US20040093622A1 - Generation of xenogeneic antibodies - Google Patents

Generation of xenogeneic antibodies Download PDF

Info

Publication number
US20040093622A1
US20040093622A1 US10/627,250 US62725003A US2004093622A1 US 20040093622 A1 US20040093622 A1 US 20040093622A1 US 62725003 A US62725003 A US 62725003A US 2004093622 A1 US2004093622 A1 US 2004093622A1
Authority
US
United States
Prior art keywords
immunoglobulin
xenogeneic
genome
producing
modified
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
US10/627,250
Inventor
Raju Kucherlapati
Aya Jakobovits
Sue Klapholz
Daniel Brenner
Daniel Capon
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.)
Amgen Fremont Inc
Original Assignee
Abgenix Inc
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
Priority claimed from PCT/US1991/000245 external-priority patent/WO1991010741A1/en
Application filed by Abgenix Inc filed Critical Abgenix Inc
Priority to US10/627,250 priority Critical patent/US20040093622A1/en
Publication of US20040093622A1 publication Critical patent/US20040093622A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/8509Vectors or expression systems specially adapted for eukaryotic hosts for animal cells for producing genetically modified animals, e.g. transgenic
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K67/00Rearing or breeding animals, not otherwise provided for; New breeds of animals
    • A01K67/027New breeds of vertebrates
    • A01K67/0271Chimeric animals, e.g. comprising exogenous cells
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K67/00Rearing or breeding animals, not otherwise provided for; New breeds of animals
    • A01K67/027New breeds of vertebrates
    • A01K67/0275Genetically modified vertebrates, e.g. transgenic
    • A01K67/0276Knockout animals
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K67/00Rearing or breeding animals, not otherwise provided for; New breeds of animals
    • A01K67/027New breeds of vertebrates
    • A01K67/0275Genetically modified vertebrates, e.g. transgenic
    • A01K67/0278Humanized animals, e.g. knockin
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/12Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from bacteria
    • C07K16/1267Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from bacteria from Gram-positive bacteria
    • C07K16/1282Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from bacteria from Gram-positive bacteria from Clostridium (G)
    • 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
    • C07K16/24Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against cytokines, lymphokines or interferons
    • C07K16/241Tumor Necrosis Factors
    • 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
    • C07K16/24Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against cytokines, lymphokines or interferons
    • C07K16/244Interleukins [IL]
    • 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
    • C07K16/24Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against cytokines, lymphokines or interferons
    • C07K16/244Interleukins [IL]
    • C07K16/248IL-6
    • 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
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2812Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against CD4
    • 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
    • C07K16/2851Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the lectin superfamily, e.g. CD23, CD72
    • C07K16/2854Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the lectin superfamily, e.g. CD23, CD72 against selectins, e.g. CD62
    • 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
    • C07K16/2875Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the NGF/TNF superfamily, e.g. CD70, CD95L, CD153, CD154
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/46Hybrid immunoglobulins
    • C07K16/461Igs containing Ig-regions, -domains or -residues form different species
    • C07K16/462Igs containing a variable region (Fv) from one specie and a constant region (Fc) from another
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/87Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/87Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation
    • C12N15/90Stable introduction of foreign DNA into chromosome
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2207/00Modified animals
    • A01K2207/15Humanized animals
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • A01K2217/05Animals comprising random inserted nucleic acids (transgenic)
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • A01K2217/07Animals genetically altered by homologous recombination
    • A01K2217/072Animals genetically altered by homologous recombination maintaining or altering function, i.e. knock in
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • A01K2217/07Animals genetically altered by homologous recombination
    • A01K2217/075Animals genetically altered by homologous recombination inducing loss of function, i.e. knock out
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • A01K2217/20Animal model comprising regulated expression system
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2227/00Animals characterised by species
    • A01K2227/10Mammal
    • A01K2227/105Murine
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2267/00Animals characterised by purpose
    • A01K2267/01Animal expressing industrially exogenous proteins
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2267/00Animals characterised by purpose
    • A01K2267/03Animal model, e.g. for test or diseases
    • 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
    • C07K2317/21Immunoglobulins specific features characterized by taxonomic origin from primates, e.g. man
    • 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

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Genetics & Genomics (AREA)
  • General Health & Medical Sciences (AREA)
  • Immunology (AREA)
  • Molecular Biology (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Medicinal Chemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Zoology (AREA)
  • Biotechnology (AREA)
  • General Engineering & Computer Science (AREA)
  • Wood Science & Technology (AREA)
  • Environmental Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biomedical Technology (AREA)
  • Animal Husbandry (AREA)
  • Physics & Mathematics (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • Microbiology (AREA)
  • Biodiversity & Conservation Biology (AREA)
  • Plant Pathology (AREA)
  • Cell Biology (AREA)
  • Mycology (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Peptides Or Proteins (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)

Abstract

The subject invention provides non-human mammalian hosts characterized by inactivated endogenous Ig loci and functional human Ig loci for response to an immunogen to produce human antibodies or analogs thereof. The hosts are produced by multiple genetic modifications of embryonic cells in conjunction with breeding. Different strategies are employed for recombination of the human loci randomly or at analogous host loci. Chimeric and transgenic mammals, particularly mice, are provided, having stably integrated large, xenogeneic DNA segments. The segments are introduced by fusion with yeast spheroplasts comprising yeast artificial chromosomes (YACs) which include the xenogeneic DNA segments and a selective marker such as HPRT, and embryonic stem cells.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application is a continuation-in-part of application Ser. No. 07/919,297 filed Jul. 24, 1992, which was a continuation-in-part of application Ser. No. 07/610,515 filed Nov. 8, 1990 which was a continuation-in-part of application Ser. No. 07/466,008 filed Jan. 12, 1990, the entire disclosures of which are all incorporated herein by reference.[0001]
  • INTRODUCTION
  • 1. Technical Field [0002]
  • The field of this invention is the production of xenogeneic specific binding proteins in a viable mammalian host. [0003]
  • 2. Background [0004]
  • The ability to produce transgenic animals has been revolutionized with the advent of the ability to culture murine embryonic stem cells, and to introduce genetic modifications in these cells for subsequent transmission to the mouse germline. Thus one has the opportunity to modify endogenous genes to produce animal strains capable of producing novel products by introduction of foreign genes into the host, particularly human genes to produce xenogeneic binding proteins. The expression of such genes in vivo in an animal model may provide for investigation of the function of the gene, the regulation of gene expression, its processing, response to various agents and the like. In addition, animals with new phenotypes, including those that mimic a variety of diseases, may be produced. For example, there is interest in introducing a dominant mutation or, complementing a recessive mutation. Depending on the particular gene, the difficulty of achieving the desired mutation will vary greatly. While some gene targets have proven to be relatively amenable to modification, other targets have proven to be extremely resistant to modification. [0005]
  • Because of the opportunity for generating transgenic animals, there is substantial interest in providing new procedures that increase the success of production of transgenic animals. Particularly, where one wishes to introduce large DNA fragments, encompassing hundreds of kilobases, there is substantial concern about the ability to introduce the large fragments in intact form into mammalian cells, the efficiency of integration, the functional capability of the gene(s) present on the fragment and transmission in the germline to the progeny. In addition, such procedures for introduction of large DNA fragments provide for determination of the function of large DNA fragments identified in the ongoing human genome project. [0006]
  • In particular, there is interest in producing xenogeneic specific binding proteins, for example human monoclonal antibodies, in small laboratory animals such as mice. Monoclonal antibodies find use in both diagnosis and therapy. Because of their ability to bind to a specific epitope, they can be uniquely used to identify molecules carrying that epitope or may be directed, by themselves or in conjunction with another moiety, to a specific site for diagnosis or therapy. [0007]
  • Monoclonal antibodies comprise heavy and light chains which join together to define a binding region for the epitope. Each of the chains is comprised of a variable region and a constant region. The constant region amino acid sequence is specific for a particular isotype of the antibody, as well as the host which produces the antibody. [0008]
  • Because of the relationship between the sequence of the constant region and the species from which the antibody is produced, the introduction of a xenogeneic antibody into the vascular system of the host can produce an immune response. Where the xenogeneic antibody is introduced repetitively, in the case of chronic diseases, it becomes impractical to administer the antibody, since it will be rapidly destroyed and may have an adverse effect. There have been, therefore, many efforts to provide a source of syngeneic or allogeneic antibodies. One technique has involved the use of recombinant DNA technology where the genes for the heavy and light chains, from a host were identified and the regions encoding the constant region isolated. These regions were then joined to the variable region encoding portion of other immunoglobulin genes from another species directed to a specific epitope. [0009]
  • While the resulting chimeric partly xenogeneic antibody is substantially more useful than using a fully xenogeneic antibody, it still has a number of disadvantages. The identification, isolation and joining of the variable and constant regions requires substantial work. In addition, the joining of a constant region from one species to a variable region from another species may change the specificity and affinity of the variable regions, so as to lose the desired properties of the variable region. Also, there are framework and hypervariable sequences specific for a species in the variable region. These framework and hypervariable sequences may result in undesirable antigenic responses. [0010]
  • It would therefore be more desirable to produce allogeneic antibodies for administration to a host by immunizing the host with an immunogen of interest. For primates, particularly humans, this approach is not practical. The human antibodies which have been produced have been based on the adventitious presence of an available spleen, from a host which had been previously immunized to the epitope of interest. While human peripheral blood lymphocytes may be employed for the production of monoclonal antibodies, these have not been particularly successful in fusions and have usually led only to IgM. Moreover, it is particularly difficult to generate a human antibody response against a human protein, a desired target in many therapeutic and diagnostic applications. There is, therefore, substantial interest in finding alternative routes to the production of allogeneic antibodies for humans. [0011]
  • Relevant Literature [0012]
  • Thomas and Capecchi (1987), [0013] Cell, 51:503-512 and Koller and Smithies (1989), Proc. Natl. Acad. Sci. USA, 86:8932-8935 describe inactivating the β2-microglobulin locus by homologous recombination in embryonic stem cells. Berman et al. (1988), EMBO J. 7:727-738 describe the human Ig VH locus. Burke, et al. (1987), Science, 236:806-812 describe yeast artificial chromosome vectors. See also, Garza et al. (1989), Science, 246:641-646 and Brownstein et al. (1989), Science, 244:1348-1351. Sakano, et al., describe a diversity segment of the immunoglobulin heavy chain genes in Sakano et al. (1981), Nature, 290:562-565. Tucker et al. (1981), Proc. Natl. Acad. Sci. USA, 78:7684-7688 describe the mouse IgA heavy chain gene sequence. Blankenstein and Kruwinkel (1987), Eur. J. Immunol., 17:1351-1357 describe the mouse variable heavy chain region. See also, Joyner et al. (1989), Nature, 338:153-155, Traver et al. (1989), Proc. Nat. Acad. Sci. USA 86:5898-5902, Pachnis et al. (1990), Proc. Nat. Acad. Sci. USA, 87:5109-5113 and PCT application PCT/US91/00245. Bruggemann et al., Proc. Nat. Acad. Sci. USA; 86:6709-6713 (1989); Behring Inst. Mitt. 87:21-24 (1990); Eur. J. Immunol. 21:1323-1326 (1991), describe monoclonal antibodies with human heavy chains. Albertsen et al., Proc. Nat. Acad. Sci. USA 87:4256-4260 (1990), describe the construction of a library of yeast artificial chromosomes containing human DNA fragments. Yeast artificial chromosome vectors are described by Burke et al., Science 236:806-812 (1987). Pavan et al., Mol. and Cell. Biol. 10(8):4163-4169 (1990) describe the introduction of a neomycin resistance cassette into the human-derived insert of a yeast artificial chromosomes using homologous recombination and transfer into an embryonal carcinoma cell line using polyethylene glycol-mediated spheroplast fusion. Pachnis et al., Proc. Nat. Acad. Sci. USA 87:5109-5113 (1990), and Gnirke et al., EMBO Journal 10(7):1629-1634 (1991), describe the transfer of a yeast artificial chromosome carrying human DNA into mammalian cells. Eliceiri et al., Proc. Nat. Acad.USA 88:2179-2183 (1991), describe the expression in mouse cells of yeast artificial chromosomes containing human genes. Huxley et al., Genomics 9:742-750 (1991) describe the expression in mouse cells of yeast artificial chromosomes containing the human HPRT gene. Mortensen et al., Mol. and Cell. Biol. 12(5):2391-2395 (1992) describe the use of high concentrations of G418 to grow heterozygous embryonic stem cells for selection of homozygous mutationally altered cells. Yeast protoplast fusion with mouse fibroblasts is described by Traver et al., Proc. Nat. Acad. Sci. USA 86:5898-5902 (1989) and Pachnis et al., Proc. Nat. Acad. Sci. USA 87:5109-5113 (1990). Davies et al., Nucl. Acids Res. 20:2693-2698 (1992) describe targeted alterations in YACs. Zachau, Biol. Chem. 371:1-6 (1990) describes the human immunoglobulin light (kappa) (IgK) locus; Matsuda et al., Nature Genetics 3:88-94 (1993) and Shin et al., EMBO 10:3641-3645 (1991) describe the cloning of the human immunoglobulin heavy (IgH) locus in YACs.
  • SUMMARY OF THE INVENTION
  • Xenogeneic specific binding proteins are produced in a non-human viable host by immunization of the host with an appropriate immunogen. [0014]
  • A preferred non-human host is characterized by: (1) being incapable of producing endogenous immunoglobulin heavy chain; (2) being substantially incapable of producing endogenous immunoglobulin light chains; and (3) capable of producing xenogeneic immunoglobulin light and heavy chains to produce a xenogeneic immunoglobulin or immunoglobulin analog. Thus, the host may have an entire endogenous immunoglobulin locus substituted by a portion of, or an entire, xenogeneic immunoglobulin locus, or may have a xenogeneic immunoglobulin locus inserted into a chromosome of the host cell and an inactivated endogenous immunoglobulin region. These various alternatives will be achieved, at least in part, by employing homologous recombination for inactivation or replacement at the immunoglobulin loci for the heavy and light chains. [0015]
  • Additionally, novel methods are provided for introducing large segments of xenogeneic DNA of at least 100 kb, particularly human DNA, into host animals, particularly mice, by introducing a yeast artificial chromosome (YAC) containing a xenogeneic DNA segment of at least 100 kb, into an embryonic stem cell for integration into the genome of the stem cell, selection of stem cells comprising the integrated YAC by means of a marker present in the YAC, introduction of the YAC-containing ES cells into embryos and generation of chimeric mice from the embryos. The chimeric animals may be mated to provide animals that are heterozygous for the YAC. The heterozygous animals may be mated to generate progeny homozygous for the integrated YAC. [0016]
  • DESCRIPTION OF THE DRAWINGS
  • FIG. 1 is a diagram of the inactivation vector for the mouse heavy chain J region, as described in Example I, infra. [0017]
  • FIG. 2 is a diagram of the DNA restriction map for the plasmid pmHδJ and the targeted mouse heavy chain J genes, as described in Example II, infra. [0018]
  • FIG. 3 is a flow cytometry plot of antibody staining for IgM allotypes in mouse strains, as described in Example II, infra. [0019]
  • FIG. 4 is a flow cytometry histogram of antibody staining for IgM allotypes in mouse strains, as described in Example II, infra. [0020]
  • FIG. 5 is a diagram of the inactivation vector for the mouse immunoglobulin kappa constant region genes, as described in Example III, infra. [0021]
  • FIG. 6 is a diagram of the derivation of the plasmid pK.TK/Neo, as described in Example III, infra. [0022]
  • FIG. 7 is a diagram of the restriction map of the light chain targeted locus, as described in Example III, infra. [0023]
  • FIG. 8 is a diagram of the targeting vector for inactivation of the kappa light chain J and constant regions and design of the targeting experiment as described in Example IV, infra. [0024]
  • FIG. 9 is a diagram of the construction of vectors for inactivating the kappa light chain J and constant regions as described in Example IV, infra. [0025]
  • FIG. 10 is a diagram of the final deletion vectors for inactivation,of the kappa light chain J and constant regions as described in Example IV, infra. [0026]
  • FIG. 11 is an illustration of the Southern analysis of light chain J and constant region deleted cells as described in Example IV, infra. [0027]
  • FIGS. [0028] 12A-E are photographs of the results of Southern blot analysis to characterize yHPRT and yeast genomic DNA integrated in ES clones as described in Example VI, infra (A=human repetitive Alu sequence; B,C=pBR322-specific sequences for the right (B) and left (C) YAC arms; D=yeast Ty repetitive sequence; E=yeast single copy gene LYS2. Shorter exposure times (12 hrs for II as compared to 48 hrs for I) of yHPRT probed with Alu and Ty sequences also are also shown. Positions of molecular weight markers are indicated. Schemes of right (a) and left (b) vector arms and the locations of pBR322-derived YAC vector fragments are shown (=telomere;=yeast-derived sequences; 0 yeast centromere;=pBR322-derived sequences;=human insert;=EcoRI cloning site; H=HindIII sites).
  • FIGS. [0029] 13A-D are photomicrographs of the results of in situ hybridization to detect integration of yHPRT and yeast genomic sequences in ES cell chromosomes as described in Example VI, infra (A, B=metaphase spreads from ESY 8-7 cells hybridized to biotinylated human genomic sequences and C=metaphase spreads or D=interphase nuclei from ESY 8-6 cells hybridized to biotinylated yeast repeated DNA sequences).
  • FIGS. 14A, B, C demonstrates the stable retention of yHPRT during in vitro ES cell differentiation and transmission through the mouse germline, as described in Example VI, infra (A: a, b=embryoid bodies; and differentiated cell types: c=blood islands; d=contracting muscle; e=neuronal cells; f=neural tubules formed by ESY clones; B: Southern blot analysis of DNA extracted from differentiated ESY 5-2, 3-6, 8-5 and 8-6 (20 μg) and yHPRT in AB1380 (40 ng) using a=human Alu probe; b=yeast Ty sequences; C: Southern blot analysis of tail DNA (20 μg) from 2 agouti offspring (4-2 and 4-3) derived from ESY chimeric male 394/95-2 using a human Alu and b=Ty sequences; shorter exposures (12 hr) of 8-6 and yHPRT probed with Ty are shown (II). [0030]
  • FIGS. 15A and B are a photograph of an electrophoresis gel showing the expression of the human HPRT gene in various mouse tissues, as described in Example VI, infra ([0031] 15A=detection of human HPRT mRNA using reverse transcription-PCR in ES, ESY 3-1 and Hut 78 cells, spleen and liver from control mice or ESY 4-3 agouti offspring; 15B=detection of mouse γ-interferon receptor mRNA by RT-PCR in samples from 15A; M=size marker).
  • FIG. 16 is a diagram of the human immunoglobulin heavy chain locus, and a human heavy chain replacement YAC vector, as described in Example VII, infra. [0032]
  • FIG. 17 is a diagram of a mouse breeding scheme, as described in Example VIII, infra. [0033]
  • FIG. 18 depicts the genotypes of some of the host animals produced by the methods of the invention. [0034]
  • DESCRIPTION OF SPECIFIC EMBODIMENTS
  • Novel transgenic non-human hosts, particularly mammalian hosts, usually murine, are provided, where the host is capable of mounting an immune response to an immunogen, where the response produces antibodies having xenogeneic, particularly primate, and more particularly human, constant and/or variable regions or such other effect or peptide sequences of interest. By “transgenic” is meant an animal that contains a genetically engineered modification, particularly, as to this invention, the introduction of a human immunoglobulin gene, in all of its cells. The hosts are characterized by being capable of producing xenogeneic immunoglobulins or analogs thereof as a result of inactivation of the endogenous immunoglobulin subunit encoding loci and introduction of xenogeneic DNA, for example DNA encoding human immunoglobulin. The modifications may retain at least a portion of the xenogeneic constant regions which provide for assembly of the variable region binding site bonded at the C-terminus to a functional peptide. The functional peptide may take many forms or conformations and may serv as an enzyme, growth factor, binding protein, ligand, cytokine, effector protein, chelating proteins, etc. The antibodies may be of any isotype, e.g., IgA, D, E, G or M or subtypes within the isotype. [0035]
  • In a first strategy, as individual steps, the xenogeneic, e.g. human, heavy and light chain immunoglobulin genes are introduced into the host germ line (e.g. sperm or oocytes) and in separate steps the corresponding host genes are rendered non-functional by inactivation using homologous recombination. Human heavy and light chain immunoglobulin genes are reconstructed in an appropriate eukaryotic or prokaryotic microorganism and the resulting DNA fragments can be introduced into the appropriate host, for example into the pronuclei of fertilized mouse oocytes or embryonic stem cells. Inactivation of the endogenous host immunoglobulin loci is achieved by targeted disruption of the appropriate loci by homologous recombination in the host cells, particularly embryonic stem cells or pronuclei of fertilized mouse oocytes. The targeted disruption can involve introduction of a lesion or deletion in the target locus, or deletion within the target locus accompanied by insertion into the locus, for example, insertion of a selectable marker. In the case of embryonic stem cells, chimeric animals are generated which are derived in part from the modified embryonic stem cells and are capable of transmitting the genetic modifications through the germ line. The mating of hosts with introduced human immunoglobulin loci to strains with inactivated endogenous loci will yield animals whose antibody production is purely xenogeneic, e.g. human. [0036]
  • In a second, alternative strategy, at least portions of the human heavy and light chain immunoglobulin loci are used to directly replace the corresponding endogenous immunoglobulin loci by homologous recombination in embryonic stem cells. This results in simultaneous inactivation and replacement of the endogenous immunoglobulin. This is followed by the generation of chimeric animals in which the embryonic stem cell-derived cells can contribute to the germ line. [0037]
  • These strategies are based on the known organization of the immunoglobulin chain loci in a number of animals, since the organization, relative location of exons encoding individual domains, and location of splice sites and transcriptional elements is understood to varying degrees in the human, the immunoglobulin heavy chain (IgH[0038] hu) locus is located on chromosome 14. In the 5′-3′ direction of transcription, the locus comprises a large cluster of variable region genes (VH), the diversity (D) region genes, followed by the joining (JH) region genes and the constant (CH) gene cluster. The size of the locus is estimated to be about from 1,500 to about 2,500 kilobases (kb). During B-cell development, discontinuous gene segments from the germ line IgH locus are juxtaposed by means of a physical rearrangement of the DNA. In order for a functional heavy chain Ig polypeptide to be produced, three discontinuous DNA segments, from the VH, D, and JH regions must be joined in a specific sequential fashion; first D to JH then VH to DJH, generating the functional unit VHDJH. Once a VHDJH has been formed, specific heavy chains are produced following transcription of the Ig locus, utilizing as a template the specific VHDJHCH unit comprising exons and introns.
  • There are two loci for immunoglobulin light chains (IgL), the kappa locus on [0039] human chromosome 2 and the lambda locus on human chromosome 22. The organization of the IgL loci is similar to that of the IgH locus, except that the D region is not present. Following IgH rearrangement, rearrangement of a light chain locus is similarly accomplished by VL to JL joining of the kappa or lambda chain. The sizes of the lambda and kappa loci are each approximately 1000 kb to 2000 kb. Expression of rearranged IgH and an. Igκ or Igλ light chain in a particular B-cell allows for the generation of antibody molecules.
  • In order to isolate, clone and transfer the IgH[0040] hu locus, a yeast artificial chromosome or “YAC” may be employed. A YAC carrying the xenogeneic DNA may be introduced into ES cells or oocytes by a variety of methods, including yeast spheroplast: ES cell fusion, microinjection and lipofection. The YAC will integrat randomly (i.e. non-homologously) into the host genome. If yeast spheroplast: ES cell fusion is employed to introduce a YAC carrying xenogeneic DNA into ES host cells, then two or more YACs in a single yeast host cell may be introduced simultaneously into the same host ES cell. The advantage of this approach is that multiple YACs each containing xenogeneic DNA, for example human heavy and light chain immunoglobulin loci, can be introduced into a single chromosome in a host cell. This eliminates the need for breeding of animals containing individual human Ig genes in order to generate a host capable of producing fully human immunoglobulins. For example, a strain of yeast containing a single YAC is targeted with a vector such as pLUTO (described infra) to introduce a mammalian selectable marker such as HPRT, and a yeast selectable marker such as LYS2 into an arm of the YAC. Chromosomal DNA from the targeted strain is then used to transform a second, usually haploid, lys2 mutant yeast strain containing a second, different YAC. Lys+ colonies are then analyzed by pulsed-field gel electrophoresis (PFGE) to identify clones harboring the two YACs and to confirm that they are unaltered in size. Additional YACs with different selectable markers, for example ADE2 (if the host is an ade2 mutant), can subsequently be added by transformation. Alternatively, a YAC-containing strain of yeast is targeted with a vector such as pLUTO to introduce a mammalian selectable marker (e.g. HPRT), as above, and then mated to a second YAC-containing strain of opposite mating type. The presence of the two YACs is then confirmed in the diploid yeast cells as described above. The diploid yeast strain is used directly for fusion or put through meiosis and ascosporogenesis (sporulation) using standard procedures. The meiotic products are then screened to identify a haploid clone containing the two YACs. With either approach described above, the second YAC can be targeted with HPRT or another selectable marker prior to introduction of the first YAC. Also, if each YAC contains a different yeast selectable marker, maintenance of both YACS during strain propagation may be genetically selected. Fusion with ES cells is then carried out in the same manner as with yeast cells containing a single YAC. Because many yeast chromosomes may integrate along with the YAC, it is expected that a substantial portion of ES clones expressing the mammalian selectable marker present in one YAC (e.g. HATR clones if the YAC marker is HPRT, and the ES cells are HPRT−), will have integrated both YACs. Methods such as Southern analysis and/or PCR may be used to identify such clones, and Southern analysis employing pulsed-field gel electrophoresis used to characterize the extent of YAC integration.
  • The entire IgH[0041] hu locus can be contained within one or a few YAC clones along with a mammalian marker such as Neo, HPRT, GPT, β-gal, etc. The same is true for the Ig light chain loci. Reconstitution of intact germ line Ig loci by homologous recombination between YACs with overlapping regions of homology can be achieved in yeast. In this manner, the isolation of DNA fragments encoding the human Ig chain is obtained. Alternatively, one can directly clone an intact germline locus in a single YAC.
  • In order to obtain a broad spectrum of high affinity antibodies, it is not necessary that one include the entire V region. Various V region gene families are interspersed within the V region cluster in humans. Thus, by obtaining a subset of the known V region genes of the human heavy and light chain Ig loci (Berman et al., EMBO J. (1988) 7:727-738) rather than the entire complement of V regions, the transgenic host may be immunized and be capable of mounting a strong immune response and provide high affinity antibodies. In this manner, relatively small DNA fragments of the chromosome may be employed. For example, a reported 670 kb fragment of the Ig[0042] Hu locus is contained on a NotI-NotI restriction fragment, which would serve to provide a variety of V regions (Berman et al., supra). Increased diversity is also provided by recombination with the various D and J regions and somatic mutation.
  • To render the host immunoglobulin loci non-functional, homologous recombination may be employed, where DNA is introduced at the endogenous host immunoglobulin heavy chain and light chain loci which inhibits the production of endogenous immunoglobulin. Because there are two heavy chain alleles and two light chain loci, kappa and lambda, each with two alleles, although one may choose to ignore the lambda loci, there will have to be multiple transformations which result in inactivation of each of the alleles. Homologous recombination may be employed to functionally inactivate each of the loci, by introduction of the homologous DNA via a construct that can disrupt or delete the target. locus into embryonic stem cells, followed by introduction of the modified cells into recipient blastocysts. Subsequent breeding allows for germ-line transmission of the inactivated locus. One can therefore choose to breed heterozygous offspring and select for homozygous offspring from the heterozygous parents. [0043]
  • In the second, alternative strategy described above, the number of steps may be reduced by providing at least a fragment of the human immunoglobulin locus within the construct used for homologous recombination with th analogous endogenous immunoglobulin, so that the human locus is substituted for at least a part of the host immunoglobulin locus, with resulting inactivation of the host immunoglobulin subunit locus. Of particular interest is the use of transformation for a single inactivation, followed by breeding of the heterozygous offspring to produce a homozygous offspring. Where the human locus is employed for substitution or insertion into the host locus for inactivation, the number of transformations may be limited to three transformations and as already indicated, one may choose to ignore the less used locus and limit the transformations to two transformations. Alternatively, one may choose to provide for inactivation. as a separate step for each locus, employing embryonic stem cells from offspring which have previously had one or more loci inactivated. In the event that only transformation is used and the human locus is integrated into the host genome in random fashion, a total of eight or more transformations may be required. [0044]
  • For inactivation, any lesion in the target locus resulting in the prevention of expression of an immunoglobulin subunit of that locus may be employed. Thus, the lesion may be in a region comprising enhancers, e.g., a 5′ or 3′ enhancer, or intron, in the V, J or C regions, and with the heavy chain, the opportunity exists in the D region, or combinations thereof. The important factor is that Ig germ line gene rearrangement is inhibited, or a functional message encoding the enodgenous immunoglobulin cannot be produced, either due to failure of transcription, failure of processing of the message, or the like. Such a lesion may take the form of a deletion in the target gene, an insertion of a foreign gene, a combination of an insertion and deletion, or a replacement using xenogeneic sequences with or without introduction of a deletion in the endogenous gene. [0045]
  • Preferably, when one is interested in inactivating the immunoglobulin subunit locus, the lesion will be introduced into one or more of the exons contained in the immunoglobulin subunit locus, for example in the constant or J region of the locus. Thus, one produces a targeting construct which lacks functional exons in this region and may comprise the sequences adjacent to and upstream and/or downstream from the J and/or C region or comprises all or part of the region with an inactivating insertion in the J or C exons. The insertion may be 50 bp or more, where such an insertion results in disruption of formation of a functional mRNA. Desirably, usually at least about 75% of the exon sequence, preferably at least about 90% of the exon sequence, is deleted. [0046]
  • Desirably, a marker gene is used in the targeting construct to replace the deleted sequences. Various markers may be employed, particularly those which allow for positive selection. Of particular interest is the use of G418 resistance, resulting from expression of the gene for neomycin phosphotransferase (“neo”). [0047]
  • In the targeting construct, upstream and/or downstream from the target gene, may be a, gene which provides for identification of whether a homologous double crossover has occurred (negative selection). For this purpose, the Herpes simplex virus thymidine kinase gene may be employed, since cells expressing the thymidine kinase gene may be killed by the use of nucleoside analogs such as acyclovir or gancyclovir, by their cytotoxic effects on cells that contain a functional HSV-tk (Mansour et al., [0048] Nature 336:348-352 (1988)). The absence of sensitivity to these nucleoside analogs indicates the absence of the HSV-thymidine kinase gene and, therefore, where homologous recombination has occurred, that a double crossover has also occurred.
  • While the presence of the marker gene in the genome will indicate that integration has occurred, it will still be necessary to determine whether homologous integration has occurred. This can be achieved in a number of ways. For the most part, DNA analysis by Southern blot hybridization will be employed to establish the location of the integration. By employing probes for the insert and the sequences at the 5′ and 3′ regions flanking the region where homologous integration would occur, one can demonstrate that homologous targeting has occurred. [0049]
  • PCR may also be used with advantage in detecting the presence of homologous recombination. PCR primers may be used which are complementary to a sequence within the targeting construct and complementary to a sequence outside the construct and at the target locus. In this way, one can only obtain DNA molecules having both the primers present in the complementary strands if homologous recombination has occurred. By demonstrating the expected size fragments, e.g. using Southern blot analysis, the occurrence of homologous recombination is supported. [0050]
  • The targeting construct may further include a replication system which is functional in the host cell. For the most part, these replication systems will involve viral replication systems, such as Simian virus 40, Epstein-Barr virus, polyoma virus, papilloma virus, and the like. Various transcriptional initiation systems may be employed, either from viruses or from mammalian genes, such as SV40, metallathionein-I and II genes, β-actin gene, adenovirus early and late genes, phosphoglycerate kinase gene, RNA polymerase II gene, or the like. In addition to promoters, wild-type enhancers may be employed to further enhance the expression of the marker gene. [0051]
  • In preparing the targeting constructs for homologous recombination, a replication system for procaryotes, particularly [0052] E. coli, may be included for preparing the targeting construct, subcloning after each manipulation, analysis such as restriction mapping or sequencing, expansion and isolation of the desired sequence. In the case of the replacement strategy, where the xenogeneic DNA insert is large, generally exceeding about 50 kbp, usually exceeding 100 kbp, and usually not more than about 1000 kbp, a yeast artificial chromosome (YAC) may be used for cloning of the targeting construct.
  • Once a targeting construct has been prepared and any undesirable sequences removed, e.g., procaryotic sequences, the construct may now be introduced into the target cell, for example an ES cell. Any convenient technique for introducing the DNA into the target cells may be employed. Techniques include protoplast fusion, e.g. yeast spheroplast:cell fusion, lipofection, electroporation, calcium phosphate-mediated DNA transfer or direct microinjection. [0053]
  • After transformation or transfection of the target cells, target cells may be selected by means of positive and/or negative markers, as previously indicated, neomycin resistance and acyclovir or gancyclovir resistance. Those cells which show the desired phenotype may then be further analyzed by restriction analysis, electrophoresis, Southern analysis, PCR, or the like. By identifying fragments which show the presence of the lesion(s) at the target locus, one can identify cells in which homologous recombination has occurred to inactivate a copy of the target locus. [0054]
  • The above described process may be performed first to inactivate a heavy chain locus in an embryonic stem cell whereby the cells are microinjected into host blastocysts which develop into a chimeric animal. The chimeric animals are bred to obtain heterozygous hosts. Then, by breeding of the heterozygous hosts, a homozygous host may be obtained or embryonic stem cells may be isolated and transformed to inactivate the second IgH locus, and the process repeated until all the desired loci have been inactivated. Alternatively, the light chain locus may be the first to be inactivated. For complete elimination of the ability to produce light chain immunoglobulin, it is desirable to inactivate both the lambda and the kappa light chain immunoglobulin loci. At any stage, the xenogeneic loci may be introduced. [0055]
  • As already indicated, the target locus may be substituted with the analogous xenogeneic locus. In this way, the xenogeneic locus will be placed substantially in the same region as the analogous host locus, so that any regulation associated with the position of the locus will be substantially the same for the xenogeneic immunoglobulin locus. For example, by isolating the variable region of the human IgH locus (including V, D, and J sequences), or portion thereof, and flanking the human locus with sequences from the murine locus, preferably sequences separated by at least about 5 kbp, in the host locus, preferably at least about 10 kbp in the host locus, one may insert the human fragment into this region in a recombinational event(s), substituting the human immunoglobulin locus for the endogenous variable region of the host immunoglobulin locus. In this manner, one may disrupt the ability of the host to produce an endogenous immunoglobulin subunit, while allowing for the promoter of the human immunoglobulin locus to be activated by the host enhancer and regulated by the regulatory system of the host. [0056]
  • In order to provide for the production of xenogeneic binding proteins in a host, it is necessary that the host be competent to provide the necessary enzymes and other factors involved with the production of antibodies, while lacking competent endogenous genes for the expression of heavy and light subunits of immunoglobulins. Thus, those enzymes and other factors associated with germ line rearrangement, splicing, somatic mutation, and the like will be functional in the host. What will be lacking is a functional natural region comprising the various exons associated with the production of endogenous immunoglobulin. [0057]
  • The integration of introduced xenogeneic DNA may be random or homologous depending on the particular strategy to be employed. Thus, by using transformation, using repetitive steps or in combination with breeding, transgenic animals may be obtained which are able to produce xenogeneic binding proteins in the substantial absence of light or heavy endogenous immunoglobulin. By transformation is intended any technique for introducing DNA into a viable cell, such as conjugation, PEG-mediated cell fusion, transformation, transfection, transduction, electroporation, lipofection, biolistics, or the like. [0058]
  • Once the xenogeneic loci, have been introduced into the host genome, either by homologous recombination or random integration, and host animals have been produced with the endogenous immunoglobulin loci inactivated by appropriate breeding of the various transgenic animals or animals derived from chimeric animals, one can produce a host which lacks the native capability to produce endogenous immunoglobulin, but has the capacity to produce xenogeneic immunoglobulins with at least a significant portion of the repertoire of the xenogeneic source. [0059]
  • The functional inactivation of the two copies of each of the three host Ig loci (heavy, kappa and lambda), where the host then contains the human IgH and the human Ig kappa and/or lambda loci would allow for the production of purely human antibody molecules without the production of host or host/human chimeric antibodies. Such a host strain, by immunization with specific antigens, would respond by the production of murine B-cells producing specific human antibodies, which B-cells could be fused with murine myeloma cells or be immortalized in any other manner for the continuous stable production of human monoclonal antibodies. Methods are well known in the art for obtaining continuous stable production of monoclonal antibodies. [0060]
  • The subject methodology and strategies need not be limited to producing complete immunoglobulins, but provides the opportunity to provide for regions joined to a portion of the constant region, e.g., C[0061] H1, CH2, CH3, or CH4, or combination thereof. Alternatively, one or more of the exons of the CH and Cκ or Cλ regions may be replaced or joined to a sequence encoding a different protein, such as an enzyme, e.g., plasminogen activator, superoxide dismutase, etc.; toxin, e.g., ricin, abrin, diphtheria toxin, etc.; growth factor; cytotoxic agent, e.g., TNF; receptor ligand, or the like. See, for example, WO 89/07142; WO 89/09344; and WO 88/03559. By inserting the protein of interest into a constant region exon and providing for splicing of the variable region to the modified constant region exon, the resulting binding protein may have a different C-terminal region from the immunoglobulin. By providing for a stop sequence with the inserted gene, the protein product will have the inserted protein as the C-terminal region. If desired, the constant region may be entirely substituted by the other protein, by providing for a construct with the appropriate splice sites for joining the variable region to the other protein.
  • The B-cells from the transgenic host producing immunoglobulin or immunoglobulin analog may be used for fusion to a murine myeloid cell to produce hybridomas or immortalized by other conventional process, e.g., transfection with oncogenes. These immortalized cells may then be grown in continuous culture or introduced into the peritoneum of a compatible host for production of ascites. [0062]
  • The subject invention provides for the production of polyclonal human anti-serum or human monoclonal antibodies or antibody analogs. Where the mammalian host has been immunized with an immunogen, the resulting human antibodies may be isolated from other proteins by using an affinity column, having an Fc binding moiety, such as protein A, or the like. [0063]
  • The invention includes the following embodiments of non-human hosts (see also FIG. 18): [0064]
  • I. Animals heterozygous for an inactive endogenous light chain immunoglobulin gene (homozygous animals are obtained by interbreeding); [0065]
  • II. Animals heterozygous for an inactive endogenous heavy chain immunoglobulin gene (homozygous animals are obtained by interbreeding); [0066]
  • III. Animals homozygous for functional endogenous light and heavy chain immunoglobulin genes and hemizygous for (i.e. containing one copy of) foreign, preferably human, heavy chain immunoglobulin genes (homozygous animals are obtained by interbreeding); [0067]
  • IV. Animals homozygous for functional endogenous light and heavy chain immunoglobulin genes and hemizygous for foreign, preferably human, light chain immunoglobulin genes (homozygous animals are obtained by interbreeding); [0068]
  • V. Animals heterozygous for inactive endogenous heavy and light chain immunoglobulin genes obtained by crossbreeding animals of category I with animals from category II (homozygous animals are obtained by interbreeding); [0069]
  • VI. Animals heterozygous for inactive endogenous heavy and light chain immunoglobulin genes and hemizygous for foreign, preferably human, heavy chain immunoglobulin genes obtained by crossbreeding animals of category III with animals from category V (animals homozygous for the inactive endogenous loci and homo- or hemizygous for the foreign gene are obtained by interbreeding); [0070]
  • VII. Animals heterozygous for inactive endogenous heavy and light chain immunoglobulin genes and hemizygous for foreign, preferably human, light chain immunoglobulin genes obtained by crossbreeding animals of category IV with animals from category V (animals homozygous for th inactive endogenous loci and homo- or hemizygous for the foreign gene are obtained by interbreeding); [0071]
  • VIII. Animals homozygous or heterozygous for inactive endogenous heavy and light chain immunoglobulin genes and hemizygous for foreign, preferably human, light and heavy chain immunoglobulin genes, obtained by crossbreeding animals of category VI and VII (animals homozygous for the inactive endogenous loci and homo- or hemizygous for the foreign gene are obtained by interbreeding); [0072]
  • In a preferred embodiment, the homozygous animals of category VIII are used to produce human antibodies. [0073]
  • IX. Animals homozygous for functional endogenous heavy and light chain immunoglobulin genes and hemizygous for foreign, preferably human, heavy and light chain immunoglobulin genes, obtained by crossbreeding animals of category III and IV (homozygous animals are obtained by interbreeding); [0074]
  • X. Animals heterozygous for an inactive endogenous heavy chain immunoglobulin gene and hemizygous for foreign, preferably human, heavy and light chain immunoglobulin genes, obtained by crossbreeding animals of category II and IX (animals homozygous for the inactive endogenous loci and homo- or hemizygous for the foreign gene are obtained by interbreeding). [0075]
  • XI. Animals heterozygous for an inactive endogenous light chain immunoglobulin gene and hemizygous for foreign, preferably human, heavy and light chain immunoglobulin genes, obtained by crossbreeding animals of category I and IX (animals homozygous for the inactive endogenous loci and homo- or hemizygous for the foreign gene are obtained by interbreeding). [0076]
  • The invention also provides a method for introducing large continuous, xenogeneic DNA sequences into a nonhuman, e.g. mammalian, host. Usually, the sequences will be at least 100 kb, more usually at least about 200 kb, generally ranging from about 200 to 1000 kb. Thus, one may wish to transfer a locus of interest, such as the immunoglobulin locus, T-cell receptor locus, major histocompatibility locus; regions of an xenogeneic chromosome, which may include one or more genes of interest, which may or may not have been characterized, such as the Low Density Lipoprotein (LDL) receptor, Apolipoprotein (Apo) B, Apo E, cystic fibrosis transmembrane conductor regulator, dystrophin, or regions of xenogeneic chromosomes that may be involved in partial chromosome trisomy ([0077] e.g. chromosomes 21, 7 and 10); and viruses. The DNA may comprise wild type or defective genes for studying a variety of diseases by creating dominant mutations or complementing recessive mutations, for example the LDL receptor and Apo B genes can be introduced for the study of hypercholesterolemia, hyperlipoproteinemia and atherosclerosis, Factor VIII or IX can be introduced for hemophilia, cystic fibrosis transmembrane conductance regulator can be introduced for cystic fibrosis and the dystrophin gene for muscular dystrophy. The xenogeneic DNA to be introduced using a YAC is from a mammalian source, particularly primates, more particularly human, other vertebrates or invertebrates and the like. One can thus impart numerous novel capabilities to the host, create genetic responses related to the xenogeneic source of the DNA, provide for the production of antibodies, provide for specific combinations of transcription factors, provide for metabolic systems, introduce dominant mutations or complement recessive mutations. The xenogeneic DNA may be modified when present in a YAC. Because homologous recombination is efficient in yeast, giving a high ratio of site-specific integration of homologous DNA, where the homologous DNA flanks other DNA of interest, one is able to modify the xenogeneic DNA before introduction into an ES cell. In this way, one can introduce defective genes into the host which express defective proteins to mimic diseased states of the xenogeneic host, to study various mechanisms of the interaction of defective proteins with other xenogeneic proteins or endogenous proteins, or to study genes or gene systems.
  • In general, to transfer large DNA segments, as described in detail herein, YACs are employed which comprise a yeast centromere, an origin of replication and telomeres bounding the DNA of interest. Various centromeres or telomeres may be used, particularly the centromeres from [0078] yeast chromosomes 4 and 5. The YAC has a marker which allows for selection or screening of cells into which the YAC becomes integrated. Not all markers allow for efficient selection. Particularly, the HPRT gene, more particularly human HPRT, is found to permit efficient selection of HPRT-deficient ES cells carrying the YAC. Other known selectable or screenable markers include hygromycin, neomycin, β-gal, and GPT. The ES cell may be derived from any non-human host, from which ES cells are available, and can be expanded in culture, which remain viable and functional, for which a marker for selection exists, and where the ES cell can be introduced into an embryo and can repopulate the host, including the germline. For the most part this capability has been established with rodents, e.g. mice and rats, and to a lesser extent with guinea pigs. Mice have been used for the production of antibodies or B-lymphocytes for immortalization for the production of antibodies. Because mice are easy to handle, can be produced in large quantities, and are known to have an extensive immune repertoire, mice will usually be the animal of choice. As other species of ES cells become available, these may also be employed in accordance with the subject invention. Of particular interest will be small laboratory animals, or domestic animals particularly rodents, including mice, rats, rabbits, cows, pigs, hamsters, horses, dogs, sheep and guinea pigs, or birds such as chickens, turkeys, etc. The ES cells may have one or more mutations, for example lacking a particular activity. Of particular interest in this invention are ES cells that are deficient in HPRT. In addition, fertilized eggs of certain species may find use in accordance with the invention.
  • The YAC may be obtained by screening existing human YAC libraries such as those available from the Centre d'Etude du Polymorphisme Human (C.E.P.H.), Paris, France, and Washington University, St. Louis, Mo., using standard procedures. Alternatively, the YAC is readily prepared as described in detail herein, by joining the yeast flanking segments comprising one arm with a centromere and telomere and another with a telomere together with the DNA of interest. Usually there will also be one or more markers present that allow for selection in the yeast host cells. For yeast selection, of particular interest are markers which complement mutations of the yeast host, such as genes involved in the production of amino acids, purines or pyrimidines, URA3, TRP1, LYS2, ADE2 on the YAC to complement ura3, trp1, lys2 and Ade2 mutations in the host. By providing for complementation, for the most part only yeast cells carrying the entire YAC will be able to survive in a selective medium. In addition to genetic verification that both YAC arms have been retained, it is desirable to confirm the integrity of the YAC using a method such as pulsed-field gel electrophoresis. [0079]
  • Those yeast hosts carrying the YAC may then be used as a source of the YAC for introduction into the ES cell. Transfer of the, YAC is efficiently achieved by preparing yeast spheroplasts in accordance with conventional ways. By degrading the outer wall, under mild conditions, in an isotonic medium, spheroplasts are produced in high yield. Exponentially growing ES cells are protease-treated, e.g. trypsinized, and combined with the spheroplasts. Conveniently, a pellet of yeast spheroplasts can be prepared and the ES cells are spun with the pellet and exposed to a fusogenic agent such as PEG for 1-2 minutes. The cells are then resuspended and incubated in appropriate serrum-free medium. The cells are then plated onto feeder cells, followed by selection in accordance with the selective marker. For the HPRT gene, HAT medium may be employed for selection. Surviving fusion colonies are then, picked, expanded and analyzed. Analysis may be performed by restriction enzyme analysis, combined with Southern blotting or pulsed-field gel electrophoresis, or by the polymerase chain reaction (PCR), employing appropriate primers, at least one of which is complementary to the DNA insert, and probing with repetitive sequences present in the xenogeneic DNA, such as Alu, for detection of human DNA sequences. Ty, Y′, rDNA, delta sequences are used to probe for for yeast sequences. Probes for YAC ends are used to confirm integrity of the YAC. Those cells that demonstrate the intact or substantially intact YAC DNA integrated into the host genome are then used in the next steps. In some clones, only a portion or little or none of the yeast DNA becomes integrated into the mouse genome. The integrated yeast DNA ranges from more than about 90% of the original yeast genome to less than about 10%. [0080]
  • In a preferred embodiment, efficient production of transgenic non-human hosts is provided using a process which integrates large, at least 100 kb, xenogeneic DNA fragments, in substantially intact form, into a host embryonic stem (ES) cell or fertilized egg (zygote). The introduction of the xenogeneic DNA is efficiently achieved by fusion of the ES cell with yeast spheroplasts that contain YACs carrying the 100 kb DNA and a selectable marker, under conditions allowing for integration of the YAC DNA containing the marker into the ES cell genome, or by transfection of a purified YAC into ES cells. ES cells comprising the YAC integrated into the genome are then selected by means of the marker, which is functional in the ES cell. For example, the hypoxanthine phosphoribosyl transferase (HPRT) gene may be used as a marker in HPRT deficient (HPRT−) ES cells. For producing animals from embryonic stem cells, after transformation, the cells may be plated onto a feeder layer in an appropriate medium, e.g. fetal bovine serum enhanced DMEM. The ES cell may have a single targeted locus (heterozygous), or may be manipulated by the process of homogenotization to have both loci targeted (homozygous). The process of homogenotization (formation of homozygotes) uses selective pressure to grow out those cells which have the gene targeting event on both chromosomes. Cells containing the two targeted alleles may be detected by employing a selective medium and after sufficient time for colonies to grow, colonies may be picked and analyzed for the occurrence of integration or homologous recombination. As described previously, the PCR may be used, with primers within or outside of the construct sequence, but at the target locus. [0081]
  • Those colonies which show homologous recombination may then be used for embryo manipulation and blastocyst injection. The selected ES cells are then introduced into embryos, by microinjection or other means, into the appropriate host. For example, murine blastocyts may be obtained from female animals by flushing the uterus 3.5 days after ovulation. The modified ES cells are then trypsinized and at least 1 and up to 15 cells may be injected into the blastocoel of the blastocyst. After injection, at least 1 and no more than about 10 of the blastocysts are returned to each uterine horn of pseudo-pregnant females. The females proceed to term and the resulting chimeric animals can be analyzed for the presence of the YAC in their somatic cells. By “chimeric” is meant an animal that carries cells derived from more than one source, e.g. from the host and another animal. For example, in the present invention a chimeric murine animal contains a genetically engineered modification, particularly a human gene, in some of its cells, e.g. in cells that develop from the modified embryonic stem cells. The presence of the integrated YAC in chimeric hosts that are generated is then analyzed. The chimeric hosts are evaluated for germline transmission of the ES cell genome by mating, for example chimeric mice are mated with C57BL/6J mice. Chimeric hosts may be bred with non-chimeric hosts, either syngeneic or allogeneic, to screen for chimeras that carry the YAC in their germ cells. Offspring that are heterozygous for the genetic modification are then interbred to produce progeny that are homozygous for the modification, stably transmitting the functioning YAC construct to their progeny. [0082]
  • The method of the invention for introduction of large xenogeneic DNA segments into a non-human host, particularly a rodent and usually a murine animal, provides for stable integration of the DNA. Genes in the inserted DNA are found to be functional and the resulting chimeric hosts are able to provide for germline transmission of the integrated DNA. After breeding of the chimeric host, transgenic heterozygous hosts are produced and are mated to produce a homozygous animal that may be used for a wide variety of purposes, including production of products, such as binding proteins, for example immunoglobulins, for screening of various drugs, for gene therapy, for example to complement for recessive genetic disorders, to study various diseases, to study the function and regulation of poorly mapped large DNA fragments. [0083]
  • The following examples are offered by way of illustration and not by way of limitation. [0084]
  • EXPERIMENTAL Example I I. Inactivation of the Mouse Heavy Chain J (JH) Genes
  • A. Construction of the Targeting Inactivation Vector [0085]
  • A 6.4 kb EcoRI fragment, containing the mouse heavy chain J genes and flanking sequences, is cloned from a Balb/c mouse embryo genomic library using the probes described in Sakano et al. (1981), [0086] Nature 290:562-565. This fragment (mDJ) is inserted into EcoRI-digested pUC19 plasmid (pmDJ). A 2.9 Kb fragment, containing the 4 J genes, is deleted by XhoI-ScaI digestion (pmDδJNeo, see FIG. 1). An 1150 bp Xhol-BamHI fragment, containing a neomycin-resistance gene driven by the Herpes simplex virus thymidine kinase gene (HSV-tk) promoter and a polyoma enhancer is isolated from pMC1Neo (Thomas and Capecchi (1987), Cell, 5, 503-512). A synthetic adaptor is added onto this fragment to convert the BamHI end into a ScaI end and the resulting fragment is joined to the XhoI-ScaI pmDδJ to form the inactivation vector (pmDδJ.Neo) in which the 5′ to 3′ orientation of the neomycin and the heavy chain promoters is identical. This plasmid is linearized by NdeI digestion before transfection to ES cells. The sequences driving the homologous recombination event are 3 kb and 0.5 kb fragments, located 5′ and 3′ to the neomycin gene, respectively.
  • B. Culturing Electroporation and Selection of ES Cells [0087]
  • The ES cell line E14TG2a (Hooper et al. (1987), [0088] Nature, 326:292-295) is cultured on mitomycin treated primary embryonic fibroblast-feeder layers essentially as described (Doetschman et al. (1985), J. Embryol. Exp. Morphol. 87:27-45). The embryonic fibroblasts are prepared from embryos from C57BL/6 females that are mated 14 to 17 days earlier with a male homozygous for a neomycin transgene (Gossler et al. (1986), PNAS 83:9065-9069). These cells are capable of growth in media containing G418. Electroporation conditions are described by (Boggs et al. (1986), Ex. Hematol. (NY) 149:988-994). ES cells are trypsinized, resuspended in culture media at a concentration of 4×107/ml and electroporated in the presence of the targeting DNA construct at a concentration of 12 nM in the first experiment and 5 nM DNA in the second. A voltage of 300 V with a capacitance of 150-250 μF is found optimal with an electroporation cell of 5 mm length and 100 mm2 cross-section. 5×106 electroporated cells are plated onto mitomycin-treated fibroblasts in 100 mm dishes in the presence of Dulbecco's modified Eagle's media (DMEM) supplemented with 15% fetal bovine serum (FBS) and 0.1 mM 2-mercaptoethanol. The media is replaced 24 hrs after electroporation with media containing 200 μg/ml G418.
  • ES colonies resulting 10-14 days after electroporation are picked with drawn out capillary pipettes for analysis using PCR. Half of each picked colony is saved in 24-well plates already seeded with mitomycin-treated feeder cells. The other halves, combined in pools of 3-4, are transferred to Eppendorf tubes containing approximately 0.5 ml of PBS and analyzed for homologous recombination by PCR. Conditions for PCR reactions are essentially as described (Kim and Smithies (1988), Nucleic Acids Res. 16:8887-8893). After pelleting, the ES cells are resuspended in 5 μl of PBS and are lysed by the addition of 55 μl of H[0089] 2O to each tube. DNAses are inactivated by heating each tube at 95° C. for 10 min. After treatment with proteinase K at 55° C. for 30 min, 30 μl of each lysate is transferred to a tube containing 20 μl of a reaction mixture including PCR buffer: 1.5 μg of each primer, 3U of Taq polymerase, 10% DMSO, and dNTPs, each at 0.2 mM. The PCR expansion employs 55 cycles using a thermocycler with 65 seconds melt at 92° C. and a 10 min annealing and extension time at 65° C. The two priming, oligonucleotides are TGGCGGACCGCTATCCCCCAGGAC and TAGCCTGGGTCCCTCCTTAC, which correspond respectively to a region 650 bases 3′ of the start codon of the neomycin gene and sequences located in the mouse heavy chain gene, 1100 bases 3′ of the insertion site. 20 μl of the reaction mix is electrophoresed on agarose gels and transferred to nylon membranes (Zeta Bind). Filters are probed with a 32P-labelled fragment of the 991 bp XbaI fragment of the J-C region.
  • Example II II. Deletion of the Mouse Ig Heavy Chain J (JH) Genes in ES Cells
  • A. Construction of the Replacement Targeting Vector [0090]
  • A 6.1-Kb EcoRI fragment, containing the mouse immunoglobulin heavy chain J region genes and flanking sequences, cloned from a BALB/c mouse embryo genomic library and inserted into pUC18 (pJ[0091] H), was digested with XhoI and NaeI to delete an about 2.3 kb fragment containing the four J genes (see FIG. 2A). An about 1.1 kb XhoI-BamHI fragment, blunted at the BamHI site, containing a neomycin resistance gene driven by the Herpes simplex virus thymidine kinase gene (HSV-tk) promoter and polyoma enhancer was isolated from pMC1Neo (Thomas and Capecchi (1987), Cell, 51, 503-512). This fragment was inserted into the XhoI-NaeI deleted pJH to form the deletion vector (pmHδJ, see FIG. 2B), in which the transcriptional orientation of the neomycin and the heavy chain genes is the same. This plasmid was linearized by NdeI digestion before transfection to ES cells. The sequences driving the homologous recombination event are about 2.8 kb and about 1.1 kb fragments, located 5′ and 3′ to the neomycin gene, respectively.
  • B. Culturing, Electroporation, and Selection of ES Cells [0092]
  • The ES cell line E14TG2a (Koller and Smithies (1989), [0093] PNAS USA, 86:8932-8935) was cultured on mitomycin C-treated embryonic fibroblast feeder layers as described (Koller and Smithies (1989), PNAS USA, 86:8932-8935). ES cells were trypsinized, resuspended in HBS buffer (pH 7.05; 137 mM NaCl, 5 mM KCl, 2 mM CaCl2, 0.7 mM Na2HPO4, 21 mM HEPES pH 7.1) at a concentration of 2×107/ml and electroporated in the presence of 50 μg/ml of the linearized inactivation vector. Electroporation was carried out with a BioRad Gene Pulser using 240 volts and 500 μF capacitance. 5×106 electroporated cells were plated onto mitomycin C-treated fibroblasts in 100 mm dishes in the presence of Dulbecco's modified Eagle's media (DMEM) supplemented with 15% fetal bovine serum and 0.1 mM 2-mercaptoethanol. The media was replaced 24 hr after electroporation with media containing 200 μg/ml G418. G418-resistant ES colonies resulting from growth 12-14 days after electroporation were picked with drawn out capillary pipettes for analysis using the polymerase chain reaction (PCR). Half of each picked colony was transferred to an individual well of a 24-well plate, already seeded with mitomycin C-treated feeder cells. The other halves, combined in pools of four, were transferred to Eppendorf tubes containing 0.3 ml of PBS and cell lysates were prepared for PCR analysis as described by Joyner et al (1989) Nature, 338:153-155. The PCR reaction included 5-20 μl of the cell lysate, 1 μM of each primer, 1.5 U of Taq polymerase and 200 μM of dNTPs. The PCR amplification employed 45 cycles using a thermal cycler (Perkin-Elmer Cetus), with 1 min. melt at 94° C., 2 min. annealing at 55° C., and 3 min. extension at 72° C. The two priming oligonucleotides are ACGGTATCGCCGCTCCCGAT and AGTCACTGTAAAGACTTCGGGTA, which correspond respectively to about 120 bases 5′ of the BamHI site of the neomycin gene, and to the sequences located in the mouse heavy chain gene, about 160 bases 3′ of the insertion site. Successful homologous recombination gives rise to an about 1.4 kb fragment. 20 μl of the reaction mixture is electrophoresed on 1% agarose gels, stained with ethidium bromide and transferred to nylon membranes (Gene Screen). Filters were probed with a 32P-labelled EcoRI-PstI about 1.4 kb fragment located in the mouse heavy chain, 3′ of the insertion site (see FIG. 2). For further analysis, genomic DNA was prepared from ES cells, digested with restriction enzymes as recommended by the manufacturers, and fragments were separated on 1% agarose gels. DNA was transferred to nylon membranes (Gene Screen) and probed with the 32P-labelled fragment as described above.
  • C. Analysis of G418-Resistant ES Colonies [0094]
  • In the first experiment, PCR analysis of the pooled colonies detected one positive PCR signal of the expected size (about 1.4 kb) out of 34 pools representing 136 G418-resistant colonies. The four individual colonies that had contributed to this positive pool were analyzed individually by PCR, and a positive clone, ES33D5, was identified. Similar analysis of 540 G418-resistant colonies obtained in the second experiment yielded 4 additional positive clones (ES41-1, ES61-1, ES65-1, ES101-1). [0095]
  • In order to verify the targeted disruption of one copy of the J genes, (the gene is autosomal and thus present in two copies), the PCR positive clones were expanded and genomic DNA was prepared, digested with HindIII or with SacI and analyzed by Southern analysis as described using the EcoRI-PstI probe. [0096]
  • The replacement of the J genes by insertion of the neomycin gene by an homologous recombination event results in a HindIII fragment, detectable with the EcoRI-PstI probe, which is about 1.9 kb longer than the equivalent fragment in the native locus, due to the loss of two HindIII sites located in the deleted J gene region (see FIG. 2C). Southern analysis of each of the 5 positive clones by HindIII digestion gave a pattern which indicated that one of the two copies of the heavy chain J genes had been disrupted. Three labelled fragments were detected: one fragment (about 760 bp), identical in size to that present in untreated cells at the same intensity, one fragment (about 2.3 kb) identical in size to that present in untreated cells, but of decreased intensity in the PCR positive clone, and an additional fragment about 4.2 kb, the size predicted for an homologous recombination event, present only in the PCR-positive clones. Similarly, th replacement of the J genes by the neomycin gene by an homologous recombination event results in a loss of one SacI site and the appearance of a fragment, detectable with the EcoRI-PstI probe, which is about 570 bp smaller than the equivalent fragment in the native locus (see FIG. 2C). Southern analysis of the clones by SacI digestion gave the expected pattern of one native and one targeted allele: about 4.0 kb fragment, identical in size to that detected in untreated cells, but of decreased intensity in the 5 positive clones, and an additional fragment of about 3.4 kb, the size predicted for a targeted homologous recombination event, present only in the identified clones. Rehybridization of the Southern blots with a probe for the neomycin gene showed that only the 4.2 kb and 3.4 kb fragments, resulting from the HindIII and the SacI digestion, respectively, hybridized to the probe as predicted by the targeting event. [0097]
  • D. Generation of Chimeric Mice with J[0098] H Deletions
  • Three and a half day old C57BL/6J (Jackson Laboratories, Bar Harbor, Me.) blastocysts were obtained from 4-5 week old superovulated females as described by Koller, et al. 1989 (supra). ES cells were trypsinized, washed once with fresh DMEM media, and diluted to about 1×10[0099] 6/ml in DMEM medium containing 10% fetal bovine serum and 20 mM HEPES, pH 7.5. 10 to 15 cells were injected into the blastocoel of each blastocyst. ES-cell containing blastocysts were then surgically transferred to one uterine horn of C57BL/6J×DBA/2 or C57BL/6J×CBA F1 pseudopregnant females.
  • The contribution of ES cells to the offspring was judged visually by examination of the coat color of the pups. C57BL/6J mice are solid black in color. The ES cell parent line E14TG2a was isolated from 129/Ola embryos, which carry three coat color genes, the dominant A[0100] W allele at the agouti locus, the recessive pink-eyes-dilute allele at the p locus, and the recessive Cch at the c locus. Chimeric offspring in which the ES cells participated in the formation of the animal have coats containing agouti and cream hairs.
  • Germline transmission ability of the chimeric mice was evaluated by mating with a C57BL/6J mouse and scoring for F1 offspring with agouti color. 50% of these agouti mice would be expected to inherit the mutated heavy chain allele, which can be identified by Southern blot analysis of DNA isolated from tails. [0101]
  • The J[0102] H-targeted ES cell line ES65-1, carrying one targeted heavy chain allele, was injected into C57BL/6J mouse blastocysts. About 45% of the surviving pups were chimeras. Two chimeric females, 238-2 and 244-3, upon mating with C57BL/6J males, yielded germline transmission at a frequency of 100% and 15%, as determined by the percent of agouti offspring. Southern blot analysis of DNA from heterozygous offspring indicated the presence of the targeted heavy chain in addition to one native allele in 2 out of 5 agouti progeny tested.
  • Mice homozygous for the mutation were obtained by intercrossing male and female mice which were identified as J[0103] H-deleted (δJH) heterozygotes. Offspring of these matings were analyzed for the presence of the two targeted heavy chain alleles by Southern blot analysis.
  • E. Analysis of B Cells from Chimeric Mice [0104]
  • If deletion of the J[0105] H region is sufficient to inactivate the heavy chain locus, then it should result in complete block of development of IgM-expressing B cells and of antibody production. Mice which are heterozygous at the JH locus carry one intact and functional heavy chain allele, derived from the C57BL/6J parent, and one JH-deleted heavy chain allele which is derived from the ES cells (129/Ola strain). The 129 and B6 strains differ in Ig heavy chain allotypes. The ES-derived B cells (IgMa allotype) can be distinguished from B6-derived B cells (IgMb allotype) with allotype-specific monoclonal antibodies, using flow cytometry analysis of antibody expressing B.
  • The specificity of these antibodies is shown in FIGS. [0106] 3(A-C). Peripheral blood lymphocytes were stained with antibodies to the B cell specific marker, B220, and with antibodies to the IgM allotype. B cells from C57BL/6J mice stained with antibodies directed against the IgMb allotype but not the IgMa allotype (FIG. 3B) B cells derived from 129/Ola mice stained with antibody against the IgMa allotype, but not the IgMb allotype (FIG. 3A). In heterozygous (a/b F1) mice carrying one intact ES-derived heavy chain allele and one intact C57BL/6J-derived heavy chain allele, both allotypes were present in equal amounts (FIG. 3C).
  • When B cells from mice which were heterozygous for the J[0107] H deletion were analyzed, where the JH deleted heavy chain allele was from the 129/Ola parent, there were no cells positive for the IgMa allotype. All B cells were IgMb positive, from the intact C57BL/6J heavy chain allele (FIG. 3D). These results indicated that the JH-deleted heavy chain locus is inactivated and cannot encode a functional IgM antibody.
  • Mice which were homozygous for the J[0108] H deletion were also analyzed for the ability to produce functional antibodies. Peripheral blood lymphocytes from homozygous mutant mice were analyzed by flow cytometry, using antibodies to the B cell specific marker B220, and with the allotype specific markers (see FIG. 4). In contrast to the control mice (FIGS. 4D-F), no B220+ cells, or IgM producing cells could be detected in the mutant mice (FIGS. 4A-C). In addition, the mutant mice had no detectable IgM in the serum. These results indicate that the deletion of the JH region from both heavy chain alleles leads to complete inhibition of B cell development, to mature B cells and production of antibody.
  • F. Generation of Homozygous Mutant ES Cells [0109]
  • The effect of J[0110] H deletion on B cells can also be analyzed by generating ES cells with both heavy chain alleles targeted, which are then used to produce chimeric mice which contain a population of lymphoid cells homozygous for the mutation.
  • Homozygous δJ[0111] H mutant ES cells were generated by subjecting one of the heterozygous mutant ES clones, ES110-1, to elevated levels of G418 (1.4 mg/ml) thus selecting for homogenotization of the targeted allele. Seven of the surviving colonies were screened by Southern blot analysis using SacI digestion for the loss of the wild-type heavy chain allele and acquisition of a second targeted allele. One of these clones, ESDK207 was shown to have lost the native heavy chain allele, as evidenced by the inability of probes to detect the wild type 4.0 kb fragment and by the increased intensity of the 3.4 kb targeted fragment. Karyotypic analysis of ESDK207 indicated that, like the parent line ES110-1, about 80% of the cells had 40 chromosomes, suggesting that two targeted alleles were present. The homozygous mutant ES cells were microinjected into C57BL/6J blastocysts and chimeric mice were generated.
  • G. Analysis of B Cells from Homozygous Chimeras [0112]
  • B cells from chimeric mice were analyzed to determine the effect of J[0113] H deletion on B cell development and antibody production. Lymphocytes from the ES cell line (129/Ola) can be distinguished from blastocyst-derived (C57BL/6J) lymphocytes by a monoclonal antibody to the Ly-9.1 marker, which is found on lymphocytes of 129 origin, but not those of B6 origin. In addition, the two strains differ in their IgM allotype, as previously described.
  • The chimeras analyzed had been derived from wild-type E14TG2a ES cells (WT), or from ES cells that wer heterozygous (ES110-1, ES65-1) or homozygous (ESDK207) at the targeted J[0114] H region. Peripheral blood mononuclear cells were stained with antibodies to the B cell specific marker B220, and with antibodies to either Ly-9.1 or IgM allotypes, and then analyzed by two-color flow cytometry. To evaluate chimerism in the T cell lineage, the cells were stained with antibody for the T cell marker Thy 1.2, and with anti-Ly-9.1 antibody. Staining of cells from the parental mouse strains provided controls for th specificity and sensitivity of the assay.
  • Mice with similar degrees of chimerism, as judged by coat color, were compared. ES-derived B and T cells were detected in the peripheral blood of chimeric mice generated from the wild-type E14TG2a ES cells, confirming the ability of this cell line to give rise to lymphoid cells in vivo. Analysis of chimeras generated from single J[0115] H-targeted ES65-1 and ES110-1 cells demonstrated the presence of B220+/IgMa+/Ly-9.1+ B cells containing a single, intact, ES cell-derived Ig heavy chain locus.
  • In contrast to the WT and single deletion chimeras, mice generated from the homozygous mutant ESDK207 cell line lacked Ly-9.1[0116] +/B220+ or IgMa+/B220+1 B cells in peripheral blood. The observed lack of ESDK207-derived B cells was not due to a lack in lymphopoiesis, since ES-derived Ly-9.1+/B220 cells represented 12% of the total pool of peripheral blood mononuclear cells. Of these, approximately half were Thy-1.2+ T cells. Thus, deletion of the JH region from both alleles blocks development of mature Igma producing B cells. Similar observations were made for chimeric spleen cells.
  • Chimeras were also tested for the presence of serum IgM derived from the ES cells. IgM[0117] a levels were high in chimeras from wild-type ES cells and cells with a single targeted mutation, but were undetectable in mice derived from the ESDK207 cell line.
  • Further analysis showed that the bone marrow of ESDK207 mice contained normal IgM[0118] b+ B cells derived from the blastocyst host, but lacked ES-derived IgMb+ B cells. However, DK207-derived bone marrow did contain a population of cells which were B220dull/Ly-9.1+ derived from the ES cells. The bone marrow is therefore likely to contain a subpopulation of ES cell-derived B cell precursors, whose maturation is blocked by the homozygous deletion of the JH region.
  • The bone marrow cells were also analyzed with three-color flow cytometry, using antibodies to Ly-9.1, B220 and either CD43 or Thy-1.2. The results show the majority of ES-derived cells were CD43 positive, which is consistent with an early block in maturation. Many of the cells were also positive for Thy-1.2, as would be expected of very early B cell precursors. These data show that deleting the J[0119] H region results in the inability of the heavy chain locus to rearrange and produce functional IgM. Lack of IgH rearrangement results in a block of B cell maturation, restricting B cell progenitors to an early stage of development.
  • Example III Deletion of the Mouse Ig kappa Light Chain Constant Cκ) Region
  • A. Construction of the Replacement Targeting Vector [0120]
  • The kappa region was inactivated with a replacement type vector, which was designed to delete the constant region of the kappa locus, and replace it with the G418 drug resistance marker through homologous recombination. Homologous recombination was driven by regions of homology which flank the constant region (see FIG. 5). [0121]
  • A genomic library from 129/Ola mouse fetal liver DNA (Stratagene) cloned into lambda phage was screened for the presence of the mouse C[0122] κ gene with a 1.6 kb HpaI/BamHI fragment (Steinmetz and Zachau (1980) Nucleic Acids Research 8:1693-1706) that spans the mouse kappa constant region. A lambda phage clone which hybridized to this probe was identified, then purified and used as a source of Cκ DNA. Analysis of the phage DNA showed that the kappa constant region probe hybridized to a 5.6 kb SphI/BamHI fragment. This fragment contained the kappa J region genes, an intronic enhancer element and the kappa constant region. It was then isolated and subcloned into the SphI and BamHI sites of the plasmid pUC218 to give the plasmid pUC218/5.6kappa.
  • In order to construct the deletion vector, fragments containing the 5′ region of the kappa constant region, a thymidine kinase gene for negative selection, a neomycin resistance gene and a 3′ region of homology to the kappa constant region were ligated together (see FIG. 6). [0123]
  • A 4.0 kb SphI/Bsu361 fragment from the plasmid pUC218/5.6kappa was subcloned into the SphI and Bsu361 sites of the vector pSK.A to give the plasmid pSK.A/5′K. The vector pSK.A is a modification of pBluescript SK- which has a synthetic polylinker: [0124]
    5′ CATATGCCTGAGGTAAGCATGCGGTACCGAATTCTATAAGCTTGCGG
    CCGCAGGTCATGCGTATACGGACTCCATTCGTACGCCATGGCTTAAGATA
    TTCGAACGAACGCCGGGCG
    3′
  • inserted between the pBluescript KpnI and SacI sites. [0125]
  • A 2.7 kb EcoRI/HindIII fragment containing the herpes thymidine kinase (TK) gene driven by the mouse phosphoglycerate kinase gene (PGK) promoter from the plasmid pKJtk (Tybulewicz, et al. (1991) Cell 65:1153-1163) was inserted into the EcoRI and NotI sites of pSK.A/5′K by using a HindIII/NotI adapter with the sequence: [0126]
  • 5′ [0127] AGCTGGAACCCCTTGCCCTTGGGGAACGCCGG 3′.
  • In the resulting plasmid, pSK.A/5′K/TK, the 5′ end of the TK gene and the kappa constant region gene are adjacent to each other, in opposite transcriptional orientations. [0128]
  • A 1.1 kb XhoI/BamHI fragment from pMC1Neo, which contains the mammalian drug selectable marker for resistance to neomycin, was cloned into the XhoI and BamHI sites of the plasmid pSK.B to give the plasmid pSK.B/Neo. The vector pSK.B is a modification of pBluescript SK- which has a synthetic polylinker: [0129]
    5′ GAGCTCGGATCCTATCTCGAGGAATTCTATAACCTTCATATGTAGCT
    CATGCTCGAGCCTAGGATAGAGCTCCTTAAGATATTCGAAGTATACA
    3′
  • inserted between the pBluescript KpnI and SacI sites. [0130]
  • A 1.1 kb BglII/BamHI fragment from pUC218/5.6kappa, which contains homology to the 3′ end of the kappa region, was cloned into BamHI digested, alkaline phosphatase treated pSK.C vector. The vector pSK.C is a modification of pBluescript SK- which has a synthetic polylinker: [0131]
    5′ AAGCTTATAGAATTCGGTACCTGGATCCTGAGCTCATAGCGGCCGCA
    GCTCATGTTCGAATATCTTAAGCCATGGACCTAGGACTCGAGTATCGCCG
    GCG
    3′
  • inserted between the pBluescript KpnI and SacI sites. The resulting plasmid, pSK.C/3′K is oriented such that transcription proceeds from the SacI site in the plasmid polylinker in the direction of the KpnI site. [0132]
  • The final targeting plasmid was constructed with a three part ligation, using (A) 6.1 kb NotI/NdeI fragment from pSK.A/5′K/TK, (B) 1.2 kb NdeI/SacI fragment from pSK.B/Neo and (C) 4.0 kb SacI/NotI fragment from pSK.C/3′K ligated to make the plasmid pK.TK/neo. [0133]
  • B. Electroporation of Kappa Deletion Vector into ES Cells [0134]
  • Purified plasmid DNA from pK.TK/Neo was cut with PvuI, extracted with phenol/chloroform and ethanol precipitated. The DNA was resuspended after precipitation at a concentration of 1 mg/ml in 10 mM Tris-HCl, 1 mM EDTA. [0135]
  • The embryonic stem cell line E14-1, a subclone of E14 (Hooper, et al. (1987) Nature 326:292-295) was cultured in DMEM 4.5 g/l glucose (J.R.H. Biosciences) supplemented with 15% heat inactivated fetal calf serum, recombinant murine leukemia inhibitory factor (ESGRO from Gibco BRL, 1000 U/ml), 0.1 mM β-mercaptoethanol, 2 mM glutamine and 100 U/ml penicillin at 37° C. in 5% CO[0136] 2.
  • The cells were cultured on mitomycin-treated primary embryonic fibroblast feeder layers essentially as described (Koller and Smithies (1989) supra). The embryonic fibroblasts were prepared from day 14 embryos carrying the homozygous targeted mutation of β2-microglobulin (Koller and Smithies (1990) Science 248:1227-1230). These feeder cells are capable of growth in media containing G418. [0137]
  • At 80% confluency, the ES cells were prepared for electroporation by trypsinization, concentration by brief centrifugation and resuspension in HEPES-buffered saline at 2×10[0138] 7 cells/ml. The cells are equilibrated at room temperature, and linearized targeting vector DNA (20 μg) added. The mixture was electroporated at 960 μF and 250 V with a BioRad Gene Pulser. The cells were left to stand at room temperature for 10 minutes before plating onto 4×10 cm dishes of mitomycin-treated fibroblast feeders (3×106 feeder cells/plate). After incubation at 37° C. for 48 hours, the cells were fed media containing 150 μg/ml G418 to select for neomycin resistance. After a further 48 hours the cells were fed media containing 150 μg/ml G418 and 2 μM gancyclovir (Syntex) to select for loss of the thymidine kinase gene.
  • C. Analysis of Targeted ES Cells [0139]
  • After ten days of drug selection with both G418 and gancyclovir, the individual surviving colonies were picked and dissociated with a drop of trypsin in a 96 well plate, then incubated at 37° for 2 minutes. The cells from each colony were transferred into a well of a 24-well plate containing mitomycin C-treated feeder cells and selective media with G418, but not gancyclovir. After an additional 5-8 days, 20% of the cells in each well were frozen, and the remainder used to prepare genomic DNA. The cells were lysed with 0.4 ml of 10 mM Tris-HCl (pH 7.5), 100 mM NaCl, 10 mM EDTA, 1% SDS and proteinase K (1 mg/ml) by overnight incubation at 50° C. The DNA was purified by phenol extraction and ethanol precipitation, then washed with 70% ethanol and resuspended in 20 μl of 10 mM Tris-HCl, 1 mM EDTA. [0140]
  • Southern analysis was carried out using BglII digested genomic DNA from each sample. An about 1.2 kb BamHI/BglII fragment which contains the region contiguous with the 3′ homology fragment in the targeting vector was used as a probe. The native ES cell locus gave an about 2.3 kb fragment, while the targeted ES cell locus gave an about 5.7 kb fragment. The increase in size is due to the loss of a BglII site during the construction of the deletion vector. [0141]
  • A Southern analysis of 166 clones showed two cell lines which had the intended mutation. These clones were further analyzed by reprobing the filters with an about 1.1 kb fragment which spans the neo gene. As expected, the probe only hybridized to the targeted allele. [0142]
  • Further analysis of the genomic DNA from the two positive clones, 1L2-850 and 1L2-972, after being thawed and expanded, reconfirmed the initial observations. A third probe, an about 1.7 kb HindIII/BglII fragment spanning the kappa J region locus, was used to check for the correct integration pattern from the 5′ end of the targeting vector. Using this probe with EcoRI digested genomic DNA, an about 15 kb fragment is detected in the native allele, and an about 5 kb fragment from the targeted locus. The additional EcoRI site is introduced by the neo gene during homologous recombination targeting (see FIG. 7). [0143]
  • D. Generation of Germline Chimeras [0144]
  • The unmodified E14-1 cells have been found to contribute to the germline at a high frequency after injection into C57BL/6J blastocysts. To generate germline chimeras containing the targeted kappa region, the targeted cell lines 1L2-850 and 1L2-972 were grown on primary feeder cells, then trypsinized and resuspended in injection medium, which consists of DMEM supplemented with 15% fetal calf serum, 20 mM HEPES (pH 7.3), antibiotics and β-mercaptoethanol. The ES cells were injected into each blastocyst, and the injected blastocysts then transferred to one uterine horn of a pseudopregnant female mouse. Chimeric pups were identified by chimeric coat color. Chimeric males were bred to C57BL/6J females, and germline transmission of the 129/Ola derived ES cells was detected by agouti coat color of the offspring. [0145]
  • One chimeric male from cell line 1L2-972 (about 40% ES cell derived as judged by its coat color), upon mating with C57B1/6J females yielded germline transmission at a frequency of 25% as determined by the percent of agouti offspring. Chimeric males, about 40%, 70% and 90% chimeric, from cell line 1L2-850 yielded germline transmission at a frequencies of 90%, 63% and 33%, respectively. Among the agouti offspring generated from the 70% chimeric male from 1L2-850, eight F1 animals out of 12 tested were found to be heterozygous at the kappa locus for the targeted C[0146] K mutation by Southern analysis (a Bgl II digest using the 1.2 kb Bam HI/Bgl II fragment described above as a probe) using genomic DNA derived from tail samples. Further breeding of a male and female from this group of 8 F1 animals, both heterozygous for the CK mutation, yielded one male offspring found to be homozygous for this mutation as confirmed by Southern analysis.
  • E. Analysis of B Cells Obtained from Mice Targeted at the Kappa Locus [0147]
  • If the kappa (κ) light chain locus is inactivated because of deletion of the light chain constant region (Cκ), the joining region(Jκ), or both Cκ and Jκ, then a complete block in the development of κ-expressing B cells should result. Mouse embryonic stem cells containing a single copy of the complete Cκ deletion (ΔCκ) were introduced into mouse blastocysts as described above to produce chimeric mice. These chimeric mice were then bred with wild-type C57BL/6 (B6) mice, and the F1 progeny were assayed for the presence of the ΔCκ mutation by Southern blotting of tail DNA. F1 mice that carried the ΔCκ mutation were bred and F2 offspring were assayed similarly for ΔCκ. One of 5 F2 offspring was shown to carry a homozygous Cκ deletion, and another was heterozygous, bearing both ΔCκ and a wild-type Cκ allele. The 3 other offspring were wild-type. The presence or absence of κ-positive B cells was assayed by flow cytometric analysis of peripheral blood B cells stained with fluorescent antibodies that react with a pan-B cell marker (B220) or with the κ light chain. For the homozygous ΔCκ F2 mouse no κ-positive B cells were detected, and in the heterozygote, there was a reduction in the frequency of κ positive B cells, consistent with the presence of a wild-type allele and a non-functional ΔCκ allele. These results demonstrate that deletion of Cκ from the chromosome prevents κ expression by mouse B cells. [0148]
  • Example IV Inactivation of the Mouse Immunoglobulin Kappa Light Chain J and Constant Region
  • A. Design of the Targeting Experiment [0149]
  • The targeting vector was designed as a replacement type vector initially to delete the constant region as well as the J region of the kappa locus and replace it with three elements through homologous recombination using regions of homology flanking the constant region (FIG. 8). A diphtheria toxin gene (A chain) flanking either or both regions of homology was included in some cases as a negative selectable marker. The three elements consisted of the G418 resistance drug marker, an additional DNA homology (ADH) sequence of mouse DNA homologous to a region of the kappa locus located upstream of the J region, and a thymidine kinase gene. As a result of the inclusion of the ADH sequence in the vector, this initial targeting placed a second copy of the ADH in the locus. This duplication was then used to effect a defined deletion of the sequences between the segments by applying selective pressure. In this case the cell deletes the thymidine kinase gene that lies between the two segments in order to survive gancyclovir selection. [0150]
  • B. Construction of the Targeting Vector [0151]
  • The regions of homology were derived from a 129 mouse fetal liver genomic library (Stratagene) which was screened using two probes, as described above in Example III. This subclone contained the J region, an intronic enhancer element and the constant region of the kappa light chain locus. The second probe was a 0.8 kb EcoRI fragment (Van Ness et al. (1981), [0152] Cell 27:593-602) that lies 2.8 kb upstream of the J region. Phage DNA from a lambda clone positive for this probe showed that the probe hybridized to a 5.5 kb SacI fragment which was subcloned into the SacI site of pBluescript SK (Stratagene) to give the plasmid pSK.5′kappa (FIG. 8).
  • The inactivation vectors which contained a 5′ region of homology, a thymidine kinase gene, a ADH, a neomycin resistance gene and a 3′ region of homology (FIG. 9) flanked in some instances by diphtheria toxin genes were constructed from three plasmids (FIG. 8) containing: (a) the 5′ fragment of homology with or without the diphtheria toxin gene (DT) driven by the mouse phosphoglycerate kinase gene (PGK) promoter as a negative selectable marker, (b) the herpes thymidine kinase gene (tk) driven by the mouse phosphoglycerate kinase gene (PGK) promoter as a negative selectable marker along with the DSH and the G418 selectable neomycin (neo) gene from pMC1Neo (Thomas and Capecchi (1987), [0153] Cell 51:503-12), and (c) the 3′ fragment of homology with or without the PGK driven DT gene. These three plasmids (FIG. 8) were constructed from pSK.A, pSK.B, and pSK.C, respectively, all derived from the plasmid pBluescript SK by modification of the polylinker.
  • The polylinker of the plasmid pBluescript SK[0154] was modified by cloning between the KpnI and SacI sites a synthetic polylinker defined by the oligonucleotides 5′-GCATATGCCTGAGGGTAAGCATGCGGTACCGAATTCTATAAGCTTGCGGCCGCAGCT-3′ AND 5′-GCGGCCGCAAGCTTATAGAATTCGGTACCGCATGCTTACCTCAGGCATATGCGTAC-3′ to create the plasmid pSK.A, 5′-GAGCTCGGATCCTATCTCGAGGAATTCTATAAGCTTCATATGTAGCT-3′ and 5′-ACATATGAAGCTTATAGAATTCCTCGAGATAGGATCCHAGCTCGTAC-3′ to create plasmid pSK.8, 5′-AAGCTTATAGAATTCGGTACCTGGATCCTGAGCTCATAGCGGCCGCAGCT-3′ to create plasmid psK.B and 5′-GCGGCCGCTATGAGCTCAGGATCCAGGTACCGAATTCTATAAGCTTGTAC-3′ to create the plasmid pSK.C.
  • A diphtheria toxin gene cassette was created in which the gene was flanked by the PGK promoter and the bovine growth hormone polyadenylation signal (Woychik et al. (1984), [0155] Proc. Natl. Acad. Sci. U.S.A, 81:3944-3948; Pfarr et al. (1986), DNA 5:115-122). A 2.3 kb XbaI/EcoRI fragment from pTH-1 (Maxwell et al. (1986), Cancer Res. 46:4660-4664) containing the diphtheria toxin A chain driven by the human metallothionein (hMTII) promoter was cloned into pBluescript SK cut with XbaI and EcoRI to give the plasmid pSK.DT. The hMTII promoter of pSK.DT was replaced with the PGK promoter from pKJ1 (Tybulewicz et al. (1991), Cell 65:1153-1163). A 0.5 kb XbaI/PstI fragment from pKJ1 was joined to a 3.1 kb XbaI/NcoI fragment from pSK.DT using a PstI/NcoI adapter formed from the oligonucleotides 5′-GGGAAGCCGCCGC-3′ and 5′-CATGGCGGCGGCTTCCCTGCA-3′ to give the plasmid pSK.pgkDT. A 248 bp fragment containing the bovine growth hormone polyadenylation signal, obtained by PCR amplification of bovine genomic DNA using the oligonucleotide primers 5′-CAGGATCCAGCTGTGCCTTCTAGTTG-3′ and 5′-CTGAGCTCTAGACCCATAGAGCCCACCGCA-3′, was cloned into pCR1000 (Invitron Corp., San Diego, Calif.). The polyadenylation sequence was then cloned behind the DT gene as a HindIII/PvuII fragment into pSK.pgkDT cut with HindIII and HpaI to give the plasmid pSK.pgkDTbovGH. The DT gene cassette from pSK.pgkDTbovGH was moved as a 2.1 kb EcoRI/HindIII fragment into pSK.A cut with EcoRI and NotI using a HindIII/NotI adapter formed from the oligonucleotides 5′-AGCTGGAACCCCTTGC-3′ and 5′-GGCCGCAAGGGGTTCC-3′ to give the plasmid pSK.A/DT. Between the SphI and Bsu36I sites of both pSK.A and pSK.A/DT the 5′ region of homology for the kappa locus was cloned. For this purpose a 4.0 kb SphI/Bsu361 fragment resulting from a partial Bsu36I digest followed by a complete SphI digest of plasmid subclone pUC218/5.6kappa was ligated to pSK.A or pSK.A/DT to give the plasmids pSK.A/5′K and pSK.A/DT/5′K, respectively. In the plasmid, pSK.A/DT/5′K, the 5′-end of the DT gene and kappa fragment were adjacent to each other running in the opposite transcriptional orientations.
  • The PGKtk gene from the plasmid pKJtk (Tybulewicz et al. (1991), [0156] Cell 65:1153-1163) was cloned as a 2.7 kb EcoRI/HindIII between the unique EcoRI and HindIII sites of pSK.B to give pSK.B/TK. A 0.8 kb EcoRI fragment used for the ADH was cloned from pSK.5′ kappa and was ligated into the EcoRI site of pSK.B/TK to give pSK.B/(TK/0.8K) such that the 5′-end of the tk gene and kappa fragment were adjacent to each other running in opposite transcriptional orientations. The 1.1 kb neo gene from pMC1Neo was cloned as an XhoI/BamHI fragment between the same sites of pSK.B/(TK/0.8K) to give pSK.B/(TK/0.8K/Neo). The plasmid pSK.C/3′K containing the 3′ fragment of homology was constructed by ligating pSK.C digested with BamHI and treated with alkaline phosphatase to the 1.1 kb Bg1II/BamHI fragment isolated from pUC218/5.6kappa. In pSK.C/3′K, the kappa fragment was oriented such that transcription proceeded from the SacI in the plasmid polylinker in the direction of the KpnI site. The 2.1 kb DT cassette from pSK.pgkDTbovGH was cloned as an EcoRI/HindIII fragment into the same sites of pSK.C to give pSK.C/3′K/DT.
  • Three-part ligations were carried out to construct the final targeting plasmids (FIG. 9). The 4.0 kb NotI/NdeI fragment from pSK.A/5′K, the 4.8 kb NdeI/SacI fragment from pSK.B/(TK/0.8K/Neo) (obtained by a SacI partial followed by and NdeI digestion of the plasmid), and the 4.0 kb SacI/NotI fragment from pSK.C/3′K were isolated and ligated together to create pK. (TK/0.8K/Neo). The 6.1 kb NotI/NdeI fragment from pSk.A/DT/5′K, the 4.8 kb NdeI/SacI fragment from pSK.B/(TK/0.8K/Neo), and 4.0 kb SacI/NotI fragment from pSK.C/3′K were isolated and ligated together to create pK.DT/(TK/0.8K/Neo). The 6.1 kb NotI/NdeI fragment from pSK.A/DT/5′K, the 4.8 kb NdeI/SacI fragment from pSK.B/(TK/0.8K/Neo), and 6.1 kb SacI/NotI fragment from pSK.C/3′K/DT (obtained by a SacI partial followed by a NotI digestion of the plasmid) were isolated and ligate together to create pK.DT/(TK/0.8K/Neo)/DT. For electroporation, the purified plasmid DNAs were first cut with PvuI or ApaLI, then extracted with phenol/chloroform and precipitated by the addition of ethanol before centrifugation. The resultant DNA pellets were resuspended at a concentration of 1 mg/ml in 10 mM Tris-HCl, 1 mM EDTA (TE). [0157]
  • C. Introduction of DNA into Cells [0158]
  • The embryonic stem cell line E14-1 was cultured as described above in Example III. The cells were equilibrated at room temperature, and DNA (20 μg) linearized with PvuI (as described above) was added. The mixture was electroporated as described above in Example III. [0159]
  • D. Analysis of Constant Region-Targeted ES Cells [0160]
  • After 7-10 days under drug selection with G418, the individual surviving colonies were each picked and dissociated in a drop of trypsin as described above in Example III. [0161]
  • Southern analysis was carried out using BgIII digested genomic DNA from each sample. A 2.3 kb fragment was detected from the native ES cell locus, while a larger 4.9 kb fragment was detected from a targeted ES cell locus (FIG. 11), using as a probe the 1.2 kb BamHI/BgIII fragment isolated from the original phage DNA contiguous with the fragment used for the 3′ homology in the targeting vector. The fragment increased in size because the BgIII site in the BgIII/BamHI fragment was lost in the targeting plasmid due to the joining of a BgIII site to a BamHI site in the ligation, and a new BgIII site located in the thymidine kinase gene is introduced into the targeted locus. [0162]
  • From a screen by the Southern analysis described above, of a total of 103 clones derived from experiments using three different targeting plasmids, 5 cell lines were identified which carried the intended mutation (Table 1). [0163]
    TABLE 1
    CK Light Chain Targeting Result in E14-1
    Number of
    Number Screened Confirmed Clone Frequency of
    Construct by Southern Targeted Clones Designation Targeting
    pK.(TK/0.8 K/Neo) 44 2 625.691 1/22
    pK.DT(TK/0.8/Neo) 42 2 604.611 1/21
    pK.DT(TK/0.8 K/Neo)DT 17 1 653 1/17
  • Further analysis of genomic DNA produced from 4 of the positive clones (clones 625, 604, 611 and 653) after being thawed and expanded, re-confirmed the initial observations. Using a second probe, a 1.7 kb HindIII/BgIII fragment which spanned the J region of the kappa locus, the correct integration pattern was checked for homologous targeting at the 5′ end of the targeting vector. Thus, using this probe with an EcoRI digest of the genomic DNA, a 15 kb fragment was detected from the unmodified allele. In contrast, a 7.8 kb fragment from the targeted allele was observed as a result of the introduction of a new EcoRI site in the thymidine kinase gene during the homologous integration (FIG. 11). [0164]
  • E. In vitro Excision of J Region DNA from Targeted Clones [0165]
  • In order to effect the desired deletion from the homologously targeted kappa locus, cells from clone 653 were plated on feeder cells at a density of 0.5-1×10[0166] 6 cells/10 cm dish in the presence of both gancyclovir (2 μM) and G418 (150 μg/ml). After growth for 5 days in the presence of both drugs, clones were picked as described above into 24-well plates and grown under G418 selection alone. After an additional 5-8 days, 20% of the cells in each well were frozen and the remainder used to prepare genomic DNA as previously described.
  • F. Analysis of J/Constant Region Deleted ES Cells [0167]
  • Southern analysis was carried out using BamHI digested genomic DNA from each sample. Using as a probe the 0.8 kb EcoRI fragment used as the ADH in the targeting vectors, as 12.7 kb fragment was detected from the native ES cell locus, while a larger 15.8 kb fragment was detected from the constant region-targeted ES cell locus (FIG. 11) using DNA from clone 653. The fragment increased in size because of the insertion of the tk gene, the ADH, and the neo gene into the 12.7 kb BamHI fragment. There was also a new BamHI site introduced at the 3′ end of the neo gene. Using DNA from the J/constant region deleted cells, a 5.5 kb fragment was detected from the modified locus in addition to the 12.7 kb fragment from the untargeted allele as predicted from analysis of the restriction map. From this screen by Southern analysis of 2 clones produced from 1.5×10[0168] 6 ES cells plated (clone 653), one cell line (clone 653B) was identified which carried the intended deletion of the J and constant regions.
  • Further analysis of genomic DNA produced from clone 653B after being thawed and expanded re-confirmed the initial observations. Using the 0.8 kb EcoRI fragment, the deletion was checked with two other restriction digests which should cut outside of the excised region on the 5′ and 3′ ends of the targeting vector. Thus using this probe with a BgIII digest of the genomic DNA from the unexcised clone 653, a 2.6 kb fragment was detected from both the unmodified and modified alleles, whereas an additional 4.9 kb fragment was observed from the targeted allele only (FIG. 11). This 4.9 kb fragment was the same as that detected with the 1.2 kb BamHI/BgIII fragment used previously. Using DNA from clone 653B, a BgIII digest revealed a 5.8 kb fragment in addition to the 2.6 kb fragment from the unmodified allele. A SacI digest of clone 653 DNA probed with the 0.8 kb EcoRI fragment showed a 5.5 kb fragment from both the unmodified and modified alleles and a 3.1 kb fragment from the targeted allele only (FIG. 11). The 5.5 kb fragment was also detected in DNA from clone 653B and an additional 2.0 kb fragment. The 5.8 kb BgIII fragment and the 2.0 kb ScaI fragment were consistent with an analysis of the predicted restriction map for a precise excision step in which 10.3 kb of DNA were deleted including the J region, the tk gene, and one copy of the ADH. [0169]
  • G. Generation of Germline Chimeras [0170]
  • The unmodified E14-1 cells contributed to the germline at a high frequency after injection into C57BL/6J blastocysts. The cells from the targeted ES cell line 691, in which only the kappa constant region has been deleted by homologous recombination without any negative selection, were microinjected and chimeric animals were produced as described above in Example III. Cells from the targeted ES cell line 653B in which both the kappa constant and J regions were deleted are also microinjected and chimeric animals are produced as described above. Chimeric pups are identified by chimeric coat color. Germline transmission of the modified ES cell is detected by the agouti coat color of the F1 offspring. [0171]
  • Example V Cloning of Human Heavy Chain Locus Using Yeast Artificial Chromosomes
  • A. Production of Yeast Artificial Chromosome (YAC) Containing Human Heavy Chain [0172]
  • An SpeI fragment, spanning the human heavy chain VH6D-J-Cμ-Cδ region (Berman et al. (1988), [0173] EMBO J. 7: 727-738; see FIG. 15) is isolated from a human YAC library (Burke, et al., Science, 236: 806-812) using DNA probes described by Berman et al. (1988) EMBO J. 7:727-738. One clone is obtained which is estimated to be about 100 kb. The isolated YAC clone is characterized by pulsed-field gel electrophoresis (Burke et al., supra; Brownstein et al., Science, 244: 1348-1351), using radiolabelled probes for the human heavy chain (Berman et al., supra).
  • B. Introduction of YAC Clones into Embryos or ES Cells [0174]
  • High molecular weight DNA is prepared in agarose plugs from yeast cells containing the YAC of interest (i.e., a YAC containing the aforementioned SpeI fragment from the IgH locus). The DNA is size-fractionated on a CHEF gel apparatus and the YAC band is cut out of the low melting point agarose gel. The gel fragment is equilibrated with polyamines and then melted and treated with agarase to digest the agarose. The polyamine-coated DNA is then injected into the male pronucleus of fertilized mouse embryos which are then surgically introduced into the uterus of a psueudopregnant female as described above. The transgenic nature of the newborns is analyzed by a slot-blot of DNA isolated from tails and the production of human heavy chain is analyzed by obtaining a small amount of serum and testing it for the presence of Ig chains with rabbit anti-human antibodies. [0175]
  • As an alternative to microinjection, YAC DNA is transferred into murine ES cells by ES cell: yeast protoplast fusion (Traver et al., (1989) [0176] Proc. Natl. Acad. Sci., USA, 86:5898-5902; Pachnis et al., (1990), ibid 87: 5109-5113). First, the neomycin-resistance gene from pMC1Neo or HPRT or other mammalian selectable marker and a yeast selectable marker are inserted into nonessential YAC vector sequences in a plasmid. This construct is used to transform a yeast strain containing the IgH YAC, and pMC1Neo (or other selectable marker) is integrated into vector sequences of the IgH YAC by homologous recombination. The modified YAC is then transferred into an ES cell by protoplast fusion (Traver et al. (1989); Pachnis et al., 1990), and resulting G418-resistant ES cells (or exhibiting another selectable phenotype) which contain the intact human IgH sequences are used to generate chimeric mice. Alternatively, a purified. YAC is transfected, for example by lipofection or calcium phosphate-mediated DNA transfer, into ES cells.
  • Example VI Introduction of Human Ig Genes into Mice
  • A. Cloning of Human Ig Genes in Yeast [0177]
  • 1. Identification and Characterization of a Human IgH YAC Clone containing VH, D, JH, Mu and Delta Sequences: [0178]
  • PCR primers for the human VH6 gene (V6A=5′ GCA GAG CCT GCT GAA [0179] TTC TGG CTG 3′ and V6B=5′ GTA ATA CAC AGC CGT GTC CTG G 3′) were used to screen DNA pools from the Washington University human YAC library (Washington University, St. Louis, Mo.). Positive pools were subsequently screened by colony hybridization and one positive microtiter plate well, A287-C10, was identified. Two different sized (205 kb and 215 kb) VH6-containing YACS were isolated from the microtiter well. In addition to VH6, the smaller of the two IgH YACs, A287-C10 (205 kb), hybridized to probes for the following sequences: delta, mu, JH, D, VH1, VH2, and VH4. The larger of the two IgH YACs, A287-C10 (215 kb), hybridized to the following probes: delta, JH, D, VH1, VH2, and VH4, but not to mu. The YACs contained sequences from at least 5 VH genes including two VH1 genes, one VH2, one VH4 and one VH6 gene. Analysis of restriction digests indicated that the 205 kb YAC contains a deletion (about 20 kb size) that removes some, but not all of the D gene cluster, with the remainder of the YAC appearing to be intact and in germline configuration. PCR and detailed restriction digest analysis of the 205 kb YAC demonstrated the presence of several different D gene family members. The 215 kb YAC appeared to contain the complete major D gene cluster but had a deletion (about 10 kb) that removed the mu gene. This deletion does not appear to affect the JH cluster or the enhancer located between JH and mu genes.
  • The putative progenitor of the above two related IgH YACs, a YAC of about 225-230 kb containing the entire genomic region between the VH2 gene and the delta gene (Shin et al., 1991, supra) (see FIG. 15), had not been identified in the A287-C10 microtiter well. Hence, an earlier aliquot of the A287-C10 microtiter plat well was examined in order to search for the progenitor YAC under the assumption that it was lost during passaging of the library. The A287-C10 microtiter well was streaked out (Washington University, St. Louis, Mo.), and 2 of 10 clones analyzed contained a 230 kb IgH YAC with another apparently unrelated YAC. [0180] Clone 1 contained in addition the IgH YAC, an approximately 220 kb YAC and clone 3 in addition contained an approximately 400 kb YAC. The IgH YAC contained mu, the complete D profile (based on a BamHI digest, see below) and JH. The IgH YAC from clone 1 was physically separated from the unrelated YAC by meiotic segregation in a cross between A287-C10/AB1380 and YPH857 (genotype=MATα ade2 lys2 ura3 tpr1 HIS5 CAN1 his3 leu2 cyh2, to yield A287-C10 (230 kb)/MP 313 (host genotype=MATα ade2 leu2 lys2 his3 ura3 trp1 can1 cyh2).
  • 2. Targeting of the A287-C10 kb YAC with a Mammalian Selectable Marker, HPRT: [0181]
  • A YAC right arm targeting vector called pLUTO (15.6 kb) was generated by subcloning a human HPRT minigene contained on a 6.1 kb BamHI fragment (Reid et al., [0182] Proc. Natl. Acad. Sci. USA 87:4299-4303 (1990)) into the BamHI site in the polylinker of pLUS (Hermanson et al., Nucleic Acids Research 19:4943-4938 (1991)). A culture of A287-C10/AB1380 containing both the 230 kb IgH YAC and an unrelated YAC was transformed with linearized pLUTO and Lys+ transformants were selected. The Lys+ clones were screened by colony hybridization for the presence of mu. One clone was identified which contained a single YAC of approximately 245 kb which hybridized to probes for mu, HPRT and LYS2.
  • Southern analysis of the 230 kb A287-C10 YAC targeted with pLUTO was carried out using a variety of probes to demonstrate the intact, unrearranged nature of the cloned, human IgH sequences. In most cases, the results of BamHI, HindIII and EcoRI digests were compared to restriction data for WI38 (a human embryonic fetal lung-derived cell line), the 205 kb and 215 kb deletion-derivatives of A287-C10 and to published values. The diversity (D) gene profile determined by hybridization with a D region probe (0.45 NcoI/PstI fragment; Berman et al., 1988) demonstrated the expected four D gene segments (D1-D4 (Siebenlist et al., 1981[0183] ; Nature 294:631-635). For example, with BamHI, four restriction fragments, 3.8 kb, 4.5 kb, 6.9 kb and 7.8 kb, were observed in A287-C10 and WI38. WI38 had one additional larger band, presumed to originate from the chromosome 16 D5 region (Matsuda et al., 1988, EMBO 7:1047-1051). PCR and Southern analysis with D family-specific primers and probes demonstrated in the 215 kb deletion-derivative YAC (which appeared to have an intact D region with the same restriction pattern as the 230 kb YAC) the presence of 2 to 4 members of each of the following D gene families: DM, DN, DK, DA, DXP and DLR. The J-mu intronic enhancer, which was sequenced from cloned PCR products from the A287-C10 230 kb YAC (primers EnA=5′ TTC CGG CCC CGA TGC GGG ACT GC 3′ and EnB1=5′ CCT CTC CCT AAG ACT 3′) and determined to be intact, also generated single restriction fragments of approximately the predicted sizes with BamHI, EcoRI and HindIII when probed with the 480 bp PCR product. The JH region was evaluated with an approximately 6 kb BamHI/HindIII fragment probe spanning DHQ52 and the entire JH region (Ravetch et al., 1981, Cell 27:583-591). A287-C10 generated restriction fragments of approximately the expected sizes. Furthermore, the same-sized restriction fragments were detected with the enhancer and the JH probes (Ravetch et al., supra; Shin et al., 1991, supra). The approximately 18 kb BamHI JH fragment detected in A287-C10 and WI38 also hybridized to a 0.9 kb mu probe sequence (Ravetch et al., supra). Hybridization with the 0.9 kb EcoRI fragment mu probe (Ravetch et al., supra) showed restriction fragments of approximately the expected sizes (Ravetch et al., supra; Shin et al., supra):>12 kb BamHI (approximately 17 kb expected); 0.9 kb EcoRI (0.9 kb expected) and approximately 12 kb HindIII (approximately 11 kb expected). WI38 gave the same-sized BamHI fragment as A287-C10. The JH and DHQ52 regions were sequenced from both of the deletion derivative YACs and both were in germline configuration. Delta was analyzed with an exon 1 PCR product (containing the approximately 160 bp region between primers D1B=5′ CAA AGG ATA ACA GCC CTG 3′ and D1D 5′ AGC TGG CTG CTT GTC ATG 3′); restriction fragments for A287-C10 were close to those expected from the literature (Shin et al., supra) and to those determined for WI38. The 3′ cloning site of the YAC may be the first EcoRI site 3′ of delta (Shin et al., supra) or another EcoRI site further 3′. VH gene probes for VH1, VH4 and VH6 (Berman et al., supra), and for VH2 (Takahashi et al., 1984, Proc. Nat. Acad. Sci. USA 81:5194-5198) were used to evaluate the variable gene content of the YAC. A287-C10 contains two VH1 genes that approximate the predicted sizes (Shin et al., supra; Matsuda et al., 1993, supra); restriction analysis with the three enzymes gave close to the expected fragment sies; e.g. with EcoRI observed bands are 3.4 and 7.8 kb (expected are 3.4 and 7.2 kb). The predicted size EcoRI fragments for VH4 (5.3 kb observed, 5.1 kb expected) and for VH6 (0.8 kb observed, 0.9 kb expected) (Shin et al., supra; Matsuda et al., supra) were present in A287-C10. The expected size EcoRI fragment was seen for VH2 (5.5 kb observed, 5.4 kb expected), but the BamHI and HindIII fragments were different from those predicted. Coincident hybridization of the BamHI and HindIII fragments with a pBR322 probe suggested that the EcoRI site which is at the 5′ end of the VH2 gene (Shin et al., supra) is the 5′ cloning site, thus eliminating the natural 5′ HindIII site and BamHI sites. The overall size of the YAC insert (estimated to be approximately 220 kb) fits well with the predicted size for an intact, unrearranged segment starting at the 5′ end of the 3′-most VH2 gene and extending to an EcoRI site 3′ of the delta locus (Shin et al., supra).
  • 3. Identification and Characterization of IgK YACs Containing CK and VK Sequences: [0184]
  • Two YACs were identified in a screen of pulsed-field gel (PFG) pools from the Washington University (St. Louis, Mo.) human YAC library with a probe from the human kappa constant region (CK) gene (2.5 kb EcoRI fragment ATCC No. 59173, Parklawn Dr., Rockville, Md.). The YACs, designated A80-C7 (170 kb) and A276-F2 (320 kb), contain the kappa deleting element kde, CK, JK and the C-J intronic enhancer and extend 3′ beyond kde. Extending 5′ from JK, the YACs also contain the B1, B2 and B3 VK genes determined by hybridization and/or PCR, and possibly other VK sequences, The A80-C7/AB1380 strain housed, in addition to the IgK YAC, an unrelated YAC of similar size. Therefore, meiotic segregation was used to separate these YACS; A80-C7 was crossed to YPH857 and a meiotic product was obtained which contained only the IgK YAC (MP8-2; host genotype=α ade2 leu2 his3 his5 lys2 ura3 trp1 can1 cyh2). The A80-C7 and A276-F2 YACs have been targeted with pLUTO to incorporate the human HPRT minigene into the YAC right vector arm. [0185]
  • Restriction analysis of the IgK YACs A80-C7 and A276-F2 using a number of enzymes supports the conclusion that both YACs are unrearranged (i.e., in germline configuration). For example, BamHI digestion followed by hybridization with the CK probe demonstrates the expected 13 kb restriction fragment (Klobeck et al., [0186] Biol. Chem. Hoppe-Seyler 370:1007-1012 (1989)). The same-sized band hybridizes to a JK probe (a 1.2 kb PCR product using primer set to amplify the JK1-5 region), as predicted from the genomic map (Klobeck et al., supra). The. B3 class IV gene (probe is a 123 bp PCR product from the B3 gene) gives a 4.9 kb BamHI and a 2.2 kb BglII fragment, close to the published values of 4.6 kb and 2.3 kb, respectively (Lorenz et al., Molec. Immunol. 25:479-484 (1988)). PCR analysis of both IgK YACs as well as human genomic DNA for the following kappa locus sequences revealed the predicted band sizes: Kde (120 bp), CK (304 bp), C-J intronic enhancer (455 bp), JK1-5 (1204 bp), B3 VK (123 bp) and B1 VK pseudogene (214 bp). Sequences used to design PCR primers for the CK, JK and C-J enhancer regions are from Whitehurst et al., Nucl. Acids. Res. 20:4929-4930 (1992); Kde is from Klobeck and Zachau, Nucl. Acids. Res. 14:4591-4603 (1986); B3 is from Klobeck et al., Nucl. Acids. Res. 13:6515-6529 (1985); and B1 is from Lorenz et al., supra.
  • B. Introduction of 680 kb yHPRT YAC into ES Cells [0187]
  • 1. Culture of yHPRT Yeast Strain and Preparation of Yeast Spheroplasts [0188]
  • The 680 kb yHPRT is a YAC containing a functional copy of the human hypoxanthine phosphoribosyltransferase (HPRT) gene cloned from a YAC library, as described in Huxley, et al. (1991) [0189] Genomics 9:742-750. The yeast strain containing the yHPRT was grown in uracil and tryptophan deficient liquid media, as described in Huxley, et al. (1991) supra.
  • To prepare the yeast spheroplasts, a 400 ml culture of yeast containing yHPRT was spun down and the yeast pellet was washed once with water and once with 1 M sorbitol. The yeast pellet was resuspended in SPEM (1 M sorbitol, 10 mM sodium phosphate pH 7.5, 10 mM EDTA pH 8.0, 30 mM β-mercaptoethanol) at a concentration of 5×10[0190] 8 yeast cells/ml. Zymolase 20T was added at a concentration of 150 μg/ml of yeast cells, and the culture was incubated at 30° C. until 90% of the cells were spheroplasts (usually for 15-20 minutes). The cells were washed twice in STC (1 M sorbitol, 10 mM Tris pH 7.5, 10 mM CaCl2) and resuspended in STC at a concentration of 2.5×108/ml.
  • 2. Culture of E14TG2a ES Cells [0191]
  • HPRT-negative ES cell line E14TG2a was cultured as previously described. [0192]
  • 3. Fusion of ES Cells and Yeast Spheroplasts [0193]
  • Exponentially growing E14TG2a ES cells growing on gelatin-coated dishes were trypsinized and washed three times with serum-free DMEM. A pellet of 2.5×10[0194] 8 yeast spheroplasts was carefully overlaid with 5×106 ES cells which were spun down onto the yeast pellet. The combined pellet was resuspended in 0.5 ml of either 50% polyethylene glycol (PEG) 1500 or 50% PEG 4000 (Boeringer Mannheim) containing 10 mM CaCl2. After 1.5 minutes incubation at room temperature or at 37° C., 5 ml of serum-free DMEM were added slowly, and the cells were left at room temperature for 30 minutes. The cells were then pelleted and resuspended in 10 ml of ES cell complete medium (as previously described) and were plated onto one 100 mm plate coated with feeder cells. After 24 hours the medium was replaced with fresh medium. Forty-eight hours post-fusion, HAT (ES media containing 1×10−4 M hypoxanthine, 4×10−7 M aminopterin, 1.6×10−5 thymidine) selection was imposed. HAT-resistant ES colonies were observed 7-10 days post-fusion in the plates from both the different fusion conditions used. yHPRT-ES (“ESY”) fusion colonies were picked and plated onto feeder-coated wells, and expanded for further analysis.
  • 4. Analysis of YAC DNA Integrated into yHPRT-ES Fusion Clones [0195]
  • DNA extracted form 23 yHPRT-ES fusion colonies was digested with HindIII and subjected to Southern blot analysis (FIG. 12) using the probes: a human repetitive Alu sequence (A); pBR322-specific sequences for the right (B) and left (C) YAC vector arms; yeast Ty repetitive sequence (D); yeast single copy gene LYS2 (E). The human HPRT probe, a 1.6 kb full length cDNA (Jolly et al., [0196] Proc. Natl. Acad. Sci. USA 80:477-481 (1983)) was used to confirm the presence of the human HPRT gene in ESY clones. The Alu probe was a 300 bp BamHI fragment from the BLURB Alu element in pBP63A (Pavan et al., Proc. Natl. Acad. Sci. USA 78:1300-1304 (1990)). The right and left vector arm probes were pBR322-derived BamHI-PvuII 1.7 and 2.7 kb fragments, respectively, which correspond to the vector sequences in pYAC4 (scheme a, b (Burke et al., in: Guide to Yeast Genetics and Molecular Biology, Methods in Enzymology, Guthrie and Fink, eds., Academic Press, 194:251-270 (1991)). The 4.5 kb fragment, detected by the right arm probe, spans the region between the HindIII site at the telomere 5′ end and the first HindIII site within the human insert. (scheme a). The 3 kb and 4.1 kb fragments detected by the left end probe correspond to the region between the HindIII site at the telomere end and the HindIII site 5′ of the yeast sequences, and the region spanning from the HindIII site 3′ of the centromere into the human insert, respectively (scheme b). The difference in the hybridization intensity of these two bands relates to the difference in the amount of homology between these fragments and the probe. The yeast Ty repetitive probe (Philippsen et al., in Gene Expression in Yeast, Proceedings of the Alko Yeast Symposium, Helsinki, Korhola and Vaisanen, eds., Foundation for Biotechnical and Industrial Fermentation Research, 1:189-200 (1983)) was a 5.6 kb XhoI fragment isolated from Ty1-containing pJEF742 which could also detect the 3′ HindIII fragment of Ty2, due to the homology between the two elements. The LYS2 gene probe was a 1.7 BamHI fragment from pLUS (Hermanson et al., Nuc. Acids. Res. 19:4943-4948 (1991)).
  • Hybridization with a human HPRT probe (full length 1.6 kb cDNA probe) demonstrated that all the clones analyzed contained the same 15, 7 and 5 kb exon-containing fragments of the human HPRT gene as the yHPRT YAC. Reprobing the same blots with a human [0197] repetitive Alu sequence 300 bp probe indicated that all the clones analyzed contained most, if not all, the Alu-containing fragments present in yHPRT (FIG. 12A). These data indicate that in most of the clones analyzed the 680 kb human insert had not been detectably rearranged or deleted upon integration into the ES cell genome. Integration of YAC vector sequences was examined using probes specific for the vector arms. Rehybridization of the same blots with a probe for the right YAC vector arm, detecting a 4.5 kb HindIII fragment, indicated that in 10 out of 23 of the clones analyzed, the right YAC arm up to the telomere was still intact and unrearranged and linked to the human insert (FIG. 12B) thus providing further evidence for the integrity of the YAC in these clones. The left arm probe detected the 3 kb and 4.1 kb HindIII yHPRT fragments in 18 out of the 20 clones analyzed (FIG. 12C), indicating a high frequency of left arm retention.
  • The structural integrity of yHPRT in ESY clones was further evaluated for two clones (ESY 5-2 and 8-7) using pulsed-field gel restriction analysis. In yeast carrying yHPRT, five Sfi fragments of the following approximate sizes were defined by different probes: 315 kb (Alu, left arm), 145 kb (Alu, HPRT); 95 kb (Alu, right arm), 70 and 50 kb (Alu only). In both ES clones, the internal HPRT and Alu-specific fragments were similar in size to the yHPRT fragments. The end fragments detected for both clones were larger than those in yHPRT, as expected for YACs integrated within a mouse chromosome: 185 and 200 kb for the right end fragment, respectively, and over 800 kb for the left end fragment for both clones. These data, together with the Alu profile, provide additional evidence for the retention of the structural integrity of the YAC in these clones. These studies were complemented by fluorescence in-situ hybridization carried out on ESY 8-7 (FIGS. 13A, B) and ESY 8-6 metaphase chromosome spreads in which a single integration site was detected for the human sequences. Photomicrographs of representative metaphase spreads (FIGS. 13A, B, C) or interphase nuclei (FIG. 13D) from ESY 8-7 cells (FIGS. 13A, B) hybridized with biotinylated human genomic sequences and ESY 8-6 cells (FIGS. 13C, D) hybridized with biotinylated yeast repeated DNA sequences. The human probe was generated from human genomic placental DNA (Clontech, Palo Alto, Calif.). The yeast probe consisted of a mix of DNA fragments encoding the yeast repeated elements; delta (a 1.08 kb Sau3A fragment of pdelta6 (Gafner et al., [0198] EMBO J. 2:583-591 (1983)) and Ty (a 1.35 kb EcoRI-SaII fragment of p29 (Hermanson et al., Nuc. Acids. Res. 19:4943-4948 (1991)), the rDNAs (a 4.6 kb BgIIIk-A L90 and a 4.4 kb BgIII-B L92 fragment (Keil and Roeder, Cell 39:377-386 (1984)), and the Y′ telomere elements (2.0 and 1.5 kb BgIII-HindIII fragments of p198 (Chan and Tye, Cell 33:563-573 (1983)). Hybridization of sequences on chromosome metaphase spreads with biotinylated probes and detection by Avidin-FITC followed by biotin-anti-Avidin-and Avidin-FITC amplification was carried as described by Trask and Pinkel, Methods Cell Biol. 30:383-400 (1990), using a Zeiss Axiophot microscope. Chromosomes were counterstained with propidium iodide. The photomicrographs shown are representative of 95% of the metaphase spreads or interphase nuclei scanned in three independent experiments carried out with the human or the yeast probes. A single integration site was detected for the human sequences.
  • The same blots were also probed with the yeast Ty repetitive element sequence to detect the presence of yeast genomic DNA sequences in the ESY clones (FIG. 12D). Whereas some of the clones were found to contain most of the Ty-containing fragments present in the parental yeast strain, some of the clones were found to have a very small fraction, if at all, of the Ty-containing fragments. These results indicate that in some ES clones, although the YAC DNA is integrated intact, little or no yeast genomic DNA was integrated. To determine if the yeast chromosomal DNA was integrated at single or multiple sites within the ES cell genome, fluorescent in-situ hybridization was performed on ESY clone 8-6 which had a complete Ty profile. A single integration site was detected using a combined yeast repetitive probe (FIGS. 13C, D), indicating that within the limits of resolution, all yeast DNA fragments integrated in one block. [0199]
  • Using the ability of ES cells to undergo in vitro orderly differentiation, YAC stability and the effect of integrated DNA on the pluripotency of ES cells was investigated. Four ES clones, containing different amounts of yeast DNA (ESY 5-2, 3-6, 8-6 and 8-7) exhibited a differentiation pattern indistinguishable from that of unfused ES cells: formation of embryoid bodies giving rise to a variety of differentiated cell types (FIG. 14A). Southern blot analysis was performed on DNA extracted from differentiated ESY 5-2, 3-6, 8-5 and 8-6 (20 μg) and yHPRT in AB1380 (40 ng) using (a) a human Alu probe; (b) yeast Ty sequences. ES clones were induced to form embryoid bodies by culturing them as aggregates in suspension for 10-14 days as described by Martin and Evans, [0200] Cell 6:467-474 (1975) Following their reattachment to tissue culture substratum, ESY-derived embryoid bodies gave rise to differentiated cell types. YAC and yeast DNA sequences were stably retained by the differentiated ES clones during 40 days of culture in non-selective medium, demonstrating that the stably integrated foreign DNA did not impair the pluripotency of the ES cells (FIG. 14B). The differentiated cultures maintained a functional human HPRT gene as evidenced by their normal growth and differentiation when transferred to HAT-selective medium.
  • 5. Generation of Chimeric Mice from yHPRT-ES Cell Lines [0201]
  • The ability of ESY cells to repopulate mice, including the germline, was demonstrated by microinjection of ES cells into mouse blastocysts and the generation of chimeric mice. ESY cells were microinjected into C57BL/6J mouse blastocysts, and chimeric mice were generated as previously described. Chimeric males were mated with C57BL/6J females and germline transmission was determined by the presence of agouti offspring. Genomic DNA prepared from the tails of the chimeric mice were analyzed for the presence of the yHPRT DNA in the mouse genome by PCR analysis. The presence of the YAC left arm was analyzed using the two priming oligonucleotides, 5′ TTCTCGGAGCACTGTCCGACC and 5′ CTTGCGCCTTAAACCAACTTGGTACCG, which were derived, respectively, from the pBR322 sequences and the SUP4 gene within the YAC left vector arm. A 259 bp PCR product was obtained from the analysis of the yeast containing yHPRT and the ESY cell lines. PCR analysis of tail DNA prepared from 18 chimeric mice generated from ESY cell lines ESY3-1 ESY3-6 and ESY5-2, gave rise to the expected PCR product, thus indicating the presence of the YAC left vector arm in the genome of the chimeric mice. [0202]
  • 6. Germline Transmission of yHPRT [0203]
  • Chimeric males, with coat color chimerism of 30-60%, derived from the ESY cell lines ESY3-1 and ESY5-2 were set up for mating for germline transmission evaluation, i.e. to determine whether the genetic modification was passed via the germ cells (sperm or oocytes) to the progeny of the animals. Three of the chimeric ESY3-1 derived males, 394/95-1, 394/95-2 and 411-1 transmitted the ES cell genome to their offspring at a frequency of 20%, 30% and 30%, respectively. Southern blot analysis of tail DNA from the agouti pups indicated the presence of the yHPRT in the genome of three mice, 4-2, 4-3 and 5-1, derived from the 394/395-2 chimera. The Alu profile obtained from such analysis was indistinguishable from that of the parent ES3-1 cell line (FIG. 14C), demonstrating that the 680 kb human insert was transmitted faithfully through the mouse germline. [0204]
  • Using a human HPRT-specific PCR assay on mRNA-derived cDNAs from a yHPRT-containing offspring, the expression of the human HPRT gene in all the tissues tested was detected (FIGS. 15A and B), thus demonstrating the transmitted YAC retained its function with fidelity. In this experiment, human HPRT mRNA was detected by reverse transcription (RT)-PCR in ES, ESY 3-1 and Hut 78 (human) cells, spleen and liver from a control mouse (C) or the 4-3 agouti offspring (derived from the 394/95-2 chimera) and a sample containing no template DNA (indicated as “−” in FIG. 15A). Reverse transcription of poly (A+) RNA and PCR amplification of specific cDNA sequences were performed using the cDNA Cycle Kit (Invitrogen). Specific amplification of a 626 bp fragment from human HPRT cDNA in the presence of murine HPRT cDNA was performed as outlined by Huxley et al, supra. Integrity of all RNA samples was demonstrated by PCR amplification of cDNAs for the mouse γ-interferon receptor. The primers used to amplify a 359 bp fragment were: GTATGTGGAGCATAACCGGAG and CAGGTTTTGTCTCTAACGTGG. The human HPRT and the γ-interferon receptor primers were designed to eliminate the possibility of obtaining PCR products from genomic DNA contamination. PCR products were analyzed by electrophoresis and visualized with ethidium bromide. The size markers are 1 kb ladder (BRL). The results of detection of mouse γ-interferon receptor mRNA by RT-PCR in the samples described above are shown in FIG. 15B. The specific human HPRT mRNA was also detected in the other tissues tested (brain, kidney and heart) derived from the 4-3 mouse. Comparable steady-state levels of mouse and human HPRT mRNA were detected in the liver of yHPRT-containing progeny. These results indicate that the uptake of as much as 13 megabases of yeast genomic DNA was not detrimental to proper development, germline transmission or gene expression. [0205]
  • The above results demonstrate that yeast spheroplasts are an effective vehicle for the delivery of a single copy large molecular weight DNA fragment into ES cells and that such molecules are stably and functionally transmitted through the mouse germline. The Alu profiles, complemented by PFGE analysis and in situ hybridization for some of the ES clones, strongly argue that the majority of the clones contained virtually all the human insert in unrearranged form (i.e. in “germline configuration”), with a high frequency of clones (40%) also retaining both YAC arms. The significant uptake of yeast genomic DNA was not detrimental to proper. differentiation of ES cells in vitro and in vivo and did not prevent germline transmission or gene expression. these methods, one can transmit large fragments of genomic DNA as inserts into non-human animal genomes, where the inserts may be transmitted intact by germline transmission. Therefore, a wide variety of xenogeneic DNA can be introduced into non-human hosts such as mammals, particularly small laboratory animals, that may impart novel phenotypes or novel genotypes. For example, one can provide in small laboratory animals genes of a mammal, such as a human, to study the etiology of a disease, the response to human genes to a wide variety of agents. Alternatively, one can introduce large loci into a mammalian host to produce products of other species, for example humans, to provide human protein sequences of proteins such as immunoglobulins, T-cell receptors, major histocompatibility complex antigens, etc. [0206]
  • Introduction of Heavy Chain YAC A287-C10 and Kappa Chain YAC A80-C into ES Cells and Embryos
  • Yeast containing the human heavy chain YAC A287-C10 targeted with pLUTO (yA287-C10) were spheroplasted and fused with the HPRT-deficient ES cell line E14.1TG3B1 as described above. Ten HAT-resistant ES (ESY) clones (2B, 2C, 2D, 3A, 3B, 5C, 1125A, 1125E, 100/1500 and 100/4000) were picked and were expanded for DNA analysis. Evaluation of the integrated YAC was performed by Southern blot analysis of HindIII-digested DNA from these clones, using human heavy chain probes for the D, J[0207] H, μ, and VH2 regions, decribed above. All ESY clones were found to contain the expected >10 kb JH and μ fragments. All ESY clones except 2D and 5C clones, were found to contain the 4.8 kb VH2 kb fragment. All ESY clones, except 2D and 3B were found to contain the expected 10 and 7.6 kb D gene fragments. Yeast genomic sequences were detected by hybridization to the yeast repetitive Ty element in all ESY clones except 2B, 2D, 100/1500 and 5C. ESY clones 2B, 3A and 5C were microinjected into C57B/6 blastocysts as described above and chimeric mice (10 from 2B clone, 1 from 3A clone and 1 from 5C clone) were generated. Southern blot, analysis of tail DNA from 10 of these chimeric animals, indicated the presence of most, if not all, of the apparent 10 Alu fragments, detected in yA287-C10 in yeast, as well as the presence of VH2 and D gene fragments. The generated chimeric mice were bred with C57BL16J mice for germline transmission evaluation. A chimeric male 78K-3 derived from the 2B clone transmitted the ES cell genome to its offspring at a frequency of 100%. Southern blot analysis of tail DNA from 4 out of 6 agouti mice pups indicated the presence of human heavy chain sequences.
  • Fusion experiments with yeast containing the human kappa chain YAC A80-C7 targeted with pLUTO (yA80-C7) with E14.1TG3B1 ES cells generated 2 HAT-resistant ESY clones: M4.4.1 and M5.2.1. Southern blot analysis of HindIII-digested DNAs from these clones revealed the presence of all the apparent 10 Alu fragments detected in yA80-C7 in yeast. In both clones yeast genomic sequences were integrated. ESY clones were microinjected into C57B1/6J blastocysts and chimeric mice were generated. [0208]
  • Example VII Production of Human Ig by Chimeric Mice by Introduction of Human Ig Using Homologous Recombination
  • As an alternative approach to that set forth in Examples I-VI, human Ig genes are introduced into the mouse Ig locus by replacing mouse heavy and light chain immunoglobulin loci directly with fragments of the human heavy and light chain loci using homologous recombination. This is followed by the generation of chimeric transgenic animals in which the embryonic stem-cell derived cells contribute to the germ line. [0209]
  • A. Construction of Human Heavy Chain Replacement Vector. [0210]
  • The replacing human sequences include the [0211] SpeI 100 kb fragment of genomic DNA which encompasses the human VH6-D-J-Cμ-Cδ heavy chain region isolated from a human-YAC library as described before. The flanking mouse heavy chain sequences, which drive the homologous recombination replacement event, contain a 10 kb BamHI fragment of the mouse Cε-Cα heavy chain and a 5′ J558 fragment comprising the 5′ half of the J558 fragment of the mouse heavy chain variable region, at the 3′ and 5′ ends of the human sequences, respectively (FIG. 16). These mouse sequences are isolated from a mouse embryo genomic library using the probes described in Tucker et al. (1981), PNAS USA, 78: 7684-7688 and Blankenstein and Krawinkel (1987, supra), respectively. The 1150 bp XhoI to BamHI fragment containing a neomycin-resistance gene driven by the Herpes simplex virus thymidine kinase gene (HSV-tk) promoter and a polyoma enhancer is isolated from pMC1Neo (Koller and Smithies, 1989, supra). A synthetic adaptor is added onto this fragment to convert the XhoI end into a BamHI end and the resulting fragment is joined to the BamHI mouse Cε-Cα in a plasmid.
  • From the YAC clone containing the human heavy chain locus, DNA sequences from each end of the insert are recovered either by inverse PCR (Silverman et al. (1989) [0212] PNAS, 86:7485-7489), or by plasmid rescue in E. coli, (Burke et al., (1987); Garza et al. (1989) Science, 246:641-646; Traver et al., 1989) (see FIG. 8). The isolated human sequence from the 5′V6 end of the YAC is ligated to the mouse J558 sequence in a plasmid and likewise, the human sequence derived from the 3′Cd end of the YAC is ligated to the Neo gene in the plasmid containing Neo and mouse Cε-Cα described above. The human V6-mouse J558 segment is now subcloned into a half-YAC cloning vector that includes a yeast selectable marker (HIS3) not present in the original IgH YAC, a centromere (CEN) and a single telomere (TEL). The human Cδ-Neo-mouse Cε-Cα is likewise subcloned into a separate half-YAC vector with a different yeast selectable marker (LEU2) and a single TEL. The half-YAC vector containing the human V6 DNA is linearized and used to transform a yeast strain that is deleted for the chromosomal HIS3 and LEU2 loci and which carries the IgH YAC. Selection for histidine-prototrophy gives rise to yeast colonies that have undergone homologous recombination between the human V6 DNA sequences and contain a recombinant YAC. The half-YAC vector containing the human Cδ DNA is then linearized and used to transform the yeast strain generated in the previous step. Selection for leucine-prototrophy results in a yeast strain containing the complete IgH replacement YAC (see FIG. 16). Preferably, both targeting events are performed in a single transformation step, selecting simultaneously for leucine and histidine prototrophy. This is particularly useful when the original centric and acentric YAC arms are in opposite orientation to that shown in FIG. 16. This YAC is isolated and introduced into ES cells by microinjection as described previously for embryos.
  • Example VIII Crossbreeding of Transgenic Mice
  • A. Generation of Human Monoclonal Antibody Producing Mice [0213]
  • Mice containing the human immunoglobulin locus are mated to mice with inactivated murine immunoglobulin genes to generate mice that produce only human antibodies. Starting with four heterozygous strains, three generations of breeding are required to create a mouse that is homozygous for inactive murine kappa and heavy chain immunoglobulins, and heterozygous for human heavy and kappa chain immunoglobulin loci. The breeding scheme is shown in FIG. 17. [0214]
  • Example IX Production of Human Monoclonal Antibodies
  • A. Immunization of Mice [0215]
  • Germline chimeric mice containing integrated human DNA from the immunoglobulin loci are immunized by injection of an antigen in adjuvant. The mice are boosted with antigen 14 days after the primary immunization, repeated after 35 and 56 days. A bleed is done on the immunized animals to test the titer of serum antibodies against the immunizing antigen. The mouse with the highest titer is sacrificed, and the spleen removed. [0216]
  • B. Fusion of Splenocytes [0217]
  • Myeloma cells used as the fusion partner for the spleen cells are thawed 6 days prior to the fusion, and grown in tissue culture. One day before the fusion, the cells are split into fresh medium containing 10% fetal calf serum at a concentration of 5×10[0218] 5 cells/ml. On the morning of the fusion the cells are diluted with an equal volume of medium supplemented with 20% fetal calf serum and 2×OPI (3 mg/ml oxaloacetate, 0.1 mg/ml sodium pyruvate and 0.4 IU/ml insulin) solution.
  • After sacrificing the mouse, the spleen is aseptically removed, and placed in a dish with culture medium. The cells are teased apart until the spleen is torn into fine pieces and most cells have been removed. The cells are washed in fresh sterile medium, and the clumps allowed to settle out. [0219]
  • The splenocytes are further washed twice by centrifugation in medium without serum. During the second wash, the myeloma cells are also washed in a separate tube. After the final wash the two cell pellets are combined, and centrifuged once together. [0220]
  • A solution of 50% polyethylene glycol (PEG) is slowly added to the cell pellet while the cells are resuspended, for a total of two minutes. 10 ml of prewarmed medium is added to the cell solution, stirring slowly for 3 minutes. The cells are centrifuged and the supernatant removed. The cells are resuspended in 10 ml of medium supplemented with 20% fetal calf serum, 1×OPI solution and 1×AH solution (58 μM azaserine, 0.1 mM hypoxanthine). The fused cells are aliquoted into 96-well plates, and cultured at 37° for one week. [0221]
  • Supernatant is aseptically taken from each well, and put into pools. These pools are tested for reactivity against the immunizing antigen. Positive pools are further tested for individual wells. When a positive well has been identified, the cells are transferred from the 96-well plate to 0.5 ml of medium supplemented with 20% fetal calf serum, 1×OPI, and 1×AH in a 24-well plate. When that culture becomes dense, the cells are expanded into 5 ml, and then into 10 ml. At this stage the cells are sub-cloned so that a single antibody producing cell is in the culture. [0222]
  • In accordance with the above procedures, a chimeric non-human host, particularly a murine host, may be produced which can be immunized to produce human antibodies or analogs specific for an immunogen. In this manner, the problems associated with obtaining human monoclonal antibodies are avoided, because the transgenic host can be immunized with immunogens which could not be used with a human host. Furthermore, one can provide for booster injections and adjuvants which would not be permitted with a human host. The resulting B-cells may then be used for immortalization for the continuous production of the desired antibody. The immortalized cells may be used for isolation of the genes encoding the immunoglobulin or analog, and be, subjected to further molecular modification by methods such as in-vitro mutagenesis or other techniques to modify the properties of the antibodies. These modified genes may then be returned to the immortalized cells by transfection to provide for a continuous mammalian cellular source of the desired antibodies. The subject invention provides for a convenient source of human antibodies, where the human antibodies are produced in analogous manner to the production of antibodies in a human host. The animal host cells conveniently provide for the activation and rearrangement of human DNA in the host cells for production of human antibodies. [0223]
  • In accordance with the subject invention, human antibodies can be produced to human immunogens, eg. proteins, by immunization of the subject host mammal with human immunogens. The resulting antisera will be specific for the human immunogen and may be harvested from the serum of the host. The immunized host B cells may be used for immortalization, eg. myeloma cell fusion, transfection, etc. to provide immortal cells, eg. hybridomas, to produce monoclonal antibodies. The antibodies, antiserum and monoclonal antibodies will be glycosylated in accordance with the species of the cell producing the antibodies. Rare variable regions of the Ig locus may be recruited in producing the antibodies, so that antibodies having rare variable regions may be obtained. [0224]
  • All publications and patent applications cited in this specification are herein incorporated by reference as if each individual publication or patent application were specifically and individually indicated to be incorporated by reference. [0225]
  • Although the foregoing invention has been described in some detail by way of illustration and example for purposes of clarity of understanding, it will be readily apparent to those of ordinary skill in the art in light of the teachings of this invention that certain changes and modifications may be made thereto without departing from the spirit or scope of the appended claims. [0226]
  • 1 33 1 24 DNA Artificial Sequence Description of Artificial Sequence Primer 1 tggcggaccg ctatccccca ggac 24 2 20 DNA Artificial Sequence Description of Artificial Sequence Primer 2 tagcctgggt ccctccttac 20 3 20 DNA Artificial Sequence Description of Artificial Sequence Primer 3 acggtatcgc cgctcccgat 20 4 23 DNA Artificial Sequence Description of Artificial Sequence Primer 4 agtcactgta aagacttcgg gta 23 5 56 DNA Artificial Sequence Description of Artificial Sequence Synthetic polylinker 5 gcatatgcct gaggtaagca tgcggtaccg aattctataa gcttgcggcc gcagct 56 6 56 DNA Artificial Sequence Description of Artificial Sequence Synthetic polylinker 6 gcggccgcaa gcttatagaa ttcggtaccg catgcttacc tcaggcatat gcgtac 56 7 32 DNA Artificial Sequence Description of Artificial Sequence Synthetic adapter 7 agctggaacc ccttgccctt ggggaacgcc gg 32 8 47 DNA Artificial Sequence Description of Artificial Sequence Synthetic polylinker 8 gagctcggat cctatctcga ggaattctat aagcttcata tgtagct 47 9 47 DNA Artificial Sequence Description of Artificial Sequence Synthetic polylinker 9 acatatgaag cttatagaat tcctcgagat aggatccgag ctcgtac 47 10 50 DNA Artificial Sequence Description of Artificial Sequence Synthetic polylinker 10 aagcttatag aattcggtac ctggatcctg agctcatagc ggccgcagct 50 11 50 DNA Artificial Sequence Description of Artificial Sequence Synthetic polylinker 11 gcggccgcta tgagctcagg atccaggtac cgaattctat aagcttgtac 50 12 47 DNA Artificial Sequence Description of Artificial Sequence Synthetic polylinker 12 acatatgaag cttatagaat tcctcgagat aggatcchag ctcgtac 47 13 13 DNA Artificial Sequence Description of Artificial Sequence Synthetic oligonucleotide 13 gggaagccgc cgc 13 14 21 DNA Artificial Sequence Description of Artificial Sequence Synthetic oligonucleotide 14 catggcggcg gcttccctgc a 21 15 26 DNA Artificial Sequence Description of Artificial Sequence Synthetic oligonucleotide 15 caggatccag ctgtgccttc tagttg 26 16 30 DNA Artificial Sequence Description of Artificial Sequence Primer 16 ctgagctcta gacccataga gcccaccgca 30 17 16 DNA Artificial Sequence Description of Artificial Sequence Synthetic oligonucleotide 17 agctggaacc ccttgc 16 18 16 DNA Artificial Sequence Description of Artificial Sequence Synthetic oligonucleotide 18 ggccgcaagg ggttcc 16 19 24 DNA Artificial Sequence Description of Artificial Sequence Primer 19 gcagagcctg ctgaattctg gctg 24 20 22 DNA Artificial Sequence Description of Artificial Sequence Primer 20 gtaatacaca gccgtgtcct gg 22 21 23 DNA Artificial Sequence Description of Artificial Sequence Primer 21 ttccggcccc gatgcgggac tgc 23 22 15 DNA Artificial Sequence Description of Artificial Sequence Primer 22 cctctcccta agact 15 23 18 DNA Artificial Sequence Description of Artificial Sequence Primer 23 caaaggataa cagccctg 18 24 18 DNA Artificial Sequence Description of Artificial Sequence Primer 24 agctggctgc ttgtcatg 18 25 21 DNA Artificial Sequence Description of Artificial Sequence Primer 25 gtatgtggag cataaccgga g 21 26 21 DNA Artificial Sequence Description of Artificial Sequence Primer 26 caggttttgt ctctaacgtg g 21 27 52 DNA Artificial Sequence Description of Artificial Sequence Synthetic polylinker 27 aagcttatag aattcggtac ctggatcctg agctcatata gcggccgcag ct 52 28 50 DNA Artificial Sequence Description of Artificial Sequence Synthetic polylinker 28 gccgccgcta tgagctcagg atccaggtac cgaattctat aagcttgtac 50 29 16 DNA Artificial Sequence Description of Artificial Sequence Synthetic polylinker 29 agctggaacc ccttgc 16 30 16 DNA Artificial Sequence Description of Artificial Sequence Synthetic polylinker 30 ggccgcaagg ggttcc 16 31 47 DNA Artificial Sequence Description of Artificial Sequence Synthetic polylinker 31 acatatgaag cttatagaat tcctcgagat aggatccgag ctcctac 47 32 21 DNA Artificial Sequence Description of Artificial Sequence Primer 32 ttctcggagc actgtccgac c 21 33 27 DNA Artificial Sequence Description of Artificial Sequence Primer 33 cttgcgcctt aaaccaactt ggtaccg 27

Claims (78)

What is claimed is:
1. A method for producing a xenogeneic immunoglobulin or analog thereof in a non-human animal host, said method comprising:
immunizing said host with an immunogen under conditions to stimulate an immune response to said immunogen, whereby said host mounts an immune response to said immunogen and produces B-cells producing immunoglobulin specific for said immunogen, and isolating xenogeneic immunoglobulin produced bys aid host,
wherein said host is characterized by 1) being substantially incapable of producing endogenous immunoglobulin heavy chain; (2) being substantially incapable of producing endogenous immunoglobulin light chains; and 3) being capable of producing a xenogeneic immunoglobulin or analog thereof.
2. A method according to claim 1, wherein said host is rendered substantially incapable of producing endogenous immunoglobulin heavy and light chains by inactivation of at least a portion of said endogenous immunoglobulin heavy and light chain loci by homologous recombination.
3. A method according to claim 2, wherein said inactivation is a result of introduction of a lesion into the endogenous immunoglobulin loci.
4. A method according to claim 1, wherein said analog comprises a variable region joined by a peptide bond to a peptide other than solely the immunoglobulin constant region.
5. A method according to claim 1, wherein said xenogeneic immunoglobulin is human immunoglobulin.
6. A method according to claim 1, including the additional step of immortalizing said B-cells.
7. A method according to claim 1, wherein said host comprises B-cells comprising a functional immunoglobulin locus comprising a xenogeneic variable region and at least one human constant region.
8. A method according to claim 1, wherein said non-human host is a rodent.
9. A method according to claim 1, wherein said xenogeneic immunoglobulin is chimeric immunoglobulin.
10. A method according to claim 9, wherein said chimeric immunoglobulin is mouse/human immmunoglobulin.
11. An immortalized non-human cell line genetically modified so as to lack the ability to produce immunoglobulin endogenous to the cell line and comprising xenogeneic immunoglobulin loci encoding at least one xenogeneic immunoglobulin heavy chain and a light chain;
wherein said xenogeneic immunoglobulin heavy and light chain loci are expressed.
12. An immortalized cell line according to claim 11, wherein said cell line is a B cell hybridoma.
13. An immortalized cell line according to claim 11, wherein said non-human cell line is a murine cell line, and said xenogeneic immunoglobulin loci are human immunoglobulin loci.
14. A method of making a xenogeneic immunoglobulin comprising culturing the immortalized cell line of claim 11 under suitable culture conditions and recovering the xenogeneic immunoglobulin.
15. A xenogeneic immunoglobulin produced by the method according to claim 1 or 14.
16. A genetically modified non-human animal comprising a modified genome selected from the group consisting of:
a genome heterozygous or homozygous for a modification that results in the inability of at least one locus to produce endogenous immunoglobulin heavy or light chains;
a genome heterozygous or homozygous for a modification that results in the inability of at least one locus to produce endogenous immunoglobulin heavy and light chains;
a genome heterozygous for a modification that results in the inability of at least one locus to produce endogenous immunoglobulin heavy and light chains and hemizygous for the ability to produce xenogeneic immunoglobulin heavy chains;
a genome heterozygous for a modification that results in the inability of at least one locus to produce endogenous immunoglobulin heavy and light chains and hemizygyous for the ability to produce xenogeneic immunoglobulin light chains;
a genome homozygous for a modification that results in the inability to produce endogenous immunoglobulin heavy and light chains and homozygous for the ability to produce xenogeneic immunoglobulin heavy or light chains;
a genome homozygous for a modification that results in the inability to produce endogenous immunoglobulin heavy and light chains and hemizygous for the ability to produce xenogeneic immunoglobulin heavy or light chains;
a genome homozygous or heterozygous for a modification that results in the inability of at least one locus to produce endogenous immunoglobulin heavy and light chains and hemizygous for the ability to produce xenogeneic immunoglobulin heavy and light chains;
a genome heterozygous for a modification that results in the inability of at least one locus to produce endogenous immunoglobulin heavy or light chain and hemizygous for a modification that results in the ability to produce xenogeneic immunoglobulin heavy and light chains;
a genome homozygous for a modification that results in the inability to produce endogenous immunoglobulin heavy or light chain and homozygous for a modification that results in the ability to produce xenogeneic immunoglobulin heavy and light chains; and
a genome homozygous for a modificatio that results in the inability to produce endogenous immunoglobulin heavy or light chain and hemizygous for a modification that results in the ability to produce xenogeneic immunoglobulin heavy and light chains.
17. A non-human animal according to claim 16, wherein the animal is murine.
18. A non-human animal according to claim 16, wherein the xenogeneic immunoglobulin is human.
19. A non-human animal according to claim 16, wherein the inability to produce endogenous immunoglobulin is a result of inactivation of at least a portion of the endogenous immunoglobulin loci by homologous recombination.
20. A non-human animal according to claim 19, wherein at least a portion of the endogenous immunoglobulin light and heavy chain loci are replaced with at least one locus capable of producing xenogeneic immunoglobulin.
21. A non-human animal according to claim 16, wherein said inactivation comprises introduction of a lesion in the loci encoding said heavy and/or light immunoglobulin chains.
22. A non-human animal according to claim 21, wherein said lesion is in the constant and/or J region.
23. A non-human animal according to claim 21, wherein said light chain loci are kappa immunoglobulin chain loci.
24. A non-human animal according to claim 21, wherein said light chain loci are lambda immunoglobulin chain loci.
25. A transgenic murine animal comprising a genome lacking the ability to produce endogenous immunoglobulin, said genome comprising a lesion in the J region of the heavy chain immunoglobulin loci, and a lesion in the constant and/or J regions of the light chain immunoglobulin loci.
26. A murine animal according to claim 25, wherein said genome further comprises xenogeneic heavy and light chain immunoglobulin loci and said murine animal has the ability to produce xenogeneic immunoglobulin.
27. A method for producing a modified non-human animal, said animal having a xenogeneic DNA segment of at least 100 kb stably integrated into the genome of said animal, said method comprising:
combining under fusing conditions yeast spheroplasts, said spheroplasts comprising a YAC having said xenogeneic DNA segment and a marker for selection, with embryonic stem cells of said animal, whereby said xenogeneic DNA segment becomes integrated into the genome of said embryonic stem cells;
selecting for embryonic stem cells carrying said xenogeneic DNA segment by means of the marker;
transferring said embryonic cells into a host blastocyst and implanting said blastocyst in a pseudopregnant animal recipient, and allowing said blastocyst to develope to term to produce a chimeric animal carrying said xenogeneic DNA segment; and
mating said chimeric animal with an animal of the same species to produce said modified animal carrying said xenogeneic DNA segment.
28. A method according to claim 27, wherein said marker is the HPRT gene and said embryonic stem cell is HPRT deficient.
29. A method according to claim 27, wherein said step of mating produces heterozygous progeny and the heterozygous progeny are mated to produce homozygous progeny.
30. A method according to claim 27, wherein said animal is a rodent.
31. A method according to claim 30, wherein said animal is a murine animal.
32. A method according to claim 27, wherein said xenogeneic DNA is human DNA.
33. A method according to claim 32, wherein said xenogeneic DNA is human immunoglobulin DNA in substantially intact form.
34. The modified animal produced by the method according to claim 27.
35. A non-human animal heterozygous or homozygous for a xenogeneic genomic mammalian DNA segment of at least 100 kb, stably integrated in substantially intact form into the genome of said animal.
36. A non-human animal according to claim 35, comprising a HPRT gene and wherein said xenogeneic DNA is human DNA.
37. A non-human animal according to claim 36, wherein said human DNA is human immunoglobulin DNA in substantially intact form.
38. A non-human animal according to claim 35, wherein said animal is a rodent.
39. A non-human animal according to claim 38, wherein said animal is a murine animal.
40. An embryonic stem cell comprising a genome having endogenous immunoglobulin heavy chain loci, and immunoglobulin light chain loci, said genome comprising a lesion in said endogenous immunoglobulin heavy chain and/or light loci, resulting in the incapacity of the immunoglobulin locus comprising said lesion to rearrange.
41. An embryonic stem cell according to claim 40, wherein said lesion is in the J and/or constant regions of said endogenous immunoglobulin loci.
42. An embryonic stem cell according to claim 40, wherein said lesion is insertion of a xenogeneic sequence.
43. An embryonic stem cell according to claim 42 wherein said xenogeneic sequence is immunoglobulin DNA or a selectable marker.
44. An embryonic stem cell according to claim 43, wherein said marker is neomycin.
45. An embryonic stem cell according to claim 42, wherein said lesion further comprises deletion of endogenous immunoglobulin DNA.
46. An embryonic stem cell according to claim 40 wherein said stem cell is homozygous for the lesion.
47. An embryonic stem cell according to claim 40 wherein the lesion is in the heavy chain immunoglobulin J region loci.
48. An embryonic stem cell according to claim 40 wherein the lesion is in th light chain immunoglobulin J region loci.
49. An embryonic stem cell according to claim 40 wherein said lesion comprises replacement of at least a portion of the immunoglobulin light and heavy chain loci comprising said endogenous immunoglobulin loci with loci capable of producing xenogeneic immunoglobulin by homologous recombination.
50. A murine embryonic stem cell comprising homozygotic alleles of immunoglobulin heavy chain loci, said loci comprising a lesion resulting in the incapacity of the immunoglobulin loci comprising said lesion to rearrange.
51. A murine embryonic stem cell according to claim 50, wherein said lesion is in the J region of said immunoglobulin heavy chain loci.
52. A murine embryonic stem cell comprising homozygotic alleles of immunoglobulin light chain loci, said loci comprising a lesion resulting in the incapacity of the immunoglobulin loci comprising said lesion to rearrange.
53. A murine embryonic stem cell according to claim 52, wherein said lesion is in the constant and/or J regions of said immunoglobulin light chain loci.
54. A murine embryonic stem cell of a murine host said stem cell comprising a genome having immunoglobulin loci comprising J regions, said stem cell comprising a lesion in at least one of the J regions of the immunoglobulin locus resulting in the incapacity of said immunoglobulin locus to rearrange, said embryonic stem cell produced by the method comprising introducing homologous DNA into a murine stem cell in culture, wherein said homologous DNA comprises a region homologous with the J region of an immunoglobulin locus and a marker gene for insertion into said locus; and selecting for embryonic stem cells having undergone homologous recombination with said homologous DNA.
55. A murine embryonic stem cell according to claim 54 wherein said marker is the neomycin gene.
56. A murine embryonic stem cell according to claim 54 wherein said lesion is in at least one of the J regions of an endogenous heavy chain immunoglobulin locus.
57. A murine embryonic stem cell comprising at least 100 kb of xenogeneic DNA.
58. A murine embryonic stem cell according to claim 57, wherein said xenogeneic DNA is immunoglobulin heavy and/or light chain immunoglobulin DNA.
59. A murine embryonic stem cell according to claim 58, wherein said xenogeneic DNA is human immunoglobulin DNA in substantially intact form.
60. A method for modifying a genome of a recipient murine embryonic stem cell by homologous recombination with a large xenogeneic DNA genomic fragment previously manipulated in a yeast artificial chromosome (YAC), the improvement which comprises:
introducing at least one YAC into said murine embryonic stem cell by spheroplast fusion, and selecting recipient cells comprising said genomic fragment, wherein said YAC comprises a mammalian selectable or screenable gene, wherein said YAC is faithfully transmitted through the host germline, and said xenogeneic DNA fragment is transmitted in substantially intact form.
61. A method according to claim 60, wherein said selectable or screenable gene is HPRT and the recipient cells are selected with HAT medium and are negative for HPRT.
62. A method according to claim 60, wherein said selectable or screenable gene is a HPRT minigene.
63. A method according to claim 60 wherein said selectable or screenable gene is cDNA encoding a gene, selected from the group consisting of neomycin, hygromycin, HPRT, GPT and βgal.
64. A method according to claim 60, wherein said YAC comprises at least 100 kb of a human immunoglobulin DNA locus in substantially intact form.
65. A modified YAC according to claim 64, further comprising a mammalian selectable or screenable marker.
66. A modified YAC according to claim 65, wherein the selectable marker is HPRT.
67. A murine embryonic stem cell comprising a genome modified according to the method of claim 60.
68. A murine animal heterozygous for a xenogeneic unrearranged mammalian DNA segment of at least 100 kb stably integrated into the genome of said murine animal.
69. A murine animal according to claim 68 comprising a xenogeneic HPRT gene and wherein said DNA segment is human immunoglobulin.
70. A human antibody molecule characterized by;
comprising the protein sequences of the human immunoglobulin heavy and light chains;
specificity for an immunogen; and
having other than human glycosylation.
71. A human antibody molecule according to claim 70, wherein said antibody is monoclonal.
72. A method for producing a genetically modified non-human animal, comprising interbreeding a first parent and a second parent, and recovering the progeny thereof, wherein the parents and progeny are selected from the group consisting of:
first and second parents heterozygous for a genome modified to be incapable of producing endogenous immunoglobulin light chain, and progeny homozygous for said modified genome;
first and second parents heterozygous for a genome modified to be incapable of producing an endogenous immunoglobulin heavy chain, and progeny homozygous for said modified genome;
a first parent heterozygous for a genome modified to be incapable of producing an endogenous immunoglobulin light chain, a second parent heterozygous for a genome modified to be incapable of producing an endogenous immunoglobulin heavy chain and progeny heterozygous for said modified genome so as to be incapable of producing endogenous immunoglobulin heavy and light chains;
first and second parents heterozygous for a genome modified to be incapable of producing endogenous immunoglobulin light and heavy chains, and progeny homozygous for said modified genome;
a first parent hemizgyous for a genome modified to be capable of producing xenogeneic immunoglobulin heavy chain, a second parent heterozygous for a genome modified to be incapable of producing endogenous immunoglobulin light and heavy chains, and progeny heterozygous for said modified genome so as to be incapable of producing endogenous immunoglobulin light and heavy chains and hemizygous for a modified genome so as to be capable of producing xenogeneic immunoglobulin heavy chain;
first and second parents heterozygous for a genome modified to be incapable of producing endogenous immunoglobulin light and heavy chains and hemizygous for a genome modified to be capable of producing xenogeneic immunoglobulin heavy chain, and progeny 1) homozygous for said modified genome and 2) homozygous for said modification of being incapable of producing endogenous immunoglobulin light and heavy chains and also hemizygous for the modification of being capable of producing xenogeneic immunoglobulin heavy chain;
a first parent hemizygous for a genome modified to be capable of producing xenogeneic immunoglobulin light chain, a second parent heterozygous for a genome modified to be incapable of producing immunoglobulin heavy and light chains, and progeny heterozygous for said genome modified to be incapable of producing endogenous immunoglobulin light and heavy chains and hemizygous for said genome modified to be capable of producing xenogeneic immunoglobulin light chain;
first and second parents heterozygous for a genome modified to be incapable of producing endogenous immunoglobulin light and heavy chains and hemizygous for a genome modified to be capable of producing xenogeneic immunoglobulin light chain, and progeny 1) homozygous for said modified genome and 2) homozygous for said modification of being incapable of producing endogenous immunoglobulin light and heavy chains and also hemizygous for the modification of being capable of producing xenogeneic immunoglobulin light chain;
a first parent heterozygous for a genome modified to be incapable of producing endogenous immunoglobulin light and heavy chains and hemizygous for xenogeneic immunoglobulin heavy chain, a second parent heterozygous for a genome modified to be incapable of producing endogenous immunoglobulin light and heavy chains and hemizygous for the modification of being capable of producing xenogeneic immunoglobulin light chain, and progeny homozygous and heterozygous for said genome modified to be incapable of producing endogenous immunoglobulin light and heavy chains and hemizygous for the modification of being capable of producing xenogeneic immunoglobulin light and heavy chains,
first and second parents heterozygous for a genome modified to be incapable of producing endogenous immunoglobulin light and heavy chains and hemizgyous for a genome modified to be capable of producing xenogeneic immunoglobulin light and heavy chains, and progeny 1) homozygous for said modified genome, and 2) homozygous for a genome modified to be incapable of producing endogenous immunoglobulin heavy and light chains and hemizygyous for a genome modified to be capable of producing xenogeneic immunoglobulin light and heavy chains;
first and second parents homozygous for a genome modified to be incapable of producing endogenous immunoglobulin light and heavy chains and hemizygous for a genome modified to be capable of producing xenogeneic immunoglobulin light and heavy chains, and progeny homozygous for said modified genome;
a first parent heterozygous for a genome modified to be incapable of producing endogenous immunoglobulin heavy chain, a second parent hemizygyous for a genome modified to be capable of producing xenogeneic immunoglobulin light and heavy chains, and progeny heterozygous for a genome modified to be incapable of producing endogenous immunoglobulin heavy chain and hemizygyous for a genome modified to be capable of producing xenogeneic immunoglobulin light and heavy chains;
first and second parents heterozygous for a genome modified to be incapable of producing endogenous immunoglobulin heavy chain and hemizygous for a genome modified to be capable of producing xenogeneic immunoglobulin light and heavy chains, and progeny 1) homozygous for said modified genome and 2) homozygous for a genome modified to be incapable of producing endogenous immunoglobulin heavy chain and hemizygous for a genome modified to be capable of producing xenogeneic immunoglobulin light and heavy chains;
a first parent heterozygous for a genome modified to be incapable of producing endogenous immunoglobulin light chain, a second parent hemizygyous for a genome modified to be capable of producing xenogeneic immunoglobulin light and heavy chains, and progeny heterozygous for a genome modified to be incapable of producing endogenous immunoglobulin light chain and hemizygyous for a genome modified to be capable of producing xenogeneic immunoglobulin light and heavy chain; and
first and second parents heterozygous for a genome modified to be incapable of producing endogenous immunoglobulin light chain and hemizgyous for a genome modified to be capable of producing xenogeneic immunoglobulin light and heavy chains, and progeny 1) homozygous for said modified genome, and 2) homozygous for a genome modified to be incapable of producing endogenous immunoglobulin light chain and hemizygous for a genome modified to be capable of producing xenogeneic immunoglobulin light and heavy chains.
73. A method according to claim 72, wherein said modification of a genome so as to be incapable of producing endogenous immunoglobulin light and/or heavy chain is inactivation of the endogenous immunoglobulin loci as a result of homologous recombination.
74. A method according to claim 73 wherein said inactivation is a result of introduction of a lesion into the endogenous immunoglobulin loci.
75. The genetically modified non-human animal produced by the method according to claim 72.
76. The animal according to claim 75, wherein the animal is a rodent.
77. The animal according to claim 76 wherein the animal is a murine animal.
78. The animal according to claim 75, wherein the xenogeneic immunoglobulin is human immunoglobulin.
US10/627,250 1990-01-12 2003-07-24 Generation of xenogeneic antibodies Abandoned US20040093622A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US10/627,250 US20040093622A1 (en) 1990-01-12 2003-07-24 Generation of xenogeneic antibodies

Applications Claiming Priority (7)

Application Number Priority Date Filing Date Title
US46600890A 1990-01-12 1990-01-12
US61051590A 1990-11-08 1990-11-08
WOPCT/US91/00245 1991-01-11
PCT/US1991/000245 WO1991010741A1 (en) 1990-01-12 1991-01-11 Generation of xenogeneic antibodies
US91929792A 1992-07-24 1992-07-24
US08/031,801 US6673986B1 (en) 1990-01-12 1993-03-15 Generation of xenogeneic antibodies
US10/627,250 US20040093622A1 (en) 1990-01-12 2003-07-24 Generation of xenogeneic antibodies

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US08/031,801 Continuation US6673986B1 (en) 1990-01-12 1993-03-15 Generation of xenogeneic antibodies

Publications (1)

Publication Number Publication Date
US20040093622A1 true US20040093622A1 (en) 2004-05-13

Family

ID=29740869

Family Applications (4)

Application Number Title Priority Date Filing Date
US08/031,801 Expired - Fee Related US6673986B1 (en) 1990-01-12 1993-03-15 Generation of xenogeneic antibodies
US08/462,513 Expired - Lifetime US6162963A (en) 1990-01-12 1995-06-05 Generation of Xenogenetic antibodies
US10/421,011 Abandoned US20030229905A1 (en) 1990-01-12 2003-04-21 Generation of xenogeneic antibodies
US10/627,250 Abandoned US20040093622A1 (en) 1990-01-12 2003-07-24 Generation of xenogeneic antibodies

Family Applications Before (3)

Application Number Title Priority Date Filing Date
US08/031,801 Expired - Fee Related US6673986B1 (en) 1990-01-12 1993-03-15 Generation of xenogeneic antibodies
US08/462,513 Expired - Lifetime US6162963A (en) 1990-01-12 1995-06-05 Generation of Xenogenetic antibodies
US10/421,011 Abandoned US20030229905A1 (en) 1990-01-12 2003-04-21 Generation of xenogeneic antibodies

Country Status (1)

Country Link
US (4) US6673986B1 (en)

Cited By (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006029005A2 (en) * 2004-09-02 2006-03-16 Oklahoma Medical Research Foundation Promoter substitution for immunoglobulin therapy
WO2010054403A1 (en) 2008-11-10 2010-05-14 Alexion Pharmaceuticals, Inc. Methods and compositions for treating complement-associated disorders
WO2011085343A1 (en) 2010-01-11 2011-07-14 Alexion Pharmaceuticals, Inc Biomarkers of immunomodulatory effects in humans treated with anti-cd200 antibodies
WO2011137395A1 (en) 2010-04-30 2011-11-03 Rother Russell P Anti-c5a antibodies and methods for using the antibodies
WO2012020096A1 (en) 2010-08-13 2012-02-16 Medimmune Limited Monomeric polypeptides comprising variant fc regions and methods of use
WO2012022734A2 (en) 2010-08-16 2012-02-23 Medimmune Limited Anti-icam-1 antibodies and methods of use
EP2463305A1 (en) 2006-01-12 2012-06-13 Alexion Pharmaceuticals, Inc. Antibodies to OX-2/CD200 and uses thereof
WO2012106634A1 (en) 2011-02-03 2012-08-09 Alexion Pharmaceuticals, Inc. Use of an anti-cd200 antibody for prolonging the survival of allografts
US8398975B2 (en) 2006-08-03 2013-03-19 Medimmune Limited Antibodies directed to αVβ6 and uses thereof
WO2015050959A1 (en) 2013-10-01 2015-04-09 Yale University Anti-kit antibodies and methods of use thereof
WO2015175874A2 (en) 2014-05-16 2015-11-19 Medimmune, Llc Molecules with altered neonate fc receptor binding having enhanced therapeutic and diagnostic properties
US9815890B2 (en) 2010-06-22 2017-11-14 The Regents Of The University Of Colorado, A Body Corporate Antibodies to the C3d fragment of complement component 3
EP3524626A1 (en) 2007-03-22 2019-08-14 Biogen MA Inc. Binding proteins, including antibodies, antibody derivatives and antibody fragments, that specifically bind cd154 and uses thereof

Families Citing this family (663)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7041871B1 (en) * 1995-10-10 2006-05-09 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
US6632976B1 (en) 1995-08-29 2003-10-14 Kirin Beer Kabushiki Kaisha Chimeric mice that are produced by microcell mediated chromosome transfer and that retain a human antibody gene
US6090382A (en) 1996-02-09 2000-07-18 Basf Aktiengesellschaft Human antibodies that bind human TNFα
HU228630B1 (en) * 1996-02-09 2013-04-29 Abbott Biotech Ltd Use of human anti bodies that bind human tnf-alpha and process for inhibiting of human tnf-alpha activity
KR20080059467A (en) 1996-12-03 2008-06-27 아브게닉스, 인크. Transgenic mammals having human ig loci including plural vh and vk regions and antibodies produced therefrom
US6235883B1 (en) 1997-05-05 2001-05-22 Abgenix, Inc. Human monoclonal antibodies to epidermal growth factor receptor
US7193128B2 (en) * 1997-06-03 2007-03-20 Chromatin, Inc. Methods for generating or increasing revenues from crops
US7119250B2 (en) * 1997-06-03 2006-10-10 The University Of Chicago Plant centromere compositions
US7235716B2 (en) 1997-06-03 2007-06-26 Chromatin, Inc. Plant centromere compositions
US6900012B1 (en) * 1997-06-03 2005-05-31 The University Of Chicago Plant artificial chromosome compositions and methods
US7227057B2 (en) 1997-06-03 2007-06-05 Chromatin, Inc. Plant centromere compositions
GB9823930D0 (en) * 1998-11-03 1998-12-30 Babraham Inst Murine expression of human ig\ locus
EE05627B1 (en) * 1998-12-23 2013-02-15 Pfizer Inc. Human monoclonal antibodies to CTLA-4
US7109003B2 (en) * 1998-12-23 2006-09-19 Abgenix, Inc. Methods for expressing and recovering human monoclonal antibodies to CTLA-4
US7989202B1 (en) 1999-03-18 2011-08-02 The University Of Chicago Plant centromere compositions
US6914128B1 (en) 1999-03-25 2005-07-05 Abbott Gmbh & Co. Kg Human antibodies that bind human IL-12 and methods for producing
ES2529706T3 (en) 1999-06-01 2015-02-24 Biogen Idec Ma Inc. A blocking monoclonal antibody against the alpha1-I domain of VLA-1, and its use for the treatment of inflammatory disorders
CA2589418A1 (en) 1999-08-24 2001-03-01 Medarex, Inc. Human ctla-4 antibodies and their uses
US7605238B2 (en) 1999-08-24 2009-10-20 Medarex, Inc. Human CTLA-4 antibodies and their uses
US6686188B2 (en) 2000-05-26 2004-02-03 Amersham Plc Polynucleotide encoding a human myosin-like polypeptide expressed predominantly in heart and muscle
US6656700B2 (en) 2000-05-26 2003-12-02 Amersham Plc Isoforms of human pregnancy-associated protein-E
IL136459A0 (en) * 2000-05-30 2001-06-14 Galim Galil Immunology Ltd Antibody library
US6984522B2 (en) 2000-08-03 2006-01-10 Regents Of The University Of Michigan Isolation and use of solid tumor stem cells
PT1313850E (en) 2000-08-28 2008-11-18 Agensys Inc Nucleic acid and corresponding protein entitled 85p1b3 useful in treatment and detection of cancer
US8178304B2 (en) * 2000-10-06 2012-05-15 Smith Terry J Diagnostic methods relating to Graves' disease and other autoimmune disorders
US8153121B2 (en) 2000-10-06 2012-04-10 Los Angeles Biomedical Research Institute at Harbor—UCLA Medical Center Diagnosis and therapy of antibody-mediated inflammatory autoimmune disorders
JP2004517918A (en) 2000-10-18 2004-06-17 イミュネックス・コーポレーション Treatment of rheumatoid arthritis using an IL-17 antagonist
US6596541B2 (en) 2000-10-31 2003-07-22 Regeneron Pharmaceuticals, Inc. Methods of modifying eukaryotic cells
US20050144655A1 (en) 2000-10-31 2005-06-30 Economides Aris N. Methods of modifying eukaryotic cells
US7041870B2 (en) 2000-11-30 2006-05-09 Medarex, Inc. Transgenic transchromosomal rodents for making human antibodies
US6924358B2 (en) 2001-03-05 2005-08-02 Agensys, Inc. 121P1F1: a tissue specific protein highly expressed in various cancers
US7271240B2 (en) 2001-03-14 2007-09-18 Agensys, Inc. 125P5C8: a tissue specific protein highly expressed in various cancers
WO2002076406A2 (en) * 2001-03-27 2002-10-03 Gershwin M Eric Antibodies against autoantigens of primary biliary cirrhosis and methods of making and using them
CA2443123A1 (en) 2001-04-10 2002-10-24 Agensys, Inc. Nuleic acids and corresponding proteins useful in the detection and treatment of various cancers
US20030191073A1 (en) 2001-11-07 2003-10-09 Challita-Eid Pia M. Nucleic acid and corresponding protein entitled 161P2F10B useful in treatment and detection of cancer
WO2002083854A2 (en) * 2001-04-13 2002-10-24 Biogen, Inc. Antibodies to vla-1
US20070111201A1 (en) * 2001-04-30 2007-05-17 Benjamin Doranz Reverse transfection of cell arrays for structural and functional analyses of proteins
AU2002309063B8 (en) 2001-05-11 2008-04-24 Kyowa Kirin Co., Ltd. Artificial human chromosome containing human antibody lambda light chain gene
CN1463270A (en) * 2001-05-31 2003-12-24 梅达莱克斯公司 Disulfide prodrugs and linkers and stablizers useful therefore
GB0115256D0 (en) * 2001-06-21 2001-08-15 Babraham Inst Mouse light chain locus
US20040078837A1 (en) * 2001-08-02 2004-04-22 Shannon Mark E. Four human zinc-finger-containing proteins: MDZ3, MDZ4, MDZ7 and MDZ12
AU2002336446B2 (en) 2001-09-06 2008-03-06 Agensys, Inc. Nucleic acid and corresponding protein entitled STEAP-1 useful in treatment and detection of cancer
AR039067A1 (en) * 2001-11-09 2005-02-09 Pfizer Prod Inc ANTIBODIES FOR CD40
CA2847885C (en) 2001-11-30 2022-03-22 Amgen Fremont Inc. Transgenic animals bearing human ig.lambda. light chain genes
US7491530B2 (en) * 2001-12-18 2009-02-17 Whitehead Institute For Biomedical Research Fusion partner cells and uses thereof
US6578724B1 (en) * 2001-12-29 2003-06-17 United States Can Company Connector for use in packaging aerosol containers
EP2287198A3 (en) 2002-03-13 2011-05-25 Biogen Idec MA Inc. Anti-alpha V beta 6 antibodies
WO2003076651A2 (en) * 2002-03-14 2003-09-18 Qlt Inc. Cancer associated araf1 protein kinase and its uses
SI1503794T1 (en) 2002-04-12 2012-09-28 Medarex Inc Methods of treatement using ctla-4 antibodies
US20040081653A1 (en) 2002-08-16 2004-04-29 Raitano Arthur B. Nucleic acids and corresponding proteins entitled 251P5G2 useful in treatment and detection of cancer
US7396913B2 (en) * 2002-10-14 2008-07-08 Abbott Laboratories Erythropoietin receptor binding antibodies
DE60336406D1 (en) 2002-10-16 2011-04-28 Purdue Pharma Lp ANTIBODIES BINDING TO CELL ASSOCIATED CA 125 / 0722P, AND METHOD FOR THEIR USE
EP1578799B8 (en) 2002-12-02 2011-03-23 Amgen Fremont Inc. Antibodies directed to tumor necrosis factor and uses thereof
NZ541050A (en) 2002-12-16 2010-06-25 Genmab As Human monoclonal antibodies against interleukin 8 (IL-8)
US20060135940A1 (en) * 2003-01-06 2006-06-22 The Trustees Of Columbia Programmed pulsed infusion methods and devices
US20060047261A1 (en) * 2004-06-28 2006-03-02 Shailendra Joshi Intra-arterial catheter for drug delivery
US20080014594A1 (en) * 2003-01-31 2008-01-17 Kevin Hestir Lung-Expressed Polypeptides
EP2343315A3 (en) 2003-02-10 2011-11-23 Agensys, Inc. Nucleic acid and corresponding protein named 158P1D7 useful in the treatment and detection of bladder and other cancers
US20050008618A1 (en) * 2003-02-27 2005-01-13 Howard Kaufman Composition for delivering an agent to a target cell and uses thereof
CA2519528C (en) 2003-03-19 2016-01-26 Abgenix, Inc. Antibodies against t cell immunoglobulin domain and mucin domain 1 (tim-1) antigen and uses thereof
US20040241808A1 (en) * 2003-04-14 2004-12-02 Renata Pasqualini Methods for ex vivo hybridoma-free production of polyclonal and monoclonal antibodies and generation of immortalized cell populations
CA2839032A1 (en) * 2003-05-19 2005-01-06 The Trustees Of Columbia University In The City Of New York Compositions and methods for treating and preventing heart tissue degeneration, and uses thereof
ATE541052T1 (en) 2003-05-30 2012-01-15 Agensys Inc VARIANTS OF PROSTATE STEM CELL ANTIGEN (PSCA) AND PARTIAL SEQUENCES THEREOF
WO2005003175A2 (en) 2003-06-13 2005-01-13 Biogen Idec Ma Inc. Aglycosyl anti-cd154 (cd40 ligand) antibodies and uses thereof
EP3037105B1 (en) 2003-06-27 2021-03-17 Amgen Fremont Inc. Antibodies directed to the deletion mutants of epidermal growth factor receptor and uses thereof
BRPI0412153B8 (en) 2003-07-02 2021-05-25 Innate Pharma Sas monoclonal antibody and monoclonal antibody antigen-binding fragment
BRPI0412890B8 (en) 2003-07-24 2021-05-25 Innate Pharma method of selecting an anti-kir2dl1 antibody or antigen-binding antibody fragment
HN2004000285A (en) * 2003-08-04 2006-04-27 Pfizer Prod Inc ANTIBODIES DIRECTED TO c-MET
BR0318454A (en) 2003-08-08 2006-09-12 Abgenix Inc parathyroid hormone (pth) targeting antibodies and their uses
AR045563A1 (en) 2003-09-10 2005-11-02 Warner Lambert Co ANTIBODIES DIRECTED TO M-CSF
US7741449B2 (en) 2003-12-10 2010-06-22 Medarex, Inc. Anti-interferon alpha antibodies
CN102838675B (en) 2003-12-10 2014-07-30 梅达雷克斯有限责任公司 Ip-10 antibodies and their uses
CN1950518A (en) 2004-02-03 2007-04-18 密执安州立大学董事会 Compositions and methods for characterizing, regulating, diagnosing, and treating cancer
MXPA06009225A (en) * 2004-02-23 2007-03-08 Chromatin Inc Plants modified with mini-chromosomes.
US7625549B2 (en) * 2004-03-19 2009-12-01 Amgen Fremont Inc. Determining the risk of human anti-human antibodies in transgenic mice
AU2005227313A1 (en) 2004-03-19 2005-10-06 Amgen Inc. Reducing the risk of human and anti-human antibodies through V gene manipulation
US7794713B2 (en) 2004-04-07 2010-09-14 Lpath, Inc. Compositions and methods for the treatment and prevention of hyperproliferative diseases
US20050227289A1 (en) 2004-04-09 2005-10-13 Reilly Edward B Antibodies to erythropoietin receptor and uses thereof
US20080038265A1 (en) * 2004-04-09 2008-02-14 Reilly Edward B Antibodies to Erythropoietin Receptor and Uses Thereof
US20060018902A1 (en) * 2004-04-09 2006-01-26 Reilly Edward B Antibodies to erythropoietin receptor and uses thereof
US7691962B2 (en) 2004-05-19 2010-04-06 Medarex, Inc. Chemical linkers and conjugates thereof
NZ550934A (en) 2004-05-19 2010-05-28 Medarex Inc Chemical linkers and conjugates thereof
EP2583981A3 (en) 2004-05-28 2013-07-31 Agensys, Inc. Antibodies and related molecules that bind to PSCA proteins
US20050281782A1 (en) * 2004-06-21 2005-12-22 Howard Kaufman Novel recombinant poxvirus composition and uses thereof
CA2570823C (en) 2004-06-21 2015-02-24 Medarex, Inc. Interferon alpha receptor 1 antibodies and their uses
CA2601417C (en) 2004-07-01 2018-10-30 Novo Nordisk A/S Human anti-kir antibodies
EP2311874B1 (en) 2004-07-22 2017-05-31 Erasmus University Medical Center Rotterdam Binding molecules
KR20070047327A (en) 2004-07-26 2007-05-04 비오겐 아이덱 엠에이 아이엔씨. Anti-cd154 antibodies
AU2005270918B2 (en) 2004-08-03 2011-03-03 Innate Pharma S.A. Therapeutic and diagnostic methods and compositions targeting 4Ig-B7-H3 and its counterpart NK cell receptor
US7709611B2 (en) 2004-08-04 2010-05-04 Amgen Inc. Antibodies to Dkk-1
US7423128B2 (en) * 2004-11-03 2008-09-09 Amgen Fremont Inc. Anti-properdin antibodies, and methods for making and using same
DE602005025525D1 (en) 2004-11-17 2011-02-03 Amgen Inc COMPLETE HUMAN MONOCLONAL ANTIBODIES AGAINST IL-13
DE602005022928D1 (en) 2004-11-30 2010-09-23 Abgenix Inc ANTIBODIES AGAINST GPNMB AND ITS USES
EP1838737B1 (en) 2004-12-20 2011-04-06 Amgen Fremont Inc. Binding proteins specific for human matriptase
AU2005319382B2 (en) 2004-12-21 2011-04-07 Astrazeneca Ab Antibodies directed to angiopoietin-2 and uses thereof
HUE026107T2 (en) 2004-12-28 2016-05-30 Innate Pharma Monoclonal antibodies against NKG2A
PT1836225E (en) 2005-01-06 2012-01-10 Novo Nordisk As Kir-binding agents and methods of use thereof
EP1851245B1 (en) 2005-01-26 2012-10-10 Amgen Fremont Inc. Antibodies against interleukin-1 beta
BRPI0608855A2 (en) 2005-03-08 2010-02-02 Pharmacia & Upjohn Co Llc anti - madcam antibody compositions process for their preparation and use of anti - madcam antibody
WO2007059082A1 (en) 2005-11-10 2007-05-24 Curagen Corporation Method of treating ovarian and renal cancer using antibodies against t cell immunoglobulin domain and mucin domain 1 (tim-1) antigen
WO2006104677A2 (en) 2005-03-24 2006-10-05 Millennium Pharmaceuticals, Inc. Antibodies that bind ov064 and methods of use therefor
AU2006230563B8 (en) 2005-03-31 2010-06-17 Agensys, Inc. Antibodies and related molecules that bind to 161P2F10B proteins
US7714016B2 (en) 2005-04-08 2010-05-11 Medarex, Inc. Cytotoxic compounds and conjugates with cleavable substrates
EP1874820A4 (en) * 2005-04-20 2010-09-22 Amgen Fremont Inc High affinity fully human monoclonal antibodies to interleukin-8 and epitopes for such antibodies
DOP2006000093A (en) * 2005-04-25 2007-01-31 Pfizer ANTIBODIES AGAINST MYOSTATIN
NZ562234A (en) 2005-04-26 2009-09-25 Pfizer P-cadherin antibodies
DK2161336T4 (en) 2005-05-09 2017-04-24 Ono Pharmaceutical Co Human monoclonal antibodies for programmed death 1 (PD-1) and methods for treating cancer using anti-PD-1 antibodies alone or in combination with other immunotherapies
AU2006265108C1 (en) 2005-07-01 2013-01-17 E. R. Squibb & Sons, L.L.C. Human monoclonal antibodies to programmed death ligand 1 (PD-L1)
CN102875681A (en) 2005-07-08 2013-01-16 拜奥根Idec马萨诸塞公司 Anti-alpha v beta 6 antibodies and uses thereof
US8642330B2 (en) * 2005-08-08 2014-02-04 Onconon, Llc Antibody compositions, methods for treating neoplastic disease and methods for regulating fertility
HUE025608T2 (en) * 2005-09-07 2016-03-29 Amgen Fremont Inc Human monoclonal antibodies to activin receptor-like kinase-1 (ALK-1)
EP1929019A2 (en) * 2005-09-08 2008-06-11 Chromatin, Inc. Plants modified with mini-chromosomes
US8221740B2 (en) * 2005-09-26 2012-07-17 The Trustees Of Columbia University In The City Of New York Side population cells in cardiac repair
EP1934261B1 (en) 2005-09-26 2014-10-29 Medarex, L.L.C. Human monoclonal antibodies to cd70
AU2006294554B2 (en) 2005-09-26 2013-03-21 E. R. Squibb & Sons, L.L.C. Antibody-drug conjugates and methods of use
AU2006301163B2 (en) 2005-10-14 2012-02-23 Innate Pharma Compositions and methods for treating proliferative disorders
JP5006330B2 (en) 2005-10-21 2012-08-22 ノバルティス アーゲー Human antibodies against IL13 and therapeutic uses
KR20080068084A (en) 2005-10-26 2008-07-22 메다렉스, 인코포레이티드 Methods and compounds for preparing cc-1065 analogs
US20070099246A1 (en) * 2005-11-03 2007-05-03 Sandy John D Antibodies, assays and kits to quantitate cartilage destruction
WO2007059404A2 (en) 2005-11-10 2007-05-24 Medarex, Inc. Duocarmycin derivatives as novel cytotoxic compounds and conjugates
EP1954311A4 (en) 2005-12-07 2009-12-23 Medarex Inc Ctla-4 antibody dosage escalation regimens
CN103204931B (en) 2005-12-08 2016-08-03 米德列斯公司 The human monoclonal antibodies of anti-fucosyl-GM 1 and the method for use anti-fucosyl-GM 1 antibody
US20070134249A1 (en) * 2005-12-08 2007-06-14 Genitope Corporation Combination therapy and antibody panels
RS52357B (en) 2005-12-13 2012-12-31 Medimmune Limited Binding proteins specific for insulin-like growth factors and uses thereof
AR056857A1 (en) 2005-12-30 2007-10-24 U3 Pharma Ag DIRECTED ANTIBODIES TO HER-3 (RECEIVER OF THE HUMAN EPIDERMAL GROWTH FACTOR-3) AND ITS USES
GB0603683D0 (en) 2006-02-23 2006-04-05 Novartis Ag Organic compounds
CN101415730B (en) 2006-03-30 2013-04-10 诺瓦提斯公司 Compositions and methods of use for antibodies of c-Met
EP2003960B1 (en) 2006-03-31 2015-06-10 E. R. Squibb & Sons, L.L.C. Transgenic animals expressing chimeric antibodies for use in preparing human antibodies
RU2473565C2 (en) 2006-04-07 2013-01-27 Аерпио Терапетикс, Инк. HUMAN PROTEIN TYROSINE PHOSPHATASE BETA BINDING ANTIBODIES (HPTPβ), AND USE THEREOF
CA2648846A1 (en) * 2006-04-07 2007-10-18 The Government Of The United States Of America As Represented By The Sec Retary, Department Of Health And Human Services Antibody compositions and methods for treatment of neoplastic disease
TW200813091A (en) 2006-04-10 2008-03-16 Amgen Fremont Inc Targeted binding agents directed to uPAR and uses thereof
DK2034830T3 (en) 2006-05-25 2014-10-27 Biogen Idec Inc ANTI-VLA-1 ANTIBODY FOR TREATMENT OF CASES
US7862812B2 (en) 2006-05-31 2011-01-04 Lpath, Inc. Methods for decreasing immune response and treating immune conditions
US7927590B2 (en) 2006-07-10 2011-04-19 Biogen Idec Ma Inc. Compositions and methods for inhibiting growth of smad4-deficient cancers
CL2007002225A1 (en) 2006-08-03 2008-04-18 Astrazeneca Ab SPECIFIC UNION AGENT FOR A RECEIVER OF THE GROWTH FACTOR DERIVED FROM PLATES (PDGFR-ALFA); NUCLEIC ACID MOLECULA THAT CODIFIES IT; VECTOR AND CELL GUESTS THAT UNDERSTAND IT; CONJUGADO UNDERSTANDING THE AGENT; AND USE OF THE AGENT OF A
ES2613957T3 (en) 2006-08-04 2017-05-29 Medimmune Limited Antibodies against ERBB2
PL2056858T3 (en) 2006-08-11 2015-01-30 Csl Ltd Treatment of pulmonary disease conditions
TW200817437A (en) 2006-08-11 2008-04-16 Medarex Inc Monoclonal antibodies against stromal cell derived factor-1 (SDF-1)
EP2074144A4 (en) 2006-09-05 2011-03-16 Medarex Inc Antibodies to bone morphogenic proteins and receptors therefor and methods for their use
PL2486941T3 (en) 2006-10-02 2017-08-31 E. R. Squibb & Sons, L.L.C. Human antibodies that bind CXCR4 and uses thereof
US7833527B2 (en) 2006-10-02 2010-11-16 Amgen Inc. Methods of treating psoriasis using IL-17 Receptor A antibodies
US8613925B2 (en) 2006-10-19 2013-12-24 Csl Limited Anti-IL-13Rα1 antibodies and their uses thereof
US8642031B2 (en) 2006-11-02 2014-02-04 Acceleron Pharma, Inc. Antagonists of BMP9, BMP10, ALK1 and other ALK1 ligands, and uses thereof
CN111518218A (en) * 2006-11-02 2020-08-11 阿塞勒隆制药公司 ALK1 receptor and ligand antagonists and uses thereof
US10059756B2 (en) 2006-11-02 2018-08-28 Acceleron Pharma Inc. Compositions comprising ALK1-ECD protein
MX2009005189A (en) 2006-11-15 2009-06-30 Medarex Inc Human monoclonal antibodies to btla and methods of use.
TWI447124B (en) 2006-12-01 2014-08-01 Medarex Llc Human antibodies that bind cd22 and uses thereof
CL2007003622A1 (en) 2006-12-13 2009-08-07 Medarex Inc Human anti-cd19 monoclonal antibody; composition comprising it; and tumor cell growth inhibition method.
KR20090088946A (en) 2006-12-14 2009-08-20 메다렉스, 인코포레이티드 Human antibodies that bind cd70 and uses thereof
TWI412367B (en) 2006-12-28 2013-10-21 Medarex Llc Chemical linkers and cleavable substrates and conjugates thereof
US7776331B1 (en) * 2007-01-16 2010-08-17 Abbott Laboratories Methods of treating plaque psoriasis
SG178789A1 (en) 2007-02-16 2012-03-29 Merrimack Pharmaceuticals Inc Antibodies against erbb3 and uses thereof
CN101616911A (en) 2007-02-21 2009-12-30 梅达莱克斯公司 Chemical linkers and conjugate thereof with single amino acids
EP2134855A4 (en) 2007-03-12 2011-01-05 Dana Farber Cancer Inst Inc Prognostic, diagnostic, and cancer therapeutic uses of fanci and fanci modulating agents
US8614089B2 (en) 2007-03-15 2013-12-24 Chromatin, Inc. Centromere sequences and minichromosomes
UY30994A1 (en) * 2007-04-02 2008-11-28 Amgen Fremont Inc ANTI-IGE ANTIBODIES
DK2336329T3 (en) * 2007-06-01 2013-01-07 Omt Inc COMPOSITIONS AND PROCEDURES FOR INHIBITING ENDOGENIC IMMUNGLOBULATIONS AND FOR PRODUCING TRANSGENE, HUMAN, IDIOTYPIC ANTIBODIES
US20080317768A1 (en) 2007-06-21 2008-12-25 Boeing Company Bioconjugated nanoparticles
CN101802211B (en) 2007-08-02 2014-10-01 诺维莫尼公司 Anti-RANTES antibodies and methods of use thereof
US9693539B2 (en) 2007-08-10 2017-07-04 E. R. Squibb & Sons, L.L.C. HCO32 and HCO27 and related examples
EP3330292A1 (en) 2007-08-21 2018-06-06 Amgen, Inc Human c-fms antigen binding proteins
WO2009026274A1 (en) 2007-08-22 2009-02-26 Medarex, Inc. Site-specific attachment of drugs or other agents to engineered antibodies with c-terminal extensions
EP2615114B1 (en) 2007-08-23 2022-04-06 Amgen Inc. Antigen binding proteins to proprotein convertase subtilisin kexin type 9 (PCSK9)
JOP20080381B1 (en) 2007-08-23 2023-03-28 Amgen Inc Antigen Binding Proteins to Proprotein Convertase subtillisin Kexin type 9 (pcsk9)
DK2769729T3 (en) 2007-09-04 2019-04-23 Compugen Ltd POLYPEPTIDES AND POLYNUCLEOTIDES AND APPLICATIONS THEREOF AS A PHARMACEUTICAL OBJECTIVE FOR THE PRODUCTION OF MEDICINAL PRODUCTS AND BIOTECHNOLOGICAL PRODUCTS
US7982016B2 (en) 2007-09-10 2011-07-19 Amgen Inc. Antigen binding proteins capable of binding thymic stromal lymphopoietin
TW200918553A (en) 2007-09-18 2009-05-01 Amgen Inc Human GM-CSF antigen binding proteins
US8828382B2 (en) 2007-09-26 2014-09-09 U3 Pharma Gmbh Heparin-binding epidermal growth factor-like growth factor binding proteins
TWI489993B (en) 2007-10-12 2015-07-01 Novartis Ag Compositions and methods of use for antibodies against sclerostin
EP2214706A1 (en) 2007-10-23 2010-08-11 Novartis AG Use of trkb antibodies for the treatment of respiratory disorders
EP2567709B1 (en) 2007-11-02 2017-12-27 Novartis AG Molecules and methods for modulating low-density-lipoprotein receptor-related protein 6 (LRP6)
SG10201405835TA (en) 2007-11-12 2014-10-30 U3 Pharma Gmbh Axl antibodies
EP2220247A4 (en) 2007-11-16 2011-10-26 Nuvelo Inc Antibodies to lrp6
CN110698561A (en) 2007-12-14 2020-01-17 诺沃—诺迪斯克有限公司 Anti-human NKG2D antibodies and uses thereof
SI2242771T1 (en) 2007-12-14 2013-09-30 Bristol-Myers Squibb Company Binding molecules to the human ox40 receptor
ES2618292T3 (en) 2008-01-31 2017-06-21 Inserm - Institut National De La Sante Et De La Recherche Medicale Antibodies against and use of human CD39 to inhibit the activity of regulatory T cells
BRPI0906387A2 (en) 2008-02-05 2015-07-07 Bristol Myers Squibb Co Alpha 5 - beta 1 antibodies and their uses
KR20100126515A (en) 2008-03-18 2010-12-01 아보트 러보러터리즈 Methods for treating psoriasis
CN105963695A (en) 2008-05-02 2016-09-28 阿塞勒隆制药公司 Methods and compositions based on ALK1 antagonists for modulating angiogenesis and pericyte coverage
MY159396A (en) 2008-08-05 2016-12-30 Novartis Ag Compositions and methods for antibodies targeting complement protein c5
AR072999A1 (en) 2008-08-11 2010-10-06 Medarex Inc HUMAN ANTIBODIES THAT JOIN GEN 3 OF LYMPHOCYTARY ACTIVATION (LAG-3) AND THE USES OF THESE
RU2547595C2 (en) 2008-08-18 2015-04-10 Пфайзер Инк Anti-ccr2 antibodies
DK2334703T3 (en) 2008-09-17 2015-10-05 Innate Pharma Configurations and methods for detection of tlr3
WO2010032060A1 (en) 2008-09-19 2010-03-25 Medimmune Llc Antibodies directed to dll4 and uses thereof
KR101826224B1 (en) 2008-09-30 2018-02-06 아블렉시스, 엘엘씨 Non-human mammals for the production of chimeric antibodies
NZ606283A (en) * 2008-11-28 2014-08-29 Abbvie Inc Stable antibody compositions and methods for stabilizing same
EP2865689A1 (en) 2008-12-08 2015-04-29 Compugen Ltd. FAM26F polypeptides and polynucleotides, and uses thereof as a drug target for producing drugs and biologics
CA2747534C (en) * 2008-12-18 2020-08-18 Erasmus University Medical Center Rotterdam Non-human transgenic animals expressing humanised antibodies and use therof
JP2012513194A (en) 2008-12-23 2012-06-14 アストラゼネカ アクチボラグ Targeted binding agents directed to α5β1 and uses thereof
JO3382B1 (en) 2008-12-23 2019-03-13 Amgen Inc Human cgrp receptor binding antibodies
WO2010081890A1 (en) 2009-01-19 2010-07-22 Innate Pharma Anti-kir3d antibodies
MX2011009220A (en) 2009-03-05 2011-09-28 Medarex Inc Fully human antibodies specific to cadm1.
GB0905023D0 (en) 2009-03-24 2009-05-06 Univ Erasmus Medical Ct Binding molecules
WO2010117325A1 (en) 2009-04-08 2010-10-14 Olle Hernell New methods for treatment of inflammatory diseases
EA030182B1 (en) 2009-04-20 2018-07-31 Оксфорд Байотерепьютикс Лтд. Antibodies specific to cadherin-17
WO2010124113A1 (en) 2009-04-23 2010-10-28 Infinity Pharmaceuticals, Inc. Anti-fatty acid amide hydrolase-2 antibodies and uses thereof
EP2424894A1 (en) 2009-04-27 2012-03-07 Novartis AG Composition and methods of use for therapeutic antibodies specific for the il-12 receptore betal subunit
EP3275900A1 (en) 2009-04-27 2018-01-31 Novartis AG Compositions and methods for increasing muscle growth
ES2514319T3 (en) 2009-05-04 2014-10-28 Abbvie Research B.V. Antibodies against nerve growth factor (NGF) with improved in vivo stability
AU2010244142B2 (en) 2009-05-05 2016-07-21 Novimmune Sa Anti-IL-17F antibodies and methods of use thereof
EP2445925A1 (en) 2009-06-25 2012-05-02 Bristol-Myers Squibb Company Protein purification by caprylic acid (octanoic acid) precipitation
DK2564695T3 (en) 2009-07-08 2015-05-26 Kymab Ltd Animal models and therapeutic molecules
US9445581B2 (en) 2012-03-28 2016-09-20 Kymab Limited Animal models and therapeutic molecules
US20120204278A1 (en) 2009-07-08 2012-08-09 Kymab Limited Animal models and therapeutic molecules
CN102471382A (en) 2009-07-10 2012-05-23 依奈特制药公司 TLR3 binding agents
US9096909B2 (en) 2009-07-23 2015-08-04 Chromatin, Inc. Sorghum centromere sequences and minichromosomes
US8809285B2 (en) 2009-07-31 2014-08-19 Wayne State University Monophosphorylated lipid A derivatives
US9259476B2 (en) 2009-07-31 2016-02-16 Wayne State University Monophosphorylated lipid A derivatives
WO2011019844A1 (en) 2009-08-13 2011-02-17 Crystal Bioscience Inc. Transgenic animal for production of antibodies having minimal cdrs
WO2011021146A1 (en) 2009-08-20 2011-02-24 Pfizer Inc. Osteopontin antibodies
US20110059111A1 (en) 2009-09-01 2011-03-10 Los Angeles Biomedical Research Institute At Harbor-Ucla Medical Center Mammalian receptors as targets for antibody and active vaccination therapy against mold infections
WO2011029823A1 (en) 2009-09-09 2011-03-17 Novartis Ag Monoclonal antibody reactive with cd63 when expressed at the surface of degranulated mast cells
TW201127400A (en) * 2009-09-14 2011-08-16 Abbott Lab Methods for treating psoriasis
CA3057650A1 (en) 2009-09-15 2011-03-24 Irving L. Weissman Synergistic anti-cd47 therapy for hematologic cancers
CN102630167B (en) 2009-09-15 2015-06-10 Csl有限公司 Treatment of neurological conditions
US20120178910A1 (en) 2009-09-23 2012-07-12 Medarex, Inc. Cation exchange chromatography (methods)
TW201117824A (en) 2009-10-12 2011-06-01 Amgen Inc Use of IL-17 receptor a antigen binding proteins
US20120231004A1 (en) 2009-10-13 2012-09-13 Oxford Biotherapeutic Ltd. Antibodies
US8785600B2 (en) 2009-10-23 2014-07-22 Millennium Pharmaceuticals, Inc. Anti-GCC antibody molecules and related compositions and methods
JO3244B1 (en) 2009-10-26 2018-03-08 Amgen Inc Human il-23 antigen binding proteins
CN105999263B (en) 2009-11-13 2021-06-29 第一三共欧洲有限公司 Materials and methods for treating or preventing human epidermal growth factor receptor-3 (HER-3) related diseases
NO2501822T3 (en) 2009-11-17 2018-01-13
NZ628923A (en) 2009-11-24 2016-02-26 Medimmune Ltd Targeted binding agents against b7-h1
US9428586B2 (en) 2009-12-01 2016-08-30 Compugen Ltd Heparanase splice variant
UA109888C2 (en) 2009-12-07 2015-10-26 ANTIBODY OR ANTIBODILITY ANTIBODY OR ITS BINDING TO THE β-CLOTE, FGF RECEPTORS AND THEIR COMPLEXES
EP3295957B1 (en) 2010-01-15 2019-08-07 Kirin-Amgen, Inc. Anti il-17ra antibody formulation and therapeutic regimens for treating psoriasis
US8865462B2 (en) * 2010-01-20 2014-10-21 Crystal Bioscience Inc. Sustained culture of avian gonocytes
US20130045492A1 (en) 2010-02-08 2013-02-21 Regeneron Pharmaceuticals, Inc. Methods For Making Fully Human Bispecific Antibodies Using A Common Light Chain
US9796788B2 (en) 2010-02-08 2017-10-24 Regeneron Pharmaceuticals, Inc. Mice expressing a limited immunoglobulin light chain repertoire
WO2011097603A1 (en) 2010-02-08 2011-08-11 Regeneron Pharmaceuticals, Inc. Common light chain mouse
MX360640B (en) 2010-03-01 2018-11-09 Tau Therapeutics Llc Star Cancer diagnosis and imaging.
NZ602054A (en) 2010-03-17 2014-10-31 Abbott Res Bv Anti-nerve growth factor (ngf) antibody compositions
US20150231215A1 (en) 2012-06-22 2015-08-20 Randolph J. Noelle VISTA Antagonist and Methods of Use
CN103119054B (en) 2010-03-26 2017-01-18 达特茅斯大学理事会 VISTA regulatory T cell mediator protein, VISTA binding agents and use thereof
US10745467B2 (en) 2010-03-26 2020-08-18 The Trustees Of Dartmouth College VISTA-Ig for treatment of autoimmune, allergic and inflammatory disorders
EP3248462B1 (en) 2010-03-31 2024-03-13 Ablexis, LLC Genetic engineering of mice for the production of chimeric antibodies
EP2558497A2 (en) 2010-04-15 2013-02-20 Amgen Inc. Human fgf receptor and beta-klotho binding proteins
WO2011140254A1 (en) 2010-05-04 2011-11-10 Adimab, Llc Antibodies against epidermal growth factor receptor (egfr) and uses thereof
WO2011140151A1 (en) 2010-05-04 2011-11-10 Dyax Corp. Antibodies against epidermal growth factor receptor (egfr)
SI2566517T1 (en) 2010-05-04 2019-01-31 Five Prime Therapeutics, Inc. Antibodies that bind csf1r
SG185416A1 (en) 2010-05-06 2012-12-28 Novartis Ag Compositions and methods of use for therapeutic low density lipoprotein -related protein 6 (lrp6) antibodies
CA2798390A1 (en) 2010-05-06 2011-11-10 Novartis Ag Compositions and methods of use for therapeutic low density lipoprotein - related protein 6 (lrp6) multivalent antibodies
CN104404050B (en) 2010-06-22 2018-06-08 瑞泽恩制药公司 hybrid light chain mice
WO2011163401A2 (en) 2010-06-22 2011-12-29 Neogenix Oncology, Inc. Colon and pancreas cancer specific antigens and antibodies
US10793829B2 (en) 2010-07-26 2020-10-06 Trianni, Inc. Transgenic mammals and methods of use thereof
EP2597945B1 (en) 2010-07-26 2020-07-22 Trianni, Inc. Transgenic animals and methods of use
US10662256B2 (en) 2010-07-26 2020-05-26 Trianni, Inc. Transgenic mammals and methods of use thereof
SG10201912639SA (en) 2010-08-02 2020-02-27 Regeneron Pharma Mice that make binding proteins comprising vl domains
AU2011286407A1 (en) 2010-08-06 2013-02-21 Amgen Use of HER3 binding agents in prostate treatment
CN103080134B (en) 2010-08-20 2015-11-25 诺华股份有限公司 The antibody of EGF-R ELISA 3 (HER3)
WO2012028703A1 (en) 2010-09-02 2012-03-08 INSERM (Institut National de la Santé et de la Recherche Médicale) Method for the prognosis of the progression of cancer
US8999335B2 (en) 2010-09-17 2015-04-07 Compugen Ltd. Compositions and methods for treatment of drug resistant multiple myeloma
CA3182320A1 (en) 2010-09-23 2012-03-29 Precision Biologics, Inc. Colon and pancreas cancer peptidomimetics
JP2013543384A (en) 2010-10-05 2013-12-05 ノバルティス アーゲー Anti-IL12Rbeta1 antibody and its use in the treatment of autoimmune and inflammatory diseases
CA2815181C (en) 2010-10-27 2020-09-15 William Gleason Richards Dkk1 antibodies and methods of use
EP2638155A1 (en) 2010-11-08 2013-09-18 Kymab Limited Cells & vertebrates for enhanced somatic hypermutation and class switch recombination
EP2643353A1 (en) 2010-11-24 2013-10-02 Novartis AG Multispecific molecules
WO2012075333A2 (en) 2010-12-02 2012-06-07 Prometheus Laboratories Inc. Her2delta16 peptides
CA2824313A1 (en) 2011-01-12 2012-07-19 Innate Pharma Tlr3 binding agents
AU2012205384B2 (en) 2011-01-14 2015-09-10 Five Prime Therapeutics, Inc. IL-27 antagonists for treating inflammatory diseases
SG10201600531TA (en) 2011-01-24 2016-02-26 Univ Singapore Pathogenic mycobacteria-derived mannose-capped lipoarabinomannan antigen binding proteins
ME02106B (en) 2011-02-25 2015-10-20 Regeneron Pharma Adam6 mice
EA201391248A1 (en) 2011-03-01 2014-05-30 Эмджен Инк. BISPECIFIC BINDING AGENTS
AU2012239997A1 (en) 2011-04-07 2013-10-17 Amgen Inc. Novel EGFR binding proteins
US9150644B2 (en) 2011-04-12 2015-10-06 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Human monoclonal antibodies that bind insulin-like growth factor (IGF) I and II
US9409987B2 (en) 2011-04-15 2016-08-09 Compugen Ltd Polypeptides and polynucleotides, and uses thereof for treatment of immune related disorders and cancer
EP2702077A2 (en) 2011-04-27 2014-03-05 AbbVie Inc. Methods for controlling the galactosylation profile of recombinantly-expressed proteins
JOP20200043A1 (en) 2011-05-10 2017-06-16 Amgen Inc Methods of treating or preventing cholesterol related disorders
CN107903325B (en) 2011-05-16 2021-10-29 埃泰美德(香港)有限公司 Multispecific FAB fusion proteins and methods of use thereof
KR102046666B1 (en) 2011-05-25 2019-11-19 이나뜨 파르마 Anti-kir antibodies for the treatment of inflammatory disorders
US8691231B2 (en) 2011-06-03 2014-04-08 Merrimack Pharmaceuticals, Inc. Methods of treatment of tumors expressing predominantly high affinity EGFR ligands or tumors expressing predominantly low affinity EGFR ligands with monoclonal and oligoclonal anti-EGFR antibodies
US9574002B2 (en) 2011-06-06 2017-02-21 Amgen Inc. Human antigen binding proteins that bind to a complex comprising β-Klotho and an FGF receptor
WO2012172495A1 (en) 2011-06-14 2012-12-20 Novartis Ag Compositions and methods for antibodies targeting tem8
EP2721058A2 (en) 2011-06-17 2014-04-23 Amgen Inc. Method of treating or ameliorating metabolic disorders using clec-2
EP2723362A1 (en) 2011-06-21 2014-04-30 Innate Pharma NKp46-MEDIATED NK CELL TUNING
JP6247209B2 (en) 2011-06-28 2017-12-13 オックスフォード ビオトヘラペウトイクス エルティーディー. Antibody to ADP ribosyl cyclase 2
AU2012277376B2 (en) 2011-06-30 2016-11-24 Compugen Ltd. Polypeptides and uses thereof for treatment of autoimmune disorders and infection
PL2726099T3 (en) 2011-07-01 2018-12-31 Novartis Ag Method for treating metabolic disorders
EP2731970B1 (en) 2011-07-15 2018-11-28 MorphoSys AG Antibodies that are cross-reactive for macrophage migration inhibitory factor (mif) and d-dopachrome tautomerase (d-dt)
WO2013016220A1 (en) 2011-07-22 2013-01-31 Amgen Inc. Il-17 receptor a is required for il-17c biology
IL273982B2 (en) 2011-08-05 2023-03-01 Regeneron Pharma Humanized universal light chain mice
WO2013022091A1 (en) 2011-08-11 2013-02-14 小野薬品工業株式会社 Therapeutic agent for autoimmune diseases comprising pd-1 agonist
AU2012309800B2 (en) 2011-09-12 2017-10-05 Cavion, Inc. Antagonists of products of the Hs.459642 unigene cluster for the inhibition of proliferation, development or differentiation of stem cells including cancer stem cells
CN107056936B (en) 2011-09-19 2021-09-03 科马布有限公司 Manipulation of immunoglobulin gene diversity and multi-antibody therapeutics
WO2013041845A2 (en) 2011-09-19 2013-03-28 Kymab Limited Animals, repertoires & methods
US8987422B2 (en) 2011-09-22 2015-03-24 Amgen Inc. CD27L antigen binding proteins
WO2013045916A1 (en) 2011-09-26 2013-04-04 Kymab Limited Chimaeric surrogate light chains (slc) comprising human vpreb
EP2766044B1 (en) 2011-10-13 2019-12-11 Aerpio Therapeutics, Inc. Treatment of ocular disease
WO2013056240A1 (en) 2011-10-13 2013-04-18 Aerpio Therapeutics, Inc. Methods for treating vascular leak syndrome and cancer
EP2581388A1 (en) 2011-10-14 2013-04-17 Centre National de la Recherche Scientifique (CNRS) Anti-sPLA2-V antibodies and uses thereof
CN104080471B (en) 2011-10-14 2018-08-10 诺华股份有限公司 Antibody and method for Wnt approach relevant diseases
PT3216871T (en) 2011-10-17 2022-03-15 Regeneron Pharma Restricted immunoglobulin heavy chain mice
GB2496375A (en) 2011-10-28 2013-05-15 Kymab Ltd A non-human assay vertebrate comprising human antibody loci and human epitope knock-in, and uses thereof
US9827246B2 (en) 2011-10-28 2017-11-28 Millennium Pharmaceuticals, Inc. Biomarkers of response to NAE inhibitors
CN104271599A (en) 2011-11-08 2015-01-07 辉瑞公司 Methods of treating inflammatory disorders using anti-M-CSF antibodies
EP2776043B1 (en) 2011-11-11 2018-02-21 Millennium Pharmaceuticals, Inc. Biomarkers of response to proteasome inhibitors
CN104126017A (en) 2011-11-11 2014-10-29 米伦纽姆医药公司 Biomarkers of response to proteasome inhibitors
PE20141937A1 (en) 2011-11-16 2014-12-18 Amgen Inc METHODS TO TREAT DISORDERS RELATED TO MUTANT VIII OF ELIMINATION OF EPIDERMAL GROWTH FACTOR
US9782452B2 (en) 2011-11-22 2017-10-10 Cornell University Methods for stimulating hematopoietic recovery by inhibiting TGFβ signaling
GB201122047D0 (en) 2011-12-21 2012-02-01 Kymab Ltd Transgenic animals
US9253965B2 (en) 2012-03-28 2016-02-09 Kymab Limited Animal models and therapeutic molecules
SG11201402739YA (en) 2011-12-05 2014-06-27 Novartis Ag Antibodies for epidermal growth factor receptor 3 (her3) directed to domain ii of her3
US20130251703A1 (en) 2011-12-05 2013-09-26 Novartis Ag Antibodies for epidermal growth factor receptor 3 (her3) directed to domain iii and domain iv of her3
EP2602265A1 (en) 2011-12-07 2013-06-12 Centre National de la Recherche Scientifique (CNRS) Antibodies anti-sPLA2-X and uses thereof
JP6635655B2 (en) 2011-12-08 2020-01-29 アムジエン・インコーポレーテツド Human LCAT antigen binding proteins and their use in therapy
SG10201913164YA (en) 2011-12-20 2020-03-30 Regeneron Pharma Humanized light chain mice
EP2794656B1 (en) 2011-12-21 2019-02-27 Novartis AG Compositions comprising antibodies targeting factor p and c5
AU2012356894B2 (en) 2011-12-23 2018-01-18 Innate Pharma Enzymatic conjugation of polypeptides
IN2014KN00848A (en) 2012-02-01 2015-10-02 Compugen Ltd
CN111533804A (en) 2012-02-06 2020-08-14 印希彼有限公司 CD47 antibodies and methods of use thereof
CN104271122A (en) 2012-02-16 2015-01-07 桑塔鲁斯股份有限公司 Anti-vla1 (cd49a) antibody pharmaceutical compositions
US20140013456A1 (en) 2012-03-16 2014-01-09 Regeneron Pharmaceuticals, Inc. Histidine Engineered Light Chain Antibodies and Genetically Modified Non-Human Animals for Generating the Same
RU2014141536A (en) 2012-03-16 2016-05-10 Регенерон Фармасьютикалз, Инк. MICE WHICH PRODUCE ANTIGEN-BINDING PROTEINS WITH PH-DEPENDENT BINDING CHARACTERISTICS
RS57118B1 (en) 2012-03-16 2018-06-29 Regeneron Pharma Histidine engineered light chain antibodies and genetically modified rodents for generating the same
LT2825037T (en) 2012-03-16 2019-08-12 Regeneron Pharmaceuticals, Inc. Rodents expressing ph-sensitive immunoglobulin sequences
EP2641916A1 (en) 2012-03-23 2013-09-25 Centre National de la Recherche Scientifique (C.N.R.S) Novel antibodies anti-sPLA2-IIA and uses thereof
WO2013144567A1 (en) 2012-03-28 2013-10-03 Kymab Limited Transgenic non-human vertebrate for the expression of class - switched, fully human, antibodies
GB2502127A (en) 2012-05-17 2013-11-20 Kymab Ltd Multivalent antibodies and in vivo methods for their production
US10251377B2 (en) 2012-03-28 2019-04-09 Kymab Limited Transgenic non-human vertebrate for the expression of class-switched, fully human, antibodies
EP2836514A4 (en) 2012-04-13 2015-12-30 Childrens Medical Center Tiki inhibitors
US10114023B2 (en) 2012-04-18 2018-10-30 Massachusetts Institute Of Technology Method of enhancing the efficacy of anti-hepatocyte growth factor receptor breast cancer therapy by administering an inhibitor of menaINV
WO2013158279A1 (en) 2012-04-20 2013-10-24 Abbvie Inc. Protein purification methods to reduce acidic species
US9067990B2 (en) 2013-03-14 2015-06-30 Abbvie, Inc. Protein purification using displacement chromatography
US9181572B2 (en) 2012-04-20 2015-11-10 Abbvie, Inc. Methods to modulate lysine variant distribution
PL3431492T3 (en) 2012-04-27 2021-07-05 Novo Nordisk A/S Human cd30 ligand antigen binding proteins
EA039663B1 (en) 2012-05-03 2022-02-24 Амген Инк. Use of an anti-pcsk9 antibody for lowering serum cholesterol ldl and treating cholesterol related disorders
BR112014028013A2 (en) 2012-05-11 2018-02-27 Five Prime Therapeutics Inc methods for treating a condition associated with rheumatoid arthritis, rheumatoid arthritis, skin lesions, lupus nephritis, lupus, an inflammatory condition, cd16 + disorder, method for reducing the number of cd16 + monocytes, methods for slowing the progression of a renal condition, rag and bone loss
UY34813A (en) 2012-05-18 2013-11-29 Amgen Inc ANTIGEN UNION PROTEINS DIRECTED AGAINST ST2 RECEIVER
US9249182B2 (en) 2012-05-24 2016-02-02 Abbvie, Inc. Purification of antibodies using hydrophobic interaction chromatography
EP2855527B1 (en) 2012-05-31 2018-08-01 Innate Pharma Tlr3 binding agents
CA2876172C (en) 2012-06-12 2021-01-19 Regeneron Pharmaceuticals, Inc. Humanized non-human animals with restricted immunoglobulin heavy chain loci
JP6285923B2 (en) 2012-06-22 2018-02-28 トラスティーズ・オブ・ダートマス・カレッジ Novel VISTA-Ig constructs and use of VISTA-Ig for the treatment of autoimmune, allergic and inflammatory disorders
US9890215B2 (en) 2012-06-22 2018-02-13 King's College London Vista modulators for diagnosis and treatment of cancer
SG11201408554QA (en) 2012-06-22 2015-02-27 Cytomx Therapeutics Inc Anti-jagged 1/jagged 2 cross-reactive antibodies, activatable anti-jagged antibodies and methods of use thereof
AR091649A1 (en) 2012-07-02 2015-02-18 Bristol Myers Squibb Co OPTIMIZATION OF ANTIBODIES THAT FIX THE LYMPHOCYTE ACTIVATION GEN 3 (LAG-3) AND ITS USES
WO2014009426A2 (en) 2012-07-13 2014-01-16 Innate Pharma Screening of conjugated antibodies
SG10201906328RA (en) 2012-08-31 2019-08-27 Five Prime Therapeutics Inc Methods of treating conditions with antibodies that bind colony stimulating factor 1 receptor (csf1r)
AU2013309506A1 (en) 2012-09-02 2015-03-12 Abbvie Inc. Methods to control protein heterogeneity
US9512214B2 (en) 2012-09-02 2016-12-06 Abbvie, Inc. Methods to control protein heterogeneity
AU2013312211B2 (en) 2012-09-07 2018-03-29 King's College London VISTA modulators for diagnosis and treatment of cancer
JOP20200308A1 (en) 2012-09-07 2017-06-16 Novartis Ag IL-18 binding molecules
CA2886783A1 (en) 2012-10-01 2014-04-10 Millennium Pharmaceuticals, Inc. Biomarkers and methods to predict response to inhibitors and uses thereof
AU2013342163B2 (en) 2012-11-08 2018-08-16 F. Hoffmann-La Roche Ltd IL-6 antagonists and uses thereof
EP2916872B1 (en) 2012-11-09 2019-02-27 Innate Pharma Recognition tags for tgase-mediated conjugation
ES2701076T3 (en) 2012-11-24 2019-02-20 Hangzhou Dac Biotech Co Ltd Hydrophilic linkers and their uses for the conjugation of drugs to molecules that bind to cells
WO2014084859A1 (en) 2012-11-30 2014-06-05 Novartis Ag Molecules and methods for modulating tmem16a activities
PL2928921T3 (en) 2012-12-05 2021-06-28 Novartis Ag Compositions and methods for antibodies targeting epo
TW201425336A (en) 2012-12-07 2014-07-01 Amgen Inc BCMA antigen binding proteins
WO2014099997A1 (en) 2012-12-18 2014-06-26 Novartis Ag Compositions and methods that utilize a peptide tag that binds to hyaluronan
WO2014106176A1 (en) 2012-12-28 2014-07-03 Precision Biologics, Inc. Humanized monoclonal antibodies and methods of use for the diagnosis and treatment of colon and pancreas cancer
JO3519B1 (en) 2013-01-25 2020-07-05 Amgen Inc Antibody constructs for CDH19 and CD3
WO2014114801A1 (en) 2013-01-25 2014-07-31 Amgen Inc. Antibodies targeting cdh19 for melanoma
MX2015010115A (en) 2013-02-06 2016-05-31 Inhibrx Llc Non-platelet depleting and non-red blood cell depleting cd47 antibodies and methods of use thereof.
MX371052B (en) 2013-02-08 2020-01-14 Novartis Ag Anti-il-17a antibodies and their use in treating autoimmune and inflammatory disorders.
ES2747920T3 (en) 2013-02-14 2020-03-12 Innate Pharma Anti-NKP46 antibody for diagnosis of peripheral non-cutaneous T-cell lymphoma (PTCL)
JP5993102B2 (en) 2013-02-20 2016-09-14 リジェネロン・ファーマシューティカルズ・インコーポレイテッドRegeneron Pharmaceuticals, Inc. Non-human animals having modified immunoglobulin heavy chain sequences
CN105026427B (en) 2013-02-20 2019-12-24 依奈特制药公司 Compounds that specifically bind to KIR3DL2 for use in the treatment of peripheral T cell lymphoma
WO2014143205A1 (en) 2013-03-12 2014-09-18 Abbvie Inc. Human antibodies that bind human tnf-alpha and methods of preparing the same
WO2014159764A1 (en) 2013-03-14 2014-10-02 Amgen Inc. Chrdl-1 antigen binding proteins and methods of treatment
EP2970443A4 (en) 2013-03-14 2017-01-11 Parkash S. Gill Cancer treatment using antibodies that bind cell surface grp78
EP3611189A1 (en) 2013-03-14 2020-02-19 Novartis AG Antibodies against notch 3
US9017687B1 (en) 2013-10-18 2015-04-28 Abbvie, Inc. Low acidic species compositions and methods for producing and using the same using displacement chromatography
WO2014159579A1 (en) 2013-03-14 2014-10-02 Abbvie Inc. MUTATED ANTI-TNFα ANTIBODIES AND METHODS OF THEIR USE
EP2967012B1 (en) 2013-03-14 2020-09-16 Erasmus University Medical Center Rotterdam Transgenic non-human mammal for antibody production
US9499614B2 (en) 2013-03-14 2016-11-22 Abbvie Inc. Methods for modulating protein glycosylation profiles of recombinant protein therapeutics using monosaccharides and oligosaccharides
HUE045859T2 (en) 2013-03-15 2020-01-28 Amgen Res Munich Gmbh Single chain binding molecules comprising n-terminal abp
WO2014144553A1 (en) 2013-03-15 2014-09-18 Amgen Inc. Secreted frizzle-related protein 5 (sfrp5) binding proteins and methods of treatment
US10993420B2 (en) 2013-03-15 2021-05-04 Erasmus University Medical Center Production of heavy chain only antibodies in transgenic mammals
US10035859B2 (en) 2013-03-15 2018-07-31 Biogen Ma Inc. Anti-alpha V beta 6 antibodies and uses thereof
US9505849B2 (en) 2013-03-15 2016-11-29 Amgen Research (Munich) Gmbh Antibody constructs for influenza M2 and CD3
WO2014140300A1 (en) 2013-03-15 2014-09-18 Innate Pharma Solid phase tgase-mediated conjugation of antibodies
WO2014143739A2 (en) 2013-03-15 2014-09-18 Biogen Idec Ma Inc. Anti-alpha v beta 6 antibodies and uses thereof
AU2014228924B2 (en) 2013-03-15 2019-04-18 Amgen Inc. Human PAC1 antibodies
US9788534B2 (en) 2013-03-18 2017-10-17 Kymab Limited Animal models and therapeutic molecules
US9783618B2 (en) 2013-05-01 2017-10-10 Kymab Limited Manipulation of immunoglobulin gene diversity and multi-antibody therapeutics
US11707056B2 (en) 2013-05-02 2023-07-25 Kymab Limited Animals, repertoires and methods
US9783593B2 (en) 2013-05-02 2017-10-10 Kymab Limited Antibodies, variable domains and chains tailored for human use
CA2922704A1 (en) 2013-05-24 2014-11-27 Neil R. Cashman Cell senescence markers as diagnostic and therapeutic targets
MX351127B (en) 2013-05-30 2017-10-03 Kiniksa Pharmaceuticals Ltd Oncostatin m receptor antigen binding proteins.
US10071169B2 (en) 2013-06-20 2018-09-11 Innate Pharma Enzymatic conjugation of polypeptides
JP6744212B2 (en) 2013-06-21 2020-08-19 イナート・ファルマ・ソシエテ・アノニムInnate Pharma Pharma S.A. Enzymatic binding of polypeptides
AR096601A1 (en) 2013-06-21 2016-01-20 Novartis Ag ANTIBODIES OF LEXINED OXIDATED LDL RECEIVER 1 AND METHODS OF USE
JP6267792B2 (en) 2013-06-28 2018-01-24 アムジエン・インコーポレーテツド Methods for treating homozygous familial hypercholesterolemia
SG11201600741SA (en) 2013-08-01 2016-02-26 Univ Catholique Louvain Anti-garp protein and uses thereof
EP3033356B1 (en) 2013-08-14 2020-01-15 Sachdev Sidhu Antibodies against frizzled proteins and methods of use thereof
MX2016001969A (en) 2013-08-14 2016-06-02 Novartis Ag Methods of treating sporadic inclusion body myositis.
TW201605896A (en) 2013-08-30 2016-02-16 安美基股份有限公司 GITR antigen binding proteins
EP3051942B1 (en) 2013-10-01 2020-09-02 Kymab Limited Animal models and therapeutic molecules
WO2015051293A2 (en) 2013-10-04 2015-04-09 Abbvie, Inc. Use of metal ions for modulation of protein glycosylation profiles of recombinant proteins
US8946395B1 (en) 2013-10-18 2015-02-03 Abbvie Inc. Purification of proteins using hydrophobic interaction chromatography
US9085618B2 (en) 2013-10-18 2015-07-21 Abbvie, Inc. Low acidic species compositions and methods for producing and using the same
US9181337B2 (en) 2013-10-18 2015-11-10 Abbvie, Inc. Modulated lysine variant species compositions and methods for producing and using the same
WO2015073884A2 (en) 2013-11-15 2015-05-21 Abbvie, Inc. Glycoengineered binding protein compositions
US11014987B2 (en) 2013-12-24 2021-05-25 Janssen Pharmaceutics Nv Anti-vista antibodies and fragments, uses thereof, and methods of identifying same
EP3087098B1 (en) 2013-12-24 2020-04-08 Janssen Pharmaceutica NV Anti-vista antibodies and fragments
SG11201605585SA (en) 2014-01-08 2016-08-30 Univ Leland Stanford Junior Targeted therapy for small cell lung cancer
CN114262344A (en) 2014-02-28 2022-04-01 杭州多禧生物科技有限公司 Charged linkers and their use in conjugation reactions
JP2017510273A (en) 2014-03-21 2017-04-13 リジェネロン・ファーマシューティカルズ・インコーポレイテッドRegeneron Pharmaceuticals, Inc. VL antigen binding protein showing different binding properties
RU2016141307A (en) 2014-03-21 2018-04-24 Регенерон Фармасьютикалз, Инк. EXCELLENT HUMAN ANIMALS THAT MAKE SINGLE-DOMAIN BINDING PROTEINS
TW201622746A (en) 2014-04-24 2016-07-01 諾華公司 Methods of improving or accelerating physical recovery after surgery for hip fracture
PE20170441A1 (en) 2014-06-06 2017-04-26 Bristol Myers Squibb Co ANTIBODIES AGAINST THE GLUCOCORTICOID-INDUCED TUMOR NECROSIS FACTOR RECEPTOR (GITR) AND ITS USES
CN107073109B (en) 2014-06-11 2021-08-06 凯西·A·格林 Use of VISTA agonists and antagonists to inhibit or enhance humoral immunity
TWI695011B (en) 2014-06-18 2020-06-01 美商梅爾莎納醫療公司 Monoclonal antibodies against her2 epitope and methods of use thereof
WO2015200089A1 (en) 2014-06-23 2015-12-30 Five Prime Therapeutics, Inc. Methods of treating conditions with antibodies that bind colony stimulating factor 1 receptor (csf1r)
EP3160991A2 (en) 2014-06-25 2017-05-03 Novartis AG Compositions and methods for long acting proteins
US20170291939A1 (en) 2014-06-25 2017-10-12 Novartis Ag Antibodies specific for il-17a fused to hyaluronan binding peptide tags
US10519234B2 (en) 2014-06-27 2019-12-31 Innate Pharma NKp46 binding proteins
WO2015197598A2 (en) 2014-06-27 2015-12-30 Innate Pharma Multispecific antigen binding proteins
AR101669A1 (en) 2014-07-31 2017-01-04 Amgen Res (Munich) Gmbh ANTIBODY CONSTRUCTS FOR CDH19 AND CD3
TW201609812A (en) 2014-07-31 2016-03-16 安美基研究(慕尼黑)公司 Optimized cross-species specific bispecific single chain antibody constructs
AR101400A1 (en) 2014-07-31 2016-12-14 Amgen Res (Munich) Gmbh INDIVIDUAL CHAIN INDIVIDUAL CHAIN ANTIBODY CONSTRUCTION WITH IMPROVED FABRIC DISTRIBUTION
JP6692343B2 (en) 2014-08-01 2020-05-13 アンスティチュ ナショナル ドゥ ラ サンテ エ ドゥ ラ ルシェルシュ メディカル Anti-CD45RC antibody for use as a drug
CU24453B1 (en) 2014-08-07 2019-11-04 Novartis Ag ANTI-PROTEIN ANTIBODIES SIMILAR TO ANGIOPOYETIN 4
EP3194437B1 (en) 2014-08-07 2021-01-20 Novartis AG Angiopoietin-like 4 (angptl4) antibodies and methods of use
WO2016030488A1 (en) 2014-08-27 2016-03-03 Innate Pharma Treatment of celiac disease
WO2016040767A2 (en) 2014-09-12 2016-03-17 Amgen Inc. Chrdl-1 epitopes and antibodies
MX2017003247A (en) 2014-09-15 2017-11-30 Amgen Inc Bi-specific anti-cgrp receptor/pac1 receptor antigen binding proteins and uses thereof.
WO2016044588A1 (en) 2014-09-19 2016-03-24 The Regents Of The University Of Michigan Staphylococcus aureus materials and methods
EP3201226A2 (en) 2014-10-03 2017-08-09 Massachusetts Institute of Technology Antibodies that bind ebola glycoprotein and uses thereof
WO2016131950A1 (en) 2015-02-20 2016-08-25 Innate Pharma Cd73 blockade
EP3736294A3 (en) 2014-10-10 2021-02-17 Innate Pharma Cd73 blockade
WO2016059220A1 (en) 2014-10-16 2016-04-21 INSERM (Institut National de la Santé et de la Recherche Médicale) Tcr-activating agents for use in the treatment of t-all
WO2016069727A1 (en) 2014-10-29 2016-05-06 Five Prime Therapeutics, Inc. Combination therapy for cancer
US10738078B2 (en) 2014-11-03 2020-08-11 Bristol-Myers Squibb Company Use of caprylic acid precipitation for protein purification
WO2016073894A1 (en) 2014-11-07 2016-05-12 Eleven Biotherapeutics, Inc. Therapeutic agents with increased ocular retention
EP3632931A1 (en) 2014-11-07 2020-04-08 Sesen Bio, Inc. Improved il-6 antibodies
EP3221358B1 (en) 2014-11-18 2021-07-21 Janssen Pharmaceutica, N.V. Cd47 antibodies, methods, and uses
CN113773388A (en) 2014-11-21 2021-12-10 百时美施贵宝公司 anti-CD 73 antibodies and uses thereof
EP3226900A4 (en) 2014-12-05 2018-09-19 Immunext, Inc. Identification of vsig8 as the putative vista receptor and its use thereof to produce vista/vsig8 modulators
UY36449A (en) 2014-12-19 2016-07-29 Novartis Ag COMPOSITIONS AND METHODS FOR ANTIBODIES DIRECTED TO BMP6
CN113563468A (en) 2014-12-19 2021-10-29 雷根尼桑斯公司 Antibodies that bind to human C6 and uses thereof
BR112017013111A2 (en) 2014-12-22 2018-05-15 Five Prime Therapeutics Inc methods of treating a disorder and treating synovitis, use of an antibody and antibody
KR102644115B1 (en) 2014-12-23 2024-03-05 브리스톨-마이어스 스큅 컴퍼니 Antibodies to tigit
EP3253796A1 (en) 2015-02-03 2017-12-13 Université Catholique de Louvain Anti-garp protein and uses thereof
WO2016149088A1 (en) 2015-03-13 2016-09-22 Bristol-Myers Squibb Company Use of alkaline washes during chromatography to remove impurities
AU2016232715A1 (en) 2015-03-19 2017-09-28 Regeneron Pharmaceuticals, Inc. Non-human animals that select for light chain variable regions that bind antigen
WO2016153983A1 (en) 2015-03-20 2016-09-29 Bristol-Myers Squibb Company Use of dextran for protein purification
US20180105554A1 (en) 2015-03-20 2018-04-19 Bristol-Myers Squibb Company Use of dextran sulfate to enhance protein a affinity chromatography
BR112017020952A2 (en) 2015-04-13 2018-07-10 Five Prime Therapeutics Inc cancer treatment method, composition and use of composition
CN116063541A (en) 2015-04-17 2023-05-05 安进研发(慕尼黑)股份有限公司 Bispecific antibody constructs for CDH3 and CD3
JOP20200116A1 (en) 2015-04-24 2017-06-16 Amgen Inc Methods for treating or preventing migraine headache
HUE061253T2 (en) 2015-05-29 2023-06-28 Bristol Myers Squibb Co Antibodies against ox40 and uses thereof
BR112017021484A2 (en) 2015-06-05 2018-07-03 Novartis Ag antibodies targeting bone morphogenetic protein 9 (bmp9) and methods for them
CA2990518A1 (en) 2015-06-23 2016-12-29 Innate Pharma Multispecific nk engager proteins
WO2016207717A1 (en) 2015-06-24 2016-12-29 Janssen Pharmaceutica Nv Anti-vista antibodies and fragments
CN107922500A (en) 2015-06-29 2018-04-17 洛克菲勒大学 The anti-CD 40 antibodies of agonist activity with enhancing
NZ739830A (en) 2015-07-12 2021-12-24 Hangzhou Dac Biotech Co Ltd Bridge linkers for conjugation of cell-binding molecules
US9839687B2 (en) 2015-07-15 2017-12-12 Suzhou M-Conj Biotech Co., Ltd. Acetylenedicarboxyl linkers and their uses in specific conjugation of a cell-binding molecule
TWI744242B (en) 2015-07-31 2021-11-01 德商安美基研究(慕尼黑)公司 Antibody constructs for egfrviii and cd3
TWI796283B (en) 2015-07-31 2023-03-21 德商安美基研究(慕尼黑)公司 Antibody constructs for msln and cd3
TWI717375B (en) 2015-07-31 2021-02-01 德商安美基研究(慕尼黑)公司 Antibody constructs for cd70 and cd3
TW202346349A (en) 2015-07-31 2023-12-01 德商安美基研究(慕尼黑)公司 Antibody constructs for dll3 and cd3
TWI829617B (en) 2015-07-31 2024-01-21 德商安美基研究(慕尼黑)公司 Antibody constructs for flt3 and cd3
CN108350072B (en) 2015-08-03 2022-05-24 诺华股份有限公司 Methods of treating FGF 21-associated disorders
IL257411B1 (en) 2015-08-24 2024-01-01 Trianni Inc Enhanced production of immunoglobulins
TN2018000076A1 (en) 2015-09-09 2019-07-08 Novartis Ag Thymic stromal lymphopoietin (tslp)-binding molecules and methods of using the molecules
HUE053914T2 (en) 2015-09-09 2021-07-28 Novartis Ag Thymic stromal lymphopoietin (tslp)-binding antibodies and methods of using the antibodies
AU2016323440B2 (en) 2015-09-15 2023-07-13 Amgen Inc. Tetravalent bispecific and tetraspecific antigen binding proteins and uses thereof
RU2638457C2 (en) 2015-09-28 2017-12-13 Общество С Ограниченной Ответственностью "Онкомакс" Antibodies specifically binding type 1 receptor of fibroblast growth factor, antibodies application for oncological disease treatment, method for antibodies production
US11130817B2 (en) 2015-10-12 2021-09-28 Innate Pharma CD73 blocking agents
KR20180082563A (en) 2015-11-19 2018-07-18 브리스톨-마이어스 스큅 컴퍼니 Antibodies to glucocorticoid-induced tumor necrosis factor receptor (GITR) and uses thereof
RU2021107536A (en) 2015-11-23 2021-07-02 Файв Прайм Терапьютикс, Инк. FGFR2 INHIBITORS SEPARATELY OR IN COMBINATION WITH IMMUNOSTIMULATING AGENTS IN THE TREATMENT OF CANCER
WO2017095823A1 (en) 2015-11-30 2017-06-08 The Regents Of The University Of California Tumor-specific payload delivery and immune activation using a human antibody targeting a highly specific tumor cell surface antigen
EP3383903A1 (en) 2015-11-30 2018-10-10 Bristol-Myers Squibb Company Anti human ip-10 antibodies and their uses
WO2017095939A1 (en) 2015-12-03 2017-06-08 Trianni, Inc. Enhanced immunoglobulin diversity
EP3390447A1 (en) 2015-12-15 2018-10-24 Amgen Inc. Pacap antibodies and uses thereof
CR20180365A (en) 2015-12-16 2018-09-28 Amgen Inc PROTEINS OF UNION TO THE ANTI-TL1A / ANTI-TNF-a BISPECTIVE ANTIGEN AND ITS USES
WO2017103895A1 (en) 2015-12-18 2017-06-22 Novartis Ag Antibodies targeting cd32b and methods of use thereof
NZ743474A (en) 2015-12-23 2023-03-31 Amgen Inc Method of treating or ameliorating metabolic disorders using binding proteins for gastric inhibitory peptide receptor (gipr) in combination with glp-1 agonists
WO2017125897A1 (en) 2016-01-21 2017-07-27 Novartis Ag Multispecific molecules targeting cll-1
DK3411402T3 (en) 2016-02-03 2022-02-07 Amgen Res Munich Gmbh BCMA AND CD3 BISPECIFIC T-CELL INVOLVING ANTIBODY CONSTRUCTIONS
WO2017134158A1 (en) 2016-02-03 2017-08-10 Amgen Research (Munich) Gmbh Psma and cd3 bispecific t cell engaging antibody constructs
EA039859B1 (en) 2016-02-03 2022-03-21 Эмджен Рисерч (Мюник) Гмбх Bispecific antibody constructs binding egfrviii and cd3
IL260743B2 (en) 2016-02-04 2024-03-01 Trianni Inc Enhanced production of immunoglobulins
MX2018009800A (en) 2016-02-12 2018-11-09 Janssen Pharmaceutica Nv Anti-vista (b7h5) antibodies.
IL307425A (en) 2016-03-04 2023-12-01 Univ Rockefeller Antibodies to cd40 with enhanced agonist activity
AU2017228470A1 (en) 2016-03-04 2018-08-30 Bristol-Myers Squibb Company Combination therapy with anti-CD73 antibodies
US10443054B2 (en) 2016-03-06 2019-10-15 Massachusetts Institute Of Technology Methods for identifying and treating invasive/metastatic breast cancers
JP7270379B2 (en) 2016-03-08 2023-05-10 イナート・ファルマ・ソシエテ・アノニム Siglec neutralizing antibody
CN109476756B (en) 2016-03-15 2022-05-31 埃泰美德(香港)有限公司 Multi-specificity Fab fusion protein and application thereof
CA3016474A1 (en) 2016-03-15 2017-09-21 Mersana Therapeutics, Inc. Napi2b-targeted antibody-drug conjugates and methods of use thereof
EP3442579A4 (en) 2016-04-15 2019-12-18 Immunext Inc. Anti-human vista antibodies and use thereof
JOP20170091B1 (en) 2016-04-19 2021-08-17 Amgen Res Munich Gmbh Administration of a bispecific construct binding to CD33 and CD3 for use in a method for the treatment of myeloid leukemia
EA039084B1 (en) 2016-05-09 2021-12-01 Бристол-Майерс Сквибб Компани Tl1a antibodies and uses thereof
JP7231411B2 (en) 2016-06-15 2023-03-01 ノバルティス アーゲー Methods of treating diseases using inhibitors of bone morphogenetic protein 6 (BMP6)
CN109641967A (en) 2016-07-01 2019-04-16 戊瑞治疗有限公司 With the combined antitumor therapy of GITR agonist and CpG
CN109757103B (en) 2016-07-14 2024-01-02 百时美施贵宝公司 Antibodies against TIM3 and uses thereof
CN115925780A (en) 2016-07-22 2023-04-07 美国安进公司 Method for purifying Fc-containing protein
US11878060B2 (en) 2016-08-07 2024-01-23 Novartis Ag mRNA-mediated immunization methods
JP2019530646A (en) 2016-08-12 2019-10-24 ブリストル−マイヤーズ スクイブ カンパニーBristol−Myers Squibb Company Protein purification method
WO2018044970A1 (en) 2016-08-31 2018-03-08 University Of Rochester Human monoclonal antibodies to human endogenous retrovirus k envelope (herv-k) and uses thereof
TW201825674A (en) 2016-09-09 2018-07-16 美商艾斯合顧問有限公司 Oncolytic virus expressing bispecific engager molecules
WO2018065552A1 (en) 2016-10-06 2018-04-12 Innate Pharma Anti-cd39 antibodies
EP3525815A2 (en) 2016-10-13 2019-08-21 Massachusetts Institute of Technology Antibodies that bind zika virus envelope protein and uses thereof
WO2018073363A1 (en) 2016-10-21 2018-04-26 Innate Pharma Treatment with anti-kir3dl2 agents
KR20220147721A (en) 2016-11-14 2022-11-03 항저우 디에이씨 바이오테크 씨오, 엘티디 Conjugation linkers, cell binding molecule-drug conjugates containing the likers, methods of making and uses such conjugates with the linkers
US11339209B2 (en) 2016-11-14 2022-05-24 Novartis Ag Compositions, methods, and therapeutic uses related to fusogenic protein minion
WO2018090052A1 (en) 2016-11-14 2018-05-17 Amgen Inc. Bispecific or biparatopic antigen binding proteins and uses thereof
EP3559032A1 (en) 2016-12-23 2019-10-30 Innate Pharma Heterodimeric antigen binding proteins
CA3049110A1 (en) 2017-01-06 2018-07-12 Abl Bio Inc. Anti-.alpha.-syn antibody and use thereof
US20180244785A1 (en) 2017-01-09 2018-08-30 Merrimack Pharmaceuticals, Inc. Anti-fgfr antibodies and methods of use
JOP20190177A1 (en) 2017-01-17 2019-07-16 Amgen Inc Method of treating or ameliorating metabolic disorders using glp-1 receptor agonists conjugated to antagonists for gastric inhibitory peptide receptor (gipr)
WO2018138032A2 (en) 2017-01-24 2018-08-02 Innate Pharma NKp46 BINDING AGENTS
US11267885B2 (en) 2017-01-26 2022-03-08 Zlip Holding Limited CD47 antigen binding unit and uses thereof
JOP20190189A1 (en) 2017-02-02 2019-08-01 Amgen Res Munich Gmbh Low ph pharmaceutical composition comprising t cell engaging antibody constructs
WO2018141959A1 (en) 2017-02-06 2018-08-09 Innate Pharma Immunomodulatory antibody drug conjugates binding to a human mica polypeptide
AU2018218557B9 (en) 2017-02-08 2021-06-24 Novartis Ag FGF21 mimetic antibodies and uses thereof
CA3051839A1 (en) 2017-02-17 2018-08-23 Bristol-Myers Squibb Company Antibodies to alpha-synuclein and uses thereof
MX2019010848A (en) 2017-03-16 2019-10-30 Innate Pharma Compositions and methods for treating cancer.
CA3054837A1 (en) 2017-03-24 2018-09-27 Novartis Ag Methods for preventing and treating heart disease
KR20230148855A (en) 2017-03-30 2023-10-25 더 존스 홉킨스 유니버시티 A supramolecular high affinity protein-binding system for purification of biomacromolecules
JP2020512357A (en) 2017-03-31 2020-04-23 ファイブ プライム セラピューティクス, インコーポレイテッド Combination therapy for cancer using anti-GITR antibody
TWI788340B (en) 2017-04-07 2023-01-01 美商必治妥美雅史谷比公司 Anti-icos agonist antibodies and uses thereof
UY37726A (en) 2017-05-05 2018-11-30 Amgen Inc PHARMACEUTICAL COMPOSITION THAT INCLUDES BISPECTIFIC ANTIBODY CONSTRUCTIONS FOR IMPROVED STORAGE AND ADMINISTRATION
GB201707561D0 (en) 2017-05-11 2017-06-28 Argenx Bvba GARP-TGF-beta antibodies
CN110621336A (en) 2017-05-16 2019-12-27 戊瑞治疗有限公司 Combination of an anti-FGFR 2 antibody with a chemotherapeutic agent in the treatment of cancer
JOP20190259A1 (en) 2017-05-31 2019-10-31 Amgen Inc Anti-jagged1 antigen binding proteins
WO2018229715A1 (en) 2017-06-16 2018-12-20 Novartis Ag Compositions comprising anti-cd32b antibodies and methods of use thereof
EP3642239A1 (en) 2017-06-20 2020-04-29 Amgen Inc. Method of treating or ameliorating metabolic disorders using binding proteins for gastric inhibitory peptide receptor (gipr) in combination with glp-1 agonists
CA3063659A1 (en) 2017-06-28 2019-01-03 Novartis Ag Methods for preventing and treating urinary incontinence
US11447545B2 (en) 2017-07-10 2022-09-20 Innate Pharma Combination therapy using antibody to human Siglec-9 and antibody to human NKG2A for treating cancer
US11623954B2 (en) 2017-07-10 2023-04-11 Innate Pharma Siglec-9-neutralizing antibodies
EP3625250B1 (en) 2017-07-21 2021-04-14 Trianni, Inc. Single chain vh and heavy chain antibodies
WO2019020807A1 (en) 2017-07-28 2019-01-31 Gene Signal International Sa Cd9p-1-targeting antibody and uses thereof
RS63101B1 (en) 2017-08-03 2022-04-29 Amgen Inc Interleukin-21 muteins and methods of treatment
MX2020001327A (en) 2017-08-04 2020-03-20 Amgen Inc Method of conjugation of cys-mabs.
US20200256880A1 (en) 2017-08-16 2020-08-13 The Broad Institute, Inc. Neuronal Assay Method Involving Calcineurin
CN111263659A (en) 2017-08-18 2020-06-09 约翰霍普金斯大学 Supramolecular filamentous assemblies for protein purification
TWI731264B (en) 2017-09-08 2021-06-21 美商安進公司 Inhibitors of kras g12c and methods of using the same
BR112020004879A2 (en) 2017-09-13 2020-09-15 Five Prime Therapeutics, Inc. methods to treat pancreatic cancer, to treat cancer and to determine the responsiveness of a subject with cancer
US20200317783A1 (en) 2017-10-06 2020-10-08 Ono Pharmaceutical Co., Ltd. Bispecific antibody
US20210040205A1 (en) 2017-10-25 2021-02-11 Novartis Ag Antibodies targeting cd32b and methods of use thereof
EP3476942B1 (en) 2017-10-27 2022-01-26 Trianni, Inc. Long germline dh genes and long hcdr3 antibodies
CA3079363A1 (en) 2017-11-21 2019-05-31 Innate Pharma Multispecific antigen binding proteins
US20200377958A1 (en) 2017-12-01 2020-12-03 Millennium Pharmaceuticals, Inc. Biomarkers and methods for treatment with nae inhibitors
EP3724229A1 (en) 2017-12-11 2020-10-21 Amgen Inc. Continuous manufacturing process for bispecific antibody products
EP3498293A1 (en) 2017-12-15 2019-06-19 Institut National De La Sante Et De La Recherche Medicale (Inserm) Treatment of monogenic diseases with an anti-cd45rc antibody
UY38041A (en) 2017-12-29 2019-06-28 Amgen Inc CONSTRUCTION OF BIESPECFIC ANTIBODY DIRECTED TO MUC17 AND CD3
JP2021510697A (en) 2018-01-12 2021-04-30 ブリストル−マイヤーズ スクイブ カンパニーBristol−Myers Squibb Company Combination therapy with anti-IL-8 antibody and anti-PD-1 antibody for cancer treatment
JP7339262B2 (en) 2018-01-12 2023-09-05 アムジェン インコーポレイテッド PAC1 antibody and uses thereof
EP3737694B1 (en) 2018-01-12 2023-03-01 Amgen Inc. Anti-pd-1 antibodies and methods of treatment
US20220089720A1 (en) 2018-01-12 2022-03-24 Bristol-Myers Squibb Company Antibodies against tim3 and uses thereof
WO2019150309A1 (en) 2018-02-02 2019-08-08 Hammack Scott Modulators of gpr68 and uses thereof for treating and preventing diseases
TWI804572B (en) 2018-02-09 2023-06-11 日商小野藥品工業股份有限公司 Bispecific antibody
AU2019236865A1 (en) 2018-03-23 2020-10-01 Bristol-Myers Squibb Company Antibodies against MICA and/or MICB and uses thereof
WO2019191416A1 (en) 2018-03-29 2019-10-03 Bristol-Myers Squibb Company Methods of purifying monomeric monoclonal antibodies
IT201800004853A1 (en) 2018-04-24 2019-10-24 Methods of treating cancer
AU2019271149B2 (en) 2018-05-14 2023-07-13 Werewolf Therapeutics, Inc. Activatable interleukin 12 polypeptides and methods of use thereof
ES2955511T3 (en) 2018-05-14 2023-12-04 Werewolf Therapeutics Inc Activatable interleukin 2 polypeptides and methods of use thereof
WO2019225787A1 (en) 2018-05-24 2019-11-28 에이비엘바이오 주식회사 Anti-b7-h3 antibody and use thereof
TW202015726A (en) 2018-05-30 2020-05-01 瑞士商諾華公司 Entpd2 antibodies, combination therapies, and methods of using the antibodies and combination therapies
CA3099893A1 (en) 2018-06-18 2019-12-26 Innate Pharma Compositions and methods for treating cancer
BR112020027095A2 (en) 2018-07-09 2021-03-30 Five Prime Therapeutics, Inc. ILT4 BINDING ANTIBODIES
MA53381A (en) 2018-07-24 2021-06-02 Amgen Inc ASSOCIATION OF LILRB1 / 2 PATH INHIBITORS AND PD-1 PATH INHIBITORS
MX2021001221A (en) 2018-07-30 2021-06-23 Amgen Res Munich Gmbh Prolonged administration of a bispecific antibody construct binding to cd33 and cd3.
KR20210042117A (en) 2018-08-03 2021-04-16 암젠 리서치 (뮌헨) 게엠베하 Antibody constructs against CLDN18.2 and CD3
AU2019327155A1 (en) 2018-08-27 2021-03-18 Affimed Gmbh Cryopreserved NK cells preloaded with an antibody construct
US20210371537A1 (en) 2018-09-18 2021-12-02 Merrimack Pharmaceuticals, Inc. Anti-tnfr2 antibodies and uses thereof
EP3626265A1 (en) 2018-09-21 2020-03-25 INSERM (Institut National de la Santé et de la Recherche Médicale) Anti-human cd45rc antibodies and uses thereof
WO2020069398A1 (en) 2018-09-27 2020-04-02 Akrevia Therapeutics Inc. Masked cytokine polypeptides
AU2019356564A1 (en) 2018-10-11 2021-04-29 Amgen Inc. Downstream processing of bispecific antibody constructs
UY38407A (en) 2018-10-15 2020-05-29 Novartis Ag TREM2 STABILIZING ANTIBODIES
JP2022509942A (en) 2018-11-16 2022-01-25 ブリストル-マイヤーズ スクイブ カンパニー Anti-NKG2A antibody and its use
CN113330033A (en) 2018-12-26 2021-08-31 先天制药公司 Leukocyte immunoglobulin-like receptor 2 neutralizing antibodies
BR112021014236A2 (en) 2019-01-22 2021-09-28 Innate Pharma ANTIBODY, PHARMACEUTICAL COMPOSITION, KIT AND METHOD TO PREDICT OR EVALUATE EFFECTIVENESS
WO2020154293A1 (en) 2019-01-22 2020-07-30 Bristol-Myers Squibb Company Antibodies against il-7r alpha subunit and uses thereof
CN113614107A (en) 2019-01-23 2021-11-05 Encefa公司 CD31 competitors and uses thereof
CN116063520A (en) 2019-01-30 2023-05-05 真和制药有限公司 anti-GAL 3 antibodies and uses thereof
CA3131953A1 (en) 2019-03-01 2020-09-10 Merrimack Pharmaceuticals, Inc. Anti-tnfr2 antibodies and uses thereof
CN113646630A (en) 2019-03-29 2021-11-12 百时美施贵宝公司 Method for measuring hydrophobicity of chromatographic resin
TW202102544A (en) 2019-04-04 2021-01-16 日商小野藥品工業股份有限公司 Bispecific antibody
JP2022532217A (en) 2019-05-14 2022-07-13 ウェアウルフ セラピューティクス, インコーポレイテッド Separation part and how to use it
WO2020234399A1 (en) 2019-05-20 2020-11-26 INSERM (Institut National de la Santé et de la Recherche Médicale) Novel anti-cd25 antibodies
MX2021014534A (en) 2019-05-30 2022-02-11 Amgen Inc Engineering the hinge region to drive antibody dimerization.
CR20210607A (en) 2019-06-12 2022-01-21 Novartis Ag Natriuretic peptide receptor 1 antibodies and methods of use
AU2020290573A1 (en) 2019-06-13 2021-11-04 Amgen Inc. Automated biomass-based perfusion control in the manufacturing of biologics
AU2020304671A1 (en) 2019-06-28 2022-01-20 Amgen Inc. Anti-CGRP receptor/anti-PAC1 receptor bispecific antigen binding proteins
CN114502725A (en) 2019-07-01 2022-05-13 特里安尼公司 Transgenic mammals and methods of use
JP2022538674A (en) 2019-07-01 2022-09-05 トニックス ファーマ リミテッド ANTI-CD154 ANTIBODY AND USES THEREOF
CN114554841A (en) 2019-07-01 2022-05-27 特里安尼公司 Transgenic mammals and methods of use thereof
EP3998081A4 (en) 2019-07-05 2023-07-12 Ono Pharmaceutical Co., Ltd. Treatment of hematologic cancer with pd-1/cd3 dual specificity protein
CN114144436A (en) 2019-07-24 2022-03-04 H.隆德贝克有限公司 anti-mGluR 5 antibodies and uses thereof
CA3148591A1 (en) 2019-07-26 2021-02-04 Amgen Inc. Anti-il13 antigen binding proteins
WO2021020416A1 (en) 2019-07-30 2021-02-04 小野薬品工業株式会社 Bispecific antibody
WO2021025140A1 (en) 2019-08-08 2021-02-11 小野薬品工業株式会社 Dual-specific protein
EP3785536B1 (en) 2019-08-28 2022-01-26 Trianni, Inc. Adam6 knockin mice
MX2022002981A (en) 2019-09-10 2022-04-06 Amgen Inc Purification method for bispecific antigen-binding polypeptides with enhanced protein l capture dynamic binding capacity.
TW202124446A (en) 2019-09-18 2021-07-01 瑞士商諾華公司 Combination therapies with entpd2 antibodies
US20220348651A1 (en) 2019-09-18 2022-11-03 Novartis Ag Entpd2 antibodies, combination therapies, and methods of using the antibodies and combination therapies
EP4034561A2 (en) 2019-09-27 2022-08-03 Starkage Therapeutics Senescent cell-associated antigen-binding domains, antibodies and chimeric antigen receptors comprising the same, and uses thereof
CN112646038A (en) 2019-10-11 2021-04-13 迈威(上海)生物科技股份有限公司 Anti-human Trop-2 antibody and application thereof
TW202128160A (en) 2019-10-18 2021-08-01 美國加利福尼亞大學董事會 Methods and compositions for treating pathogenic blood vessel disorders
JP2023500506A (en) 2019-11-04 2023-01-06 アストラゼネカ・アクチエボラーグ Combination therapy to treat cancer
MX2022005566A (en) 2019-11-08 2022-07-19 Amgen Inc Engineering charge pair mutations for pairing of hetero-igg molecules.
AU2020381536A1 (en) 2019-11-13 2022-04-21 Amgen Inc. Method for reduced aggregate formation in downstream processing of bispecific antigen-binding molecules
WO2021102049A1 (en) 2019-11-19 2021-05-27 Amgen Inc. Novel multispecific antibody format
UY38995A (en) 2019-12-20 2021-06-30 Amgen Inc MESOTHELIN TARGETING CD40 AGONIST MULTI-SPECIFIC ANTIBODY CONSTRUCTS FOR THE TREATMENT OF SOLID TUMORS
CA3160927A1 (en) 2019-12-27 2021-07-01 Affimed Gmbh Method for the production of bispecific fcyriii x cd30 antibody construct
CA3166898A1 (en) 2020-01-06 2021-07-15 Hifibio (Hk) Limited Anti-tnfr2 antibody and uses thereof
AU2021205143A1 (en) 2020-01-07 2022-07-28 Hifibio, Inc. Anti-Galectin-9 antibody and uses thereof
EP4093771A1 (en) 2020-01-22 2022-11-30 Amgen Research (Munich) GmbH Combinations of antibody constructs and inhibitors of cytokine release syndrome and uses thereof
CN114845740A (en) 2020-01-31 2022-08-02 先天制药公司 Treatment of cancer
US20230146593A1 (en) 2020-03-12 2023-05-11 Amgen Inc. Method for treatment and prophylaxis of crs in patients comprising a combination of bispecific antibodies binding to cds x cancer cell and tnf alpha or il-6 inhibitor
JP2023518225A (en) 2020-03-19 2023-04-28 アムジエン・インコーポレーテツド Antibodies against mucin 17 and uses thereof
CN115461363A (en) 2020-05-01 2022-12-09 诺华股份有限公司 Immunoglobulin variants
CN116096758A (en) 2020-05-01 2023-05-09 诺华股份有限公司 Engineered immunoglobulins
EP3909601A1 (en) 2020-05-11 2021-11-17 LeukoCom GmbH A novel antibody binding specifically to human ceacam1/3/5 and use thereof
WO2021231732A1 (en) 2020-05-15 2021-11-18 Bristol-Myers Squibb Company Antibodies to garp
CA3183756A1 (en) 2020-05-19 2021-11-25 Amgen Inc. Mageb2 binding constructs
CA3183693A1 (en) 2020-05-29 2021-12-02 Amgen Inc. Adverse effects-mitigating administration of a bispecific construct binding to cd33 and cd3
CN115803062A (en) 2020-06-03 2023-03-14 博泰康医药公司 Antibodies to trophoblast cell surface antigen 2 (TROP-2)
MX2022016548A (en) 2020-06-26 2023-03-14 Amgen Inc Il-10 muteins and fusion proteins thereof.
WO2022006562A1 (en) 2020-07-03 2022-01-06 Dana-Farber Cancer Institute, Inc. Multispecific coronavirus antibodies
US20230256114A1 (en) 2020-07-07 2023-08-17 Bionecure Therapeutics, Inc. Novel maytansinoids as adc payloads and their use for the treatment of cancer
TW202216778A (en) 2020-07-15 2022-05-01 美商安進公司 Tigit and cd112r blockade
TW202216761A (en) 2020-07-16 2022-05-01 瑞士商諾華公司 Anti-betacellulin antibodies, fragments thereof, and multi-specific binding molecules
JP2023534987A (en) 2020-07-24 2023-08-15 アムジエン・インコーポレーテツド Immunogen derived from SARS-COV2 spike protein
CN116419747A (en) 2020-08-07 2023-07-11 福蒂斯治疗公司 CD46 targeting immunoconjugates and methods of use thereof
KR20230077722A (en) 2020-08-10 2023-06-01 지브이20 테라퓨틱스 엘엘씨 Compositions and methods for targeting IGSF8 to treat autoimmune diseases and cancer
MX2023002125A (en) 2020-08-20 2023-04-26 Amgen Inc Antigen binding proteins with non-canonical disulfide in fab region.
WO2022043496A2 (en) 2020-08-28 2022-03-03 INSERM (Institut National de la Santé et de la Recherche Médicale) Use of mait cells as biomarkers and biotargets in covid-19
US20230374162A1 (en) 2020-10-07 2023-11-23 Amgen Inc. Rational selection of building blocks for the assembly of multispecific antibodies
AU2021357841A1 (en) 2020-10-08 2023-06-15 Affimed Gmbh Trispecific binders
CA3198456A1 (en) 2020-10-14 2022-04-21 Five Prime Therapeutics, Inc. Anti-c-c chemokine receptor 8 (ccr8) antibodies and methods of use thereof
EP4240767A1 (en) 2020-11-06 2023-09-13 Amgen Inc. Polypeptide constructs binding to cd3
CN116472288A (en) 2020-11-06 2023-07-21 诺华股份有限公司 Antibody Fc variants
CN116323671A (en) 2020-11-06 2023-06-23 安进公司 Multi-targeting bispecific antigen binding molecules with increased selectivity
EP4240407A1 (en) 2020-11-06 2023-09-13 Amgen Inc. Antigen binding domain with reduced clipping rate
KR20230098334A (en) 2020-11-06 2023-07-03 암젠 리서치 (뮌헨) 게엠베하 Polypeptide constructs that selectively bind to CLDN6 and CD3
EP4244246A1 (en) 2020-11-10 2023-09-20 Amgen Inc. Novel linkers of multispecific antigen binding domains
AR124123A1 (en) 2020-11-20 2023-02-15 Inst Nat Sante Rech Med ANTI-CD25 ANTIBODIES
JP2024504547A (en) 2020-11-20 2024-02-01 インセルム(インスティチュート ナショナル デ ラ サンテ エ デ ラ リシェルシェ メディカル) Anti-CD25 antibody
CA3196198A1 (en) 2020-11-25 2022-06-02 Manel KRAIEM Treatment of cancer
EP4258865A1 (en) 2020-12-09 2023-10-18 Trianni, Inc. Heavy chain-only antibodies
WO2022130182A1 (en) 2020-12-14 2022-06-23 Novartis Ag Reversal binding agents for anti-natriuretic peptide receptor 1 (npr1) antibodies and uses thereof
AU2022205313A1 (en) 2021-01-06 2023-07-20 Tonix Pharma Limited Methods of inducing immune tolerance with modified anti-cd154 antibodies
BR112023019138A2 (en) 2021-03-26 2023-10-24 Innate Pharma MULTI-SPECIFIC PROTEIN, PHARMACEUTICAL COMPOSITION, RECOMBINANT CELL, METHOD OF PREPARING AN NK CELL COMPOSITION, NK CELL COMPOSITION, USE OF A PROTEIN OR COMPOSITION, METHODS AND USE
JP2024513376A (en) 2021-04-02 2024-03-25 アムジエン・インコーポレーテツド MAGEB2 binding construct
AU2022263406A1 (en) 2021-04-20 2023-10-19 Amgen Inc. Balanced charge distribution in electrostatic steering of chain pairing in multi-specific and monovalent igg molecule assembly
EP4330281A1 (en) 2021-04-29 2024-03-06 Amgen Inc. Methods for reducing low molecular weight species of recombinantly-produced proteins
CA3216988A1 (en) 2021-05-05 2022-11-10 Werner Mueller Transgenic rodents expressing chimeric equine-rodent antibodies and methods of use thereof
IL307672A (en) 2021-05-06 2023-12-01 Amgen Res Munich Gmbh Cd20 and cd22 targeting antigen-binding molecules for use in proliferative diseases
WO2022235940A1 (en) 2021-05-06 2022-11-10 Dana-Farber Cancer Institute, Inc. Antibodies against alk and methods of use thereof
WO2022258691A1 (en) 2021-06-09 2022-12-15 Innate Pharma Multispecific proteins binding to nkg2d, a cytokine receptor, a tumour antigen and cd16a
WO2022258678A1 (en) 2021-06-09 2022-12-15 Innate Pharma Multispecific proteins binding to nkp30, a cytokine receptor, a tumour antigen and cd16a
CN117616050A (en) 2021-06-09 2024-02-27 先天制药公司 Multispecific proteins that bind to NKP46, cytokine receptor, tumor antigen and CD16A
WO2022272018A1 (en) 2021-06-25 2022-12-29 Amgen Inc. Treatment of cardiovascular disease with trem-1 antigen binding proteins
IL308154A (en) 2021-07-30 2023-12-01 Affimed Gmbh Duplexbodies
CA3232212A1 (en) 2021-10-01 2023-04-06 Ping XIANG Transgenic rodents for cell line identification and enrichment
TW202334221A (en) 2021-11-03 2023-09-01 德商安富美德有限公司 Bispecific cd16a binders
WO2023079493A1 (en) 2021-11-03 2023-05-11 Affimed Gmbh Bispecific cd16a binders
CA3235395A1 (en) 2021-11-10 2023-05-19 Peter Burrows Transgenic mammals and methods of use thereof
WO2023097024A1 (en) 2021-11-24 2023-06-01 Dana-Farber Cancer Institute, Inc. Antibodies against ctla-4 and methods of use thereof
WO2023114544A1 (en) 2021-12-17 2023-06-22 Dana-Farber Cancer Institute, Inc. Antibodies and uses thereof
WO2023114543A2 (en) 2021-12-17 2023-06-22 Dana-Farber Cancer Institute, Inc. Platform for antibody discovery
WO2023137161A1 (en) 2022-01-14 2023-07-20 Amgen Inc. Triple blockade of tigit, cd112r, and pd-l1
WO2023170207A1 (en) 2022-03-09 2023-09-14 Alderaan Biotechnology Anti-cd160 transmembrane isoform antibodies
US20230357381A1 (en) 2022-04-26 2023-11-09 Novartis Ag Multispecific antibodies targeting il-13 and il-18
TW202346368A (en) 2022-05-12 2023-12-01 德商安美基研究(慕尼黑)公司 Multichain multitargeting bispecific antigen-binding molecules of increased selectivity
WO2024039672A2 (en) 2022-08-15 2024-02-22 Dana-Farber Cancer Institute, Inc. Antibodies against msln and methods of use thereof
WO2024039670A1 (en) 2022-08-15 2024-02-22 Dana-Farber Cancer Institute, Inc. Antibodies against cldn4 and methods of use thereof
WO2024059675A2 (en) 2022-09-14 2024-03-21 Amgen Inc. Bispecific molecule stabilizing composition
WO2024062019A1 (en) 2022-09-21 2024-03-28 Synabs Anti-ccr8 antibodies and uses thereof

Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4950599A (en) * 1987-01-29 1990-08-21 Wolf Bertling Method for exchanging homologous DNA sequences in a cell using polyoma encapsulated DNA fragments
US4959313A (en) * 1987-06-22 1990-09-25 The Jackson Laboratory Cellular enhancer for expressing genes in undifferentiated stem cells
US5204244A (en) * 1987-10-27 1993-04-20 Oncogen Production of chimeric antibodies by homologous recombination
US5545807A (en) * 1988-10-12 1996-08-13 The Babraham Institute Production of antibodies from transgenic animals
US5545806A (en) * 1990-08-29 1996-08-13 Genpharm International, Inc. Ransgenic non-human animals for producing heterologous antibodies
US5569825A (en) * 1990-08-29 1996-10-29 Genpharm International Transgenic non-human animals capable of producing heterologous antibodies of various isotypes
US5591669A (en) * 1988-12-05 1997-01-07 Genpharm International, Inc. Transgenic mice depleted in a mature lymphocytic cell-type
US6114598A (en) * 1990-01-12 2000-09-05 Abgenix, Inc. Generation of xenogeneic antibodies

Family Cites Families (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR2637613B1 (en) 1987-06-19 1991-09-27 Transgene Sa PROCESS FOR THE PREPARATION OF STABLE CELL LINES FOR THE PRODUCTION OF DETERMINED PROTEINS FROM TRANSGENIC ANIMALS; TUMOR CELL LINES AND PROTEINS OBTAINED
EP0322240B1 (en) 1987-12-23 1995-03-01 The Board Of Trustees Of The Leland Stanford Junior University Chimeric immunocompromised mammals and their use
DE69034263D1 (en) * 1989-07-25 2009-04-02 Cell Genesys Inc Homologous recombination for universal donor cells and mammalian chimeric cells
GB8920211D0 (en) * 1989-09-07 1989-10-18 Ici Plc Diagnostic method
JP3008208B2 (en) 1990-06-01 2000-02-14 武田薬品工業株式会社 Novel hybridoma, its production method and production method of bioactive substance
GB9119338D0 (en) 1991-09-10 1991-10-23 Inst Of Animal Physiology And Control of gene expression
EP0648265A4 (en) 1992-06-18 1996-12-04 Genpharm Int Methods for producing transgenic non-human animals harboring a yeast artificial chromosome.
ES2301158T3 (en) 1992-07-24 2008-06-16 Amgen Fremont Inc. XENOGENIC ANTIBODY PRODUCTION.
DE19745212A1 (en) * 1997-10-13 1999-04-15 Clariant Gmbh Process for the preparation of fluorinated aromatic compounds

Patent Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4950599A (en) * 1987-01-29 1990-08-21 Wolf Bertling Method for exchanging homologous DNA sequences in a cell using polyoma encapsulated DNA fragments
US4959313A (en) * 1987-06-22 1990-09-25 The Jackson Laboratory Cellular enhancer for expressing genes in undifferentiated stem cells
US5204244A (en) * 1987-10-27 1993-04-20 Oncogen Production of chimeric antibodies by homologous recombination
US5545807A (en) * 1988-10-12 1996-08-13 The Babraham Institute Production of antibodies from transgenic animals
US5591669A (en) * 1988-12-05 1997-01-07 Genpharm International, Inc. Transgenic mice depleted in a mature lymphocytic cell-type
US6114598A (en) * 1990-01-12 2000-09-05 Abgenix, Inc. Generation of xenogeneic antibodies
US5545806A (en) * 1990-08-29 1996-08-13 Genpharm International, Inc. Ransgenic non-human animals for producing heterologous antibodies
US5569825A (en) * 1990-08-29 1996-10-29 Genpharm International Transgenic non-human animals capable of producing heterologous antibodies of various isotypes

Cited By (33)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006029005A3 (en) * 2004-09-02 2006-06-22 Oklahoma Med Res Found Promoter substitution for immunoglobulin therapy
US20060194319A1 (en) * 2004-09-02 2006-08-31 Webb Carol F Promoter substitution for immunoglobulin therapy
WO2006029005A2 (en) * 2004-09-02 2006-03-16 Oklahoma Medical Research Foundation Promoter substitution for immunoglobulin therapy
EP2463305A1 (en) 2006-01-12 2012-06-13 Alexion Pharmaceuticals, Inc. Antibodies to OX-2/CD200 and uses thereof
US8894998B2 (en) 2006-08-03 2014-11-25 Medimmune Limited Antibodies directed to αVβ6 and uses thereof
US8398975B2 (en) 2006-08-03 2013-03-19 Medimmune Limited Antibodies directed to αVβ6 and uses thereof
EP3524626A1 (en) 2007-03-22 2019-08-14 Biogen MA Inc. Binding proteins, including antibodies, antibody derivatives and antibody fragments, that specifically bind cd154 and uses thereof
EP2894166A1 (en) 2008-11-10 2015-07-15 Alexion Pharmaceuticals, Inc. Methods and compositions for treating complement-associated disorders
EP2894165A1 (en) 2008-11-10 2015-07-15 Alexion Pharmaceuticals, Inc. Methods and compositions for treating complement-associated disorders
EP3974448A1 (en) 2008-11-10 2022-03-30 Alexion Pharmaceuticals, Inc. Methods and compositions for treating complementassociated disorders
WO2010054403A1 (en) 2008-11-10 2010-05-14 Alexion Pharmaceuticals, Inc. Methods and compositions for treating complement-associated disorders
EP3121197A1 (en) 2008-11-10 2017-01-25 Alexion Pharmaceuticals, Inc. Methods and compositions for treating complement-associated disorders
EP3101031A1 (en) 2008-11-10 2016-12-07 Alexion Pharmaceuticals, Inc. Methods and compositions for treating complement-associated disorders
WO2011085343A1 (en) 2010-01-11 2011-07-14 Alexion Pharmaceuticals, Inc Biomarkers of immunomodulatory effects in humans treated with anti-cd200 antibodies
US9371378B1 (en) 2010-04-30 2016-06-21 Alexion Pharmaceuticals, Inc. Anti-C5a antibodies
EP2824111A2 (en) 2010-04-30 2015-01-14 Alexion Pharmaceuticals, Inc. Anti-C5A Antibodies and Methods for Using the Antibodies
US11407821B2 (en) 2010-04-30 2022-08-09 Alexion Pharmaceuticals, Inc. Anti-C5A antibodies
US10450370B2 (en) 2010-04-30 2019-10-22 Alexion Pharmaceuticals, Inc. Anti-C5a antibodies
US9221901B2 (en) 2010-04-30 2015-12-29 Alexion Pharmaceuticals, Inc. Methods of treating complement-associated disorders with anti-C5a antibodies
US9309310B2 (en) 2010-04-30 2016-04-12 Alexion Pharmaceuticals, Inc. Nucleic acids encoding anti-C5a antibodies
US9963503B2 (en) 2010-04-30 2018-05-08 Alexion Pharmaceuticals, Inc. Methods of producing anti-C5a antibodies
US9434784B1 (en) 2010-04-30 2016-09-06 Alexion Pharmaceuticals, Inc. Nucleic acids encodng anti-C5A antibodies
US9469690B2 (en) 2010-04-30 2016-10-18 Alexion Pharmaceuticals, Inc. Methods of treating complement-associated disorders with anti-C5a antibodies
US9011852B2 (en) 2010-04-30 2015-04-21 Alexion Pharmaceuticals, Inc. Anti-C5a antibodies
WO2011137395A1 (en) 2010-04-30 2011-11-03 Rother Russell P Anti-c5a antibodies and methods for using the antibodies
US9815890B2 (en) 2010-06-22 2017-11-14 The Regents Of The University Of Colorado, A Body Corporate Antibodies to the C3d fragment of complement component 3
USRE49339E1 (en) 2010-06-22 2022-12-20 The Regents Of The University Of Colorado, A Body Corporate Antibodies to the C3D fragment of complement component 3
WO2012020096A1 (en) 2010-08-13 2012-02-16 Medimmune Limited Monomeric polypeptides comprising variant fc regions and methods of use
WO2012022734A2 (en) 2010-08-16 2012-02-23 Medimmune Limited Anti-icam-1 antibodies and methods of use
WO2012106634A1 (en) 2011-02-03 2012-08-09 Alexion Pharmaceuticals, Inc. Use of an anti-cd200 antibody for prolonging the survival of allografts
WO2015050959A1 (en) 2013-10-01 2015-04-09 Yale University Anti-kit antibodies and methods of use thereof
WO2015175874A2 (en) 2014-05-16 2015-11-19 Medimmune, Llc Molecules with altered neonate fc receptor binding having enhanced therapeutic and diagnostic properties
EP3888690A2 (en) 2014-05-16 2021-10-06 MedImmune, LLC Molecules with altered neonate fc receptor binding having enhanced therapeutic and diagnostic properties

Also Published As

Publication number Publication date
US6162963A (en) 2000-12-19
US20030229905A1 (en) 2003-12-11
US6673986B1 (en) 2004-01-06

Similar Documents

Publication Publication Date Title
US6673986B1 (en) Generation of xenogeneic antibodies
US6114598A (en) Generation of xenogeneic antibodies
EP0652950B1 (en) Generation of xenogeneic antibodies
US7049426B2 (en) Transgenic animals for producing specific isotypes of human antibodies via non-cognate switch regions
US5789650A (en) Transgenic non-human animals for producing heterologous antibodies
CA2124967C (en) Transgenic non-human animals capable of producing heterologous antibodies
JP2938569B2 (en) Method for producing xenogeneic immunoglobulin and transgenic mouse
US5814318A (en) Transgenic non-human animals for producing heterologous antibodies
US20040010810A1 (en) Generation of xenogeneic antibodies
CA2274105A1 (en) Transgenic non-human animals capable of producing heterologous antibodies
CA2702329A1 (en) Generation of xenogeneic antibodies
AU743883B2 (en) Transgenic non-human animals capable of producing heterologous antibodies

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

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