, body weight and effect on prolongation of survival time.
  • the human oral cavity carcinoma OCC-1 was passaged in vivo using BALB/c-nu/nu nude mice (Japan CLEA Co., Inc.).
  • BALB/c-nu/nu nude mice Japan CLEA Co., Inc.
  • 6-weeks-old male BALB/c-nu/nu nude mice (Japan CLEA Co., Inc.) were purchased and acclimatized for 1 week to give 7-weeks-old mice, which were provided for use in the evaluation.
  • the cachexia model mice were prepared and divided into groups in the following manner.
  • the passaged human oral cavity carcinoma OCC-1 was removed from the nude mouse, and then finely cut into 3-mm cube of blocks.
  • the resultant tumor blocks were subcutaneously implanted into each of the mice at the lateral region at one piece per mouse.
  • a mouse monoclonal antibody was administered to the mice of a test group twice a week, and the survival time of each of the mice was observed.
  • a single dose of an existing hypercalcemia-treating agent, pamidronate (pamidronate disodium; Aredia) was administered to the mice of another test group via tail vein at a dose amount of 15 mg/kg.
  • pamidronate pamidronate disodium
  • PBS phosphate-buffered saline
  • the mouse monoclonal antibody against PTHrP was administered to the cachexia model mice of a test group twice at intervals of two days via tail vein at a dose amount of either 10 ⁇ g or 100 ⁇ g per mouse for each administration.
  • a single dose of an existing hypercalcemia-treating agent, pamidronate (pamidronate disodium; Aredia) was administered to the mice of another test group via tail vein at a dose amount of 15 mg/kg.
  • phosphate-buffered saline (PBS) was administered to the mice of a control group via tail vein twice at intervals of two days at a dose amount of 0.2 ml/mouse for each administration.
  • the blood calcium level of each of the mice was determined to evaluate the pharmacological efficacy of the antibody.
  • the blood calcium level was determined as whole blood ionized calcium level, by drawing blood from each of the mice via the orbit using a hematocrit tube and applying the blood to 643 Automatic Ca/pH Analyzer (CIBA-CORNING). The body weight of each mouse was weighed everyday till four days after the administration of the antibody. The results are shown in FIGS. 2 and 3.
  • the tumor volume was determined four days after the administration of the antibody, by measuring the longest axis (a mm) and the shortest axis (b mm) of the tumor and applying the both measured values to Galant's equation [ab 2 /2]. The results are shown in FIG. 4.
  • mice administered with the antibody at a dose amount of 10 ⁇ g showed blood calcium levels equivalent to those of the mice administered with pamidronate, weight loss in antibody-administered mice was observed to be inhibited, as weight loss was not as pronounced as that in pamidronate-administered mice.
  • the mice administered with the antibody at a dose amount of 100 ⁇ g prevented the increase in blood calcium level and inhibited weight loss to a higher degree, when compared to pamidronate-administered mice and the control mice.
  • the neutralizing mouse monoclonal antibody against PTHrP has excellent effects that any existing hypercalcemia-treating agents cannot exhibit, such as prevention of weight loss and prolongation of survival time.
  • a nude mouse implanted with human oral cavity carcinoma OCC-1 [purchased from the Central Institute for Experimental Animals] was used. It has been known that a nude mouse implanted with human oral cavity carcinoma OCC-1 exhibits an increased blood calcium level as increasing the tumor volume and develops cachexia symptoms such as weight loss and decrease in movements. In this test, improvement of such human oral cavity carcinoma OCC-1-induced cachexia symptoms by the humanized antibody version “q” was evaluated with respect to blood calcium level, body weight and effect on prolongation of survival time.
  • the cachexia model mice were prepared and divided into groups in the following manner.
  • the passaged human oral cavity carcinoma OCC-1 was removed from the nude mouse, and then finely cut into 3-mm cube of blocks.
  • the resultant tumor blocks were subcutaneously implanted into each of the mice at the lateral region at one piece per mouse.
  • the humanized antibody version “q” was administered to the cachexia model mice of a test group twice at intervals of two days via tail vein at a dose amount of either 10 ⁇ g or 100 ⁇ g per mouse for each administration.
  • phosphate-buffered saline (PBS) was administered to the mice of a control group via tail vein twice at intervals of two days at a dose amount of 0.1 ml/mouse for each administration.
  • the blood calcium level of each of the mice was determined to evaluate the pharmacological efficacy of the antibody.
  • the blood calcium level was determined as whole blood ionized calcium level, by drawing blood from each of the mice via the orbit using a hematocrit tube and applying the blood to 643 Automatic Ca/pH Analyzer (CIBA-CORNING). The body weight of each mouse was weighed everyday till four days after the administration of the antibody. The results are shown in FIGS. 17 and 18.
  • the tumor volume was determined four days after the first administration of the antibody, by measuring the longest axis (a mm) and the shortest axis (b mm) of the tumor and applying the both measured values to Galant's equation [ab 2 /2]. The results are shown in FIG. 19.
  • mice administered with the humanized antibody version “q” at a dose amount of either 10 ⁇ g or 100 ⁇ g prevented the increase in blood calcium level and weight loss in antibody-administered mice was observed to be inhibited, as weight loss was not as pronounced as that in control mice.
  • the efficacy of the humanized antibody version “q” on the model animals with malignancy-associated cachexia was similar to that of the above-tested mouse monoclonal antibody.
  • Hybridomas capable of producing a monoclonal antibody against human PTHrP (1-34) (SEQ ID NO: 75), #23-57-154 and #23-57-137-1, were prepared in accordance with the method reported by Kanji Sato et al. (Sato, K. et al., J. Bone Miner. Res. 8, 849-860, 1993).
  • the immunogen used was PTHrP (1-34) (Peninsula), to which a carrier protein thyroglobulin was conjugated with carbodiimide (Dojinn).
  • the thycloglobulin-conjugated PTHrP (1-34) was dialyzed to obtain a solution having a protein concentration of 2 ⁇ g/ml.
  • the resultant solution was mixed with Freund's adjuvant (Difco) at a mixing ratio of 1:1 to give an emulsion.
  • This emulsion was injected to 16 female BALB/C mice 11 times dorsal-subcutaneously or intraperitoneally at a dose amount of 100 ⁇ g/mouse for each injection, thereby immunizing the mice.
  • Freund's complete adjuvant was used for the priming immunization; while for the boosting immunization, Freund's incomplete adjuvant was used.
  • each of the immunized mice was determined for its antibody titer in the serum in the following manner. That is, each of the mice was blood-drawn via its tail vein, and the anti-serum is separated from the blood. The anti-serum was diluted with a RIA buffer and mixed with 125 I-labeled PTHrP (1-34) to determine the binding activity. The mice that were confirmed to have a sufficiently increased titer were injected with PTHrP (1-34) without a carrier protein intraperitoneally at a dose amount of 50 ⁇ g/mouse for the final immunization.
  • mice Three days after the final immunization, the mouse is sacrificed and the spleen was removed therefrom.
  • the spleen cells were subjected to cell fusion with mouse myeloma cell line P3x63Ag8U.1 in accordance with any conventional known method using 50% polyethylene glycol 4000.
  • the fused cells thus prepared were seeded to each well of 85 of 96-well plates at 2 ⁇ 10 4 /well.
  • Hybridomas were screened in HAT medium as follows.
  • hybridomas were obtained by determining the presence of PTHrP-recognition antibodies in the culture supernatant of the wells in which cell growth had been observed in HAT medium, by a solid phase RIA method.
  • the hybridomas were collected from the wells in which the binding ability to the PTHrP-recognition antibodies had been confirmed.
  • the hybridomas thus obtained was suspended into RPMI-1640 medium containing 15% FCS supplemented with OPI-supplement (Sigma), followed by unification of the hybridomas by a limiting dilution method.
  • two types of hybridoma clones, #23-57-154 and #23-57-137-1 could be obtained, both which had a strong binding ability to PTHrP (1-34).
  • Hybridoma clone #23-57-137-1 was designated “mouse-mouse hybridoma #23-57-137-1”, and has been deposited under the terms of the Budapest Treaty on Aug. 15, 1996 at the National Institute of Bioscience and Human-technology, Agency of Industrial Science and Technology, Japan (1-3, Higashi 1-chome, Tsukuba-shi, Ibaraki, Japan) under the accession No. FERM BP-5631.
  • mRNA from hybridoma #23-57-137-1 was prepared using Quick Prep mRNA Purification Kit (Pharmacia Biotech). That is, cells of hybridoma #23-57-137-1 were fully homogenized with an extraction buffer, and mRNA was isolated and purified therefrom on an oligo(dT)-Cellulose Spun Column in accordance with the instructions included in the column. The resultant solution was subjected to ethanol precipitation to obtain the mRNA as a precipitate. The mRNA precipitate was dissolved in an elution buffer.
  • a gene encoding H-chain V-region of the mouse monoclonal antibody against human PTHrP was cloned by a 5′-RACE method (Frohman, M. A. et al., Proc. Natl. Acad. Sci. USA, 85, 8998-9002, 1988; Belyavsky, A. et al., Nucleic Acids Res. 17, 2919-2932, 1989).
  • the 5′-RACE method was performed using 5′-Ampli FINDER RACE Kit (CLONETECH) in accordance with the instructions included in the kit.
  • the primer used for synthesis of cDNA was MHC2 primer (SEQ ID NO: 1) which is capable of hybridizing to mouse H-chain C-region.
  • the above-prepared mRNA (about 2 ⁇ g), which was a template for the cDNA synthesis, was mixed with MHC2 primer (10 pmoles).
  • the resultant mixture was reacted with a reverse transcriptase at 52° C. for 30 minuets to effect the reverse transcription of the mRNA into cDNA.
  • the resultant reaction solution was added with 6N NaOH to hydrolyze any RNA remaining therein (at 65° C. for 30 min.) and then subjected to ethanol precipitation to isolate and purify the cDNA as a precipitate.
  • the purified cDNA was ligated to Ampli FINDER Anchor (SEQ ID NO: 42) at the 5′ end by reacting with T4 RNA ligase at 37° C. for 6 hours and additionally at room temperature for 16 hours.
  • Anchor primer SEQ ID NO: 2
  • MHC-G1 primer SEQ ID NO: 3
  • the PCR solution comprised (per 50 ⁇ l) 10 mM Tris-HCl (pH 8.3), 50 mM KCl, 0.25 mM dNTPs (dATP, dGTP, dCTP, dTTP), 1.5 mM MgCl 2 , 2.5 units of TaKaRa Taq (Takara Shuzo Co., Ltd.), 10 pmoles Anchor primer, and 1 ⁇ l of the reaction mixture of the cDNA to which MHC-G1 primer and Ampli FINDER Anchor primer had been ligated, over which mineral oil (50 ⁇ l) was layered.
  • the PCR was performed in Thermal Cycler Model 480J (Perkin Elmer) for 30 cycles under the conditions: 94° C. for 45 sec.; 60° C. for 45 sec.; and 72° C. for 2 min.
  • a gene encoding L-chain V-region of the mouse monoclonal antibody against human PTHrP was cloned by the 5′-RACE method (Frohman, M. A. et al., Proc. Natl. Acad. Sci. USA, 85, 8998-9002, 1988; Belyavsky, A. et al., Nucleic Acids Res. 17, 2919-2932, 1989).
  • the 5′-RACE method was performed using 5′-Ampli Finder RACE Kit (Clonetech) in accordance with the instructions included in the kit. In this method, oligo-dT primer was used as the primer for synthesizing cDNA.
  • the above-prepared mRNA (about 2 ⁇ g), which was a template for the cDNA synthesis, was mixed with oligo-dT primer.
  • the resultant mixture was reacted with a reverse transcriptase at 52° C. for 30 min. to effect the reverse transcription of the mRNA into cDNA.
  • the resultant reaction solution was added with 6N NaOH to hydrolyze any RNA remaining therein (at 65° C. for 30 min.).
  • the resultant solution was subjected to ethanol precipitation to isolate and purified the cDNA as a precipitate.
  • the cDNA thus synthesized was ligated to Ampli FINDER Anchor at the 5′ end by reacting with T4 RNA ligase at 37° C. for 6 hours and additionally at room temperature for 16 hours.
  • a PCR primer MLC (SEQ ID NO: 4) was designed based on the conserved sequence of mouse L-chain ⁇ chain C-region and then synthesized using 394 DNA/RNA Synthesizer (ABI).
  • the PCR solution comprised (per 100 ⁇ l) 10 mM Tris-HCl (pH 8.3), 50 mM KCl, 0.25 mM dNTPs (DATP, dGTP, dCTP, dTTP), 1.5 mM MgCl 2 , 2.5 units of AmpliTaq (PERKIN ELMER), 50 pmoles of Anchor primer (SEQ ID NO: 2), and 1 ⁇ l of the reaction mixture of the cDNA to which MLC (SEQ ID NO: 4) and Ampli FINDER Anchor were ligated, over which mineral oil (50 ⁇ l) was layered.
  • the PCR reaction was performed in Thermal Cycler Model 480J (Perkin Elmer) for 35 cycles under the conditions: 94° C. for 45 sec.; 60
  • each of the DNA fragments amplified by the PCR methods described above was separated by agarose gel electrophoresis on a 3% Nu Sieve GTG agarose (FMC Bio. Products).
  • an agarose gel segment containing a DNA fragment of about 550 bp was excised from the gel.
  • Each of the gel segments was subjected to purification of the DNA fragment of interest using GENECLEAN II Kit (BIO101) in accordance with the instructions included in the kit.
  • the purified DNA was precipitated with ethanol, and the DNA precipitate was dissolved in 20 ⁇ l of a solution containing 10 mM Tris-HCl (pH 7.4) and 1 mM EDTA.
  • a portion (1 ⁇ l) of the DNA solution was digested with a restriction enzyme XmaI (New England Biolabs) at 37° C. for 1 hour and further digested with a restriction enzyme EcoRI (Takara Shuzo Co., Ltd.) at 37° C. for 1 hour.
  • the digestion solution was extracted with phenol and chloroform and then precipitated with ethanol to collect the DNA.
  • the resultant cell mixture was added with 300 ⁇ l of SOC medium (Molecular Cloning: A Laboratory Manual, Sambrook, et al., Cold Spring Harbor Laboratory Press, 1989) and then incubated at 37° C. for 30 min.
  • the resultant cell solution was plated on LB agar medium or 2 ⁇ YT agar medium (Molecular Cloning: A Laboratory Manual, Sambrook, et al., Cold Spring Harbor Laboratory Press, 1989) containing either 100 or 50 ⁇ g/ml of ampicillin, 0.1 mM of IPTG and 20 ⁇ g/ml of X-gal, and then incubated at 37° C. overnight. In this manner, E. Coli transformants were prepared.
  • the transformants were cultured at 37° C. overnight in 2 ml of LB or 2 ⁇ YT medium containing either 100 or 50 ⁇ g/ml of ampicillin.
  • the cell fraction was applied to Plasmid Extracter PI-100 ⁇ (Kurabo Industries, Ltd.) or QIAprep Spin Plasmid Kit (QIAGEN) to give plasmid DNA.
  • Plasmid Extracter PI-100 ⁇ Kelabo Industries, Ltd.
  • QIAprep Spin Plasmid Kit QIAGEN
  • the nucleotide sequence of the cDNA coding region carried on the plasmid was determined in DNA Sequencer 373A (ABI; Perkin-Elmer) using Dye Terminator Cycle Sequencing Kit (Perkin-Elmer). In this sequencing, M13 Primer M4 (Takara Shuzo Co., Ltd.) (SEQ ID NO: 5) and M13 Primer RV (Takara Shuzo Co., Ltd.) (SEQ ID NO: 6) were used, and the nucleotide sequence was confirmed in the both directions.
  • the plasmid containing a gene encoding mouse H-chain V-region derived from hybridoma #23-57-137-1 was designated “MBC1H04”, and plasmid containing a gene encoding mouse L-chain V-region derived from hybridoma #23-57-137-1 was designated “MBC1L24”.
  • the nucleotide sequences (including the corresponding amino acids sequences) of the DNA encoding the mouse #23-57-137-1 antibody-derived H-chain V-region in plasmid MBC1H04 and gene encoding the mouse #23-57-137-1 antibody-derived L-chain V-region in plasmid MBC1H24 were shown in SEQ. ID Nos: 57 and 65, respectively.
  • Both of the polypeptides for the H-chain V-region fragment and for the L-chain V-region fragment were starting from the 58th nucleotide (which encoding glutamine) in the DNA sequences shown in SEQ ID Nos: 57 and 65, respectively.
  • the amino acid sequences of the polypeptides for the H-chain V-region and the L-chain V-region were also shown in SEQ. ID NOs: 46 and 45, respectively.
  • the E. coli strain containing plasmid MBC1H04 and the E. coli strain containing plasmid MBC1L24 were designated “ Escherichia coli JM109 (MBC1H04)” and “ Escherichia coli JM109 (MBC1L24)”, respectively.
  • These E. coli strains have been deposited under the terms of the Budapest Treaty at the National Institute of Bioscience and Human-Technology, Agency of Industrial Science and Technology, Japan (1-3, Higashi 1-chome, Tsukuba-shi, Ibaraki, Japan) on Aug. 15, 1996, under the Accession No. FERM BP-5628 for Escherichia coli JM109 (MBC1H04) and FERM BP-5627 for Escherichia coli JM109 (MBC1L24), respectively.
  • the H-chain V-region and the L-chain V-region have general structures similar to each other, in which there are four framework regions (FRs) linked through three hypervariable regions (i.e., complementarity determining regions; CDRs).
  • the amino acid sequences of the FRs are relatively well conserved, while the amino acid sequence of the CDRs have an extremely high variability (Kabat, E. A. et al., “Sequence of Proteins of Immunological Interest”, U.S. Dept. Health and Human Services, 1983).
  • the homology in amino acid between the V-regions of the mouse monoclonal antibody against human PTHrP was determined with reference to the database of amino acid sequences for antibodies established by Kabat et al.
  • the CDRs of the V-regions were determined as shown in Table 1.
  • a backward primer MBC1-S1 (SEQ ID NO: 7) was designed to hybridize to a DNA sequence encoding the 5′ region of the leader sequence for the V-region and to have both a Kozak consensus sequence (Kozak, M. et al., J. Mol. Biol., 196, 947-950, 1987) and a HindIII-recognition sequence.
  • a forward primer MBC1-a (SEQ ID NO: 8) was designed to hybridize to a DNA sequence encoding the 3′ region of the J region and to have both a donor splice sequence and a BamHI-recognition sequence.
  • the PCR reaction was performed using TaKaRa Ex Taq (Takara Shuzo Co., Ltd.) and a buffer appended thereto.
  • the PCR solution comprised (per 50 ⁇ l) 0.07 ⁇ g of plasmid MBC1H04 as a template DNA, 50 pmoles of MBC1-a and 50 pmoles of MBC1-S1 as primers, 2.5U of TaKaRa Ex Taq and 0.25 mM dNTPs in the buffer, over which 50 ⁇ l of mineral oil was layered.
  • the PCR was run for 30 cycles under the conditions: 94° C. for 1 min.; 55° C. for 1 min.; 72° C. for 2 min.
  • the DNA fragments thus amplified by the PCR method were separated by agarose gel electrophoresis on a 3% Nu Sieve GTG Agarose (FMC Bio. Products).
  • an agarose gel segment containing a DNA fragment of 437 bp was excised, and the DNA fragment was purified therefrom using GENECLEAN II Kit (BIO101) in accordance with the instructions included in the kit.
  • the purified DNA was collected by ethanol precipitation, and then dissolved in 20 ⁇ l of a solution containing 10 mM Tris-HCl (pH 7.4) and 1 mM EDTA.
  • a portion (1 ⁇ l) of the resultant DNA solution was digested with restriction enzymes BamHI and HindIII (Takara Shuzo Co., Ltd.) at 37° C. for 1 hour. The digestion solution was extracted with phenol and chloroform and then precipitated with ethanol to collect the DNA of interest.
  • HindIII-BamHI DNA fragment which containing a gene encoding the mouse H-chain V-region, was subcloned into pUC19 vector that had been digested with HindIII and BamHI.
  • the resultant plasmid was sequenced in DNA Sequencer 373A (Perkin-Elmer) using M13 Primer M4 and M13 Primer RV as primers and Dye Terminator Cycle Sequencing Kit (Perkin-Elmer).
  • a plasmid which carried a gene of correct nucleotide sequence encoding the mouse H-chain V-region derived from hybridoma #23-57-137-1 and had a HindIII-recognition sequence and a Kozak sequence on its 5′ region and a BamHI-recognition sequence on its 3′ region was obtained, which was designated “MBC1H/pUC19”.
  • a forward primer MBC1HVR2 (SEQ ID NO: 10) for the H-chain V-region was designed to hybridize to the DNA sequence encoding the 3′ region of the J region, to encoding the 5′ region of the C-region and to have ApaI- and SmaI-recognition sequences.
  • the PCR reaction was performed using TaKaRa Ex Taq (Takara Shuzo Co., Ltd.) and a buffer appended thereto.
  • the PCR solution comprised (per 50 ⁇ l) 0.6 ⁇ g of plasmid MBC1H/pUC19 as a template DNA, 50 pmoles of MBC1HVS2 and 50 pmoles of MBC1HVR2 as primers, 2.5U of TaKaRa Ex Taq and 0.25 mM of dNTPs in the buffer, over which 50 ⁇ l of mineral oil was layered.
  • the PCR reaction was run for 30 cycles under the conditions: 94° C. for 1 min.; 55° C. for 1 min.; 72° C. for 1 min.
  • the DNA fragments amplified by the PCR reaction were separated by agarose gel electrophoresis on a 1% Sea Kem GTG Agarose (FMC Bio. Products). Then, an agarose gel segment containing a DNA fragment of 456 bp was excised and the DNA fragment was purified therefrom using GENECLEAN II Kit (BIO101) in accordance with the instructions included in the kit. The purified DNA was precipitated with ethanol and then dissolved in 20 ⁇ l of a solution containing 10 mM Tris-HCl (pH 7.4) and 1 mM EDTA.
  • the resultant DNA solution (1 ⁇ g) was digested with restriction enzymes EcoRI and SmaI (Takara Shuzo Co., Ltd.) at 37° C. for 1 hour. The digestion solution was extracted with phenol and chloroform and then precipitated with ethanol to collect the DNA. The obtained EcoRI-SmaI DNA fragment, which containing a gene encoding the mouse H-chain V-region, was subcloned into pUC19 vector that had been digested with EcoRI and SmaI. The resultant plasmid was sequenced in DNA Sequencer 373A (Perkin-Elmer) using M13 Primer M4 and M13 Primer RV, and Dye Terminator Cycle Sequencing Kit (Perkin-Elmer).
  • a plasmid which contained a gene encoding mouse H-chain V-region derived from hybridoma #23-57-137-1 of correct nucleotide sequence and had EcoRI- and HindIII-recognition sequences and a Kozak sequence on its 5′ region and ApaI- and SmaI-recognition sequences on its 3′ region was obtained, which was designated “MBC1Hv/pUC19”.
  • cDNA containing the DNA for human antibody H-chain C-region C ⁇ 1 was prepared as follows.
  • mRNA was prepared from a CHO cell into which both an expression vector DHFR- ⁇ E-RVh-PM-1-f (see WO 92/19759) encoding the genomic DNAs of humanized PM1 antibody H-chain V-region and human antibody H-chain C-region IgG1 (N. Takahashi et al., Cell 29, 671-679, 1982) and an expression vector RV1-PM1a (see WO 92/19759) encoding the genomic DNAs of humanized PM1 antibody L-chain V-region and human antibody L-chain K chain C-region had been introduced.
  • cDNA containing the humanized PM1 antibody H-chain V-region and the human antibody C-region C ⁇ 1 was cloned by a RT-PCR method, and then subcloned into plasmid pUC19 on the HindIII-BamHI site. After sequencing, a plasmid which had the correct nucleotide sequence was obtained, which was designated “pRVh-PM1f-cDNA”.
  • the plasmid obtained (pRVh-PM1f-cDNA) was digested with BamHI, blunt-ended with Klenow fragment, and further digested with HindIII, thereby obtaining a blunt-ended HindIII-BamHI fragment.
  • the blunt-ended HindIII-BamHI fragment was ligated to the above-mentioned HindIII site- and EcoRI site-deleted expression vector DHFR- ⁇ E-RVh-PM1-f that had been digested with HindIII and BamHI.
  • an expression vector RVh-PM1f-cDNA was constructed which contained cDNA encoding the humanized PM1 antibody H-chain V-region and the human antibody C-region C ⁇ 1.
  • the expression vector RVh-PM1f-cDNA containing the cDNA encoding the humanized PM1 antibody H-chain V-region and the human antibody C-region C ⁇ 1 was digested with ApaI and BamHI, and a DNA fragment containing the H-chain C-region was collected therefrom.
  • the resultant DNA fragment was introduced into the above-mentioned plasmid MBC1Hv/pUC19 that had been digested with ApaI and BamHI.
  • the plasmid thus prepared was designated “MBC1HcDNA/pUC19”.
  • This plasmid contained cDNA encoding the mouse antibody H-chain V-region and the human antibody C-region C ⁇ 1, and had EcoRI- and HindIII-recognition sequences on its 5′ region and a BamHI-recognition sequence on its 3′ region.
  • the plasmid MBC1HcDNA/pUC19 was digested with EcoRI and BamHI to give a DNA fragment comprising a nucleotide sequence encoding the chimeric antibody H-chain.
  • the resultant DNA fragment was introduced into an expression vector pCOS1 that had been digested with EcoRI and BamHI, thereby giving an expression vector for the chimeric antibody, which was designated “MBC1HcDNA/pCOS1”.
  • the expression vector pCOS1 was constructed using HEF-PMh-g ⁇ 1 (see WO 92/19759) by deleting therefrom an antibody genes by digestion with EcoRI and SmaI, and then ligating it to EcoRI-NotI-BamHI Adaptor (Takara Shuzo Co., Ltd.).
  • the plasmid MBC1HcDNA/pUC19 was digested with EcoRI and BamHI to obtain a DNA fragment containing a gene for the chimeric antibody H-chain.
  • the DNA fragment was then introduced into an expression plasmid pCHO1 that had been digested with EcoRI and BamHI to give an expression plasmid for the chimeric antibody, which was designated “MBC1HcDNA/pCHO1”.
  • the expression vector pCHO1 was constructed using DHFR-AE-rvH-PM1-f (see WO 92/19759) by deleting therefrom an antibody gene by digestion with EcoRI and SmaI, and then ligating it to EcoRI-NotI-BamHI Adaptor (Takara Shuzo Co., Ltd.).
  • pUC19 vector containing a gene for human L-chain C-region a HindIII site-deleted pUC19 vector was prepared.
  • pUC19 vector (2 ⁇ g) was digested in 20 ⁇ l of a reaction solution containing 20 mM Tris-HCl (pH 8.5), 10 mM MgCl 2 , 1 mM DTT, 100 mM KCl, 8 U of HindIII (Takara Shuzo Co., Ltd.) at 37° C. for 1 hour.
  • the resultant digestion solution was extracted with phenol and chloroform, and then subjected to ethanol precipitation to collect the DNA of interest.
  • the DNA collected was reacted in 50 ⁇ l of a reaction solution containing 50 mM Tris-HCl (pH 7.5), 10 mM MgCl 2 , 1 mM DTT, 100 mM NaCl, 0.5 mM dNTPs and 6U of Klenow fragment (GIBCO BRL) at room temperature for 20 min., thereby rendering the terminal ends of the DNA blunt.
  • This reaction mixture was extracted with phenol and chloroform and then subjected to ethanol precipitation to collect the vector DNA.
  • the vector DNA thus collected was reacted in 10 ⁇ l of a reaction solution containing 50 mM Tris-HCl (pH 7.6), 10 mM MgCl 2 , 1 mM ATP, 1 mM DTT, 5% (v/v) polyethylene glycol-8000 and 0.5 U of T4 DNA ligase (GIBCO BRL) at 16° C. for 2 hours, to cause self-legation of the vector DNA.
  • the reaction solution (5 ⁇ l) was added to 100 ⁇ l of a solution containing competent cells of E. coli , JM109 (Nippon Gene Co., Ltd.), and the resultant solution was allowed to stand on ice for 30 min., at 42° C.
  • the transformant was cultured in 2 ⁇ YT medium (20 ml) containing ampicillin (50 ⁇ g/ml) at 37° C. overnight. From the cell fraction of the culture medium, a plasmid DNA was isolated and purified using Plasmid Mini Kit (QIAGEN) in accordance with the instructions included in the kit. The purified plasmid was digested with HindIII. The plasmid that was confirmed to have a HindIII site-deletion was designated “pUC19 ⁇ HindIII”.
  • Human antibody L-chain ⁇ chain C-region has been known to have at least four isotypes including Mcg + Ke + Oz ⁇ , Mcg ⁇ Ke ⁇ Oz ⁇ , Mcg ⁇ Ke ⁇ Oz + and Mcg ⁇ Ke + Oz ⁇ (P. Dariavach, et al., Proc. Natl. Acad. Sci. USA, 84, 9074-9078, 1987).
  • a search was made for a human antibody L-chain ⁇ chain C-region homologous to the #23-57-137-1 mouse L-chain ⁇ chain C-region from the EMBL database. As a result, it was found that the isotype Mcg + Ke + Oz ⁇ of the human antibody L-chain ⁇ chain (Accession No.
  • a gene encoding human antibody L-chain ⁇ chain C-region was constructed by a PCR method.
  • the primer for the PCR was synthesized using 394 DNA/RNA Synthesizer (ABI).
  • the synthesized primers were as follows: HLAMB1 (SEQ ID NO: 11) and HLAMB3 (SEQ ID NO: 13), both having a sense DNA sequence; and HLAMB2 (SEQ ID NO: 12) and HLAMB4 (SEQ ID NO: 14), both having an antisense DNA sequence; each primer containing a complementary sequence of 20-23 bp on the both terminal ends.
  • External primers HLAMBS (SEQ ID NO: 15) and HLAMBR (SEQ ID NO: 16) had sequences homologous to the primers HLAMB1 and HLAMB4, respectively.
  • HLAMBS contained EcoRI-, HindIII- and BlnI-recognition sequences
  • HLAMBR contained an EcoRI-recognition sequence.
  • the reaction solution was added with the external primers HLAMBS and HLAMBR. This reaction mixture was subjected to the third PCR reaction to amplify the full length DNA.
  • Each PCR reaction was performed using TaKaRa Ex Taq (Takara Shuzo Co., Ltd.) in accordance with the instructions included in the kit.
  • 100 ⁇ l of either a reaction solution containing 5 pmoles of HLAMB1, 0.5 pmole of HLAMB2 and 5U of TaKaRa Ex Taq (Takara Shuzo Co., Ltd.) or a reaction solution containing 0.5 pmole of HLAMB3, 5 pmoles of HLAMB4 and 5U of TaKaRa Ex Taq (Takara Shuzo Co., Ltd.) was used, over which 50 ⁇ l of mineral oil was layered.
  • the PCR reaction was run for 5 cycles under the conditions: 94° C. for 1 min., 60° C. for 1 min. and 72° C. for 1 min.
  • the reaction solution to which the external primers HLAMBS and HLAMBR (50 pmoles each) were added was used.
  • the PCR reaction was run for 30 cycles under the conditions: 94° C. for 1 min., 60° C. for 1 min. and 72° C. for 1 min.
  • the DNA fragment obtained by the third PCR reaction was subjected to electrophoresis on a 3% low-melting agarose gel (NuSieve GTG Agarose, FMC), and separated and purified from the gel using GENECLEAN II Kit (BIO101) in accordance with the instructions included in the kit.
  • the DNA fragment obtained was digested in a reaction solution (20 ⁇ l) containing 50 mM Tris-HCl (pH 7.5), 10 mM MgCl 2 , 1 mM DTT, 100 mM NaCl and 8U of EcoRI (Takara Shuzo Co., Ltd.) at 37° C. for 1 hour.
  • the digestion solution was extracted with phenol and chloroform, and the DNA was collected therefrom by the ethanol precipitation.
  • the DNA was dissolved in a solution (8 ⁇ l) containing 10 mM Tris-HCl (pH 7.4) and 1 mM EDTA.
  • the above-prepared plasmid pUC19 ⁇ HindIII (0.8 ⁇ g) was digested with EcoRI in the same manner as mentioned above.
  • the digestion solution was subjected to phenol/chloroform extraction and then ethanol precipitation, thereby giving a digested plasmid pUC19 ⁇ HindIII.
  • the digested plasmid was reacted in a reaction solution (50 ⁇ l) containing 50 mM Tris-HCl (pH 9.0), 1 mM MgCl 2 and alkaline phosphatase ( E. coli C75; Takara Shuzo Co., Ltd.) at 37° C. for 30 min.
  • the reaction solution was subjected to phenol/chloroform extraction, and the DNA was collected therefrom by ethanol precipitation.
  • the DNA thus obtained was dissolved in a solution (10 ⁇ l) containing 10 mM Tris-HCl (pH 7.4) and 1 mM EDTA.
  • the BAP-treated plasmid pUC19 ⁇ HindIII (1 ⁇ l) was ligated to the above-obtained PCR product (4 ⁇ l) using DNA Ligation Kit Ver.2 (Takara Shuzo Co., Ltd.).
  • the resultant plasmid was introduced into a competent cell of E. coli, JM109, to give a transformant.
  • the transformant was cultured overnight in 2 ⁇ YT medium (2 ml) containing 50 ⁇ g/ml of ampicillin. From the cell fraction, the plasmid was isolated using QIAprep Spin Plasmid Kit (QIAGEN).
  • the plasmid obtained was sequenced for the cloned DNA portion.
  • the sequencing was performed in 373A DNA Sequencer (ABI) using M13 Primer M4 and M13 Primer RV (Takara Shuzo Co., Ltd.).
  • M13 Primer M4 and M13 Primer RV Takara Shuzo Co., Ltd.
  • the plasmid was designated “C ⁇ /pUC19”.
  • primers HCLMS (SEQ ID NO: 17) and HCLMR (SEQ ID NO: 18) were newly synthesized, and correct DNA was reconstructed using these primers by a PCR method.
  • the plasmid C ⁇ /pUC19 having the DNA deletion therein was used as a template, and the reaction was performed with each of the primer sets of HLAMBS and HCLMS and HCLMS and HLAMB4.
  • the PCR products were purified separately.
  • the PCR products were assembled together.
  • the reaction product of the second PCR reaction was added with external primers HLAMBS and HLAMB4 and amplified to give the full length DNA.
  • a reaction solution (100 ⁇ l) containing 0.1 ⁇ g of C ⁇ /pUC19 as a template, either 50 pmoles of each of the primers HLAMBS and HCLMR or 50 pmoles of each of the primers HCLMS and HLAMB4, and 5U of TaKaRa Ex Taq (Takara Shuzo Co., Ltd.) was used, over which 50 ⁇ l of mineral oil was layered.
  • the PCR reaction was run for 30 cycles under the conditions: 94° C. for 1 min., 60° C. for 1 min. and 72° C. for 1 min.
  • PCR products of the first PCR reaction HLAMBS-HCLMR (236 bp) and HCLMS-HLAMB4 (147 bp), were subjected to electrophoresis separately on a 3% low-melting agarose gel to isolate the DNA fragments.
  • the DNA fragments were collected and purified from the gels using GENECLEAN II Kit (BIO101).
  • 20 ⁇ l of a reaction solution containing 40 ng of each of the purified DNA fragments and 1U of TaKaRa Ex Taq (Takara Shuzo Co., Ltd.) was used, over which 25 ⁇ l of mineral oil was layered.
  • the PCR reaction was run for 5 cycles under the conditions: 94° C. for 1 min., 60° C. for 1 min. and 72° C. for 1 min.
  • a portion (0.1 ⁇ g) of the DNA fragment thus obtained was digested with EcoRI, and then subcloned into plasmid pUC19 ⁇ HindIII that had been BAP-treated.
  • the resultant plasmid was introduced into a competent cell of E. coli, JM109, to form a transformant.
  • the transformant was cultured overnight in 2 ml of 2 ⁇ YT medium containing 50 ⁇ g/ml of ampicillin. From the cell fraction, the plasmid was isolated and purified using QIAprep Spin Plasmid Kit (QIAGEN).
  • the purified plasmid was sequenced in 373A DNA Sequencer (ABI) using M13 Primer M4 and M13 Primer RV (Takara Shuzo Co., Ltd.). The plasmid that was confirmed to have the correct nucleotide sequence without any deletion was designated “C ⁇ /pUC19”.
  • a DNA fragment encoding the L-chain K chain C-region was cloned from plasmid HEF-PM1k-gk (WO 92/19759) by a PCR method.
  • a forward primer HKAPS (SEQ ID NO: 19) was designed to contain EcoRI-, HindIII and BlnI-recognition sequences, and a backward primer HKAPA (SEQ ID NO: 20) was designed to contain an EcoRI-recognition sequence. These primers were synthesized in 394 DNA/RNA Synthesizer (ABI).
  • a PCR reaction was performed using 100 ⁇ l of a reaction solution containing 0.1 ⁇ g of plasmid HEF-PM1k-gk as a template, 50 pmoles of each of primers HKAPS and HKAPA and 5U of TaKaRa Ex Taq (Takara Shuzo Co., Ltd.), over which 50 ⁇ l of mineral oil was layered.
  • the PCR reaction was run for 30 cycles under the conditions: 94° C. for 1 min., 60° C. for 1 min. and 72° C. for 1 min., thereby giving a PCR product of 360 bp.
  • the DNA fragment was isolated and purified by electrophoresis on a 3% low-melting agarose, and then collected and purified using GENECLEAN II Kit (BIO101).
  • the DNA fragment thus obtained was digested with EcoRI, and then cloned into plasmid pUC19 ⁇ HindIII that had been BAP-treated.
  • the resultant plasmid was introduced into a competent cell of E. coli, JM109, to form a transformant.
  • the transformant was cultured overnight in 2 ml of 2 ⁇ YT medium containing 50 ⁇ g/ml of ampicillin. From the cell fraction, the plasmid was purified using QIAprep Spin Plasmid Kit (QIAGEN).
  • the purified plasmid was sequenced in 373A DNA Sequencer (ABI) using M13 Primer M4 and M13 Primer RV (Takara Shuzo Co., Ltd.). The plasmid that was confirmed to have the correct nucleotide sequence was designated “C ⁇ /pUC19”.
  • An expression vector for the chimeric #23-57-137-1 antibody L-chain was constructed.
  • a gene encoding #23-57-137-1 L-chain V-region was ligated to the HindIII-BlnI site (located just in front of the human antibody C-region) of each of the plasmids C ⁇ /pUC19 and C ⁇ /pUC19, thereby obtaining pUC19 vectors containing the DNA encoding the chimeric #23-57-137-1 antibody L-chain V-region and either of the L-chain ⁇ chain C-region or the L-chain ⁇ region C-region, respectively.
  • Each of the resultant vectors was then digested with EcoRI to separate the gene for the chimeric antibody L-chain. The gene was subcloned into HEF expression vector.
  • a DNA fragment encoding #23-57-137-1 antibody L-chain V-region was cloned from plasmid MBC1L24 by a PCR method. Primers used in the PCR method were separately synthesized using 394 DNA/RNA Synthesizer (ABI).
  • a backward primer MBCCHL1 (SEQ ID NO: 21) was designed to contain a HindIII-recognition sequence and a Kozak sequence (Kozak, M. et al., J. Mol. Biol. 196, 947-950, 1987), and a forward primer MBCCHL3 (SEQ ID NO: 22) was designed to contain BglII- and RcoRI-recognition sequences.
  • the PCR reaction was performed using 100 ⁇ l of a reaction solution containing 10 mM Tris-HCl (pH 8.3), 50 mM KCl, 1.5 mM MgCl 2 , 0.2 mM dNTPs, 0.1 ⁇ g MBC1L24, 50 pmoles of each of primers MBCCHL1 and MBCCHL3 and 1 ⁇ l of AmpliTaq (PERKIN ELMER), over which 50 ⁇ l of mineral oil was layered.
  • the PCR reaction was run for 30 cycles under the conditions: 94° C. for 45 sec., 60° C. for 45 sec. and 72° C. for 2 min.
  • a PCR product of 444 bp was electrophoresed on a 3% low-melting agarose gel, and collected and purified using GENECLEAN II Kit (BIO101). The purified PCR product was dissolved in 20 ⁇ l of a solution containing 10 mM Tris-HCl (pH 7.4) and 1 mM EDTA.
  • the PCR product (1 ⁇ l) was digested in 20 ⁇ l of a reaction solution containing 10 mM Tris-HCl (pH 7.5), 10 mM MgCl 2 , 1 mM DTT, 50 mM NaCl, 8U of HindIII (Takara Shuzo Co., Ltd.) and 8U of EcoRI (Takara Shuzo Co., Ltd.) at 37° C. for 1 hour.
  • the digestion solution was subjected to phenol/chloroform extraction, and the DNA of interest was collected therefrom by ethanol precipitation.
  • the DNA was dissolved in 8 ⁇ l of a solution containing 10 mM Tris-HCl (pH 7.4) and 1 mM EDTA.
  • plasmid pUC19 (1 ⁇ g) was digested with HindIII and EcoRI, and subjected to phenol/chloroform extraction and then ethanol precipitation.
  • the obtained digested plasmid was BAP-treated with alkaline phosphatase ( E. coli C75; Takara Shuzo Co., Ltd.).
  • the resultant reaction solution was extracted with phenol and chloroform, and the DNA was collected therefrom by ethanol precipitation.
  • the DNA was dissolved in 10 ⁇ l of a solution containing 10 mM Tris-HCl (pH 7.4) and 1 mM EDTA.
  • the BAP-treated plasmid pUC19 (1 ⁇ l) was ligated to the above-obtained PCR product (4 ⁇ l) using DNA Ligation Kit Ver. 2 (Takara Shuzo Co., Ltd.).
  • the resultant plasmid was introduced into a competent cell of E. coli, JM109 (Nippon Gene Co., Ltd.), in the same manner as mentioned above, to form a transformant.
  • the transformant was plated on 2 ⁇ YT agar medium containing 50 ⁇ g/ml of ampicillin and cultured at 37° C. overnight. The resultant transformant was then cultured at 37° C. overnight in 2 ml of 2 ⁇ YT medium containing 50 ⁇ g/ml of ampicillin.
  • the plasmid was purified using QIAprep Spin Plasmid Kit (QIAGEN). After determining the nucleotide sequence, the plasmid that was confirmed to have the correct nucleotide sequence was designated “CHL/pUC19”.
  • Each of plasmids C ⁇ /pUC19 and C ⁇ /pUC19 (1 ⁇ g each) was digested in 20 ⁇ l of a reaction solution containing 20 mM Tris-HCl (pH 8.5), 10 mM MgCl 2 , 1 mM DTT, 100 mM KCl, 8U of HindIII (Takara Shuzo Co., Ltd.) and 2U of BlnI (Takara Shuzo Co., Ltd.) at 37° C. for 1 hour.
  • the digestion solution was extracted with phenol and chloroform, and the DNA was collected therefrom by ethanol precipitation.
  • the DNA was BAP-treated at 37° C. for 30 min.
  • the reaction solution was extracted with phenol and chloroform, and the DNA was collected therefrom by ethanol precipitation.
  • the DNA was dissolved in 10 ⁇ l of a solution containing 10 mM Tris-HCl (pH 7.4) and 1 mM EDTA.
  • the plasmid CHL/pUC19 that contained DNA encoding #23-57-137-1 L-chain V-region (8 ⁇ g) was digested with HindIII and BlnI in the same manner as mentioned above to give a DNA fragment of 409 bp.
  • the DNA fragment was electrophoresed on a 3% low-melting agarose gel, and then collected and purified using GENECLEAN II Kit (BIO101) from the gel.
  • the DNA was dissolved in 10 ⁇ l of a solution containing 10 mM Tris-HCl (pH 7.4) and 1 mM EDTA.
  • the DNA for L-chain V-region DNA (4 ⁇ l) was subcloned into 1 ⁇ l of each of the BAP-treated plasmids C ⁇ /pUC19 and C ⁇ /pUC19, and then introduced into a competent cell of E. coli, JM109, to form a transformant.
  • the transformant was cultured overnight in 3 ml of 2 ⁇ YT medium containing 50 ⁇ g/ml of ampicillin. From the cell fraction, the plasmid was isolated and purified using QIAprep Spin Plasmid Kit (QIAGEN).
  • the two plasmids thus prepared were designated “MBC1L( ⁇ )/pUC19” and “MBC1L ( ⁇ )/pUC19”, respectively.
  • Each of plasmids MBC1L( ⁇ )/pUC19 and MBC1L( ⁇ )/pUC19 was digested with EcoRI and then subjected to electrophoresis on a 3% low-melting agarose gel.
  • a DNA fragment of 743 bp was isolated and purified from the gel using GENECLEANII Kit (BIO101), and then dissolved in 10 ⁇ l of a solution containing 10 mM Tris-HCl (pH 7.4) and 1 mM EDTA.
  • An expression vector (plasmid HEF-PM1k-gk) (2.7 ⁇ g) was digested with EcoRI and then extracted with phenol and chloroform, and the DNA was collected therefrom by ethanol precipitation. The DNA fragment was BAP-treated, and then subjected to electrophoresis on a 1% low-melting agarose gel. From the gel, a DNA fragment of 6561 bp was isolated and purified therefrom using GENECLEANII Kit (BIO101). The purified DNA fragment was dissolved in 10 ⁇ l of a solution containing 10 mM Tris-HCl (pH 7.4) and 1 mM EDTA.
  • BAP-treated HEF vector (2 ⁇ l) was ligated to an EcoRI fragment (3 ⁇ l) of each of plasmid MBC1L( ⁇ )/pUC19 and MBC1L( ⁇ )/pUC19.
  • the ligation product was introduced into a competent cell of E. coli, JM109, to form a transformant.
  • the transformant was cultured in 2 ml of 2 ⁇ YT medium containing 50 ⁇ g/ml of ampicillin. From the cell fraction, the plasmid was purified using QIAprep Spin Plasmid Kit (QIAGEN).
  • the purified plasmid was digested in 20 ⁇ l of a reaction solution containing 20 mM Tris-HCl (pH 8.5), 10 mM MgCl 2 , 1 mM DTT, 100 mM KCl, 8U of HindIII (Takara Shuzo Co., Ltd.) and 2 U of PvuI (Takara Shuzo Co., Ltd.) at 37° C. for 1 hour.
  • This reaction gave digestion fragments of 5104/2195 bp if the fragment was inserted in the correct orientation, or gave digestion fragments of 4378/2926 bp if the fragment was inserted in the reverse orientation.
  • the plasmid that was confirmed to have the fragment in the correct orientation was designated “MBC1L( ⁇ )/neo” for plasmid MBC1L( ⁇ )/pUC19 or “MBC1L( ⁇ )/neo” for plasmid MBC1L( ⁇ )/PUC19.
  • the transient expression of the chimeric antibodies was performed using each of the combinations of plasmids MBC1HcDNA/pCOS1 and MBC1L ( ⁇ )/neo and plasmids MBC1HcDNA/pCOS1 and MBC1L( ⁇ )/neo, by co-transfecting a COS-7 cell with the plasmids by electroporation using Gene Pulser (Bio Rad). That is, the plasmids (10 ⁇ g each) were added to a COS-7 cell suspension (0.8 ml; 1 ⁇ 10 7 cells/ml) in PBS( ⁇ ). The resultant solution was applied with pulses at an electrostatic capacity of 1,500V and 2 ⁇ F to cause electroporation.
  • the electroporated cells were suspended in DMEM medium (GIBCO) containing 2% Ultra Low IgG fetal calf serum (GIBCO), and then cultured using a 10-cm culture dish in a CO 2 incubator. After cultivating for 72 hours, a culture supernatant was collected and centrifuged to remove cell debris and was provided for use as a sample for the subsequent ELISA. In this procedure, the purification of the chimeric antibody from the COS-7 cell culture supernatant was performed using AffiGel Protein A MAPSII Kit (Bio Rad) in accordance with the instructions included in the kit.
  • An ELISA plate for determining antibody concentration was prepared as follows. Each well of a 96-well ELISA plate (Maxisorp, NUNC) was coated with 100 ⁇ l of a coating buffer (0.1 M NaHCO 3 , 0.02% NaN 3 ) supplemented with 1 ⁇ g/ml of goat anti-human IgG antibody (TAGO), and then blocked with 200 ⁇ l of a dilution buffer [50 mM Tris-HCl, 1 mM MgCl 2 , 0.1 M NaCl, 0.05% Tween 20, 0.02% NaN 3 , 1% bovine serum albumin (BSA); pH 7.2].
  • a coating buffer 0.1 M NaHCO 3 , 0.02% NaN 3
  • Tween 20 0.05% Tween 20
  • BSA bovine serum albumin
  • Each well of the plate was added with each of the serial dilutions of the COS-7 cell culture supernatant in which each of the chimeric antibodies had been expressed, or added with each of the serial dilutions of each of the chimeric antibodies per se in a purified form.
  • the plate was incubated at room temperature for 1 hour and washed with PBS-Tween 20.
  • Each well of the plate was then added with 100 ⁇ l of a solution of alkaline phosphatase-conjugated goat anti-human IgG antibodies (TAGO).
  • TAGO alkaline phosphatase-conjugated goat anti-human IgG antibodies
  • An ELISA plate for the determination of antigen-binding ability was prepared as follows. Each well of a 96-well ELISA plate was coated with 100 ⁇ l of a coating buffer supplemented with 1 ⁇ g/ml of human PTHrP (1-34) (Peptide Research Institute), and then blocked with 200 ⁇ l of a dilution buffer. Each well was added with each of the serial dilutions of the COS-7 cell culture supernatant in which each of the chimeric antibodies had been expressed, or added with each of the serial dilutions of each of the chimeric antibodies per se in a purified form.
  • each well of the plate was added with 100 ⁇ l of a solution of alkaline phosphatase-conjugated goat anti-human IgG antibodies (TAGO). After the plate was incubated at room temperature and washed with PBS-Tween 20, each well of the plate was added with 1 mg/ml of a substrate solution (“Sigma 104”, p-nitrophenylphosphoric acid, SIGMA). The solution was measured on its absorbance at 405 nm using Microplate Reader (Bio Rad).
  • TAGO alkaline phosphatase-conjugated goat anti-human IgG antibodies
  • the chimeric antibodies had an ability to bind to human PTHrP (1-34) and the cloned mouse antibody V-regions had the correct structures (FIG. 5). It was also found that there was no difference in the ability to bind to PTHrP (1-34) between the chimeric antibody with L-chain ⁇ chain C-region and the chimeric antibody with L-chain ⁇ chain C-region. Therefore, the L-chain C-region of the humanized antibody was constructed using the humanized antibody L-chain ⁇ chain.
  • each of the plasmid DNAs (10 ⁇ g each) was added to 0.8 ml of a cell suspension of CHO cells in PBS( ⁇ ) (1 ⁇ 10 7 cells/ml).
  • the resultant solution was applied with pulses at an electrostatic capacity of 1,500V and 25 ⁇ F.
  • the electroporated cells were suspended in MEM-A medium (GIBCO) containing 10% fetal calf serum (GIBCO).
  • the resultant suspension was cultured using three 96-well plates (Falcon) in a CO 2 incubator.
  • the medium was replaced by a selective medium [ribonucleoside- or deoxyribonucleoside-free MEM- ⁇ medium (GIBCO) containing 10% fetal calf serum (GIBCO) and 500 mg/ml of GENETICIN (G418Sulfate; GIBCO)].
  • a selective medium [ribonucleoside- or deoxyribonucleoside-free MEM- ⁇ medium (GIBCO) containing 10% fetal calf serum (GIBCO) and 500 mg/ml of GENETICIN (G418Sulfate; GIBCO)].
  • GEBCO ribonucleoside- or deoxyribonucleoside-free MEM- ⁇ medium
  • GENETICIN G418Sulfate
  • the cultivation of the established cell line capable of stable production of the antibodies was scaled up in a roller bottle using ribonucleoside- or deoxyribonucleoside-free MEM medium containing 2% Ultra Low IgG fetal calf serum.
  • the culture supernatant was collected and then filtered using a 0.2- ⁇ m filter (Millipore) to remove cell debris therefrom.
  • a humanized #23-57-137-1 antibody H-chain was produced by CDR-grafting technique by means of a PCR method.
  • a humanized #23-57-137-1 antibody H-chain (version “a”) having FRs derived from human antibody S31679 (NBRF-PDB; Cuisinier, A. M. et al., Eur. J.
  • CDR-grafting primers MBC1HGP1 (SEQ ID NO: 23) and MBC1HGP3 (SEQ ID NO: 24) (both containing a sense DNA sequence) and MBC1HGP2 (SEQ ID NO: 25) and MBC1HGP4 (SEQ ID NO: 26) (both containing an antisense DNA sequence), all of which containing a 15-21 bp complementary sequence on both terminal ends thereof; and external primers: MBC1HVS1 (SEQ ID NO: 27) and MBC1HVR1 (SEQ ID NO: 28) having a homology to the CDR-grafting primers MBC1HGP1 and MBC1HGP4, respectively.
  • the CDR-grafting primers MBC1HGP1, MBC1HGP2, MBC1HGP3 and MBC1HGP4 were separated on an urea-denatured polyacrylamide gel (Molecular Cloning: A Laboratory Manual, Sambrook, et al., Cold Spring Harbor Laboratory Press, 1989), and extracted therefrom by a crush-and-soak method (Molecular Cloning: A Laboratory Manual, Sambrook, et al., Cold Spring Harbor Laboratory Press, 1989) in the following manner.
  • Each of the CDR-grafting primers (1 nmole) was separated on a 6% denatured polyacrylamide gel to give DNA fragments. From the resultant DNA fragments, one having a desired length was identified on a silica gel thin plate by irradiation of UV ray and then collected therefrom by a crush-and-soak method. The resultant DNA was dissolved in 20 ⁇ l of a solution containing 10 mM Tris-HCl (pH 7.4) and 1 mM EDTA. The PCR reaction was performed using TaKaRa Ex Taq (Takara Shuzo Co., Ltd.).
  • the PCR reaction solution (100 ⁇ l) comprised 1 ⁇ l of each of the above-mentioned CDR-grafting primers MBC1HGP1, MBC1HGP2, MBC1HGP3 and MBC1HGP4, 0.25 mM dNTPs and 2.5U of TaKaRa Ex Taq in the buffer.
  • the PCR reaction was run for 5 cycles under the conditions: 94° C. for 1 min., 55° C. for 1 min. and 72° C. for 1 min.
  • the resultant reaction solution was added with the external primers MBC1HVS1 and MBC1HVR1 (50 pmoles each). Using this reaction mixture, the PCR reaction was further run for additional 30 cycles under the same conditions.
  • the DNA fragment thus amplified was separated by agarose gel electrophoresis on a 4% Nu Sieve GTG agarose (FMC Bio. Products).
  • the DNA for the humanized H-chain V-region constructed in the above step was modified by a PCR method.
  • a backward primer MBC1HVS2 was designed to hybridize to the sequence encoding the 5′ region of the leader sequence for the V-region and to have a Kozak consensus sequence (Kozak et al., J. Mol. Biol.
  • a forward primer MBC1HVR2 was designed to hybridize to both the DNA sequence encoding the 3′ region of the J region and the DNA sequence encoding the 5′ region of the C-region and to have ApaI- and SmaI-recognition sequences.
  • the PCR reaction was performed using TaKaRa Ex Taq (Takara Shuzo Co., Ltd.) and a buffer appended thereto.
  • the PCR reaction solution comprised 0.4 ⁇ g of hMBCHv/pUC19 as a DNA template, 50 pmoles of each of MBC1HVS2 and MBC1HVR2 as primers, 2.5U of TaKaRa Ex Taq and 0.25 mM dNTPs in the buffer.
  • the PCR reaction was run for 30 cycles under the conditions: 94° C. for 1 min., 55° C. for 1 min. and 72° C. for 1 min.
  • the DNA fragment thus amplified was separated by agarose gel electrophoresis on a 3% Nu Sieve GTG agarose (FMC Bio. Products).
  • a gel segment containing a DNA fragment of 456 bp was excised, and the DNA fragment was purified therefrom using GENECLEANII Kit (BIO101) in accordance with the instructions included in the kit.
  • the DNA fragment thus purified was precipitated with ethanol and then dissolved in 20 ⁇ l of a solution containing 10 mM Tris-HCl (pH 7.4) and 1 mM EDTA.
  • the PCR reaction solution thus obtained was used for subcloning of the DNA fragment into plasmid pUC19 that had been digested with EcoRI and SmaI, and then the resultant plasmid was sequenced.
  • a plasmid which contained DNA encoding mouse H-chain V-region derived from hybridoma #23-57-137-1 and also contained EcoRI- and HindIII-recognition sequences and a Kozak sequence on the 5′ region and ApaI- and SmaI-recognition sequences on the 3′ region, which was designated “hMBC1Hv/pUC19”.
  • Plasmid RVh-PM1f-cDNA carrying a cDNA sequence for hPM1 antibody H-chain was digested with ApaI and BamHI to give a DNA fragment containing a DNA fragment containing DNA encoding the H-chain C-region.
  • the DNA fragment was introduced into plasmid hMBC1Hv/PUC19 that had been digested with ApaI and BamHI.
  • the obtained plasmid was designated “hMBC1HcDNA/pUC19”.
  • This plasmid contained both DNA encoding the humanized #23-57-137-1 antibody H-chain V-region and DNA encoding the human H-chain C-region C ⁇ 1 and had EcoRI- and HindIII-recognition sequences on the 5′ region and a BamHI-recognition sequence on the 3′ region.
  • the nucleotide sequence and the corresponding amino acid sequence for the humanized H-chain version “a” carried on the plasmid hMBC1HcDNA/pUC19 are shown in SEQ ID NO: 58 and SEQ ID NO: 56, respectively.
  • the plasmid hMBC1HcDNA/pUC19 was digested with EcoRI and BamHI to give a DNA fragment containing DNA encoding the H-chain.
  • the DNA fragment was introduced into expression plasmid pCOS1 that had been digested with EcoRI and BamHI.
  • hMBC1HcDNA/pCOS1 an expression plasmid for a humanized antibody was obtained, which was designated “hMBC1HcDNA/pCOS1”.
  • plasmid hMBC1HcDNA/pUC19 was digested with EcoRI and BamHI to give a DNA fragment containing DNA encoding the H-chain.
  • the DNA fragment was introduced into expression vector pCHO1 that had been digested with EcoRI and BamHI.
  • hMBC1HcDNA/pCHO1 an expression plasmid for the humanized antibody was obtained, which was designated “hMBC1HcDNA/pCHO1”.
  • a gene for the FR hybrid L-chain having both FRs from a humanized antibody and FRs from a mouse (chimeric) antibody was constructed, and evaluated each region for humanization.
  • a hybrid antibody having FR1 and FR2 both derived from a human antibody and FR3 and FR4 both derived from a mouse antibody was prepared by utilizing the AflII restriction site located on CDR2.
  • Plasmids MBC1L( ⁇ )/neo and hMBC1L( ⁇ )/neo (10 ⁇ g each) were separately digested in 100 ⁇ l of a reaction solution containing 10 mM Tris-HCl (pH 7.5), 10 mM MgCl 2 , 1 mM DTT, 50 mM NaCl, 0.01% (w/v) of BSA and 10 U of AflII (Takara Shuzo Co., Ltd.) at 37° C. for 1 hour.
  • the reaction solutions were subjected to electrophoresis on a 2% low-melting agarose gel, thereby giving DNA fragments of 6282 bp (referred to as “c1”) and 1022 bp (referred to as “c2”) from the plasmid MBC1L( ⁇ )/neo or DNA fragments of 6282 bp (referred to as “h1”) and 1022 bp (referred to as “h2”) from the plasmid hMBC1L( ⁇ )/neo.
  • c1 DNA fragments of 6282 bp
  • h2 DNA fragments of 6282 bp
  • h2 DNA fragments of 6282 bp
  • h2 DNA fragments of 6282 bp
  • h2 DNA fragments of 6282 bp
  • h2 DNA fragments of 6282 bp
  • h2 DNA fragments of 6282 bp
  • h2 DNA fragments of 6282 bp
  • Each of the c1 and h1 fragments (1 ⁇ g) was BAP-treated.
  • the DNA fragment was extracted with phenol and chloroform, collected by ethanol precipitation, and dissolved in 10 ⁇ l of a solution containing 10 mM Tris-HCl (pH 7.4) and 1 mM EDTA.
  • the BAP-treated c1 and h1 DNA fragments (1 ⁇ l each) were ligated to the h2 and c2 DNA fragments (4 ⁇ l each), respectively, (at 4° C. overnight).
  • Each of the ligation products was introduced into a competent cell of E. coli, JM109, to form a transformant.
  • the transformant was cultured in 2 ml of 2 ⁇ YT medium containing 50 ⁇ g/ml of ampicillin. From the cell fraction, the plasmid was purified using QIAprep Spin Plasmid Kit (QIAGEN).
  • the purified plasmid was digested in 20 ⁇ l of a reaction solution containing 10 mM Tris-HCl (pH 7.5), 10 mM MgCl 2 , 1 mM DTT, and either 2U of ApaLI (Takara Shuzo Co., Ltd.) or 8U of BamHI (Takara Shuzo Co., Ltd.) and HindIII (Takara Shuzo Co., Ltd.) at 37° C. for 1 hour. If the c1-h2 was ligated correctly, this digestion reaction gave fragments of 5560/1246/498 bp (by the ApaLI digestion) or fragments of 7134/269 bp (by the BamHI/HindIII digestion). Based on this assumption, the desired plasmids were identified.
  • the expression vector encoding the human FR1,2/mouse FR3,4 hybrid antibody L-chain was designated “h/mMBC1( ⁇ )/neo”.
  • h/mMBC1( ⁇ )/neo The expression vector encoding the human FR1,2/mouse FR3,4 hybrid antibody L-chain was designated “h/mMBC1( ⁇ )/neo”.
  • a clone for the h1-c1 could not be obtained. Therefore, recombination on a pUC vector was performed, and then the resultant recombinant product was cloned into a HEF vector.
  • plasmid hMBC1La ⁇ /pUC19 which contained DNA encoding a humanized antibody L-chain V-region without any amino acid replacements
  • plasmid hMBC1Ld ⁇ /pUC19 which contained DNA encoding a humanized antibody L-chain V-region with an amino acid replacement at the 91-position amino acid tyrosine in FR3 (i.e., the 87th amino acid in accordance with The Kabat's prescription) by isoleucine, were used as templates.
  • Plasmids MBC1L( ⁇ )/pUC19, hMBC1La ⁇ /pUC19 and hMBC1Ld ⁇ /pUC19 (10 ⁇ l each) were separately digested in 30 ⁇ l of a reaction solution containing 10 mM Tris-HCl (pH 7.5), 10 mM MgCl 2 , 1 mM DTT, 50 mM NaCl, 0.01% (w/v) of BSA, 16U of HindIII and 4U of AflII at 37° C. for 1 hour.
  • the reaction solutions were separately subjected to electrophoresis on a 2% low-melting agarose gel, thereby giving a DNA fragment 215 bp from plasmid MBC1L( ⁇ )/pUC19 (referred to as “c2′”) and a DNA fragment of 3218 bp from each of plasmids hMBC1La ⁇ /pUC19 and hMBC1Ld ⁇ /pUC19 (referred to as “ha1′” and “hd1′”, respectively).
  • c2′ DNA fragment 215 bp from plasmid MBC1L( ⁇ )/pUC19
  • ha1′ hMBC1Ld ⁇ /pUC19
  • Each of the ha1′ and hd1′ fragments was ligated to the c2′ fragment and then introduced into a competent cell of E. coli , JM109, to form a transformant.
  • the transformant was cultured in 2 ml of 2 ⁇ YT medium containing 50 ⁇ g/ml of ampicillin. From the cell fraction, the plasmid was purified using QIAprep Spin Plasmid Kit (QIAGEN).
  • the plasmids thus prepared were designated “m/hMBC1La ⁇ /pUC19” for the ha1′ fragment-containing plasmid and “m/hMBC1Ld ⁇ /pUC19” for the hd1′ fragment-containing plasmid.
  • Each of the plasmids m/hMBC1La ⁇ /pUC19 and m/hMBC1Ld ⁇ /pUC19 was digested with EcoRI.
  • the DNA fragment of 743 bp was electrophoresed on a 2% low-melting agarose gel, and then collected and purified therefrom using GENECLEANII Kit (BIO101).
  • the resultant DNA fragment was dissolved in 20 ⁇ l of a solution containing 10 mM Tris-HCl (pH 7.4) and 1 mM EDTA.
  • Each of the DNA fragments (4 ⁇ l each) was ligated to the above-obtained BAP-treated HEF vector (1 ⁇ l).
  • the ligation product was introduced into a competent cell of E. coli, JM109, to form a transformant.
  • the transformant was cultured in 2 ml of 2 ⁇ YT medium containing 50 ⁇ g/ml of ampicillin. From the cell fraction, the plasmid was purified using QIAprep Spin Plasmid Kit (QIAGEN).
  • Each of the purified plasmids was digested in 20 ⁇ l of a reaction solution containing 20 mM Tris-HCl (pH 8.5), 10 mM MgCl 2 , 1 mM DTT, 100 mM KCl, 8U of HindIII (Takara Shuzo Co., Ltd.) and 2U of PvuI (Takara Shuzo Co., Ltd.) at 37° C. for 1 hour.
  • the plasmid DNA was identified based on the expectation that if the DNA fragment was inserted in the plasmid in a correct orientation, this digestion would give a digestion fragment of 5104/2195 bp, whereas if the DNA fragment is inserted in the plasmid in the reverse orientation, this digestion would give a digestion fragment of 4378/2926 bp.
  • the plasmids thus obtained were expression vectors coding for mouse FR1,2/human FR3,4 hybrid antibody L-chain, which were designated expression vectors “m/hMBC1La ⁇ /neo” and “m/hMBC1Ld ⁇ /neo”, respectively.
  • An FR1/FR2 hybrid antibody was prepared in the same manner as mentioned above utilizing a SnaBI restriction site located on CDR1.
  • Plasmids MBC1L( ⁇ )/neo and h/mMBC1L( ⁇ )/neo (10 ⁇ g each) were separately digested in 20 ⁇ l of a reaction solution containing 10 mM Tris-HCl (pH 7.9), 10 mM MgCl 2 , 1 mM DTT, 50 mM NaCl, 0.01% (w/v) of BSA and 6U of SnaBI (Takara Shuzo Co., Ltd.) at 37° C. for 1 hour.
  • reaction solutions were further digested in 50 ⁇ l of a reaction solution containing 20 mM Tris-HCl (pH 8.5), 10 mM MgCl 2 , 1 mM DTT, 100 mM KCl, 0.01% (w/v) of BSA and 6U of PvuI at 37° C. for 1 hour.
  • the resultant reaction solutions were separately subjected to electrophoresis on a 1.5% low-melting agarose gel, thereby giving DNA fragments of 4955 bp (m1) and 2349 bp (m2) from the plasmid MBC1L( ⁇ )/neo and DNA fragments of 4955 bp (hm1) and 2349 bp (hm2) from the plasmid h/mMBC1L( ⁇ )/neo.
  • These DNA fragments were collected and purified from the gels using GENECLEANII Kit (BIO101). Each of the DNA fragments obtained was dissolved in 40 ⁇ l of a solution containing 10 mM Tris-HCl (pH 7.4) and 1 mM EDTA.
  • Each of the purified plasmids was digested in 20 ⁇ l of a reaction solution containing 10 mM Tris-HCl (pH 7.5), 10 mM MgCl 2 , 1 mM DTT and either 8U of ApaI (Takara Shuzo Co., Ltd.) or 2U of ApaLI (Takara Shuzo Co., Ltd.) at 37° C. for 1 hour.
  • the digestion reaction gave a fragment of 7304 bp (by the ApaI digestion) or fragments of 5560/1246/498 bp (by the ApaLI digestion) for m1-hm2, and gave fragments of 6538/766 bp (by the ApaI digestion) or fragments of 3535/2025/1246/498 bp (by the ApaLI digestion) for hm1-m2. Based on this assumption, the plasmids were identified.
  • an expression vector encoding a human FR1/mouse FR2,3,4 hybrid antibody L-chain (designated “hmmMBC1L( ⁇ )/neo”) and a expression vector encoding a mouse FR1/human FR2/mouse FR3,4 hybrid antibody L-chain (designated “mhmMBC1L( ⁇ )/neo”) were obtained.
  • a humanized #23-57-137-1 antibody L-chain was prepared by CDR-grafting technique by means of PCR method.
  • a humanized #23-57-137-1 antibody L-chain version “a” that contained FR1, FR2 and FR3 derived from human antibody HSU03868 (GEN-BANK, Deftos M. et al., Scand. J. Immunol., 39, 95-103, 1994) and FR4 derived from human antibody S25755 (NBRF-PDB), six PCR primers were used.
  • the six primers were as follows: CDR-grafting primers MBC1LGP1 (SEQ ID NO: 29) and MBC1LGP3 (SEQ ID NO: 30), both having a sense DNA sequence, CDR-grafting primers MBC1LGP2 (SEQ ID NO: 31) and MBC1LGP4 (SEQ ID NO: 32), both having an antisense DNA sequence, all of which had a 15-21 bp complementary sequence on the both terminal ends; and external primers MBC1LVS1 (SEQ ID NO: 33) and MBC1LVR1 (SEQ ID NO: 34) having a homology to the CDR-grafting primers MBC1LGP1 and MBC1LGP4, respectively.
  • the CDR-grafting primers MBC1LGP1, MBC1LGP2, MBC1LGP3 and MBC1LGP4 were separated on a urea-denatured polyacrylamide gel (Molecular Cloning: A Laboratory Manual, Sambrook et al., Cold Spring Harbor Laboratory Press, 1989) and extracted therefrom segment by a crush-and-soak method (Molecular Cloning: A Laboratory Manual, Sambrook et al., Cold Spring Harbor Laboratory Press, 1989). Each of the CDR-grafting primers (1 nmole) was separated with 6% denatured polyacrylamide gel. The identification of the DNA fragment of a desired length was performed on a silica gel thin plate by irradiation of UV ray. The desired DNA fragment was collected from the gel by a crush-and-soak method. The collected DNA fragment was dissolved in 20 ⁇ l of a solution containing 10 mM Tris-HCl (pH 7.4) and 1 mM EDTA.
  • the PCR reaction was performed using TaKaRa Ex Taq (Takara Shuzo Co., Ltd.) and a buffer appended thereto.
  • the PCR reaction solution comprised (per 100 ⁇ l) 1 ⁇ l of each of the CDR-grafting primers MBC1LGP1, MBC1LGP2, MBC1LGP3 and MBC1LGP4, 0.25 mM dNTPs, 2.5U of TaKaRa Ex Taq in the buffer.
  • the PCR reaction was run for 5 cycles under the conditions: 94° C. for 1 min., 55° C. for 1 min. and 72° C. for 1 min.
  • the resultant reaction mixture was added with 50 pmoles of each of the external primers MBC1LVS1 and MBC1LVR1. Using this reaction mixture, the PCR reaction was run for additional 30 cycles under the same conditions. The DNA fragment thus amplified was separated by agarose gel electrophoresis on a 3% Nu Sieve GTG agarose (FMC Bio. Products).
  • a correction primer MBC1LGP10R (SEQ ID NO: 35) was designed and synthesized.
  • the PCR reaction was performed using TaKaRa Ex Taq (Takara Shuzo Co., Ltd.) and a buffer appended thereto.
  • the PCR reaction solution comprised (per 100 ⁇ l) 0.6 ⁇ g of the plasmid hMBCL/pUC19 as a template DNA, 50 pmoles of each of the primers MBC1LVS1 and MBC1LGP10R, 2.5U of TaKaRa Ex Taq (Takara Shuzo Co., Ltd.) and 0.25 mM dNTPs in the buffer, over which mineral oil (50 ⁇ l) was layered.
  • the PCR reaction was run for 30 cycles under the conditions: 94° C. for 1 min., 55° C. for 1 min. and 72° C. for 1 min.
  • the DNA fragment thus amplified was separated by agarose gel electrophoresis on a 3% Nu Sieve GTG agarose (FMC Bio. Products).
  • a gel segment containing a DNA fragment of 421 bp was excised, and the DNA fragment was purified therefrom using GENECLEANII Kit (BIO101) in accordance with the instructions included in the kit.
  • the PCR reaction mixture thus prepared was used for subcloning of the DNA fragment into plasmid pUC19 that had been digested with BamHI and HindIII.
  • the plasmid was sequenced using M13 Primer M4 and M13 Primer RV. As a result, it was confirmed that the plasmid had the correct sequence.
  • the plasmid was then digested with HindIII and BlnI, and a DNA fragment of 416 bp was separated by electrophoresis on a 1% agarose gel.
  • the DNA fragment was purified using GENECLEANII Kit (BIO101) in accordance with the instructions included in the kit, and then introduced into plasmid C ⁇ /pUC19 that had been digested with HindIII and BlnI.
  • the resultant plasmid was designated “hMBC1La ⁇ /pUC19”.
  • This plasmid was digested with EcoRI to give a DNA fragment encoding humanized L-chain.
  • the DNA fragment was introduced into plasmid pCOS1 so that the initiation codon for the humanized L-chain was located downstream to the EF1 ⁇ promoter.
  • the plasmid thus obtained was designated “hMBC1La ⁇ /pCOS1”.
  • the DNA sequence (including the corresponding amino acid sequence) of the humanized L-chain version “a” is shown in SEQ ID NO: 66.
  • the amino acid sequence of the version “a” is also shown in SEQ ID NO: 47.
  • a humanized L-chain version “b” was prepared using a mutagenesis technique by a PCR method.
  • the version “b” was designed such that the 43-position amino acid glycine (corresponding to the 43th amino acid in accordance with the Kabat's prescription) was replaced by proline and the 49-position amino acid lysine (corresponding to the 49th amino acid accordance with the Kabat's prescription) by aspartic acid in the version “a”.
  • the PCR reaction was performed using plasmid hMBC1La ⁇ /pUC19 as a template with a mutagenic primer MBC1LGP5R (SEQ ID NO: 36) and a primer MBC1LVS1.
  • the DNA fragment obtained was digested with BamHI and HindIII, and the digestion fragment was subcloned into the BamHI-HindIII site of pUC19.
  • the plasmid was digested with HindIII and AflII, and the resultant digestion fragment was ligated to plasmid hMBC1La ⁇ /pUC19 that had been digested with HindIII and AflII.
  • the plasmid thus obtained was designated “hMBC1Lb ⁇ /pUC19”. This plasmid was digested with EcoRI to give a DNA fragment containing DNA encoding the humanized L-chain. The DNA fragment was introduced into plasmid pCOS1 such that the initiation codon for the humanized L-chain was located downstream to the EF1 ⁇ promoter. The plasmid thus obtained was designated “hMBC1Lb ⁇ /pCOS1”.
  • a humanized L-chain version “c” was prepared using a mutagenesis technique by a PCR method.
  • the version “c” was designed such that the 84-position amino acid serine (corresponding to the 80th amino acid in accordance with the Kabat's prescription) was replaced by proline.
  • the PCR reaction was performed using plasmid hMBC1La ⁇ /pUC19 as a template with a mutagenic primer MBC1LGP6S (SEQ ID NO: 37) and a primer M13 Primer RV.
  • the DNA fragment obtained was digested with BamHI and HindIII and then subcloned into pUC19 that had been digested with BamHI and HindIII.
  • the plasmid was digested with BstPI and Aor51HI, and the resultant DNA fragment was ligated to plasmid hMBC1La ⁇ /pUC19 that had been digested with BstPI and Aor51HI.
  • the plasmid thus obtained was designated “hMBC1Lc ⁇ /pUC19”.
  • This plasmid was digested with EcoRI to give a DNA fragment containing DNA encoding the humanized L-chain.
  • the fragment was introduced into the EcoRI site of plasmid pCOS1 such that the initiation codon for the humanized L-chain was located downstream to the EF1 ⁇ promoter.
  • the plasmid thus obtained was designated “hMBC1Lc ⁇ /pCOS1”.
  • Humanized L-chain versions “d”, “e” and “f” were also prepared using a mutagenesis technique by a PCR method.
  • the versions “d”, “e” and “f” were designed such that the 91-position amino acid tyrosine (corresponding to the 87th amino acid in accordance with the Kabat's prescription) was replaced by isoleucine in the versions “a”, “b” and “c”, respectively.
  • a PCR reaction was performed using each of plasmid hMBC1La ⁇ /pCOS1 (for version “d”), hMBC1Lb ⁇ /pCOS1 (for version “e”) and hMBC1Lc ⁇ /pCOS1 (for version “f”), respectively, as a template, a mutagenic primer MBC1LGP11R (SEQ ID NO: 38) and a primer M-S1 (SEQ ID NO: 44).
  • the DNA fragment thus obtained was digested with BamHI and HindIII and then subcloned into pUC19 that had been digested with BamHI and HindIII. After sequencing, the plasmid was digested with HindIII and BlnI, and the resultant digestion fragment was ligated to plasmid C ⁇ /pUC19 that had been digested with HindIII and BlnI.
  • the plasmids thus obtained were respectively designated “hMBC1Ld ⁇ /pUC19” (for version “d”), “hMBC1Le ⁇ /pUC19” (for version “e”) and “hMBC1Lf ⁇ /pUC19” (for version “f”).
  • Each of these plasmids was digested with EcoRI to give a DNA fragment containing DNA encoding the humanized L-chain.
  • the DNA fragment was introduced into the EcoRI site of plasmid pCOS1 such that the initiation codon for the humanized L-chain was located downstream to the EF1 ⁇ promoter of the plasmid.
  • hMBC1Ld ⁇ /pCOS1 for version “e”
  • hMBC1Le ⁇ /pCOS1 for version “e”
  • hMBC1Lf ⁇ /pCOS1 for version “f”.
  • Humanized L-chain versions “g” and “h” were also prepared using a mutagenesis technique by a PCR method.
  • the versions “g” and “h” were designed such that the 36-position amino acid histidine (corresponding to the 36th amino acid in accordance with the Kabat's prescription) was replaced by tyrosine in the versions “a” and “d”, respectively.
  • the PCR reaction was performed using a mutagenic primer MBC1LGP9R (SEQ ID NO: 39), M13 Primer RV and plasmid hMBC1La ⁇ /pUC19 as a template.
  • the PCR fragment obtained was digested with ApaI and HindIII and then introduced into either of plasmids hMBC1La ⁇ /pUC19 and hMBC1Ld ⁇ /pUC19 that had been digested with ApaI and HindIII.
  • the plasmids obtained were sequenced. Plasmids that were confirmed to contain the correct sequence were designated “hMBC1Lg ⁇ /pUC19” (for version “g”) and “hMBC1Lh ⁇ /pUC19” (for version “h”). Each of these plasmids was digested with EcoRI to give a DNA fragment containing DNA encoding the humanized L-chain.
  • the DNA fragment was introduced into the EcoRI site of plasmid pCOS1 such that the initiation codon for the humanized L-chain was located downstream to the EF1 ⁇ promoter.
  • the plasmids thus obtained were respectively designated “hMBC1Lg ⁇ /pCOS1” (for version “g”) and “hMBC1Lh ⁇ /pCOS1” (for version “h”).
  • Humanized L-chain versions “i”, “j”, “k”, “l”, “m”, “n” and “o” were also prepared using a mutagenesis technique by a PCR method.
  • the PCR reaction was performed using plasmid hMBC1La ⁇ /pUC19 as a template with a mutagenic primer MBC1LGP14S (SEQ ID NO: 41) and a primer V1RV ( ⁇ ) (SEQ ID NO: 43).
  • the resultant DNA fragment was digested with ApaI and BlnI and then subcloned into plasmid hMBC1Lg ⁇ /pUC19 that had been digested with ApaI and BlnI.
  • the plasmid obtained was sequenced, and the clone into which the mutation for each version was introduced was selected.
  • This plasmid was digested with EcoRI to give a DNA fragment containing DNA encoding the humanized L-chain.
  • the DNA fragment was introduced into the EcoRI site of plasmid pCOS1 such that the initiation codon for the humanized L-chain was located downstream to the EF1 promoter.
  • the DNA sequences (including the corresponding amino acid sequences) of the versions “j”, “l”, “m” and “o” are shown in SEQ ID NOs: 67, 68, 69 and 70, respectively.
  • the amino acid sequences of these versions are also shown in SEQ ID Nos: 48, 49, 50 and 51, respectively.
  • an Aor51HI restriction fragment (514 bp) in the expression plasmid hMBC1Lh ⁇ /pCOS, which containing CDR3 and a portion of FR3 and the entire FR4 was ligated, so that the 91-position amino acid tyrosine (corresponding to the 87th amino acid in accordance with the Kabat's prescription) was replaced by isoleucine.
  • the resultant plasmid was sequenced.
  • the DNA sequences (including the corresponding amino acids) of the versions “q”, “r”, “s” and “t” are shown in SEQ ID Nos: 71, 72, 73 and 74, respectively.
  • the amino acid sequences of these versions are also shown in SEQ ID Nos: 52, 53, 54 and 55, respectively.
  • Plasmid hMBC1Lq ⁇ /pCOS1 was digested with HindIII and EcoRI and then subcloned into plasmid pUC19 that had been digested with HindIII and EcoRI. The plasmid thus obtained was designated “hMBC1Lq ⁇ /pUC19.
  • E. coli strains each containing plasmids hMBC1HcDNA/pUC19 and hMBC1Lq ⁇ /pUC19 were designated “ Escherichia coli JM109 (hMBC1HcDNA/pUC19)” and “ Escherichia coli JM109 (hMBC1Lq ⁇ /pUC19)”, respectively, which have been deposited under the terms of Budapest Treaty at the National Institute of Bioscience and Human-Technology, Agency of Industrial Science and Technology, Japan, (1-3, Higashi 1-chome, Tsukuba-shi, Ibaraki, Japan) on Aug. 15, 1996, under the accession No.
  • FERM BP-5629 for Escherichia coli JM109 (hMBC1HcDNA/pUC19), and FERM BP-5630 for Escherichia coli JM109 (hMBC1Lq ⁇ /pUC19).
  • the above-prepared expression plasmids were expressed transiently in COS-7 cells.
  • each of the following combinations of plasmids were co-transfected into a COS-7 cell by electroporation using Gene Pulser (Bio Rad): hMBC1HcDNA/pCOS1 and h/mMBC1L( ⁇ )/neo; hMBC1HcDNA/pCOS1 and m/hMBC1La ⁇ /neo; hMBC1HcDNA/pCOS1 and m/hMBC1Ld ⁇ /neo; hMBC1HcDNA/pCOS1 and hmmMBC1L( ⁇ )/neo; and hMBC1HcDNA/pCOS1 and mhmMBC1L( ⁇ )/neo
  • a cell suspension (0.8 ml) of COS-7 cells in PBS( ⁇ ) (1 ⁇ 10 7 cells/ml) was added with each combination of the plasmid DNAs (10 ⁇ g each).
  • the resultant solution was applied with pulses at an electrostatic capacity of 1,500V and 25 ⁇ F.
  • the electroporated cells were suspended in DMEM medium containing 2% Ultra Low IgG fetal calf serum (GIBCO), and then cultured using a 10-cm culture dish in a CO 2 incubator. After cultivating for 72 hours, a culture supernatant was collected and centrifuged to remove cell debris.
  • the solutions thus prepared were provided for use in the ELISA below.
  • An ELISA plate for determining antibody concentration was prepared as follows. Each well of a 96-well ELISA plate (Maxisorp, NUNC) was coated with 100 ⁇ l of a coating buffer (0.1 M NaHCO 3 , 0.02% NaN 3 ) containing 1 ⁇ g/ml of goat anti-human IgG antibody (TAGO) and then blocked with 200 ⁇ l of a dilution buffer [50 mM Tris-HCl, 1 mM MgCl 2 , 0.1 M NaCl, 0.05% Tween 20, 0.02% NaN 3 , 1% bovine serum albumin (BSA); pH 7.2].
  • a coating buffer 0.1 M NaHCO 3 , 0.02% NaN 3
  • Tween 20 0.05% Tween 20
  • BSA bovine serum albumin
  • each of the wells was added with each of the serial dilutions of the COS cell culture supernatant in which each of the hybrid antibodies and the humanized antibodies was expressed, or added with each of the serial dilutions of each of the hybrid antibodies and humanized antibodies in a purified form.
  • the plate was incubated at room temperature for 1 hour and washed with PBS-Tween 20.
  • each of the wells was added with 100 ⁇ l of alkaline phosphatase-conjugated goat anti-human IgG antibody (TAGO). The plate was incubated at room temperature for 1 hour and washed with PBS-Tween 20.
  • TAGO alkaline phosphatase-conjugated goat anti-human IgG antibody
  • each of the wells was added with 1 mg/ml of a substrate solution (“Sigma 104”, p-nitrophenylphosphoric acid, SIGMA).
  • the solution in each well was measured on its absorbance at 405 nm using Microplate Reader (Bio Rad) to determine the antibody concentration.
  • Hu IgG1 ⁇ Purified The Binding Site was used as the standard.
  • An ELISA plate for determining antigen-binding ability was prepared as follows. Each well of a 96-well ELISA plate (Maxisorp, NUNC) was coated with 100 ⁇ l of a coating buffer containing 1 ⁇ g/ml of human PTHrP (1-34) and then blocked with 200 ⁇ l of a dilution buffer. Subsequently, each well was added with each of the serial dilutions of the COS-7 cell culture supernatant in which each of the hybrid antibodies and humanized antibodies was expressed, or added with each of the serial dilutions of each of the hybrid antibodies and humanized antibodies in a purified form. The plate was incubated at room temperature and washed with PBS-Tween 20.
  • each well was added with 100 ⁇ l of alkaline phosphatase-conjugated goat anti-human IgG antibody (TAGO). The plate was incubated at room temperature and washed with PBS-Tween 20. Subsequently, each well was added with 1 mg/ml of a substrate solution (“Sigma 104”, p-nitrophenylphosphoric acid, SIGMA). The solution was measured on its absorbance at 405 nm using Microplate Reader (Bio Rad).
  • TAGO alkaline phosphatase-conjugated goat anti-human IgG antibody
  • each of the above-prepared expression plasmids was introduced into a CHO cell (DXB11).
  • the expression vectors were separately cleaved with restriction enzyme PvuI to give linear DNA fragments.
  • the resultant DNA fragments were extracted with phenol and chloroform and then precipitated with ethanol.
  • the DNA fragments thus prepared were used in the subsequent electroporation. That is, the plasmid DNA fragments (10 ⁇ g each) were added to 0.8 ml of a cell suspension of CHO cells in PBS( ⁇ ) (1 ⁇ 10 7 cells/ml). The resultant solution was applied with pulses at an electrostatic capacity of 1,500V and 25 ⁇ F. After 10 min.
  • the cells thus treated were suspended in MEM- ⁇ medium (GIBCO) containing 10% fetal calf serum (GIBCO), and then cultured in a CO 2 incubator using 96-well plates (Falcon).
  • MEM- ⁇ medium GIBCO
  • the medium was replaced by ribonucleoside- or deoxyribonucleoside-free MEM- ⁇ selective medium containing 10% fetal calf serum (GIBCO) and 500 mg/ml of GENETICIN (G418Sulfate; GIBCO).
  • GENETICIN G418Sulfate
  • ROS17/2.8-5 cells were cultured in Ham'S F-12 medium (GIBCO) containing 10% fetal calf serum (GIBCO) in a CO 2 incubator.
  • the ROS17/2.8-5 cells were seeded in each well of a 96-well plate at 10 4 cells/100 ⁇ l/well and cultured for one day. After the cultivation was completed, the culture medium was replaced by Ham'S F-12 medium (GIBCO) containing 4 mM Hydrocortisone and 10% fetal calf serum.
  • the cultured cells were washed with 260 ⁇ l of Ham'S F-12 medium (GIBCO), and then added with 80 ⁇ l of Ham'S F-12 medium containing 1 mM isobutyl-1-methyl xanthine (IBMX, SIGMA), 10% fetal calf serum and 10 mM HEPES. The resultant mixture was incubated at 37° C. for 30 min.
  • the culture mediums of the mouse antibodies, the chimeric antibodies and the humanized antibodies to be tested for neutralizing activity were previously diluted serially in the following groups: [10 ⁇ g/ml, 3.3 ⁇ g/ml, 1.1 ⁇ g/ml and 0.37 ⁇ g/ml], [10 ⁇ g/ml, 2 ⁇ g/ml, 0.5 ⁇ g/ml and 0.01 ⁇ g/ml] and [10 ⁇ g/ml, 5 ⁇ g/ml, 1.25 ⁇ g/ml, 0.63 ⁇ g/ml and 0.31 ⁇ g/ml].
  • Each of the diluted antibody sample solutions was mixed with an equivalent amount of 4 ng/ml of PTHrP (1-34).
  • the resultant mixed solution (80 ⁇ l) was added to each well.
  • the final concentration of each antibody became a quarter of the above-mentioned concentration of the antibody, and accordingly the concentration of PTHrP (1-34) became 1 ng/ml.
  • the culture supernatant was removed and the residue was washed with PBS three times.
  • cAMP in the cells was extracted with 100 ⁇ l of a 0.3% HCl-95% ethanol and then evaporated using a water jet aspirator to remove the HCl-ethanol.
  • the residue was dissolved in 120 ⁇ l of EIA buffer appended to cAMP EIA Kit (CAYMAN CHEMICAL'S) to extract the cAMP therefrom.
  • the cAMP was determined using cAMP EIA Kit (CAYMAN CHEMICAL'S) in accordance with the instructions included in the kit.
  • the present invention provides a therapeutic agent for cachexia comprising, as an active ingredient, a substance capable of inhibiting the binding between PTHrP and a receptor thereof.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Endocrinology (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Molecular Biology (AREA)
  • Genetics & Genomics (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Immunology (AREA)
  • Zoology (AREA)
  • Toxicology (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Epidemiology (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Peptides Or Proteins (AREA)
  • Massaging Devices (AREA)
  • Valve Device For Special Equipments (AREA)
  • Confectionery (AREA)
  • Medicines Containing Plant Substances (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Polymerisation Methods In General (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
US09/423,800 1997-05-15 1998-05-13 Cachexia remedy Abandoned US20020165363A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US10/337,981 US7468184B2 (en) 1997-05-15 2003-01-08 Therapeutic agent for cachexia

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
JP12550597 1997-05-15
JP125505/1997 1997-05-15
JP19444597 1997-07-18
JP194445/1997 1997-07-18

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP1998/002116 A-371-Of-International WO1998051329A1 (fr) 1997-05-15 1998-05-13 Remede contre la cachexie

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US10/337,981 Division US7468184B2 (en) 1997-05-15 2003-01-08 Therapeutic agent for cachexia

Publications (1)

Publication Number Publication Date
US20020165363A1 true US20020165363A1 (en) 2002-11-07

Family

ID=26461935

Family Applications (2)

Application Number Title Priority Date Filing Date
US09/423,800 Abandoned US20020165363A1 (en) 1997-05-15 1998-05-13 Cachexia remedy
US10/337,981 Expired - Fee Related US7468184B2 (en) 1997-05-15 2003-01-08 Therapeutic agent for cachexia

Family Applications After (1)

Application Number Title Priority Date Filing Date
US10/337,981 Expired - Fee Related US7468184B2 (en) 1997-05-15 2003-01-08 Therapeutic agent for cachexia

Country Status (19)

Country Link
US (2) US20020165363A1 (ru)
EP (1) EP1004313B1 (ru)
KR (1) KR100508338B1 (ru)
CN (1) CN1329080C (ru)
AT (1) ATE361099T1 (ru)
AU (1) AU750021C (ru)
BR (1) BR9809629A (ru)
CA (1) CA2289910A1 (ru)
DE (1) DE69837707T2 (ru)
ES (1) ES2285769T3 (ru)
HK (1) HK1029047A1 (ru)
IL (1) IL132896A0 (ru)
NO (1) NO995558L (ru)
PL (1) PL193575B1 (ru)
SK (1) SK155799A3 (ru)
TR (1) TR199902800T2 (ru)
TW (1) TWI227140B (ru)
UA (1) UA74130C2 (ru)
WO (1) WO1998051329A1 (ru)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030190731A1 (en) * 1997-03-31 2003-10-09 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
US20090049561A1 (en) * 2001-05-10 2009-02-19 Chugai Seiyaku Kabushiki Kaisha Non-human animal exhibiting bone metastasis of tumor cells
US20090246140A1 (en) * 2008-03-26 2009-10-01 Neurosigma, Inc. Methods for identifying and targeting autonomic brain regions

Families Citing this family (41)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
SK40599A3 (en) 1996-09-26 2000-06-12 Chugai Pharmaceutical Co Ltd Antibody against human parathormone related peptides
SK155799A3 (en) 1997-05-15 2000-06-12 Chugai Pharmaceutical Co Ltd Cachexia remedy
WO2001002011A1 (fr) * 1999-07-02 2001-01-11 Chugai Seiyaku Kabushiki Kaisha REMEDES CONTRE LES MALADIES CAUSEES PAR PTH OU PTHrP
TWI255718B (en) 1999-07-02 2006-06-01 Chugai Pharmaceutical Co Ltd Ameliorative agent for low vasopressin concentration
AU2001252618A1 (en) 2000-04-28 2001-11-12 Chugai Seiyaku Kabushiki Kaisha Cell proliferation inhibitors
EP1312378A4 (en) * 2000-08-16 2004-03-17 Chugai Pharmaceutical Co Ltd AGENTS FOR THE RELIEF OF SYMPTOMS CAUSED BY JOINT DISEASES
DOP2006000277A (es) 2005-12-12 2007-08-31 Bayer Pharmaceuticals Corp Anticuerpos anti mn y métodos para su utilización
US9701747B2 (en) 2007-05-21 2017-07-11 Alderbio Holdings Llc Method of improving patient survivability and quality of life by anti-IL-6 antibody administration
US8404235B2 (en) * 2007-05-21 2013-03-26 Alderbio Holdings Llc Antagonists of IL-6 to raise albumin and/or lower CRP
US8252286B2 (en) 2007-05-21 2012-08-28 Alderbio Holdings Llc Antagonists of IL-6 to prevent or treat thrombosis
WO2008144763A2 (en) * 2007-05-21 2008-11-27 Alder Biopharmaceuticals, Inc. Antibodies to il-6 and use thereof
US8062864B2 (en) 2007-05-21 2011-11-22 Alderbio Holdings Llc Nucleic acids encoding antibodies to IL-6, and recombinant production of anti-IL-6 antibodies
US8178101B2 (en) 2007-05-21 2012-05-15 Alderbio Holdings Inc. Use of anti-IL-6 antibodies having specific binding properties to treat cachexia
US20090238825A1 (en) * 2007-05-21 2009-09-24 Kovacevich Brian R Novel rabbit antibody humanization methods and humanized rabbit antibodies
US7906117B2 (en) 2007-05-21 2011-03-15 Alderbio Holdings Llc Antagonists of IL-6 to prevent or treat cachexia, weakness, fatigue, and/or fever
US8420089B2 (en) 2008-11-25 2013-04-16 Alderbio Holdings Llc Antagonists of IL-6 to raise albumin and/or lower CRP
US9452227B2 (en) 2008-11-25 2016-09-27 Alderbio Holdings Llc Methods of treating or diagnosing conditions associated with elevated IL-6 using anti-IL-6 antibodies or fragments
US8323649B2 (en) 2008-11-25 2012-12-04 Alderbio Holdings Llc Antibodies to IL-6 and use thereof
US8992920B2 (en) 2008-11-25 2015-03-31 Alderbio Holdings Llc Anti-IL-6 antibodies for the treatment of arthritis
US9212223B2 (en) 2008-11-25 2015-12-15 Alderbio Holdings Llc Antagonists of IL-6 to prevent or treat thrombosis
US8337847B2 (en) 2008-11-25 2012-12-25 Alderbio Holdings Llc Methods of treating anemia using anti-IL-6 antibodies
US9724410B2 (en) 2009-11-24 2017-08-08 Alderbio Holdings Llc Anti-IL-6 antibodies or fragments thereof to treat or inhibit cachexia, associated with chemotherapy toxicity
US9775921B2 (en) 2009-11-24 2017-10-03 Alderbio Holdings Llc Subcutaneously administrable composition containing anti-IL-6 antibody
WO2011098449A1 (en) 2010-02-10 2011-08-18 Novartis Ag Methods and compounds for muscle growth
CA2818813C (en) 2010-11-23 2020-10-06 Alder Biopharmaceuticals, Inc. Anti-il-6 antibodies for the treatment of oral mucositis
CA2825165C (en) 2011-01-31 2018-07-03 Toray Industries, Inc. Therapeutic or prophylactic agent for cachexia
KR20210025132A (ko) 2012-05-11 2021-03-08 주식회사 젬백스앤카엘 항염증 활성을 갖는 펩티드 및 이를 포함하는 조성물
US9730984B2 (en) 2012-05-11 2017-08-15 Gemvax & Kael Co., Ltd. Composition for preventing or treating rheumatoid arthritis
US10967000B2 (en) 2012-07-11 2021-04-06 Gemvax & Kael Co., Ltd. Cell-penetrating peptide, conjugate comprising same and composition comprising same
ES2716870T3 (es) 2013-04-19 2019-06-17 Gemvax & Kael Co Ltd Composición para el tratamiento y prevención de lesión isquémica
CN104508485B (zh) 2013-06-07 2017-01-18 杰姆维克斯&凯尔有限公司 在癌症免疫疗法中有用的生物标记
KR20160039152A (ko) 2013-06-21 2016-04-08 주식회사 젬백스앤카엘 호르몬 분비 조절제, 이를 포함하는 조성물, 및 이를 사용한 호르몬 분비 조절 방법
RU2661596C2 (ru) 2013-10-23 2018-07-17 Джемвакс Энд Каэл Ко., Лтд. Композиция для лечения и профилактики доброкачественной гиперплазии простаты
ES2818921T3 (es) 2013-11-22 2021-04-14 Gemvax & Kael Co Ltd Péptido que tiene actividad inhibidora de la angiogénesis y composición que contiene el mismo
KR102314231B1 (ko) 2013-12-17 2021-10-19 주식회사 젬백스앤카엘 전립선 암 치료용 조성물
EP3130345B9 (en) 2014-04-11 2022-05-04 Gemvax & Kael Co., Ltd. Peptide having fibrosis inhibitory activity and composition containing same
CN106659149B (zh) 2014-04-30 2020-05-19 珍白斯凯尔有限公司 用于器官、组织或细胞移植的组合物、试剂盒和移植方法
KR102413243B1 (ko) 2014-12-23 2022-06-27 주식회사 젬백스앤카엘 안질환 치료 펩티드 및 이를 포함하는 안질환 치료용 조성물
US10835582B2 (en) 2015-02-27 2020-11-17 Gemvax & Kael Co. Ltd. Peptide for preventing hearing loss, and composition comprising same
JP6923453B2 (ja) 2015-07-02 2021-08-18 ジェムバックス アンド カエル カンパニー,リミティド 抗ウイルス活性効能を有するペプチド及びこれを含む組成物
KR20230028596A (ko) 2016-04-07 2023-02-28 김상재 텔로머라제 활성 증가 및 텔로미어 연장 효능을 가지는 펩티드 및 이를 포함하는 조성물

Family Cites Families (36)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0293158A3 (en) 1987-05-26 1990-05-09 Merck & Co. Inc. Parathyroid hormone antagonists
EP0293130A3 (en) 1987-05-26 1990-06-13 Merck & Co. Inc. Dimers of parathyroid hormone antagonists
US5001223A (en) 1987-05-26 1991-03-19 Merck & Co., Inc. Parathyroid hormone antagonists with enhanced metabolic properties
US4771124A (en) 1987-05-26 1988-09-13 Merck & Co., Inc. Parathyroid hormone antagonists with simplified synthetic methodology
JPH03504499A (ja) 1988-05-27 1991-10-03 セントカー・インコーポレーテツド 抗体試薬のための配合物
JPH03504605A (ja) 1988-05-27 1991-10-09 セントカー・インコーポレーテツド 抗体産生物の凍結乾燥した配合物
IL162181A (en) 1988-12-28 2006-04-10 Pdl Biopharma Inc A method of producing humanized immunoglubulin, and polynucleotides encoding the same
US5217896A (en) * 1988-12-30 1993-06-08 Oncogene Science, Inc. Monoclonal antibodies recognizing parathyroid hormone-like protein
JPH02207099A (ja) 1989-02-07 1990-08-16 Tonen Corp PTHrP関連ペプチド、その製造法及び用途
CA2035179C (en) 1990-01-31 2001-08-14 Gerald S. Brenner Pharmaceutical compositions containing insoluble salts of bisphosphonic acids
GB9009548D0 (en) 1990-04-27 1990-06-20 Celltech Ltd Chimeric antibody and method
JPH04228089A (ja) * 1990-05-15 1992-08-18 Kanegafuchi Chem Ind Co Ltd 高カルシウム血症治療・予防剤
EP0539491A1 (en) * 1990-07-13 1993-05-05 The Regents Of The University Of California PARATHYROID HORMONE ANALOGUES MODIFIED AT POSITIONS 3, 6 or 9
CA2107569C (en) 1991-04-05 2011-08-02 Gino V. Segre Parathyroid hormone receptor and dna encoding same
DE122009000019I1 (de) 1991-04-25 2009-07-16 Chugai Seiyaku K K 5 1 Rekombinierte humane antikörper gegen den humanen interleukin-6 rezeptor
WO1993013133A1 (en) 1991-12-20 1993-07-08 Yamanouchi Pharmaceutical Co., Ltd. HUMAN TYPE ANTIBODY REACTIVE WITH GPIIb/IIIa
US5648267A (en) 1992-11-13 1997-07-15 Idec Pharmaceuticals Corporation Impaired dominant selectable marker sequence and intronic insertion strategies for enhancement of expression of gene product and expression vector systems comprising same
US5849695A (en) 1993-01-13 1998-12-15 The Regents Of The University Of California Parathyroid hormone analogues useful for treatment of osteoporosis and disorders of calcium meatabolism in mammals
JP3504697B2 (ja) 1993-09-30 2004-03-08 株式会社先端生命科学研究所 PTHrPアンタゴニスト活性を有するポリペプチド及びそれを含むカルシウム代謝治療薬
JPH07316195A (ja) 1994-05-25 1995-12-05 Nippon Kayaku Co Ltd 新規なPTHrP関連ペプチド及びその用途
CO4410206A1 (es) * 1994-07-28 1997-01-09 Sandoz Ag DERIVADOS DE PTH o PTHrP, SU PREPARACION Y COMPOSICIONES FARMACEUTICAS QUE LAS COMPRENDEN
US5626845A (en) * 1995-01-23 1997-05-06 Xenotech Incorporated Method to ameliorate osteolysis and metastasis
EP0813423B1 (en) * 1995-01-23 2002-07-03 Xenotech Incorporated Composition to inhibit osteolysis and metastasis
US5993817A (en) 1995-01-23 1999-11-30 Xenotech Method to ameliorate osteolysis and metastasis
DE69637104T2 (de) 1995-02-20 2008-01-31 Yukio Kato Heilmittel für arthrosis deformans und entzündliche gelenkerkrankungen
ES2153564T3 (es) 1995-03-01 2001-03-01 Stryker Corp Regeneracion de la dentina inducida por un morfogeno.
KR100654645B1 (ko) 1995-04-27 2007-04-04 아브게닉스, 인크. 면역화된 제노마우스 유래의 인간 항체
US5660826A (en) 1995-06-06 1997-08-26 The Regents Of The University Of California Therapeutic sepsis treatment using antagonists to PTHrP
EP1166796A3 (en) 1996-02-01 2002-03-06 Chugai Seiyaku Kabushiki Kaisha Agents for preventing and treating thrombocytopenia
SK40599A3 (en) 1996-09-26 2000-06-12 Chugai Pharmaceutical Co Ltd Antibody against human parathormone related peptides
SK155799A3 (en) 1997-05-15 2000-06-12 Chugai Pharmaceutical Co Ltd Cachexia remedy
JP4372240B2 (ja) 1997-05-15 2009-11-25 中外製薬株式会社 悪液質治療剤
JP4414494B2 (ja) 1998-02-03 2010-02-10 旭化成ファーマ株式会社 白血球減少症の予防剤および治療剤
ES2302374T3 (es) 1998-05-05 2008-07-01 Societe De Conseils De Recherches Et D'applications Scientifiques S.A.S. Compuesto selectivo de receptores pth2.
JP2000080100A (ja) 1998-06-17 2000-03-21 Japan Tobacco Inc 副甲状腺ホルモン関連タンパクに対するヒトモノクローナル抗体
CA2332128A1 (en) 1998-06-26 2000-01-06 Chugai Seiyaku Kabushiki Kaisha Therapeutic agent for hypercalcemic crisis

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030190731A1 (en) * 1997-03-31 2003-10-09 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
US20090049561A1 (en) * 2001-05-10 2009-02-19 Chugai Seiyaku Kabushiki Kaisha Non-human animal exhibiting bone metastasis of tumor cells
US20090246140A1 (en) * 2008-03-26 2009-10-01 Neurosigma, Inc. Methods for identifying and targeting autonomic brain regions
US8257684B2 (en) 2008-03-26 2012-09-04 Neurosigma, Inc. Methods for identifying and targeting autonomic brain regions

Also Published As

Publication number Publication date
PL193575B1 (pl) 2007-02-28
CN1329080C (zh) 2007-08-01
PL336797A1 (en) 2000-07-17
DE69837707T2 (de) 2008-01-10
TR199902800T2 (xx) 2000-04-21
CA2289910A1 (en) 1998-11-19
AU750021C (en) 2003-10-16
EP1004313B1 (en) 2007-05-02
NO995558D0 (no) 1999-11-12
IL132896A0 (en) 2001-03-19
AU7236998A (en) 1998-12-08
BR9809629A (pt) 2000-07-04
DE69837707D1 (de) 2007-06-14
KR100508338B1 (ko) 2005-08-17
NO995558L (no) 2000-01-12
EP1004313A1 (en) 2000-05-31
TWI227140B (en) 2005-02-01
AU750021B2 (en) 2002-07-11
UA74130C2 (ru) 2005-11-15
EP1004313A4 (en) 2005-01-19
SK155799A3 (en) 2000-06-12
CN1255858A (zh) 2000-06-07
ATE361099T1 (de) 2007-05-15
US7468184B2 (en) 2008-12-23
ES2285769T3 (es) 2007-11-16
HK1029047A1 (en) 2001-03-23
KR20010012613A (ko) 2001-02-15
US20030138424A1 (en) 2003-07-24
WO1998051329A1 (fr) 1998-11-19

Similar Documents

Publication Publication Date Title
AU750021B2 (en) Cachexia remedy
US20030124119A1 (en) Stable antibody compositions and injection preparations
AU744146B2 (en) Antibody against human parathormone related peptides
AU753131B2 (en) Remedies for hypercalcemic crisis
JP4372240B2 (ja) 悪液質治療剤
US20040067231A1 (en) Agents for ameliorating symtoms caused by joint diseases
EP1197225A1 (en) REMEDIES FOR DISEASES CAUSED BY PTH OR PTHrP
US7655227B1 (en) Agents for ameliorating low vasopressin level
AU2003203622B2 (en) Cachexia remedy
US20030049211A1 (en) Remedies and preventives for dental diseases
EP1195164A1 (en) Remedies for drug-resistant hypercalcemia
MXPA99010364A (en) Cachexia remedy
CZ398499A3 (cs) Léčivo proti kachexii
AU2004222761A1 (en) Remedies for drug-resistant hypercalcemia
JP2006306895A (ja) 悪液質治療剤
CA2401357A1 (en) Tissue degradation inhibitor agent
AU2005203262A1 (en) Remedies for diseases caused by PTH or PTHrP

Legal Events

Date Code Title Description
AS Assignment

Owner name: CHUGAI SEIYAKU KABUSHIKI KAISHA, JAPAN

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:SATO, KOH;TSUNENARI, TOSHIAKI;ISHII, KIMIE;REEL/FRAME:010512/0462

Effective date: 19991102

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION