US20040236080A1 - Cell proliferation inhibitors containing anti-glypican 3 antibody - Google Patents

Cell proliferation inhibitors containing anti-glypican 3 antibody Download PDF

Info

Publication number
US20040236080A1
US20040236080A1 US10/481,524 US48152404A US2004236080A1 US 20040236080 A1 US20040236080 A1 US 20040236080A1 US 48152404 A US48152404 A US 48152404A US 2004236080 A1 US2004236080 A1 US 2004236080A1
Authority
US
United States
Prior art keywords
antibody
glypican
cells
cell
antibodies
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
US10/481,524
Other languages
English (en)
Inventor
Hiroyuki Aburatani
Tetsuo Nakamura
Masayuki Tsuchiya
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
Perseus Proteomics Inc
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 ABURATANI, HIROYUKI, CHUGAI SEIYAKU KABUSHIKI KAISHA reassignment ABURATANI, HIROYUKI ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: ABURATANI, HIROYUKI, NAKAMURA, TETSUO, TSUCHIYA, MASAYUKI
Publication of US20040236080A1 publication Critical patent/US20040236080A1/en
Assigned to PERSEUS PROTEOMICS INC. reassignment PERSEUS PROTEOMICS INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: ABURATANI, HIROYUKI
Assigned to CHUGAI SEIYAKU KABUSHIKI KAISHA reassignment CHUGAI SEIYAKU KABUSHIKI KAISHA ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: MATSUDA, KAZUYUKI
Assigned to CHUGAI SELYAKU KABUSHIKI KAISHA reassignment CHUGAI SELYAKU KABUSHIKI KAISHA CORRECTIVE ASSIGNMENT TO CORRECT THE ASSIGNOR'S NAME. DOCUMENT PREVIOUSLY RECORDED AT REEL 018428 FRAME 0185 Assignors: PERSEUS PROTEOMICS INC.
Priority to US11/702,780 priority Critical patent/US7744880B2/en
Priority to US12/799,491 priority patent/US8263077B2/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • C07K16/3076Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells against structure-related tumour-associated moieties
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/34Identification of a linear epitope shorter than 20 amino acid residues or of a conformational epitope defined by amino acid residues
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • C07K2317/732Antibody-dependent cellular cytotoxicity [ADCC]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • C07K2317/734Complement-dependent cytotoxicity [CDC]

Definitions

  • the present invention relates to a cell growth inhibitor containing an anti-glypican 3 antibody as an active ingredient:
  • the glypican family has been reported to be present as a new family of heparan sulfate proteoglycans that are present on cell surfaces.
  • the 5 types of glypicans (glypican 1, glypican 2, glypican 3, glypican 4 and glypican 5) have been reported as members of the glypican family.
  • These members of the family have core proteins of uniform size (approximately 60 kDa), share specific and well-conserved cysteine sequences, and bind to cell membranes via glycosyl phosphatidyl inositol (GPI) anchors.
  • GPI glycosyl phosphatidyl inositol
  • a Dally (division abnormally delayed) gene has been identified by screening for genes of a variant of Drosophila melanogaster having an abnormal cell division pattern in the development of the central nervous system.
  • the cDNA of Dally is known to represent an open reading frame (ORF) encoding a product having a sequence that shows homology (24 to 26% homology) with a membrane-spanning proteoglycan (GRIPs) of a vertebrate containing all the features of a glypican.
  • ORF open reading frame
  • GRIPs membrane-spanning proteoglycan
  • the dally gene plays a role in regulating the dpp (decapentaplegia) receptor mechanism.
  • the glypican of a mammal may regulate the signal transduction between TGF and BMP.
  • glypican may function as a common receptor for some of heparin-binding growth factors (e.g., EGF, PDGF, BMP2 and FGF's).
  • Glypican 3 has been isolated as a transcript under developmental regulation in rat intestine (Filmus, J., Church, J. G., and Buick, R. n. (1988) Mol. Cell Biol.8, 4243-4249), and then identified as OCT-5, a GPI-linked heparan sulfate proteoglycan of the glypican family, which has a core protein with a molecular weight of 69 kDa (Filmus, J., Shi, W., Wong, Z.-M., and Wong, M. J. (1995) Biochem. J. 311, 561-565).
  • glypican 3 has been isolated as MXR-7 from a human gastric cancer cell line (Hermann Lü et al., Gene 188 (1997) 151-156). Glypican 3 has been reported to form a protein-protein complex with an insulin-like growth factor-2 so as to regulate the action of the growth factor (Pilia, G. et al, (1996) Nat. Genet.12, 241-247). This report suggests that glypican 3 does not always interact with growth factor having the heparan sulfate chain.
  • glypican may function as a receptor for endostatin that may act as a vascularization inhibitor (Molecular Cell (2001), 7, 811-822).
  • endostatin may act as a vascularization inhibitor
  • An object of the present invention is to provide a cell growth inhibitor containing an anti-glypican 3 antibody as an active ingredient.
  • anti-glypican 3 antibody exerts cell proliferation-inhibiting activity by ADCC (antibody-dependent cell-mediated cytotoxicity) activity and CDC (complement-dependent cytotoxicity) activity. Furthermore, it is also predicted that anti-glypican 3 antibodies also exert cell proliferation-inhibiting activity by inhibiting the action of a growth factor. Furthermore, anti-glypican 3 antibodies can also exert cell proliferation-inhibiting activity by binding with cytotoxic substances such as a radioactive isotope, a chemotherapeutant or toxin derived from bacteria.
  • cytotoxic substances such as a radioactive isotope, a chemotherapeutant or toxin derived from bacteria.
  • the present invention is as follows:
  • a cell growth inhibitor containing an anti-grlypican 3 antibody as an active ingredient
  • the cell growth inhibitor of (4) wherein the cells are selected from the group consisting of hepatic cancer cells, lung cancer cells, colon cancer cells, mammary cancer cells, prostate cancer cells, leukemia cells, lymphoma cells and pancreatic cancer cells;
  • the present invention is a cell growth inhibitor containing an anti-glypican antibody as an active ingredient. Furthermore, the present invention is a cell growth inhibitor containing the anti-glypican antibody as an active ingredient that can be used for the therapy against diseases based on abnormal cell proliferation, and particularly against cancer.
  • Examples of the anti-glypican 3 antibody of the present invention include a known antibody such as a humanized antibody, a human antibody (WO96/33735), a chimeric antibody (JP Patent Publication (Kokai) No. 4-228089 A (1992) ) or a mouse antibody, as well as antibodies in the present invention.
  • an antibody may be a polyclonal antibody, and is preferably a monoclonal antibody.
  • the anti-glypican 3 antibody used in the present invention may be derived from any origin, may be of any type (monoclonal or polyclonal) and may be in any form, as long as it is capable of inhibiting cell proliferation.
  • the anti-glypican 3 antibody used in the present invention can be obtained by a known means as a polyclonal or a monoclonal antibody.
  • a particularly preferred anti-glypican 3 antibody used in the present invention is a monoclonal antibody derived from a mammal.
  • Examples of the monoclonal antibody derived from a mammal include an antibody produced by a hybridoma and an antibody produced by a host transformed using an expression vector containing the antibody gene by genetic engineering techniques. This antibody binds to glypican 3 so as to inhibit cell proliferation.
  • An example of such an antibody is a monoclonal antibody produced by the hybridoma clone of the present invention.
  • a monoclonal antibody-producing hybridoma can be basically prepared using known techniques as follows. That is, the hybridoma can be prepared by performing immunization using glypican 3 as an immunogen according to a standard immunization method, causing the thus obtained immunocytes to fuse with known parent cells by a standard cell fusion method, and then screening for monoclonal antibody-producing cells by a standard screening method.
  • monoclonal antibodies can be prepared as follows.
  • Human glypican 3 to be used as an immunogen to obtain antibodies is first obtained by expressing the glypican 3 (MXR7) gene (amino acid sequence) as disclosed by Lü, H. et al (Gene 188 (1997), 151-156). Specifically, the gene sequence encoding glypican 3 is inserted in a known expression vector system, an appropriate host cell is transformed, and then a target human glypican 3 protein is purified by a known method from the host cells or the culture supernatant.
  • MXR7 amino acid sequence
  • this purified glypican 3 protein is used as an immunogen.
  • the partial peptide of glypican 3 can be used as a sensitization antigen.
  • the partial peptide can be obtained by chemical synthesis from the amino acid sequence of human glypican 3.
  • Anti-glypican 3 antibody inhibits cell proliferation activity with the ADCC action, the CDC action and the activity of a growth factor. Moreover, the anti-glypican 3 antibody can also inhibit cell proliferation by binding with a cytotoxic substance such as a radioisotope, a chemotherapeutant or a toxin derived from bacteria.
  • a cytotoxic substance such as a radioisotope, a chemotherapeutant or a toxin derived from bacteria.
  • an epitope on a glypican 3 molecule which is recognized by the anti-glypican 3 antibody is not limited to a particular epitope.
  • the anti-glypican 3 antibody may recognize any epitope, as long as the epitope is present on a glypican 3 molecule. Accordingly, any fragment can be used as an antigen for preparing the anti-glypican 3 antibody of the present invention, as long as it contains the epitope on a glypican 3 molecule.
  • a mammal to be immunized with an immunogen is not specifically limited, and is preferably selected in consideration of compatibility with a parent cell to be used for cell fusion.
  • rodents such as mice, rats, hamsters or rabbits, or monkeys are generally used.
  • Animals are immunized with an immunogen according to a known method.
  • immunization is performed by a general method wherein a mammal is injected intraperitoneally or subcutaneously with an immunogen.
  • an immunogen is diluted with or suspended in an appropriate volume of PBS (Phosphate-Buffered Saline), physiological saline or the like; an appropriate volume of a standard adjuvant such as a Freund's complete adjuvant is mixed with the product if necessary; emulsification is performed; and then the solution is administered to mammals several times every 4 to 21 days.
  • PBS Phosphate-Buffered Saline
  • physiological saline physiological saline
  • an appropriate volume of a standard adjuvant such as a Freund's complete adjuvant
  • emulsification is performed; and then the solution is administered to mammals several times every 4 to 21 days.
  • an appropriate carrier can also be used upon immunization with an immunogen.
  • Mammals are immunized as described above, and then an increased titer of a desired antibody in the serum is confirmed. Subsequently, immunocytes are collected from the mammals, and then subjected to cell fusion.
  • a preferred immunocyte is particularly a splenocyte.
  • a mammalian myeloma cell As a partner cell to be fused with the above immunocyte, a mammalian myeloma cell is used.
  • a cell line of a myeloma cell that is preferably used herein include various known cell lines such as P3 (P3 ⁇ 63 Ag8.653) (J. Immnol. (1979) 123, 1548-1550), P3 ⁇ 63 Ag8U.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 above immunocytes with myeloma cells can be basically performed according to a known method, for example, the method of Kohler and Milstein et al (Kohler. G. and Milstein, C., Methods Enzymol. (1981) 73, 3-46).
  • the above cell fusion is performed in a standard nutrition culture solution in the presence of, for example, a cell-fusion accelerator.
  • a cell-fusion accelerator for example, polyethylene glycol (PEG), hemagglutinating virus of Japan (HVJ) or the like is used.
  • PEG polyethylene glycol
  • HVJ hemagglutinating virus of Japan
  • an adjuvant such as dimethylsulfoxide can also be used by addition to further enhance fusion efficiency.
  • any ratio of immunocytes to myeloma cells may be set for use herein.
  • the number of immunocytes be 1 to 10 times greater than that of myeloma cells.
  • a culture solution to be used for the above cell fusion for example, a RPM11640 culture solution or a MEM culture solution which is appropriate for the growth of the above myeloma cell line, or other standard culture solutions that are used for this type of cell culture can be used.
  • a serum fluid such as fetal calf serum (FCS) can be used in combination therewith.
  • Cell fusion is performed by mixing sufficiently certain amounts of the above immunocytes and myeloma cells in the above culture solution, adding a PEG (e.g., with an average molecular weight of approximately 1000 to 6000) solution (a general concentration of 30 to 60% (w/v)) pre-heated at approximately 37° C. with, and then mixing the solution, so as to form target fused cells (hybridomas). Subsequently, an appropriate culture solution is added successively, and then a step of removing the supernatant by centrifugation is repeated, so that reagents for cell fusion or the like that is unfavorable for the growth of the hybridomas is removed.
  • a PEG e.g., with an average molecular weight of approximately 1000 to 6000
  • a general concentration of 30 to 60% (w/v) a general concentration of 30 to 60% (w/v)
  • hybridomas are selected by culturing the hybridomas in a standard selective culture solution such as a HAT culture solution (a culture solution containing hypoxanthine, aminopterin and thymidine). Culture in the above HAT culture solution is continued for a time period sufficient for the cells (unfused cells) other than the target hybridomas to die (normally, several days to several weeks). Subsequently, a standard limiting dilution method is conducted, so that screening for and monocloning of hybridomas that produce a target antibody are performed.
  • a standard selective culture solution such as a HAT culture solution (a culture solution containing hypoxanthine, aminopterin and thymidine). Culture in the above HAT culture solution is continued for a time period sufficient for the cells (unfused cells) other than the target hybridomas to die (normally, several days to several weeks). Subsequently, a standard limiting dilution method is conducted, so that screening for and monocloning of hybridomas that produce
  • desired human antibodies having binding activity to glypican 3 can also be obtained (see Japanese Patent Publication (Kokoku) No. 1-59878 B (1989)) by sensitizing in vitro human lymphocytes with glypican 3, and causing the sensitized lymphocytes to fuse with the human-derived myeloma cells having a permanent division potential.
  • glypican 3 as an antigen is administered to a transgenic animal having all the repertories of a human antibody gene to obtain anti-glypican 3 antibody-producing cells, and then human antibodies for glypican 3 may be obtained from the immortalized anti-glypican 3 antibody-producing cells (see International Patent Publication Nos. WO 94/25585, WO 93/12227, WO 92/03918 and WO 94/02602).
  • the thus prepared hybridomas producing monoclonal antibodies can be passage-cultured in a standard culture solution, or can be stored for a long period in liquid nitrogen.
  • One example of a method employed to obtain monoclonal antibodies from the hybridomas involves culturing - the hybridomas and obtaining monoclonal antibodies in the culture supernatant according to a standard method. Another method involves administering the hybridomas to mammals that are compatible with the hybridomas to cause them to proliferate, and obtaining monoclonal antibodies in the ascites. The former method is suitable to obtain antibodies of high purity. On the other hand, the latter method is suitable for the mass production of antibodies.
  • a monoclonal antibody that can be used in the present invention is a recombinant monoclonal antibody that is prepared by cloning the antibody gene from the hybridoma, incorporating the gene into an appropriate vector, introducing the vector into a host, and then causing the host to produce the recombinant monoclonal antibodies by genetic engineering techniques (e.g., see Vandamme, A. M. et al., Eur. J. Biochem. (1990) 192, 767-775, 1990). Specifically, mRNA encoding the variable (V) region of an anti-glypican 3 antibody is isolated from a hybridoma producing the anti-glypican 3 antibody.
  • mRNA is isolated by a known method such as a guanidine ultracentrifugal method (Chirgwin, J. M. et al., Biochemistry (1979) 18, 5294-5299) or an AGPC method (Chomczynski, P et al., Anal. Biochem. (1987) 162, 156-159), thereby preparing total RNA.
  • Target mRNA is then prepared using an mRNA Purification Kit (Pharmacia) or the like.
  • mRNA can also be directly prepared using a QuickPrep mRNA Purification Kit (Pharmacia).
  • the cDNA of the antibody V region is synthesized using reverse transcriptase from the thus obtained mRNA.
  • cDNA is synthesized using an AMV Reverse Transcriptase First-strand cDNA Synthesis Kit (SEIKAGAKU CORPORATION) or the like.
  • SEIKAGAKU CORPORATION AMV Reverse Transcriptase First-strand cDNA Synthesis Kit
  • a 5′-Ampli FINDER RACE Kit (Clontech) and the 5′-RACE method using PCR (Frohman, M. A. et al., Proc. Natl. Acad. Sci. USA (1988) 85, 8998-9002, Belyavsky, A. et al., Nucleic Acids Res. (1989) 17, 2919-2932) can be employed.
  • a target DNA fragment is purified from the thus obtained PCR product, and then ligated to a vector DNA. Furthermore, a recombinant vector is prepared from the product, and then the vector is introduced into Escherichia coli or the like, consecutively, colonies are selected, thereby preparing a desired recombinant vector. The nucleotide sequence of a target DNA is then confirmed by a known method, such as a dideoxynucleotide chain termination method.
  • the antibody gene is incorporated into an expression vector so that the gene is expressed under the regulation of the gene expression control region including, for example, an enhancer and a promoter.
  • a host cell is transformed with the expression vector, causing the host to express the antibody.
  • An antibody gene can be expressed by incorporating a DNA encoding the antibody heavy chain (H-chain) or a DNA encoding the antibody light chain (L-chain) separately into expression vectors, and then simultaneously transforming a host cell with the vectors; or by incorporating DNAs encoding the H-chain and the L-chain into a single expression vector, and then transforming a host cell with the vector (see WO 94/11523).
  • a transgenic animal can also be used to produce a recombinant antibody.
  • an antibody gene is inserted in a gene encoding a protein (e.g., goat ⁇ casein) uniquely produced in milk, thereby preparing as a fused gene.
  • a DNA fragment containing the fused gene into which the antibody gene has been inserted is injected into a goat embryo, and then such embryo is introduced into a female goat.
  • Desired antibodies can be obtained from the milk produced by the transgenic goat or the offspring born from the goat that has accepted the embryo.
  • hormones can be administrated for the transgenic goat (Ebert, K. M. et al., Bio/Technology (1994) 12, 699-702).
  • artificially altered gene recombinant antibodies such as chimeric antibodies or humanized antibodies can be used for, for example, lowering heteroantigenicity against a human.
  • altered antibodies can be produced using a known method.
  • Chimeric antibodies can be obtained by ligating the DNA encoding the above antibody V-region to a DNA encoding a human antibody C-region, incorporating the product into an expression vector, and then introducing the vector into a host to cause the host to produce the antibodies. Using this known method, chimeric antibodies useful in the present invention can be obtained.
  • Humanized antibodies are also referred to as reshaped human antibodies, which are prepared by grafting an antibody CDR (complementarity determining region) of a mammal other than a human, such as a mouse, to the CDR of a human antibody.
  • the general gene recombination technique thereof is also known (see European Patent Application Publication No. EP 125023 and WO 96/02576).
  • the DNA sequence is synthesized by the PCR method using as primers several oligonucleotides that have been prepared to have a portion overlapping the terminal regions of both mouse antibody CDR and the framework region (FR) of a human antibody (see the method as described in WO 98/13388).
  • the framework region ligated to the CDR. having a good antigen binding site is selected. Amino acids in the framework region in the antibody variable region may be substituted as required, so that the CDR of a reshaped human antibody forms an appropriate antigen-binding site (Sato, K. et al., Cancer Res. (1993) 53, 851-856).
  • Regions of a human antibody are used for the C regions of a chimeric antibody and a humanized antibody.
  • C ⁇ 1, C ⁇ 2, C ⁇ 3 or C ⁇ 4C, and for the L-chain C ⁇ or C ⁇ can be used.
  • the human antibody C-region may be modified.
  • a chimeric antibody consists of the variable region of an antibody derived from a mammal other than a human, and a constant region derived from a human antibody.
  • a humanized antibody consists of the CDR of an antibody derived from a mammal other than a human, and the framework region and C region derived from a human antibody. Since the antigenicity of the humanized antibody is designed to be low in a human body, it is useful as an active ingredient of a therapeutic agent of the present invention.
  • the antibody used in the present invention is not limited to the whole molecule as long as it binds to glypican 3 and suppresses cell proliferation, and may be a fragment of the antibody or the modified product thereof. Both a bivalent antibody and a monovalent antibody are included. Examples of the fragment of an antibody include Fab, F(ab′) 2 , Fv, Fab/c having one Fab and a complete Fc, and a single chain Fv (scFv) wherein the Fv of the H-chain or the L-chain is ligated with an appropriate linker.
  • an antibody fragment is synthesized by treating the antibody with an enzyme such as papain or pepsin, or genes encoding these antibody fragments are constructed, the genes are introduced into expression vectors, and the genes are then expressed by appropriate host cells (see e.g., 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. & Skerra, A.
  • scFv is obtained by linking the H-chain V-region and the L-chain V-region of antibodies.
  • the H-chain V-region and the L-chain V-region are linked via a linker, or preferably a peptide linker (Huston, J. S. et al., Proc. Natl. Acad. Sci. U.S.A. (1988) 85, 5879-5883).
  • the H-chain V-region and the L-chain V-region in scFv may be derived from any of those described as antibodies in this specification.
  • As a peptide linker to link the V-regions for example, any single-stranded peptide comprising 12 to 19 amino acid residues is used.
  • a DNA encoding scFv can be obtained as follows. Amplification is performed by the PCR method using as templates the entire or DNA portions encoding desired amino acid sequences (of a DNA encoding the H-chain or the H-chain V-region of the above antibody, and a DNA encoding the L-chain or the L-chain V-region), and using a primer pair that specifies both ends. Amplification is then further performed by a combined use of a DNA encoding a peptide linker portion and a primer pair that specify to cause both ends to ligate respectively to the H-chain and L-chain.
  • expression vectors containing the DNAs, and hosts transformed with the expression vectors can be obtained according to the standard method.
  • scFv can be obtained according to the standard method.
  • antibody fragments can be produced using hosts by obtaining the genes thereof in a manner similar to the above method, and then causing the expression of the genes.
  • the “antibody” in the present invention also encompasses these antibody fragments.
  • a modified antibody anti-glypican antibodies bound to polyethylene glycol (PEG) or one of various molecules such as a cytotoxic substance can be used.
  • PEG polyethylene glycol
  • the “antibody” in the present invention also encompasses these modified antibodies.
  • Such a modified antibody can be obtained by chemically modifying the obtained antibody.
  • an antibody modification method has already been established in the art.
  • the antibody used in the present invention may be a bispecific antibody.
  • the bispecific antibody may have antigen-binding sites that recognize a different epitope on a glypican 3 molecule.
  • one antigen-binding site may recognize glypican 3 and the other antigen-binding site may recognize a cytotoxic substance such as a chemotherapeutant, toxin derived from cells, radioactive substance or the like.
  • a cytotoxic substance is allowed to directly act on a cell expressing glypican 3 to specifically damage tumor cells, so that tumor cell proliferation can be suppressed.
  • a bispecific antibody can be prepared by binding H-L pairs of two types of antibodies, and it can also be obtained by fusing hybridomas producing different monoclonal antibodies to prepare bispecific antibody-producing fused cells. Furthermore, a bispecific antibody can also be prepared by genetic engineering techniques.
  • Antibody genes constructed as described above can be expressed and thus obtained by a known method.
  • a gene can be expressed by operably linking a useful promoter that is generally employed and the antibody gene to be expressed, and by linking a polyA signal downstream on the 3′ side thereof.
  • a promoter/enhancer is, for example, a human cytomegalovirus immediate early promoter/enhancer.
  • examples of another promoter/enhancer that can be used in the present invention for antibody expression include a virus promoter/enhancer such as a retrovirus, a polyoma virus, an adenovirus or a simian virus 40 (SV40), or a promoter/enhancer derived from a mammalian cell such as human elongation factor 1a (HEF1a).
  • a virus promoter/enhancer such as a retrovirus, a polyoma virus, an adenovirus or a simian virus 40 (SV40), or a promoter/enhancer derived from a mammalian cell such as human elongation factor 1a (HEF1a).
  • gene expression can be readily performed by the method of Mulligan et al (Nature (1979) 277, 108) and when a HEF1a promoter/enhancer is used, gene expression can be readily performed by the method of Mizushima et al (Nucleic Acids Res. (1990) 18, 5322).
  • a useful promoter that is generally used, a signal sequence for antibody secretion, and an antibody gene to be expressed are operably linked, so that the gene can be expressed.
  • a promoter include a lacz promoter and an araB promoter.
  • the antibody gene can be expressed by the method of Ward et al (Nature (1098) 341, 544-546; FASEB J. (1992) 6, 2422-2427), or when the araB promoter is used, the antibody gene can be expressed by the method of Better et al (Science (1988) 240, 1041-1043).
  • a pelB signal sequence (Lei, S. P. et al J. Bacteriol. (1987) 169, 4379) may be used when the antibody is produced in the periplasm of Escherichia coli. After antibodies produced in the periplasm are isolated, the structure of the antibody is appropriately refolded and used.
  • a replication origin that can be used is derived from a SV40, a polyoma virus, an adenovirus, a bovine papilloma virus (BPV) or the like.
  • an expression vector can contain an aminoglycoside transferase (APH) gene, a thymidine kinase (TK) gene, an Escherichia coli xanthine guanine phosphoribosyltransferase (Ecogpt) gene, a dihydrofolate reductase (dhfr) gene or the like as a selection marker.
  • APH aminoglycoside transferase
  • TK thymidine kinase
  • dhfr dihydrofolate reductase
  • any expression systems such as a eukaryotic cell system or a prokaryotic cell system can be used.
  • eukaryotic cells include animal cells such as cells of an established mammalian cell system or an insect cell system, and filamentous fungus cells and yeast cells.
  • prokaryotic cells include bacterial cells such as Escherichia coli cells.
  • antibodies used in the present invention are expressed in mammalian cells such as CHO, COS, myeloma, BHK, Vero or HeLa cells.
  • a transformed host cell is cultured in vitro or in vivo, so as to cause the host cell to produce a target antibody.
  • Host cells are cultured according to a known method.
  • DMEM fetal calf serum
  • MEM fetal calf serum
  • IMDM fetal calf serum
  • the antibodies expressed and produced as described above can be isolated from the cells or host animals, and purified to a uniform level. Isolation and purification of the antibodies to be used in the present invention can be performed using affinity columns.
  • An example of a column using a protein A column is a Hyper D, POROS, Sepharose F. F. (Pharmacia).
  • Other standard isolation and purification methods that are employed for proteins may be used, and there is no limitation regarding their use.
  • a chromatography column other than the above affinity column, a filter, ultrafiltration, a method of salting out, dialyses and the like may be appropriately selected and combined for use, so that antibodies can be isolated and purified (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 a sample containing anti-glypican 3 antibodies, such as the culture supernatant of anti-glypican 3 antibody-producing cells or purified antibodies are added to a plate coated with glypican 3.
  • a secondary antibody labeled with an enzyme such as alkaline phosphatase is added.
  • the plate is then incubated and then washed, an enzyme substrate such as p-nitrophenyl phosphoric acid is added, and then absorbance is measured, so that antigen-binding activity can be evaluated.
  • the antibodies used in the present invention have ADCC activity or CDC activity as cytotoxic activity.
  • ADCC activity can be measured by mixing effector cells, target cells and anti-glypican 3 antibodies, and then examining the degree of ADCC.
  • effector cells for example, mouse splenocytes, human peripheral blood or monocytes isolated from the bone marrow can be used.
  • target cells for example, human established cell lines such as HuH-7 human hepatic cancer cell line can be used.
  • Target cells are previously labeled with 51 Cr, anti-glypican 3 antibodies are added to the cells, and then incubation is performed.
  • effector cells are added at an appropriate ratio of the cells to the target cells, and then incubation is performed. After incubation, the supernatant is collected and radioactivity in the supernatant is counted, so that ADCC activity can be measured.
  • CDC activity can be measured by mixing the above labeled target cells and anti-glypican 3 antibodies, adding complements, performing incubation, culturing, and then counting radioactivity in the supernatant.
  • Antibodies need an Fc portion to exert cytotoxic activity.
  • the anti-glypican 3 antibody used in the present invention is required to contain the Fc portion.
  • the anti-glypican 3 antibody of the present invention can be used to inhibit vascularization.
  • the cell growth inhibitor of the present invention is used to treat or improve conditions arising from disease based on abnormal cell proliferation, and particularly, cancer.
  • Preferred examples of the target carcinoma cells of the cell growth inhibitor of the present invention include, but are not specifically limited to, hepatic cancer cells, lung cancer cells, colon cancer cells, mammary cancer cells, prostate cancer cells, leukemia cells, lymphoma cells and pancreatic cancer cells. Hepatic cancer cells are particularly preferred.
  • Effective dose is selected from the range of 0.001 mg to 1000 mg per kg in body weight per administration. Alternatively, a dose in the range of 0.01 to 100000 mg/body per patient can be selected. However, the dose of the therapeutic agent containing anti-glypican 3 antibodies of the present invention is not limited to these doses.
  • the agent can be administered either before or after the onset of the clinical symptoms of a disease.
  • the therapeutic agent containing anti-glypican 3 antibodies as an active ingredient of the present invention can be formulated according to standard methods (Remington's Pharmaceutical Science, latest edition, Mark Publishing Company, Easton, U.S.A.), and may contain a pharmaceutically acceptable carrier and an additive together.
  • Examples of such carriers and pharmaceutical additives include water, a pharmaceutically acceptable organic solvent, collagen, polyvinyl alcohol, polyvinylpyrrolidone, carboxyvinyl polymer, sodium carboxymethylcellulose, sodium polyacrylate, sodium alginate, water-soluble dextran, sodium carboxymethylstarch, pectin, methyl cellulose, ethyl cellulose, xanthan 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 surfactant that is acceptable as a pharmaceutical additive.
  • a pharmaceutically acceptable organic solvent collagen
  • polyvinyl alcohol polyvinylpyrrolidone
  • carboxyvinyl polymer sodium carboxymethylcellulose, sodium polyacrylate, sodium alginate, water-soluble dextran, sodium carboxymethylstarch, pe
  • the agent that can be used as a pharmaceutical preparation for injection can be prepared by dissolving purified anti-glypican 3 antibodies in a solvent such as a physiological saline, a buffer or a glucose solution, and then adding an adsorption inhibitor such as Tween80, Tween20, gelatine or human serum albumin to the solution.
  • the freeze-dried agent may be used to prepare a dosage form that is dissolved for reconstitution before use.
  • sugar alcohol or saccharides such as mannitol or glucose can be used.
  • FIG. 1 shows ADCC activity on HuH-7 cells of anti-glypican 3 antibodies (K6534).
  • FIG. 2 shows CDC activity on HuH-7 cells of anti-glypican 3 antibodies (K6511).
  • FIG. 3 shows the expression of glypican on HuH-7 cells.
  • FIG. 4A shows the results of FACS analysis of GPC3 expression by human lung cancer cell lines. They are the results of analyses with anti-glypican 3 antibodies (K6534).
  • FIG. 4B shows the results of FACS analysis of GPC3 expression by human leukemia cell lines. They are the results of analyses with anti-glypican 3 antibodies (K6534).
  • FIG. 4C shows the results of FACS analysis of GPC3 expression by human lymphoma cell lines. They are the results of analyses with anti-glypican 3 antibodies (K6534).
  • FIG. 4D shows the results of FACS analysis of GPC3 expression by human colon cancer cell lines. They are the results of analyses with anti-glypican 3 antibodies (K6534).
  • FIG. 4E shows the results of FACS analysis of GPC3 expression by human mammary cancer cell lines. They are the results of analyses with anti-glypican 3 antibodies (K6534).
  • FIG. 4F shows the results of FACS analysis of GPC3 expression by human prostate cancer cell lines. They are the results of analyses with anti-glypican 3 antibodies (K6534).
  • FIG. 4G shows the results of FACS analysis of GPC3 expression by the human pancreatic cancer cell line and the human hepatic cancer cell line. They are the results of analyses with anti-glypican 3 antibodies (K6534).
  • a peptide having an amino acid sequence (the 355th to the 371st amino acids) (RQYRSAYYPEDLFIDKK) of human glypican-3 protein was synthesized.
  • the synthetic peptide. was bound to keyhole limpet hemocyanin (KLH) using a maleinimide benzoyloxy succinimide (MBS) type crosslinking agent, thereby preparing an immunogen.
  • KLH keyhole limpet hemocyanin
  • MMS maleinimide benzoyloxy succinimide
  • a method employed as an antibody titer assay method involves causing the diluted sera to react with the peptides (0.5 ⁇ g) immobilized on a plate, performing reaction with HRP-labeled anti-mouse antibodies, adding a substrate, and then measuring absorbance at 450 nm of the thus developed color (a peptide solid-phase ELISA method). After antibody titers were confirmed, splenocytes were collected, and then fused (Kohler, G, Milstein, C: Nature, 256: 495 (1975)) with myeloma cells (P 3 /X63-Ag8), thereby preparing hybridomas. Monoclonal antibodies produced by five types of hybridomas were then purified. The binding activity to the peptide was measured using the peptide solid-phase ELISA method, and then IgG1 antibodies (hereinafter, K6534) and IgG3 antibodies (hereinafter, K6511) having high binding activity were selected.
  • IgG1 antibodies herein
  • ADCC antibody-dependent cell-mediated cytotoxicity
  • CDC complement-dependent cytotoxicity
  • the spleen was extracted from.a CBA/N mouse (8-week-old, male), and then splenocytes were isolated in RPMI1640 media (GIBCO). The cells were washed in the same media containing 10% fetal bovine serum (FBS, HyClone), and then the cell concentration was prepared at 5 ⁇ 10 6 /mL, thereby preparing effector cells.
  • Baby Rabbit Complement (CEDARLANE) was diluted 10 times in 10% FBS-containing DMEM media (GIBCO), thereby preparing a complement solution.
  • A represents radioactivity (cpm) in each sample
  • B represents radioactivity (cpm) in a sample supplemented with 1% NP-40 (nacalai tesque)
  • C represents radioactivity (cpm) in a sample containing only target cells. The experiment was performed in duplicate, and average values were calculated.
  • FIG. 1 shows ADCC activity and FIG. 2 shows CDC activity.
  • Approximately 5 ⁇ 10 5 HuH-7 cells were suspended in 100 ⁇ L of FACS/PBS (prepared by dissolving 1 g of bovine serum albumin (SIGMA) in 1 L of CellWASH (Beckton Dickinson)). Then, anti-glypican 3 antibodies (K6511) or mouse IgG2a (Biogenesis) as a control antibody were added at 25 ⁇ g/mL, and then the solution was allowed to stand on ice for 30 minutes. After washing with FACS/PBS, the product was suspended in 100 ⁇ L of FACS/PBS. 4 ⁇ L of Goat Anti-Mouse Ig FITC (Becton Dickinson) was added, and then the solution was allowed to stand on ice for 30 minutes.
  • FACS/PBS prepared by dissolving 1 g of bovine serum albumin (SIGMA) in 1 L of CellWASH (Beckton Dickinson)
  • anti-glypican 3 antibodies K6511
  • mouse IgG2a Biogenesis
  • FIG. 3 shows the result of flow cytometry. Glypican 3 was expressed on HuH-7 cells, suggesting that anti-glypican 3 antibodies bind to glypican 3 expressed on the cell so as to inhibit cell proliferation (FIG. 3).
  • GPC3 glypican 3
  • the cells were cultured for 2 days and then subjected to assay.
  • the adhered cells were collected using Trypsin-EDTA (Cat. No. 25300-054, Lot 14210, GIBCO) that had been diluted 10 times with cell dissociation buffer (Cat. No. 13150-016, Lot 1098554, GIBCO).
  • the collected cells were allowed to react on ice with anti-glypican 3 antibodies (K6534, 600 ⁇ g/ml) or mIgG2a antibodies as a negative control (Biogenesis, M-IgG2a-i, Lot EA990719A, 1 mg/ml) (final antibody concentration of 10 ⁇ g/ml).
  • the cells After being washed, the cells were allowed to react with FITC-labeled anti-mouse Ig antibody (Cat. No. 349031, BD PharMingen) on ice (2 ⁇ l/test). After the cells were washed, fluorescence intensity was measured using a flow cytometer (EPICS XL, BECKMAN COULTER).
  • FITC-labeled anti-mouse Ig antibody Cat. No. 349031, BD PharMingen
  • fluorescence intensity was measured using a flow cytometer (EPICS XL, BECKMAN COULTER).
  • the expression of GPC3 was confirmed in the lung cancer cell lines (A549, NCI-H460, NCI-H23, NCI-H226, DMS114, EKVX, HOP-62 and NCI-H322 M; the leukemia cell lines P30/OHK, BALL-1, THP-1 and P39/TSU), the lymphoma cell lines (MLMA, Ramos and U937)-,; the colon cancer cell lines (SW480, COLO205, LoVo and SW837), the mammary cancer cell lines (MDA-MB-231, SK-BR-3, and MDA-MB-468), the prostate cancer cell lines (LNCaP and 22Rv1), the pancreatic cancer cell line (MIAPaCa-2), and the hepatic (liver) cancer cell line (HepG2). Based on these results, it is concluded that the cell growth inhibitor of the present invention is useful in treating lung cancer, colon cancer, mammary cancer, prostate cancer, leukemia, lymphoma, pan
  • a cell growth inhibitor containing an anti-glypican 3 antibody as an active ingredient is provided.
  • a carcinoma cell growth inhibitor containing an anti-glypican 3 antibody as an active ingredient is provided.
  • the anti-glypican 3 antibody binds to glypican 3 that is expressed on HuH-7 cells of a human hepatic cancer cell line, so as to inhibit cell proliferation.
  • the agent containing the anti-glypican 3 antibody is useful as a cell growth inhibitor, and in particular a carcinoma cell inhibitor.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Immunology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Biochemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Cell Biology (AREA)
  • Oncology (AREA)
  • Hematology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Microbiology (AREA)
  • Mycology (AREA)
  • Epidemiology (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Peptides Or Proteins (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Heterocyclic Carbon Compounds Containing A Hetero Ring Having Oxygen Or Sulfur (AREA)
US10/481,524 2001-06-22 2002-06-21 Cell proliferation inhibitors containing anti-glypican 3 antibody Abandoned US20040236080A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US11/702,780 US7744880B2 (en) 2001-06-22 2007-02-05 Cell growth inhibitors containing anti-glypican 3 antibody
US12/799,491 US8263077B2 (en) 2001-06-22 2010-04-26 Cell growth inhibitors containing anti-glypican 3 antibody

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
JP2001-189443 2001-06-22
JP2001189443 2001-06-22
PCT/JP2002/006237 WO2003000883A1 (en) 2001-06-22 2002-06-21 Cell proliferation inhibitors containing anti-glypican 3 antibody

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US11/702,780 Continuation US7744880B2 (en) 2001-06-22 2007-02-05 Cell growth inhibitors containing anti-glypican 3 antibody

Publications (1)

Publication Number Publication Date
US20040236080A1 true US20040236080A1 (en) 2004-11-25

Family

ID=19028362

Family Applications (3)

Application Number Title Priority Date Filing Date
US10/481,524 Abandoned US20040236080A1 (en) 2001-06-22 2002-06-21 Cell proliferation inhibitors containing anti-glypican 3 antibody
US11/702,780 Expired - Lifetime US7744880B2 (en) 2001-06-22 2007-02-05 Cell growth inhibitors containing anti-glypican 3 antibody
US12/799,491 Expired - Lifetime US8263077B2 (en) 2001-06-22 2010-04-26 Cell growth inhibitors containing anti-glypican 3 antibody

Family Applications After (2)

Application Number Title Priority Date Filing Date
US11/702,780 Expired - Lifetime US7744880B2 (en) 2001-06-22 2007-02-05 Cell growth inhibitors containing anti-glypican 3 antibody
US12/799,491 Expired - Lifetime US8263077B2 (en) 2001-06-22 2010-04-26 Cell growth inhibitors containing anti-glypican 3 antibody

Country Status (15)

Country Link
US (3) US20040236080A1 (ko)
EP (3) EP1780273B1 (ko)
JP (7) JP4281869B2 (ko)
KR (3) KR20080077295A (ko)
CN (2) CN101240022B (ko)
AT (2) ATE407204T1 (ko)
AU (2) AU2002315857B2 (ko)
CA (1) CA2451493C (ko)
CY (1) CY1113854T1 (ko)
DE (2) DE60237929D1 (ko)
DK (2) DK1411118T3 (ko)
ES (3) ES2353335T3 (ko)
HK (2) HK1079547A1 (ko)
PT (2) PT1411118E (ko)
WO (1) WO2003000883A1 (ko)

Cited By (46)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060167232A1 (en) * 2002-09-04 2006-07-27 Chugai Seiyaku Kabushiki Kaisha Antibody against n-terminal peptide or c-terminal peptide of gpc3 solubilized in blood
US20070087005A1 (en) * 2005-10-14 2007-04-19 Lazar Gregory A Anti-glypican-3 antibody
US20070190599A1 (en) * 2004-07-09 2007-08-16 Kiyotaka Nakano Anti-glypican 3 antibody
WO2007081790A3 (en) * 2006-01-04 2007-09-27 Picobella Llc Methods for diagnosing and treating prostate cancer
US20070269444A1 (en) * 2004-08-24 2007-11-22 Chugai Seiyaku Kabushiki Kaisha Adjuvant Therapy with the Use of Anti-Glypican 3 Antibody
US20080124330A1 (en) * 2004-10-26 2008-05-29 Chugai Seiyaku Kabushiki Kaisha Anti-Glypican 3 Antibody Having Modified Sugar Chain
US20090263392A1 (en) * 2006-03-31 2009-10-22 Chugai Seiyaku Kabushiki Kaisha Methods of modifying antibodies for purification of bispecific antibodies
US20100015133A1 (en) * 2005-03-31 2010-01-21 Chugai Seiyaku Kabushiki Kaisha Methods for Producing Polypeptides by Regulating Polypeptide Association
US20100209432A1 (en) * 2007-07-17 2010-08-19 Medarex, Inc. Monoclonal antibodies against glypican-3
US20100298542A1 (en) * 2007-09-26 2010-11-25 Chugai Seiyaku Kabushiki Kaisha Modified Antibody Constant Region
US20110098450A1 (en) * 2008-09-26 2011-04-28 Chugai Seiyaku Kabushiki Kaisha Antibody Molecules
US20110104157A1 (en) * 2008-04-04 2011-05-05 Chugai Seiyaku Kabushiki Kaisha Liver cancer drug
US20110111406A1 (en) * 2008-04-11 2011-05-12 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule capable of binding to two or more antigen molecules repeatedly
US20110129459A1 (en) * 2007-12-05 2011-06-02 Chugai Seiyaku Kabushiki Kaisha Anti-nr10 antibody and use thereof
US9096651B2 (en) 2007-09-26 2015-08-04 Chugai Seiyaku Kabushiki Kaisha Method of modifying isoelectric point of antibody via amino acid substitution in CDR
US9228017B2 (en) 2009-03-19 2016-01-05 Chugai Seiyaku Kabushiki Kaisha Antibody constant region variant
US9334331B2 (en) 2010-11-17 2016-05-10 Chugai Seiyaku Kabushiki Kaisha Bispecific antibodies
US9765135B2 (en) 2014-12-19 2017-09-19 Chugai Seiyaku Kabushiki Kaisha Anti-C5 antibodies
US9969800B2 (en) 2015-02-05 2018-05-15 Chugai Seiyaku Kabushiki Kaisha IL-8 antibodies
US9975966B2 (en) 2014-09-26 2018-05-22 Chugai Seiyaku Kabushiki Kaisha Cytotoxicity-inducing theraputic agent
US10000560B2 (en) 2014-12-19 2018-06-19 Chugai Seiyaku Kabushiki Kaisha Anti-myostatin antibodies, polypeptides containing variant Fc regions, and methods of use
US10150808B2 (en) 2009-09-24 2018-12-11 Chugai Seiyaku Kabushiki Kaisha Modified antibody constant regions
US20190046659A1 (en) * 2015-08-03 2019-02-14 Carsgen Therapeutics Ltd Antibody against glypican-3 and application thereof
US10253091B2 (en) 2009-03-19 2019-04-09 Chugai Seiyaku Kabushiki Kaisha Antibody constant region variant
US10253100B2 (en) 2011-09-30 2019-04-09 Chugai Seiyaku Kabushiki Kaisha Therapeutic antigen-binding molecule with a FcRn-binding domain that promotes antigen clearance
US10435458B2 (en) 2010-03-04 2019-10-08 Chugai Seiyaku Kabushiki Kaisha Antibody constant region variants with reduced Fcgammar binding
US10451627B2 (en) 2012-12-21 2019-10-22 Chugai Seiyaku Kabushiki Kaisha Method for assaying soluble GPC3 protein
US10618965B2 (en) 2011-02-25 2020-04-14 Chugai Seiyaku Kabushiki Kaisha Method for altering plasma retention and immunogenicity of antigen-binding molecule
WO2020086758A1 (en) 2018-10-23 2020-04-30 Dragonfly Therapeutics, Inc. Heterodimeric fc-fused proteins
US10752697B2 (en) 2015-10-29 2020-08-25 Mogam Institute For Biomedical Research Anti-glypican 3 antibody and pharmaceutical composition containing the same
US10774148B2 (en) 2015-02-27 2020-09-15 Chugai Seiyaku Kabushiki Kaisha Composition for treating IL-6-related diseases
US11046784B2 (en) 2006-03-31 2021-06-29 Chugai Seiyaku Kabushiki Kaisha Methods for controlling blood pharmacokinetics of antibodies
US11053308B2 (en) 2016-08-05 2021-07-06 Chugai Seiyaku Kabushiki Kaisha Method for treating IL-8-related diseases
US11066483B2 (en) 2010-11-30 2021-07-20 Chugai Seiyaku Kabushiki Kaisha Cytotoxicity-inducing therapeutic agent
US11072666B2 (en) 2016-03-14 2021-07-27 Chugai Seiyaku Kabushiki Kaisha Cell injury inducing therapeutic drug for use in cancer therapy
US11124576B2 (en) 2013-09-27 2021-09-21 Chungai Seiyaku Kabushiki Kaisha Method for producing polypeptide heteromultimer
US11142587B2 (en) 2015-04-01 2021-10-12 Chugai Seiyaku Kabushiki Kaisha Method for producing polypeptide hetero-oligomer
WO2021216916A1 (en) 2020-04-22 2021-10-28 Dragonfly Therapeutics, Inc. Formulation, dosage regimen, and manufacturing process for heterodimeric fc-fused proteins
US11359009B2 (en) 2015-12-25 2022-06-14 Chugai Seiyaku Kabushiki Kaisha Anti-myostatin antibodies and methods of use
US11376326B2 (en) 2015-07-01 2022-07-05 Chugai Seiyaku Kabushiki Kaisha GPC3-targeting therapeutic agent which is administered to patient for whom the GPC3-targeting therapeutic agent is effective
US11649262B2 (en) 2015-12-28 2023-05-16 Chugai Seiyaku Kabushiki Kaisha Method for promoting efficiency of purification of Fc region-containing polypeptide
US11760807B2 (en) 2014-05-08 2023-09-19 Chugai Seiyaku Kabushiki Kaisha GPC3-targeting drug which is administered to patient responsive to GPC3-targeting drug therapy
US11767362B1 (en) 2016-03-15 2023-09-26 Chugai Seiyaku Kabushiki Kaisha Methods of treating cancers using PD-1 axis binding antagonists and anti-GPC3 antibodies
US11827699B2 (en) 2011-09-30 2023-11-28 Chugai Seiyaku Kabushiki Kaisha Methods for producing antibodies promoting disappearance of antigens having plurality of biological activities
US11851486B2 (en) 2017-05-02 2023-12-26 National Center Of Neurology And Psychiatry Method for predicting and evaluating therapeutic effect in diseases related to IL-6 and neutrophils
US11891434B2 (en) 2010-11-30 2024-02-06 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule capable of binding to plurality of antigen molecules repeatedly

Families Citing this family (32)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7361336B1 (en) * 1997-09-18 2008-04-22 Ivan Bergstein Methods of cancer therapy targeted against a cancer stem line
AU2002315857B2 (en) * 2001-06-22 2007-07-26 Kaisha, Chugai Seiyaku Kabushiki Cell proliferation inhibitors containing anti-glypican 3 antibody
MXPA04011132A (es) * 2002-05-23 2005-08-15 Sunnybrook & Womens College Diagnostico de carcinoma hepatocelular.
EP1671645A4 (en) * 2003-09-04 2007-07-25 Chugai Pharmaceutical Co Ltd REMEDIES AND MEDIUM FOR DETECTING GALLENGIFT CANCER
JPWO2007015529A1 (ja) 2005-08-03 2009-02-19 パナソニック株式会社 基地局装置、通信端末装置、およびマルチキャリア通信方法
MY148637A (en) 2007-09-28 2013-05-15 Chugai Pharmaceutical Co Ltd Anti-glypican-3 antibody having improved kinetics in plasma
US20120190049A1 (en) * 2010-12-10 2012-07-26 University Of Washington Through Its Center For Commercialization Glypican-3 targeting of liver cancer cells using multifunctional nanoparticles
WO2012133782A1 (ja) 2011-03-30 2012-10-04 中外製薬株式会社 抗原結合分子の血漿中滞留性と免疫原性を改変する方法
AU2012317395B2 (en) 2011-09-30 2017-06-29 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule inducing immune response to target antigen
TW201726745A (zh) 2011-09-30 2017-08-01 中外製藥股份有限公司 促進抗原消失的抗原結合分子
WO2013051294A1 (ja) 2011-10-05 2013-04-11 中外製薬株式会社 糖鎖受容体結合ドメインを含む抗原の血漿中からの消失を促進する抗原結合分子
DK2818183T3 (da) 2012-02-24 2020-06-15 Chugai Pharmaceutical Co Ltd Antigen-bindende molekyle til at fremme forsvinden af antigen ved hjælp af Fc RIIB
CN107964042B (zh) 2012-05-30 2022-04-19 中外制药株式会社 靶组织特异性抗原结合分子
SG11201407972RA (en) * 2012-06-01 2015-01-29 Us Health High-affinity monoclonal antibodies to glypican-3 and use thereof
EP4119947A1 (en) 2012-12-21 2023-01-18 Chugai Seiyaku Kabushiki Kaisha Gpc3-targeting drug which is administered to patient responsive to gpc3-targeting drug therapy
AU2014358191B2 (en) 2013-12-04 2020-12-24 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecules, the antigen-binding activity of which varies according to the concentration of compounds, and libraries of said molecules
US9926377B2 (en) * 2014-05-22 2018-03-27 Genentech, Inc. Anti-GPC3 antibodies and immunoconjugates
CN104829704B (zh) * 2014-12-15 2016-08-17 河北省科学院生物研究所 一种磷脂酰肌醇蛋白聚糖gpc3蛋白片段及其应用和制备的杂交瘤细胞株
ES2805085T3 (es) * 2015-01-16 2021-02-10 Glyp Holdings Pty Ltd Epítopos de glipicano y usos de éstos
EP3373968B1 (en) * 2015-11-09 2024-04-17 The Children's Hospital of Philadelphia Glypican 2 as a cancer marker and therapeutic target
JP7125347B2 (ja) 2016-08-22 2022-08-24 中外製薬株式会社 ヒトgpc3ポリペプチドを発現する遺伝子改変非ヒト動物
JPWO2018097308A1 (ja) 2016-11-28 2019-10-17 中外製薬株式会社 リガンド結合活性が調整可能なリガンド結合分子
EP3684413A1 (en) 2017-09-20 2020-07-29 Chugai Seiyaku Kabushiki Kaisha Dosage regimen for combination therapy using pd-1 axis binding antagonists and gpc3 targeting agent
CA3083346A1 (en) 2017-11-28 2019-06-06 Chugai Seiyaku Kabushiki Kaisha Ligand-binding molecule having adjustable ligand-binding activity
AR114112A1 (es) 2018-02-15 2020-07-22 Seattle Genetics Inc Anticuerpos de glipicano 3 y conjugados de los mismos
JPWO2019230868A1 (ja) 2018-05-30 2021-06-24 中外製薬株式会社 単ドメイン抗体含有リガンド結合分子
MX2020013977A (es) 2018-06-22 2021-06-15 Kite Pharma Inc Proteínas quiméricas transmembrana y usos de las mismas.
AU2019354395A1 (en) 2018-10-01 2021-05-06 Adicet Therapeutics, Inc. Compositions and methods regarding engineered and non-engineered γδ -T cells for treatment of solid tumors
CN113613676A (zh) 2019-03-19 2021-11-05 中外制药株式会社 包含对抗原的结合活性因mta而变化的抗原结合结构域的抗原结合分子及用于获得该抗原结合结构域的文库
JPWO2020246563A1 (ko) 2019-06-05 2020-12-10
CN114127277A (zh) 2019-06-05 2022-03-01 中外制药株式会社 蛋白酶底物和包含蛋白酶切割序列的多肽
CN116194124A (zh) 2020-07-31 2023-05-30 中外制药株式会社 包含表达嵌合受体的细胞的药物组合物

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6214973B1 (en) * 1993-11-19 2001-04-10 Chugai Seiyaku Kabushiki Kaisha Reshaped human antibody to human medulloblastoma cells
US20030045691A1 (en) * 1996-10-04 2003-03-06 Koichiro Ono Reshaped human anti-hm 1.24 antibody
US20060008456A1 (en) * 1997-10-03 2006-01-12 Chugai Seiyaku Kabushiki Kaisha Natural humanized antibody
US20060193828A1 (en) * 1999-08-23 2006-08-31 Chugai Seiyaku Kabushiki Kaisha Expression enhancer for HM1.24 antigen

Family Cites Families (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
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
ATE390933T1 (de) 1995-04-27 2008-04-15 Amgen Fremont Inc Aus immunisierten xenomäusen stammende menschliche antikörper gegen il-8
JP3587664B2 (ja) 1996-10-04 2004-11-10 中外製薬株式会社 再構成ヒト抗hm1.24抗体
JP4228089B2 (ja) 1997-03-11 2009-02-25 株式会社ニコン 過電流検知機構およびそれを用いた電子閃光装置
US7361336B1 (en) 1997-09-18 2008-04-22 Ivan Bergstein Methods of cancer therapy targeted against a cancer stem line
JP3445153B2 (ja) 1998-06-04 2003-09-08 富士通株式会社 電話番号一時的使用装置及び方法
AU2002315857B2 (en) 2001-06-22 2007-07-26 Kaisha, Chugai Seiyaku Kabushiki Cell proliferation inhibitors containing anti-glypican 3 antibody

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6214973B1 (en) * 1993-11-19 2001-04-10 Chugai Seiyaku Kabushiki Kaisha Reshaped human antibody to human medulloblastoma cells
US20030045691A1 (en) * 1996-10-04 2003-03-06 Koichiro Ono Reshaped human anti-hm 1.24 antibody
US20060008456A1 (en) * 1997-10-03 2006-01-12 Chugai Seiyaku Kabushiki Kaisha Natural humanized antibody
US20060193828A1 (en) * 1999-08-23 2006-08-31 Chugai Seiyaku Kabushiki Kaisha Expression enhancer for HM1.24 antigen

Cited By (92)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090060907A1 (en) * 2002-09-04 2009-03-05 Chugai Seiyaku Kabushiki Kaisha Antibody against secreted N-terminal peptide of GPC3 present in blood or C-terminal peptide of GPC3
US20060167232A1 (en) * 2002-09-04 2006-07-27 Chugai Seiyaku Kabushiki Kaisha Antibody against n-terminal peptide or c-terminal peptide of gpc3 solubilized in blood
US20070190599A1 (en) * 2004-07-09 2007-08-16 Kiyotaka Nakano Anti-glypican 3 antibody
US7919086B2 (en) 2004-07-09 2011-04-05 Chugai Seiyaku Kabushiki Kaisha Anti-glypican 3 antibody
US20100248359A1 (en) * 2004-07-09 2010-09-30 Chugai Seiyaku Kabushiki Kaisha Anti-Glypican 3 Antibody
US20070269444A1 (en) * 2004-08-24 2007-11-22 Chugai Seiyaku Kabushiki Kaisha Adjuvant Therapy with the Use of Anti-Glypican 3 Antibody
US7871613B2 (en) 2004-08-24 2011-01-18 Chugai Seiyaku Kabushiki Kaisha Adjuvant therapy with the use of anti-glypican 3 antibody
US20080124330A1 (en) * 2004-10-26 2008-05-29 Chugai Seiyaku Kabushiki Kaisha Anti-Glypican 3 Antibody Having Modified Sugar Chain
US7867734B2 (en) * 2004-10-26 2011-01-11 Chugai Seiyaku Kabushiki Kaisha Anti-glypican 3 antibody having modified sugar chain
US20100015133A1 (en) * 2005-03-31 2010-01-21 Chugai Seiyaku Kabushiki Kaisha Methods for Producing Polypeptides by Regulating Polypeptide Association
US11168344B2 (en) 2005-03-31 2021-11-09 Chugai Seiyaku Kabushiki Kaisha Methods for producing polypeptides by regulating polypeptide association
US10011858B2 (en) 2005-03-31 2018-07-03 Chugai Seiyaku Kabushiki Kaisha Methods for producing polypeptides by regulating polypeptide association
US20080267979A1 (en) * 2005-10-14 2008-10-30 Gregory Alan Lazar Anti-Glypican-3 Antibody
US10118959B2 (en) 2005-10-14 2018-11-06 Chugai Seiyaku Kabushiki Kaisha Anti-glypican-3 antibody
US9102739B2 (en) 2005-10-14 2015-08-11 Chugai Seiyaku Kabushiki Kaisha Anti-glypican-3 antibody
US20070087005A1 (en) * 2005-10-14 2007-04-19 Lazar Gregory A Anti-glypican-3 antibody
US20100041126A1 (en) * 2006-01-04 2010-02-18 Bruce Wang Methods for diagnosing and treating prostate cancer
US7723043B2 (en) 2006-01-04 2010-05-25 Picobella, Lp Methods for screening for prostate cancer
EP1977247A2 (en) * 2006-01-04 2008-10-08 Picobella, LLC Methods for diagnosing and treating prostate cancer
EP1977247A4 (en) * 2006-01-04 2009-07-08 Picobella Llc METHODS OF DIAGNOSING AND TREATING PROSTATE CANCER
WO2007081790A3 (en) * 2006-01-04 2007-09-27 Picobella Llc Methods for diagnosing and treating prostate cancer
US10934344B2 (en) 2006-03-31 2021-03-02 Chugai Seiyaku Kabushiki Kaisha Methods of modifying antibodies for purification of bispecific antibodies
US11046784B2 (en) 2006-03-31 2021-06-29 Chugai Seiyaku Kabushiki Kaisha Methods for controlling blood pharmacokinetics of antibodies
US20090263392A1 (en) * 2006-03-31 2009-10-22 Chugai Seiyaku Kabushiki Kaisha Methods of modifying antibodies for purification of bispecific antibodies
US9670269B2 (en) 2006-03-31 2017-06-06 Chugai Seiyaku Kabushiki Kaisha Methods of modifying antibodies for purification of bispecific antibodies
US20100209432A1 (en) * 2007-07-17 2010-08-19 Medarex, Inc. Monoclonal antibodies against glypican-3
AU2008275985B2 (en) * 2007-07-17 2013-09-19 E. R. Squibb & Sons, L.L.C. Monoclonal antibodies against Glypican-3
US9217033B2 (en) 2007-07-17 2015-12-22 E. R. Squibb & Sons, L.L.C. Monoclonal antibodies against Glypican-3
US8680247B2 (en) * 2007-07-17 2014-03-25 Medarex, L.L.C. Monoclonal antibodies against glypican-3
US9688762B2 (en) 2007-09-26 2017-06-27 Chugai Sciyaku Kabushiki Kaisha Modified antibody constant region
US11332533B2 (en) 2007-09-26 2022-05-17 Chugai Seiyaku Kabushiki Kaisha Modified antibody constant region
US20100298542A1 (en) * 2007-09-26 2010-11-25 Chugai Seiyaku Kabushiki Kaisha Modified Antibody Constant Region
US11248053B2 (en) 2007-09-26 2022-02-15 Chugai Seiyaku Kabushiki Kaisha Method of modifying isoelectric point of antibody via amino acid substitution in CDR
US9828429B2 (en) 2007-09-26 2017-11-28 Chugai Seiyaku Kabushiki Kaisha Method of modifying isoelectric point of antibody via amino acid substitution in CDR
US9096651B2 (en) 2007-09-26 2015-08-04 Chugai Seiyaku Kabushiki Kaisha Method of modifying isoelectric point of antibody via amino acid substitution in CDR
US9399680B2 (en) 2007-12-05 2016-07-26 Chugai Seiyaku Kabushiki Kaisha Nucleic acids encoding anti-NR10 antibodies
US20110229459A1 (en) * 2007-12-05 2011-09-22 Chugai Seiyaku Kabushiki Kaisha Anti-nr10 antibody and use thereof
US8575317B2 (en) 2007-12-05 2013-11-05 Chugai Seiyaku Kabushiki Kaisha Anti-NR10 antibody and use thereof
US20110129459A1 (en) * 2007-12-05 2011-06-02 Chugai Seiyaku Kabushiki Kaisha Anti-nr10 antibody and use thereof
US20110104157A1 (en) * 2008-04-04 2011-05-05 Chugai Seiyaku Kabushiki Kaisha Liver cancer drug
US9890377B2 (en) 2008-04-11 2018-02-13 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule capable of binding to two or more antigen molecules repeatedly
US11371039B2 (en) 2008-04-11 2022-06-28 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule capable of binding to two or more antigen molecules repeatedly
US9868948B2 (en) 2008-04-11 2018-01-16 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule capable of binding to two or more antigen molecules repeatedly
US11359194B2 (en) 2008-04-11 2022-06-14 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule capable of binding two or more antigen molecules repeatedly
US20110111406A1 (en) * 2008-04-11 2011-05-12 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule capable of binding to two or more antigen molecules repeatedly
US10472623B2 (en) 2008-04-11 2019-11-12 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule capable of binding two or more antigen molecules repeatedly
US20110098450A1 (en) * 2008-09-26 2011-04-28 Chugai Seiyaku Kabushiki Kaisha Antibody Molecules
US8562991B2 (en) 2008-09-26 2013-10-22 Chugai Seiyaku Kabushiki Kaisha Antibody molecules that bind to IL-6 receptor
US10662245B2 (en) 2008-09-26 2020-05-26 Chugai Seiyaku Kabushiki Kaisha Methods of reducing IL-6 activity for disease treatment
US10066018B2 (en) 2009-03-19 2018-09-04 Chugai Seiyaku Kabushiki Kaisha Antibody constant region variant
US10253091B2 (en) 2009-03-19 2019-04-09 Chugai Seiyaku Kabushiki Kaisha Antibody constant region variant
US9228017B2 (en) 2009-03-19 2016-01-05 Chugai Seiyaku Kabushiki Kaisha Antibody constant region variant
US10150808B2 (en) 2009-09-24 2018-12-11 Chugai Seiyaku Kabushiki Kaisha Modified antibody constant regions
US10435458B2 (en) 2010-03-04 2019-10-08 Chugai Seiyaku Kabushiki Kaisha Antibody constant region variants with reduced Fcgammar binding
US10450381B2 (en) 2010-11-17 2019-10-22 Chugai Seiyaku Kabushiki Kaisha Methods of treatment that include the administration of bispecific antibodies
US9334331B2 (en) 2010-11-17 2016-05-10 Chugai Seiyaku Kabushiki Kaisha Bispecific antibodies
US11891434B2 (en) 2010-11-30 2024-02-06 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule capable of binding to plurality of antigen molecules repeatedly
US11066483B2 (en) 2010-11-30 2021-07-20 Chugai Seiyaku Kabushiki Kaisha Cytotoxicity-inducing therapeutic agent
US10618965B2 (en) 2011-02-25 2020-04-14 Chugai Seiyaku Kabushiki Kaisha Method for altering plasma retention and immunogenicity of antigen-binding molecule
US11718678B2 (en) 2011-02-25 2023-08-08 Chugai Seiyaku Kabushiki Kaisha Method for altering plasma retention and immunogenicity of antigen-binding molecule
US11827699B2 (en) 2011-09-30 2023-11-28 Chugai Seiyaku Kabushiki Kaisha Methods for producing antibodies promoting disappearance of antigens having plurality of biological activities
US10253100B2 (en) 2011-09-30 2019-04-09 Chugai Seiyaku Kabushiki Kaisha Therapeutic antigen-binding molecule with a FcRn-binding domain that promotes antigen clearance
US10451627B2 (en) 2012-12-21 2019-10-22 Chugai Seiyaku Kabushiki Kaisha Method for assaying soluble GPC3 protein
US10782300B2 (en) 2012-12-21 2020-09-22 Chugai Seiyaku Kabushiki Kaisha GPC3-targeting drug which is administered to patient responsive to GPC3-targeting drug therapy
US11124576B2 (en) 2013-09-27 2021-09-21 Chungai Seiyaku Kabushiki Kaisha Method for producing polypeptide heteromultimer
US11760807B2 (en) 2014-05-08 2023-09-19 Chugai Seiyaku Kabushiki Kaisha GPC3-targeting drug which is administered to patient responsive to GPC3-targeting drug therapy
US11001643B2 (en) 2014-09-26 2021-05-11 Chugai Seiyaku Kabushiki Kaisha Cytotoxicity-inducing therapeutic agent
US9975966B2 (en) 2014-09-26 2018-05-22 Chugai Seiyaku Kabushiki Kaisha Cytotoxicity-inducing theraputic agent
US10023630B2 (en) 2014-12-19 2018-07-17 Chugai Seiyaku Kabushiki Kaisha Methods of neutralizing C5 with anti-C5 antibodies
US9765135B2 (en) 2014-12-19 2017-09-19 Chugai Seiyaku Kabushiki Kaisha Anti-C5 antibodies
US11454633B2 (en) 2014-12-19 2022-09-27 Chugai Seiyaku Kabushiki Kaisha Anti-myostatin antibodies, polypeptides containing variant Fc regions, and methods of use
US10385122B2 (en) 2014-12-19 2019-08-20 Chugai Seiyaku Kabushiki Kaisha Nucleic acids encoding anti-C5 antibodies
US11597760B2 (en) 2014-12-19 2023-03-07 Chugai Seiyaku Kabushiki Kaisha Method of detecting the presence of complement C5
US10738111B2 (en) 2014-12-19 2020-08-11 Chugai Seiyaku Kabushiki Kaisha Anti-myostatin antibodies, polypeptides containing variant Fc regions, and methods of use
US10000560B2 (en) 2014-12-19 2018-06-19 Chugai Seiyaku Kabushiki Kaisha Anti-myostatin antibodies, polypeptides containing variant Fc regions, and methods of use
US11180548B2 (en) 2015-02-05 2021-11-23 Chugai Seiyaku Kabushiki Kaisha Methods of neutralizing IL-8 biological activity
US9969800B2 (en) 2015-02-05 2018-05-15 Chugai Seiyaku Kabushiki Kaisha IL-8 antibodies
US10519229B2 (en) 2015-02-05 2019-12-31 Chugai Seiyaku Kabushiki Kaisha Nucleic acids encoding IL-8 antibodies
US10774148B2 (en) 2015-02-27 2020-09-15 Chugai Seiyaku Kabushiki Kaisha Composition for treating IL-6-related diseases
US11142587B2 (en) 2015-04-01 2021-10-12 Chugai Seiyaku Kabushiki Kaisha Method for producing polypeptide hetero-oligomer
US11376326B2 (en) 2015-07-01 2022-07-05 Chugai Seiyaku Kabushiki Kaisha GPC3-targeting therapeutic agent which is administered to patient for whom the GPC3-targeting therapeutic agent is effective
US20190046659A1 (en) * 2015-08-03 2019-02-14 Carsgen Therapeutics Ltd Antibody against glypican-3 and application thereof
US10752697B2 (en) 2015-10-29 2020-08-25 Mogam Institute For Biomedical Research Anti-glypican 3 antibody and pharmaceutical composition containing the same
US11359009B2 (en) 2015-12-25 2022-06-14 Chugai Seiyaku Kabushiki Kaisha Anti-myostatin antibodies and methods of use
US11649262B2 (en) 2015-12-28 2023-05-16 Chugai Seiyaku Kabushiki Kaisha Method for promoting efficiency of purification of Fc region-containing polypeptide
US11072666B2 (en) 2016-03-14 2021-07-27 Chugai Seiyaku Kabushiki Kaisha Cell injury inducing therapeutic drug for use in cancer therapy
US11767362B1 (en) 2016-03-15 2023-09-26 Chugai Seiyaku Kabushiki Kaisha Methods of treating cancers using PD-1 axis binding antagonists and anti-GPC3 antibodies
US11780912B2 (en) 2016-08-05 2023-10-10 Chugai Seiyaku Kabushiki Kaisha Composition for prophylaxis or treatment of IL-8 related diseases
US11053308B2 (en) 2016-08-05 2021-07-06 Chugai Seiyaku Kabushiki Kaisha Method for treating IL-8-related diseases
US11851486B2 (en) 2017-05-02 2023-12-26 National Center Of Neurology And Psychiatry Method for predicting and evaluating therapeutic effect in diseases related to IL-6 and neutrophils
WO2020086758A1 (en) 2018-10-23 2020-04-30 Dragonfly Therapeutics, Inc. Heterodimeric fc-fused proteins
WO2021216916A1 (en) 2020-04-22 2021-10-28 Dragonfly Therapeutics, Inc. Formulation, dosage regimen, and manufacturing process for heterodimeric fc-fused proteins

Also Published As

Publication number Publication date
EP1411118A4 (en) 2005-01-26
AU2007201255B2 (en) 2010-03-11
EP1780273A1 (en) 2007-05-02
DK2208784T3 (da) 2013-03-18
HK1079547A1 (en) 2006-04-07
CN1688692A (zh) 2005-10-26
PT1411118E (pt) 2008-12-09
DK1411118T3 (da) 2008-11-17
US20070172488A1 (en) 2007-07-26
AU2007201255A1 (en) 2007-04-19
EP1780273B1 (en) 2010-10-06
JP2005225892A (ja) 2005-08-25
HK1120522A1 (en) 2009-04-03
WO2003000883A1 (en) 2003-01-03
CA2451493A1 (en) 2003-01-03
ES2353335T3 (es) 2011-03-01
JP2008120828A (ja) 2008-05-29
JP2007238632A (ja) 2007-09-20
US8263077B2 (en) 2012-09-11
JP4347402B2 (ja) 2009-10-21
CY1113854T1 (el) 2016-07-27
CA2451493C (en) 2016-08-23
ES2312586T3 (es) 2009-03-01
ATE407204T1 (de) 2008-09-15
JP4281869B2 (ja) 2009-06-17
EP1411118A1 (en) 2004-04-21
CN101240022A (zh) 2008-08-13
KR20080077295A (ko) 2008-08-21
US7744880B2 (en) 2010-06-29
CN101240022B (zh) 2012-07-18
US20110002922A1 (en) 2011-01-06
KR100877176B1 (ko) 2009-01-07
JP2007051159A (ja) 2007-03-01
KR20090107091A (ko) 2009-10-12
KR20040030699A (ko) 2004-04-09
EP2208784B1 (en) 2013-01-02
CN1314803C (zh) 2007-05-09
EP2208784A1 (en) 2010-07-21
JP5596935B2 (ja) 2014-09-24
KR100953520B1 (ko) 2010-04-21
DE60237929D1 (de) 2010-11-18
EP1411118B1 (en) 2008-09-03
AU2002315857B2 (en) 2007-07-26
JPWO2003000883A1 (ja) 2004-10-07
ATE483801T1 (de) 2010-10-15
DE60228721D1 (en) 2008-10-16
PT2208784E (pt) 2013-04-03
JP2009024020A (ja) 2009-02-05
JP2009161565A (ja) 2009-07-23
ES2399028T3 (es) 2013-03-25

Similar Documents

Publication Publication Date Title
EP1780273B1 (en) Cell proliferation inhibitors containing anti-glypican 3 antibody
EP2050466B1 (en) Diagnosis and treatment of cancer using anti-desmoglein-3 antibodies
US8546546B2 (en) Anti-Muc 17 antibody
WO2011093097A1 (ja) 抗dll3抗体
US9139647B2 (en) Diagnosis and treatment of cancer using anti-TM4SF20 antibody
US9079957B2 (en) Diagnosis and treatment of cancer using anti-TMPRSS11E antibody
US20080008710A1 (en) Therapeutic Agent And Diagnostic Agent For Cholangiocarcinoma

Legal Events

Date Code Title Description
AS Assignment

Owner name: CHUGAI SEIYAKU KABUSHIKI KAISHA, JAPAN

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:ABURATANI, HIROYUKI;NAKAMURA, TETSUO;TSUCHIYA, MASAYUKI;REEL/FRAME:015517/0111

Effective date: 20040614

Owner name: ABURATANI, HIROYUKI, JAPAN

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:ABURATANI, HIROYUKI;NAKAMURA, TETSUO;TSUCHIYA, MASAYUKI;REEL/FRAME:015517/0111

Effective date: 20040614

AS Assignment

Owner name: PERSEUS PROTEOMICS INC., JAPAN

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:ABURATANI, HIROYUKI;REEL/FRAME:018191/0638

Effective date: 20060630

AS Assignment

Owner name: CHUGAI SEIYAKU KABUSHIKI KAISHA, JAPAN

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:MATSUDA, KAZUYUKI;REEL/FRAME:018428/0185

Effective date: 20060930

AS Assignment

Owner name: CHUGAI SELYAKU KABUSHIKI KAISHA, JAPAN

Free format text: CORRECTIVE ASSIGNMENT TO CORRECT THE ASSIGNOR'S NAME. DOCUMENT PREVIOUSLY RECORDED AT REEL 018428 FRAME 0185;ASSIGNOR:PERSEUS PROTEOMICS INC.;REEL/FRAME:018522/0207

Effective date: 20060930

STCB Information on status: application discontinuation

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