WO1993012227A1 - Transgenic non-human animals capable of producing heterologous antibodies - Google Patents

Transgenic non-human animals capable of producing heterologous antibodies Download PDF

Info

Publication number
WO1993012227A1
WO1993012227A1 PCT/US1992/010983 US9210983W WO9312227A1 WO 1993012227 A1 WO1993012227 A1 WO 1993012227A1 US 9210983 W US9210983 W US 9210983W WO 9312227 A1 WO9312227 A1 WO 9312227A1
Authority
WO
WIPO (PCT)
Prior art keywords
human
transgene
gene
transgenic
animal
Prior art date
Application number
PCT/US1992/010983
Other languages
French (fr)
Inventor
Nils Lonberg
Robert M. Kay
Original Assignee
Genpharm International, 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 US07/810,279 external-priority patent/US5569825A/en
Priority claimed from US07/853,408 external-priority patent/US5789650A/en
Application filed by Genpharm International, Inc. filed Critical Genpharm International, Inc.
Priority to JP5511191A priority Critical patent/JPH07503132A/en
Priority to EP93901143A priority patent/EP0746609A4/en
Priority to CA002124967A priority patent/CA2124967C/en
Priority to US08/053,131 priority patent/US5661016A/en
Publication of WO1993012227A1 publication Critical patent/WO1993012227A1/en
Priority to US08/544,404 priority patent/US5770429A/en
Priority to US11/009,840 priority patent/US20060015949A1/en
Priority to US11/009,873 priority patent/US7501552B2/en
Priority to US11/009,769 priority patent/US20060026703A1/en

Links

Classifications

    • 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
    • C07K16/18Immunoglobulins [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
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • C07K16/3007Carcino-embryonic Antigens
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/42Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against immunoglobulins
    • C07K16/4283Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against immunoglobulins against an allotypic or isotypic determinant on Ig
    • C07K16/4291Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against immunoglobulins against an allotypic or isotypic determinant on Ig against IgE
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/44Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material not provided for elsewhere, e.g. haptens, metals, DNA, RNA, amino acids
    • 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
    • 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
    • A01K2217/075Animals 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
    • 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
    • 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
    • A01K2267/035Animal model for multifactorial diseases
    • A01K2267/0381Animal model for diseases of the hematopoietic system
    • 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

Definitions

  • the invention relates to transgenic non-human animals capable of producing heterologous antibodies
  • transgenic animals used to produce such transgenic animals
  • transgenes capable of functionally rearranging a heterologous D gene in V-D-J recombination, immortalized B-cells capable of producing heterologous antibodies, methods and transgenes for producing heterologous antibodies of multiple isotypes, methods and transgenes for inactivating or suppressing
  • variable region sequence comprises somatic mutation
  • transgenic nonhuman animals which produce antibodies having a human primary sequence and which bind to human antigens.
  • human immunoglobulins that are reactive with specific human antigens that are promising therapeutic and/or diagnostic targets.
  • producing human immunoglobulins that bind specifically with human antigens is problematic.
  • the present technology for generating monoclonal antibodies involves pre-exposing, or priming, an animal
  • idiotype and also screening for immunoglobulin class (isotype), it is possible to select hybridoma clones that secrete the desired antibody.
  • transgenic animals harboring a functional heterologous immunoglobulin transgene are a method by which antibodies reactive with self antigens may be
  • the transgenic animal must produce transgenic B cells that are capable of maturing through the B lymphocyte development pathway. Such maturation requires the presence of surface IgM on the transgenic B cells, however isotypes other than IgM are desired for therapeutic uses.
  • transgenes and transgenic animals preferably include cis-acting sequences that
  • sequences for V(D)J joining are reportedly a highly conserved, near-palindromic heptamer and a less well conserved AT-rich nanomer separated by a spacer of either 12 or 23 bp (Tonegawa (1983), Nature, 302, 575-581; Hesse, et al. (1989), Genes in Dev. , 3, 1053-1061). Efficient recombination reportedly occurs only between sites containing recombination signal sequences with different length spacer regions.
  • mice [Buchini, et al. (1987), Nature. 326, 409-411 (unrearranged chicken ⁇ transgene); Goodhart, et al. (1987) , Proc. Natl. Acad. Sci. USA, 84, 4229-4233) (unrearranged rabbit k gene); and Bruggemann, et al. (1989), Proc. Natl. Acad. Sci. USA, 86, 6709-6713 (hybrid mouse-human heavy chain)].
  • the results of such experiments have been variable, in some cases, producing incomplete or minimal rearrangement of the
  • Fc portion of molecules such as the interaction with mast cells or basophils through Fee, and binding of complement by Fc ⁇ or Fc ⁇ , it further is desirable to generate a functional diversity of antibodies of a given specificity by variation of isotype.
  • transgenic animals have been generated that incorporate transgenes encoding one or more chains of a heterologous antibody, there have been no reports of hererologous transgenes that undergo successful isotype switching.
  • Transgenic animals that cannot switch isotypes are limited to producing h-iterologous antibodies of a single isotype, and more specifically are limited to producing an isotype that is essential for B cell maturation, such as IgM and possibly IgD, which may be of limited therapeutic utility.
  • IgM and possibly IgD an isotype that is essential for B cell maturation
  • transgenes and transgenic animals that are capable of
  • heterologous antibodies e.g. antibodies encoded by genetic sequences of a first species that are produced in a second species. More particularly, there is a need in the art for heterologous immunoglobulin transgenes and transgenic animals that are capable of undergoing functional V-D-J gene rearrangement that incorporates all or a portion of a D gene segment which contributes to recombinational diversity. Further, there is a need in the art for transgenes and transgenic animals that can support V-D-J recombination and isotype switching so that (1) functional B cell development may occur, and (2)
  • therapeutically useful heterologous antibodies may be any therapeutically useful heterologous antibodies.
  • transgenic nonhuman animals which are capable of producing a heterologous antibody, such as a human antibody.
  • heterologous antibodies wherein such B-cells are immortalized to provide a source of a monoclonal antibody specific for a particular antigen.
  • a further object of the invention is to provide methods to generate an immunoglobulin variable region gene segment repertoire that is used to construct one or more transgenes of the invention.
  • Transgenic nonhuman animals are provided which are capable of producing a heterologous antibody, such as a human antibody.
  • heterologous antibodies may be of various isotypes, including: IgG1, IgG2, IgG3, IgG4, IgM, IgA1, IgA2, IgA sec , IgD, of IgE.
  • the transgenic B cells and pre-B cells to produce surface-bound immunoglobulin, particularly of the IgM (or possibly IgD) isotype, in order to effectuate B cell development and
  • a cell of the B-cell lineage will produce only a single isotype at a time, although cis or trans
  • RNA splicing such as occurs naturally with the ⁇ s (secreted ⁇ ) and ⁇ M (membrane-bound ⁇ ) forms, and the ⁇ and ⁇ immunoglobulin chains, may lead to the contemporaneous
  • isotype switching may be classical class- switching or may result from one or more non-classical isotype switching mechanisms.
  • the invention provides heterologous immunoglobulin transgenes and transgenic nonhuman animals harboring such transgenes, wherein the transgenic animal is capable of producing heterologous antibodies of multiple isotypes by undergoing isotype switching.
  • Classical isotype switching occurs by recombination events which involve at least one switch sequence region in the transgene.
  • Non-classical isotype switching may occur by, for example, homologous recombination between human ⁇ and human ⁇ ⁇ sequences ( ⁇ - associated deletion).
  • Alternative non-classical switching mechanisms such as intertransgene and/or interchromosomal recombination, among others, may occur and effectuate isotype switching.
  • transgenic nonhuman animals produce a first immunoglobulin isotype that is necessary for antigen-stimulated B cell maturation and can switch to encode and produce one or more subsequent heterologous isotypes that have therapeutic and/or diagnostic utility.
  • nonhuman animals of the invention are thus able to produce, in one embodiment, IgG, IgA, and/or IgE antibodies that are encoded by human immunoglobulin genetic sequences and which also bind specific human antigens with high affinity.
  • the invention also encompasses B-cells from such transgenic animals that are capable of expressing heterologous antibodies of various isotypes, wherein such B-cells are immortalized to provide a source of a monoclonal antibody specific for a particular antigen.
  • Hybridoma cells that are derived from such B-cells can serve as one source of such heterologous monoclonal antibodies.
  • the invention provides heterologous unrearranged and rearranged immunoglobulin heavy and light chain transgenes capable of undergoing isotype switching in vivo in the
  • isotype switching may occur spontaneously or be induced by treatment of the transgenic animal or explanted B- lineage lymphocytes with agents that promote isotype
  • T-cell-derived lymphokines e.g., IL-4 and IFN ⁇
  • the invention includes methods to induce heterologous antibody production in the aforementioned transgenic non-human animal, wherein such antibodies may be of various isotypes.
  • These methods include producing an antigen- stimulated immune response in a transgenic nonhuman animal for the generation of heterologous antibodies, particularly heterologous antibodies of a switched isotype (i.e., IgG, IgA, and IgE).
  • heterologous immunoglobulins produced in the transgenic animal and monoclonal antibody clones derived from the B-cells of said animal may be of various isotypes.
  • This invention further provides methods that facilitate isotype switching of the transgene, so that
  • Switch regions may be grafted from various C H genes and ligated to other C H genes in a transgene construct; such grafted switch sequences will typically function independently of the
  • ⁇ -associated deletion sequences may be linked to various C H genes to effect non-classical switching by deletion of sequences between two ⁇ -associated deletion sequences.
  • a transgene may be constructed so that a particular C H gene is linked to a different switch sequence and thereby is switched to more frequently than occurs when the naturally associated switch region is used.
  • This invention also provides methods to determine whether isotype switching of transgene sequences has occurred in a transgenic animal containing an immunoglobulin transgene.
  • the invention provides immunoglobulin transgene constructs and methods for producing immunoglobulin transgene constructs, some of which contain a subset of germline
  • immunoglobulin loci sequences (which may include deletions).
  • the invention includes a specific method for facilitated cloning and construction of immunoglobulin transgenes,
  • restriction sites flanked by two unique NotI sites. This method exploits the complementary termini of Xhol and Sall restrictions sites and is useful for creating large constructs by ordered concatemerization of restriction fragments in a vector.
  • the transgenes of the invention include a heavy chain transgene comprising DNA encoding at least one variable gene segment, one diversity gene segment, one joining gene segment and one constant region gene segment.
  • immunoglobulin light chain transgene comprises DNA encoding at least one variable gene segment, one joining gene segment and one constant region gene segment.
  • the gene segments encoding the light and heavy chain gene segments are heterologous to the transgenic non-human animal in that they are derived from, or correspond to, DNA encoding immunoglobulin heavy and light chain gene segments from a species not consisting of the transgenic non-human animal.
  • the transgene is constructed such that the individual gene segments are unrearranged, i.e., not rearranged so as to encode a functional immunoglobulin light or heavy chain.
  • Such unrearranged transgenes permit recombination of the gene segments (functional rearrangement) and expression of the resultant rearranged immunoglobulin heavy and/or light chains within the transgenic non-human animal when said animal is exposed to antigen.
  • heterologous heavy and light immunoglobulin transgenes comprise relatively large fragments of unrearranged heterologous DNA. Such fragments typically comprise a substantial portion of the C, J (and in the case of heavy chain, D) segments from a heterologous immunoglobulin locus. In addition, such fragments also comprise a substantial portion of the variable gene segments.
  • regulatory sequences e.g. promoters, enhancers, class switch regions, recombination signals and the like, corresponding to sequences derived from the heterologous DNA.
  • regulatory sequences may be incorporated into the transgene from the same or a related species of the non-human animal used in the invention.
  • human immunoglobulin gene segments may be combined in a transgene with a rodent immunoglobulin enhancer sequence for use in a transgenic mouse.
  • a transgenic non-human animal containing germline unrearranged light and heavy immunoglobulin transgenes - that undergo VDJ joining during D-cell differentiation - is contacted with an antigen to induce production of a heterologous antibody in a secondary repertoire B-cell.
  • vectors and methods to disrupt the endogenous immunoglobulin loci in the non-human animal to be used in the invention utilize a transgene, preferably positive-negative selection vector, which is constructed such that it targets the functional disruption of a class of gene segments encoding a heavy and/or light immunoglobulin chain endogenous to the non-human animal used in the invention.
  • endogenous gene segments include diversity, joining and constant region gene segments.
  • the positive-negative selection vector is contacted, with at least one embryonic stem cell of a non-human animal after which cells are selected wherein the positive-negative selection vector has integrated into the genome of the non-human animal by way of homologous recombination.
  • the resultant transgenic non-human animal is substantially incapable of mounting an immunoglobulin-mediated immune response as a result of homologous integration of the vector into chromosomal DNA.
  • Such immune deficient non-human animals may thereafter be used for study of immune deficiencies or used as the recipient of heterologous immunoglobulin heavy and light chain transgenes.
  • the invention also provides vectors, methods, and compositions useful for suppressing the expression of one or more species of immunoglobulin chain(s), without disrupting an endogenous immunoglobulin locus. Such methods are useful for suppressing expression of one or more endogenous immunoglobulin chain(s), without disrupting an endogenous immunoglobulin locus. Such methods are useful for suppressing expression of one or more endogenous immunoglobulin chain(s), without disrupting an endogenous immunoglobulin locus. Such methods are useful for suppressing expression of one or more endogenous
  • immunoglobulin chains while permitting the expression of one or more transgene-encoded immunoglobulin chains.
  • suppression of immunoglobulin chain expression does not require the time-consuming breeding that is needed to
  • Ig chain suppression may be accomplished with: (1) transgenes encoding and expressing antisense RNA that specifically hybridizes to an endogenous Ig chain gene sequence, (2) antisense oligonucleotides that specifically hybridize to. an endogenous Ig chain gene
  • immunoglobulins that bind specifically to an endogenous Ig chain polypeptide.
  • Fig. 1 depicts the complementarity determining regions CDR1, CDR2 and CDR3 and framework regions FR1, FR2, FR3 and FR4 in unrearranged genomic DNA and mRNA expressed from a rearranged immunoglobulin heavy chain gene
  • Fig. 2 depicts the human ⁇ chain locus
  • Fig. 3 depicts. the human ⁇ chain locus
  • Fig. 4 depicts the human heavy chain locus
  • Fig. 5 depicts a transgene construct containing a rearranged IgM gene ligated to a 25 kb fragment that contains human ⁇ 3 and ⁇ 1 constant regions followed by a 700 bp fragment containing the rat chain 3' enhancer sequence.
  • Fig. 6 is a restriction map of the human ⁇ chain locus depicting the fragments to be used to form a light chain transgene by way of in vivo homologous recombination.
  • Fig. 7 depicts the construction of pGP1.
  • Fig. 8 depicts the construction of the polylinker contained in pGP1.
  • Fig. 9 depicts the fragments used to construct a human heavy chain transgene of the invention.
  • Fig. 10 depicts the construction of pHIG1 and pCON1.
  • Fig. 11 depicts the human C ⁇ l fragments which are inserted into pRE3 (rat enhancer 3') to form pREG2.
  • Fig. 12 depicts the construction of pHIG3' and PCON.
  • Fig. 13 depicts the fragment containing human D region segments used in construction of the transgenes of the invention.
  • Fig. 14 depicts the construction of pHIG2 (D segment. containing plasmid).
  • Fig. 15 depicts the fragments covering the human J ⁇ and human C ⁇ gene segments used in constructing a transgene of the invention.
  • Fig. 16 depicts the structure of pE ⁇ .
  • Fig. 17 depicts the construction of pKapH.
  • Figs. 18A through 18D depict the construction of a positive-negative selection vector for functionally disrupting the endogenous heavy chain immunoglobulin locus of mouse.
  • Figs. 19A through 19C depict the construction of a positive-negative selection vector for functionally disrupting the endogenous immunoglobulin light-chain loci in mouse.
  • Figs. 20 a through e depict the structure of a kappa light chain targeting vector.
  • Figs. 21 a through f depict the structure of a mouse heavy chain targeting vector.
  • Fig. 22 depicts the map of vector pGPe.
  • Fig. 23 depicts the structure of vector pJM2.
  • Fig. 24 depicts the structure of vector pCOR1.
  • Fig. 25 depicts the transgene constructs for pIGM1, pHC1 and pHC2.
  • Fig. 26 depicts the structure of p ⁇ e2.
  • Fig. 27 depicts the structure of pVGE1.
  • Fig. 28 depicts the assay results of human Ig expression in a pHC1 transgenic mouse.
  • Fig,. 29 depicts the structure of pJCK1.
  • Fig. 30 depicts the construction of a synthetic heavy chain variable region.
  • Fig. 31 is a schematic representation of the ' heavy chain minilocus constructs pIGM 1 , pHC1, and pHC2.
  • Fig. 32 is a schematic representation of the heavy chain minilocus construct pIGG1 and the ⁇ light chain
  • Fig. 33 depicts a scheme to reconstruct functionally rearranged light chain genes.
  • Fig. 34 depicts serum ELISA results
  • Fig. 35 depicts the results of an ELISA assay of serum from 8 transgenic mice.
  • Fig. 36 is a schematic representation of plasmid pBCE1.
  • Fig. 37 depicts the immune response of transgenic mice of the present invention against KLH-DNP, by measuring IgG and IgM levels specific for KLH-DNP (37A), KLH (37B) and BSA-DNP (37C).
  • Fig. 38 shows ELISA data demonstrating the presence of antibodies that bind human carcinoembryonic antigen (CEA) and comprise human ⁇ chains; each panel shows reciprocal serial dilutions from pooled serum samples obtained from mice on the indicated day following immunization.
  • CEA carcinoembryonic antigen
  • Fig. 39 shows ELISA data demonstrating the presence of antibodies that bind human carcinoembryonic antigen (CEA) and comprise human ⁇ chains; each panel shows reciprocal serial dilutions from pooled serum samples obtained from mice on the indicated day following immunization.
  • CEA carcinoembryonic antigen
  • Fig. 40 shows aligned variable region sequences of 23 randomly-chosen cDNAs generated from mRNA obtained from lymphoid tissue of HCI transgenic mice immunized, with human carcinoembryonic antigen (CEA) as compared to the germline transgene sequence (top line); on each line nucleotide changes relative to germline sequence are shown above the alteration in deduced amino acid sequence (if any); the regions
  • Non-germline encoded nucleotides are shown in capital letters.
  • Germline V H 251 and J H are shown in lower case letters.
  • Deduced amino acid changes are given beneath
  • Fig. 41 shows the data from Fig. 40 in histogram format; deduced amino acid residue position is shown as the ordinate (left is the amino-terminal direction, right is in the direction towards the carboxy-terminus) and frequency of sequence variation is shown as the abscissa.
  • Fig. . 42 show the nucleotide sequence of a human DNA fragment, designated vk65.3, containing a V ⁇ gene segment; the deduced amino acid sequences of the V ⁇ coding regions are also shown; splicing and recombination signal sequences
  • Fig. 43 show the nucleotide sequence of a human DNA fragment, designated vk65.5, containing a V ⁇ gene segment; the deduced amino acid sequences of the V ⁇ coding regions are also shown; splicing and recombination signal sequences
  • Fig. 44 show the nucleotide sequence of a human DNA fragment, designated vk65.8, containing a V ⁇ gene segment; the deduced amino acid sequences of the V ⁇ coding regions are also shown; splicing and recombination signal sequences
  • Fig. 45 show the nucleotide sequence of a human DNA fragment, designated vk65.15, containing a V ⁇ gene segment; the deduced amino acid sequences of the V ⁇ coding regions are also shown; splicing and recombination signal sequences
  • Fig. 46 shows formation of a light chain minilocus by homologous recombination between two overlapping fragments which were co-injected.
  • Table 1 depicts the sequence of vector pGPe.
  • Table 2 depicts the sequence of gene V H 4 .8.
  • Table 3 depicts the detection of human IgM and IgG in the serum of transgenic mice of this invention.
  • Table 4 depicts sequences of VDJ joints.
  • Table 5 depicts the distribution of J segments incorporated into pHC1 transgene encoded transcripts to J segments found in adult human peripheral blood lymphocytes (PBL).
  • Table 6 depicts the distribution of D segments incorporated into pHC1 transgene encoded transcripts to D segments found in adult human peripheral blood lymphocytes (PBL).
  • Table 7 depicts the length of the CDR3 peptides from transcripts with in-frame VDJ joints in the pHC1 transgenic mouse and in human PBL.
  • Table 8 depicts the predicted amino acid sequences of the VDJ regions from 30 clones analyzed from a pHC1
  • Table 9 shows transgenic mice of line 112 that were used in the indicated experiments; (+) indicates the presence of the respective transgene, (++) indicates that the animal is homozygous for the J H D knockout transgene.
  • human immunoglobulins that are reactive with specific human antigens that are promising therapeutic and/or diagnostic targets.
  • producing human immunoglobulins that bind specifically with human antigens is problematic.
  • the immunized animal that serves as the source of B cells must make an immune response against the presented antigen.
  • the antigen presented In order for an animal to make an immune response, the antigen presented must be foreign and the animal must not be tolerant to the antigen.
  • self-tolerance will prevent an immunized human from making a substantial immune response to the human protein, since the only epitopes of the antigen that may be immunogenic will be those that result from polymorphism of the protein within the human population
  • B-cells for forming a hybridoma (a human in the illustrative given example) does make an immune response against an
  • One methodology that can be used to obtain human antibodies that are specifically reactive with human antigens is the production of a transgenic mouse harboring the human immunoglobulin transgene constructs of this invention.
  • transgenes containing all or portions of the human immunoglobulin heavy and light chain loci or transgenes containing synthetic "miniloci" (described infra, and in
  • transgenic nonhuman animal which comprise essential functional elements of the human heavy and light chain loci, are employed to produce a transgenic nonhuman animal.
  • a transgenic nonhuman animal will have the capacity to produce immunoglobulin chains that are encoded by human immunoglobulin genes, and additionally will be capable of making an immune response against human antigens.
  • transgenic animals can serve as a source of immune sera reactive with specified human antigens, and B-cells from such transgenic animals can be fused with myeloma cells to produce hybridomas that secrete monoclonal antibodies that are encoded by human, immunoglobulin genes and which are specifically reactive with human antigens.
  • transgenic mice containing various forms of immunoglobulin genes has been reported previously.
  • Rearranged mouse immunoglobulin heavy or light chain genes have been used to produce transgenic mice.
  • functionally rearranged human Ig genes including the ⁇ or ⁇ 1 constant region have been expressed in transgenic mice.
  • V-D-J or V-J not rearranged immunoglobulin genes have been variable, in some cases, producing incomplete or minimal rearrangement of the transgene.
  • immunoglobulin transgenes which undergo successful isotype switching between C H genes within a transgene.
  • antibody refers to a glycoprotein comprising at least two light polypeptide chains and two heavy polypeptide chains. Each of the heavy and light polypeptide chains contains a variable region (generally the amino terminal portion of the polypeptide chain) which
  • Each of the heavy and light polypeptide chains also comprises a constant region of the polypeptide chains (generally the carboxyl terminal portion) which may mediate the binding of the immunoglobulin to host tissues or factors including
  • a heterologous antibody is defined in relation to the transgenic non-human organism producing such an antibody. It is defined as an antibody having an amino acid sequence or an encoding DNA sequence corresponding to that found in an organism not consisting of the transgenic non-human animal.
  • hybrid antibody refers to an antibody having a light and heavy chains of different organismal origins.
  • an antibody having a human heavy chain associated with a murine light chain is a
  • isotype refers to the antibody class (e.g., IgM or IgG 1 ) that is encoded by heavy chain constant region genes.
  • isotype switching refers to the phenomenon by which the class, or isotype, of an antibody changes from one Ig class to one of the other Ig classes.
  • nonswitched isotype refers to the isotypic class of heavy chain that is produced when no isotype switching has taken place; the C H gene encoding the
  • nonswitched isotype is typically the first C H gene immediately downstream from the functionally rearranged VDJ gene.
  • switch sequence refers to those DNA sequences responsible for switch recombination.
  • a "switch donor” sequence typically a ⁇ switch region, will be 5' (i.e., upstream) of the construct region to be deleted during the switch recombination.
  • the "switch acceptor” region will be between the construct region to be deleted and the replacement constant region (e.g., ⁇ , ⁇ , etc.). As there is no specific site where recombination always occurs, the final gene sequence will typically not be predictable from the construct.
  • glycosylation pattern is defined as the pattern of carbohydrate units that are covalently attached to a protein, more specifically to an immunoglobulin protein.
  • a glycosylation pattern of a heterologous antibody can be characterized as being substantially similar to
  • glycosylation patterns which occur naturally on antibodies produced by the species of the nonhuman transgenic animal, when one of ordinary skill in the art- would recognize the glycosylation pattern of the heterologous antibody as being more similar to said pattern of glycosylation in the species of the nonhuman transgenic animal than to the species from which the C H genes of the transgene were derived.
  • telomere binding refers to the property of the antibody: (1) to bind to a predetermined antigen with an affinity of at least 1 x 10 7 M -1 , and (2) to preferentially bind to the predetermined antigen with an affinity that is at least twc-fold greater than its affinity for binding to a non-specific antigen (e.g., BSA, casein) other than the predetermined antigen.
  • a non-specific antigen e.g., BSA, casein
  • naturally-occurring refers to the fact that an object can be found in nature.
  • a polypeptide or polynucleotide sequence that is present in an organism (including viruses) that can be isolated from a source in nature and which has not been intentionally modified by man in the laboratory is naturally-occurring.
  • rearranged refers to a configuration of a heavy chain or light chain immunoglobulin locus wherein a V segment is positioned immediately adjacent to a D-J or J segment in a conformation encoding essentially a complete V H or V L domain, respectively.
  • immunoglobulin gene locus can be identified by comparison to germline DNA; a rearranged locus will have at least one recombined heptamer/nonamer homology element.
  • V segment configuration refers to the configuration wherein the V segment is not recombined so as to be immediately adjacent to a D or J segment.
  • the transgenes of the invention are constructed so as to produce isotype switching and one or more of the following: (1) high level and cell-type specific expression, (2) functional gene rearrangement, (3) activation of and response to allelic exclusion, (4) expression of a sufficient primary repertoire, (5) signal transduction, (6) somatic hypermutation, and (7) domination of the transgene antibody locus during the immune response.
  • the transgene need not activate allelic exclusion.
  • the transgene comprises a
  • transgenic non-human animals contain rearranged, unrearranged or a combination of rearranged and unrearranged heterologous immunoglobulin heavy and light chain transgenes in the
  • Each of the heavy chain transgenes comprises at least one C H gene.
  • the heavy chain transgene may contain functional isotype switch sequences, which are capable of supporting isotype switching of a heterologous transgene encoding multiple C H genes in B- cells of the transgenic animal.
  • Such switch sequences may be those which occur naturally in the germline immunoglobulin locus from the species that serves as the source of the transgene C H genes, or such switch sequences may be derived from those which occur in the species that is to receive the transgene construct (the transgeneic animal).
  • a human transgene construct that is used to produce a transgenic mouse may produce a higher frequency of isotype switching events if it incorporates switch sequences similar to those that occur naturally in the mouse heavy chain locus, as presumably the mouse switch sequences are optimized to
  • Switch sequences made be isolated and cloned by conventional cloning methods, or may be synthesized de novo from overlapping synthetic oligonucleotides designed on the basis of published sequence information relating to immunoglobulin switch region sequences (Mills et al., Nucl. Acids Res. 18:7305-7316 (1991);
  • heterologous heavy and light chain immunoglobulin transgenes are found in a significant fraction of the B-cells of the transgenic animal (at least 10 percent).
  • the transgenes of the invention include a heavy chain transgene comprising DNA encoding at least one variable gene segment, one diversity gene segment, one joining gene segment and at least one constant region gene segment.
  • the immunoglobulin light chain transgene comprises DNA encoding at least one variable gene segment, one joining gene segment and at least one constant region gene segment.
  • the gene segments encoding the light and heavy chain gene segments are
  • the transgene is constructed such that the individual gene segments are unrearranged, i.e., not rearranged so as to encode a functional immunoglobulin light or heavy chain.
  • unrearranged transgenes support
  • V, D, and J gene segments preferably support incorporation of all ora portion of a D region gene segment in the resultant
  • the transgenes comprise an unrearranged "mini-locus". Such transgenes typically comprise an unrearranged "mini-locus". Such transgenes typically comprise an unrearranged "mini-locus". Such transgenes typically comprise an unrearranged "mini-locus". Such transgenes typically comprise an unrearranged "mini-locus". Such transgenes typically comprise an unrearranged "mini-locus". Such transgenes typically comprise
  • the various regulatory sequences e.g. promoters, enhancers, class switch regions, splice-donor and splice- acceptor sequences for RNA processing, recombination signals and the like, comprise corresponding sequences derived from the heterologous DNA.
  • Such regulatory sequences may be incorporated into the transgene from the same or a related species of the non-human animal used in the invention.
  • human immunoglobulin gene segments may be combined in a transgene with a rodent immunoglobulin enhancer sequence for use in a transgenic mouse.
  • synthetic regulatory sequences may be incorporated into the transgene, wherein such synthetic regulatory sequences are not homologous to a
  • Synthetic regulatory sequences are designed according to consensus rules, such as, for example, those specifying the permissible sequences of a splice- acceptor site or a promoter/enhancer motif.
  • the invention also includes transgenic animals containing germ line cells having a heavy and light transgene wherein one of the said transgenes contains rearranged gene segments with the other containing unrearranged gene segments.
  • the rearranged transgene is a light chain immunoglobulin transgene and the unrearranged transgene is a heavy chain immunoglobulin transgene.
  • the basic structure of all immunoglobulins is based upon a unit consisting of two light polypeptide chains and two heavy polypeptide chains. Each light chain comprises two regions known as the variable light chain region and the constant light chain region. Similarly, the immunoglobulin heavy chain comprises two regions designated the variable heavy chain region and the constant heavy chain region.
  • the constant region for the heavy or light chain is encoded by genomic sequences referred to as heavy or light constant region gerie (C H ) segments.
  • C H heavy or light constant region gerie
  • the use of a particular heavy chain gene segment defines the class of immunoglobulin.
  • the ⁇ constant region gene segments define the IgM class of antibody whereas the use of a ⁇ , ⁇ 2, ⁇ 3 or ⁇ 4 constant region gene segment defines the IgG class of antibodies as well as the IgG subclasses IgG1 through IgG4.
  • the use of a ⁇ 1 or ⁇ 2 constant region gene segment defines the IgA class of antibodies as well as the subclasses IgA1 and IgA2.
  • the ⁇ and e constant region gene segments define the IgD and IgE antibodv classes, respectively.
  • immunoglobulin chains together contain the antigen binding domain of the antibody. Because of the need for diversity in this region of the antibody to permit binding to a wide range of antigens, the DNA encoding the initial or primary
  • repertoire variable region comprises a number of different DNA segments derived from families of specific variable region gene segments.
  • families comprise variable (V) gene segments and joining (J) gene segments.
  • V variable
  • J joining
  • the initial variable region of the light chain is encoded by one V gene segment and one J gene segment each selected from the family of V and J gene segments contained in the genomic DNA of the organism.
  • the DNA encoding the initial or primary repertoire variable region of the heavy chain comprises one heavy chain V gene segment, one heavy chain diversity (D) gene segment and one J gene segment, each selected from the appropriate V, D and J families of
  • a heavy chain transgene include cis-acting sequences that support functional V-D-J rearrangement that can incorporate all or part of a D region gene sequence in a rearranged V-D-J gene sequence. Typically, at least about 1 percent of
  • transgene-encoded heavy chains include recognizable D region sequences in the V region.
  • at least about 10 percent of transgene-encoded V regions include recognizable D region sequences, more preferably at least about 30 percent, and most preferably more than 50 percent include recognizable D region sequences.
  • a recognizable D region sequence is generally at least about eight consecutive nucleotides corresponding to a sequence present in a D region gene segment of a heavy chain transgene and/or the amino acid sequence encoded by such D region nucleotide sequence. For example, if a transgene includes the D region gene DHQ52, a transgene-encoded mRNA containing the sequence 5'-TAACTGGG-3' located in the V region between a V gene segment sequence and a J gene segment
  • a transgene includes the D region gene DHQ52
  • a transgeneencoded heavy chain polypeptide containing the amino acid sequence -DAF- located in the V region between a V gene segment amino acid sequence and a J gene segment amino acid sequence is recognizable as containing a D region sequence, specifically a DHQ52 sequence.
  • D region sequences may be recognizable but may not correspond identically to a consecutive D region sequence in the transgene.
  • CTAAXTGGGG-3' where X is A, T, or G, and which is located in a heavy chain V region and flanked by a V region gene sequence and a J region gene sequence, can be recognized as
  • polypeptide sequences -DAFDI-, -DYFDY-, or -GAFDI- located in a V region and flanked on the amino-terminal side by an amino acid sequence encoded by a transgene V gene sequence and flanked on the carboxyterminal side by an amino acid sequence encoded by a transgene J gene sequence is recognizable as a D region sequence.
  • an amino acid sequence or nucleotide sequence is recognizable as a D region sequence if: (1) the sequence is located in a V region and is flanked on one side by a V gene sequence (nucleotide sequence or deduced amino acid sequence) and on the other side by a J gene sequence (nucleotide sequence or deduced amino acid sequence) and (2) the sequence is substantially identical or substantially similar to a known D gene sequence (nucleotide sequence or encoded amino acid sequence).
  • substantially identical denotes a characteristic of a polypeptide sequence or nucleic acid sequence, wherein the polypeptide sequence has at least 50 percent sequence identity compared to a reference sequence, and the nucleic acid sequence has at least 70 percent sequence identity compared to a reference sequence.
  • the percentage of sequence identity is calculated excluding small deletions or additions which total less than 35 percent of the reference sequence.
  • the reference sequence may be a subset of a larger sequence, such as an entire D gene; however, the reference sequence is at least 8 nucleotides long in the case of
  • the reference sequence is at least 8 to 12 nucleotides or at least 3 to 4 amino acids, and preferably the reference sequence is 12 to 15 nucleotides or more, or at least 5 amino acids.
  • substantially similarity denotes a characteristic of an polypeptide sequence, wherein the
  • polypeptide sequence has at least 80 percent similarity to a reference sequence.
  • the percentage of sequence similarity is calculated by scoring identical amino acids or positional conservative amino acid substitutions as similar.
  • positional conservative amino acid substitution is one that can result from a single nucleotide substitution; a first amino acid is replaced by a second amino acid where a codon for the first amino acid and a codon for the second amino acid can differ by a single nucleotide substitution.
  • sequence -Lys-Glu-Arg-Val- is substantially similar to the sequence -Asn-Asp-Ser-Val-, since the codon sequence -AAA-GAA-AGA-GUU- can be mutated to -AAC-GAC-AGC-GUU- by introducing only 3 substitution mutations, single
  • the reference sequence may be a subset of a larger sequence. such as an entire D gene; however, the reference sequence is at least 4 amino residues long. Typically, the reference sequence is at least 5 amino acids, and preferably the
  • reference sequence is 6 amino acids or more.
  • immunoglobulin gene segments the V, D, J and constant (C) gene segments are found, for the most part, in clusters of V, D, J and C gene segments in the precursors of primary
  • RSS's recombination signal sequences
  • V, D and J segments flank recombinationally competent V, D and J segments.
  • RSS's necessary and sufficient to direct recombination comprise a dyad-symmetric heptamer, an AT-rich nonamer and an intervening spacer region of either 12 or 23 base pairs.
  • each V and D gene segment comprises the sequence CACAGTG or its analogue followed by a spacer of unconserved sequence and then a nonamer having the sequence ACAAAAACC or its analogue. These sequences are found on the J, or downstream side, of each V and D gene segment. Immediately preceding the germline D and J segments are again two recombination signal sequences, first the nonamer and then the heptamer again separated by an unconserved sequence. The heptameric and nonameric sequences following a V L , V H or D segment are complementary to those preceding the J L , D or J H segments with which they recombine. The spacers between the heptameric and nonameric sequences are either 12 base pairs long or between 22 and 24 base pairs long.
  • variable recombination between the V and J segments in the light chain and between the D and J segments of the heavy chain.
  • Such variable recombination is generated by variation in the exact place at which such segments are joined.
  • variation in the light chain typically occurs within the last codon of the V gene segment and the first codon of the J segment.
  • Similar imprecision in joining occurs on the heavy chain chromosome between the D and J H segments and may extend over as many as 10 nucleotides.
  • nucleotides may be inserted between the D and J H and between the V H and D gene segments which are not encoded by genomic DNA.
  • the addition of these nucleotides is known as N-region diversity.
  • RNA transcript After VJ and/or VDJ rearrangement, transcription of the rearranged variable region and one or more constant region gene segments located downstream from the rearranged variable region produces a primary RNA transcript which upon
  • RNA splicing results in an mRNA which encodes a full length heavy or light immunoglobulin chain.
  • heavy and light chains include a leader signal sequence to effect secretion through and/or insertion of the immunoglobulin into the transmembrane region of the B-cell.
  • the DNA encoding this signal sequence is contained within the first exon of the V segment used to form the variable region of the heavy or light immunoglobulin chain.
  • Appropriate regulatory sequences are also present in the mRNA to control translation of the mRNA to produce the encoded heavy and light immunoglobulin
  • polypeptides which upon proper association with each other form an antibody molecule.
  • variable region gene segments and the variable recombination which may occur during such joining is the production of a primary antibody repertoire.
  • each B-cell which has differentiated to this stage produces a single primary repertoire antibody.
  • cellular events occur which suppress the functional
  • allelic exclusion The process by which diploid B-cells maintain such mono-specificity is termed allelic exclusion.
  • B-cell clones expressing immunoglobulins from within the set of sequences comprising the primary repertoire are immediately available to respond to foreign antigens. Because of the limited diversity generated by simple VJ and VDJ joining, the antibodies produced by the so-called primary response are of relatively low affinity.
  • Two different types of B-cells make up this initial response: precursors of primary antibody-forming cells and precursors of secondary repertoire B-cells (Linton et al., Cell 59:1049-1059 (1989)).
  • the first type of B-cell matures into IgM-secreting plasma cells in response to certain antigens.
  • the other B-cells respond to initial exposure to antigen by entering a T-cell dependent maturation pathway.
  • the structure of the antibody molecule on the cell surface changes in two ways: the constant region switches to a non-IgM subtype and the sequence of the variable region can be modified by multiple single amino acid substitutions to produce a higher affinity antibody molecule.
  • variable region of a heavy or light Ig chain contains an antigen binding domain. It has been determined by amino acid and nucleic acid
  • CDRl, CDR2 and CDR3 also referred to as hypervariable regions 1, 2 and 3
  • the CDR1 and CDR2 are located within the variable gene segment whereas the CDR3 is largely the result of recombination between V and J gene segments or V, D and J gene segments.
  • Those portions of the variable region which do not consist of CDR1, 2 or 3 are commonly referred to as framework regions designated FR1, FR2, FR3 and FR4. See Fig. 1.
  • framework regions designated FR1, FR2, FR3 and FR4
  • rearranged DNA is mutated to give rise to new clones with altered Ig molecules.
  • Those clones with higher affinities for the foreign antigen are selectively expanded by helper
  • Transgenic non-human animals in one aspect of the invention are produced by introducing at least one of the immunoglobulin transgenes of the invention (discussed
  • non-human animals which are used in the invention generally comprise any mammal which is capable of rearranging immunoglobulin gene segments to produce a primary antibody response.
  • nonhuman transgenic animals may include, for example, transgenic pigs, transgenic rats, transgenic rabbits, transgenic cattle, and other transgenic animal species, particularly mammalian species, known in the art.
  • particularly preferred non-human animal is the mouse or other members of the rodent family.
  • mice any non-human mammal which is capable of mounting a primary and secondary antibody response may be used.
  • Such animals include non-human primates, such as chimpanzee, bovine, ovine, and porcine species, other members of the rodent family, e.g. rat, as well as rabbit and guinea pig.
  • Particular preferred animals are mouse, rat, rabbit and guinea pig, most preferably mouse.
  • various gene segments from the human genome are used in heavy and light chain transgenes in an unrearranged form.
  • such transgenes are introduced into mice.
  • the unrearranged gene segments of the light and/or heavy chain transgene have DNA sequences unique to the human species which are
  • the transgenes comprise rearranged heavy and/or light
  • transgenes corresponding to functionally rearranged VDJ or VJ segments contain immunoglobulin DNA sequences which are also clearly distinguishable from the endogenous immunoglobulin gene segments in the mouse.
  • sequences may be detected in the transgenic non-human animals of the invention with antibodies specific for immunoglobulin epitopes encoded by human immunoglobulin gene segments.
  • Transgenic B-cells containing unrearranged transgenes from human or other species functionally recombine the appropriate gene segments to form functionally rearranged light and heavy chain variable regions. It will be readily apparent that the antibody encoded by such rearranged
  • transgenes has a DNA and/or amino acid sequence which i ⁇ heterologous to that normally encountered in the nonhuman animal used to practice the indention.
  • an "unrearranged immunoglobulin heavy chain transgene” comprises DNA encoding at least one variable gene segment, one diversity gene segment, one joining gene segment and one constant region gene segment.
  • Each of the gene segments of said heavy chain transgene are derived from, or has a sequence corresponding to, DNA encoding
  • an "unrearranged immunoglobulin light chain transgene” comprises DNA encoding at least one variable gene segment, one joining gene segment and at least one constant region gene segment wherein each gene segment of said light chain tr; nsgene is derived from, or has a sequence corresponding to, DNA encoding immunoglobulin light chain gene segments from a species not consisting of the non-human animal into which said light chain transgene is introduced.
  • Such heavy and light chain transgenes in this aspect of the invention contain the above-identified gene segments in an unrearranged form.
  • interposed between the V, D and J segments in the heavy chain transgene and between the V and J segments on the light chain transgene are appropriate.
  • transgenes also include appropriate RNA splicing signals to join a constant region gene segment with the VJ or VDJ
  • switch regions are incorporated upstream from each of the constant region gene segments and downstream from the variable region gene segments to permit recombination between such constant regions to allow for immunoglobulin class switching, e.g. from IgM to IgG.
  • switch regions are incorporated upstream from each of the constant region gene segments and downstream from the variable region gene segments to permit recombination between such constant regions to allow for immunoglobulin class switching, e.g. from IgM to IgG.
  • immunoglobulin transgenes also contain transcription control sequences including promoter regions situated upstream from the variable region gene segments which typically contain TATA motifs.
  • a promoter region can be defined approximately as a DNA sequence that, when operably linked to a downstream sequence, can produce transcription of the downstream
  • Promoters may require the presence of additional linked cis-acting sequences in order to produce efficient transcription.
  • other sequences that participate in the transcription of sterile transcripts are preferably included. Examples of sequences that participate in
  • sequences typically include about at least 50 bp immediately upstream of a switch region, preferably about at least 200 bp upstream of a switch region; and more preferably about at least 200-1000 bp or more upstream of a switch region.
  • Suitable sequences occur immediately upstream of the human S ⁇ 1 , S ⁇ 2 , S ⁇ 3 , S ⁇ 4 , S ⁇ 1 , S ⁇ 2 , and S ⁇ switch regions, although the sequences immediately upstream of the human S ⁇ 1 , and S ⁇ 3 switch regions are preferable.
  • interferon (IFN) inducible transcriptional regulatory elements such as IFN-inducible enhancers, are preferably included immediately upstream of transgene switch sequences.
  • promoters In addition to promoters, other regulatory sequences which function primarily in B-lineage cells are used. Thus, for example, a light chain enhancer sequence situated
  • regulatory enhancers are used to maximize the transcription and translation of the transgene so as to induce allelic exclusion and to provide relatively high levels of transgene expression.
  • regulatory control sequences have been generically described, such regulatory sequences may be heterologous to the nonhuman animal being derived from the genomic DNA from which the heterologous transgene immunoglobulin gene segments are obtained. Alternately, such regulatory gene segments are derived from the corresponding regulatory sequences in the genome of the non-human animal, or closely related species, which contains the heavy and light transgene.
  • gene segments are derived from human beings.
  • the transgenic non-human animals harboring such heavy and light transgenes are capable of mounting an Ig-mediated immune response to a specific antigen administered to such an animal.
  • B-cells are produced within such an animal which are capable of producing heterologous human antibody.
  • an appropriate monoclonal antibody e.g. a hybridoma
  • a source of therapeutic human monoclonal antibody is provided.
  • Such human Mabs have significantly reduced immunogenicity when therapeutically administered to humans.
  • transgenic nonhuman animals contain functionally at least one rearranged
  • heterologous heavy chain immunoglobulin transgene in the germline of the transgenic animal.
  • Such animals contain primary repertoire B-cells that express such rearranged heavy transgenes.
  • B-cells preferably are capable of undergoing somatic mutation when contacted with an antigen to form a heterologous antibody having high affinity and specificity for the antigen.
  • Said rearranged transgenes will contain at least two C H genes and the associated sequences required for isotype switching.
  • the invention also includes transgenic animals containing germ line cells having heavy and light transgenes wherein one of the said transgenes contains rearranged gene segments with the other containing unrearranged gene segments.
  • the heavy chain transgenes shall have at least two C H genes and the associated sequences required for isotype switching.
  • the invention further includes methods for generating a synthetic variable region gene segment repertoire to be used in the transgenes of the invention.
  • the method comprises generating a population of immunoglobulin V segment DNAs wherein each of the V segment DNAs encodes an
  • immunoglobulin V segment contains at each end a cleavage recognition site of a restriction endonuclease.
  • Such synthetic variable region heavy chain transgenes shall have at least two C H genes and the associated sequences required for isotype switching.
  • the cell In the development of a B lymphocyte, the cell initially produces IgM with a binding specificity determined by the productively rearranged V H and V L regions.
  • each B cell and its progeny cells synthesize antibodies with the same L and H chain V regions, but they may switch the isotype of the H chain.
  • This gene rearrangement process typically occurs by recombination between so called switch segments located immediately upstream of each heavy chain gene (except S) .
  • the individual switch segments are between 2 and 10 kb in length, and consist primarily of short repeated sequences.
  • the switch (S) region of the ⁇ gene, S ⁇ is located about 1 to 2 kb 5' to the coding sequence and is composed of numerous tandem repeats of sequences of the form
  • All the sequenced S regions include numerous occurrences of the pentamers GAGCT and GGGGT that are the basic repeated elements of the S gene (T. Nikaido et al., J. Biol. Chem. 257:7322- 7329 (1982) which is incorporated herein by reference); in the other S regions these pentamers are not precisely tandemly repeated as in S ⁇ , but instead are embedded in larger repeat units.
  • the S ⁇ 1 region has an additional higher-order
  • Switch regions of human H chain genes have been found to be very similar to their mouse homologs. Indeed, similarity between pairs of human and mouse clones 5' to the C H genes has been found to be confined to the S regions, a fact that confirms the biological significance of these regions.
  • a switch recombination between ⁇ and a genes produces a composite S ⁇ -S ⁇ sequence.
  • the switch machinery can apparently accommodate different alignments of the repeated homologous regions of germline S precursors and then join the sequences at different positions within the alignment.
  • the switch machinery can apparently accommodate different alignments of the repeated homologous regions of germline S precursors and then join the sequences at different positions within the alignment.
  • cytokines might upregulate isotype-specific recombinases, it is also possible that the same enzymatic machinery catalyzes switches to all isotypes and that specificity lies in
  • T-cell-derived lymphokines IL-4 and IFN ⁇ have been shown to specifically promote the expression of certain isotypes: IL-4 decreases IgM, IgG2a, IgG2b, and IgG3
  • IgE and IgG1 expression increases IgE and IgG1 expression; while IFN ⁇ selectively stimulates IgG2a expression and antagonizes the IL-4-induced increase in IgE and IgG1 expression (Coffman et al., J. Immunol. 136:949-954 (1986) and Snapper et al.,
  • lymphokines actually promote switch recombination.
  • the observed induction of the ⁇ I sterile transcript by IL-4 and inhibition by IFN- ⁇ correlates with the observation that IL-4 promotes class switching to ⁇ 1 in B-cells in culture, while IFN- ⁇ inhibits ⁇ 1 expression. Therefore, the inclusion of regulatory sequences that affect the transcription of sterile transcripts may also affect the rate of isotype switching. For example, increasing the transcription of a particular sterile transcript typically can be expected to enhance the frequency of isotype switch
  • transgenes incorporate transcriptional regulatory sequences within about 1-2 kb upstream of each switch region that is to be utilized for isotype switching.
  • These transcriptional regulatory sequences preferably include a promoter and an enhancer element, and more preferably include the 5' flanking (i.e., upstream) region that is naturally associated (i.e., occurs in germline configuration) with a switch region. This 5' flanking (i.e., upstream) region that is naturally associated (i.e., occurs in germline configuration) with a switch region. This 5'
  • flanking region is typically about at least 50 nucleotides in length, preferably about at least 200 nucleotides in length, and more preferably at least 500-1000 nucleotides.
  • each switch region incorporated in the transgene construct have the 5' flanking region that occurs immediately upstream in the naturally occurring germline configuration.
  • transgene function An important requirement for transgene function is the generation of a primary antibody repertoire that is diverse enough to trigger a secondary immune response for a wide range of antigens.
  • V region proximal constant regions are deleted leading to the
  • RNA splicing For each heavy chain . class, alternative patterns of RNA splicing give rise to both transmembrane and secreted immunoglobulins.
  • the human heavy chain locus consists of
  • immunoglobulin heavy and light chain transgenes comprise unrearranged genomic DNA from humans.
  • a preferred transgene comprises a NotI fragment having a length between 670 to 830 kb. The length of this fragment is ambiguous because the 3' restriction site has not been accurately mapped. It is known, however, to reside between the ⁇ l and gene segments. This fragment contains members of all six of the known V H families, the D and J gene segments, as well as the ⁇ , ⁇ , ⁇ 3 , ⁇ 1 and ⁇ 1 constant regions (Berman et al., EMBO J. 7:727-738 (1988), which is incorporated herein by reference).
  • IgM B-cell development
  • IgG 1 switched heavy chain class
  • a genomic fragment containing all of the necessary gene segments and regulatory sequences from a human light chain locus may be similarly constructed. Such transgenes are constructed as described in the Examples.
  • immunoglobulin heavy chain locus may be formed in vivo in the non-human animal during transgenesis.
  • Such in vivo transgene construction is produced by introducing two or more
  • fragments have DNA sequences which are substantially identical
  • In vivo transgene construction can be used to form any number of immunoglobulin transgenes which because of their size are otherwise difficult, or impossible, to make or manipulate by present technology.
  • in vivo transgene construction is useful to generate immunoglobulin transgenes which are larger than DNA fragments which may be manipulated by YAC vectors (Murray and Szostak, Nature 305: 189-193
  • Such in vivo transgene construction may be used to introduce into a non-human animal substantially the entire immunoglobulin loci from a species not consisting of the transgenic non-human animal.
  • in vivo homologous recombination may also be utilized to form "mini-locus" transgenes as described in the Examples.
  • portions of the DNA fragments preferably comprise about 500 bp to about 2000 bp, most preferably 1.0 kb to 2.0 kb.
  • immunoglobulin minilocus refers to a DNA sequence (which may be within a longer
  • DNA sequence usually of less than about 150 kb, typically between about 25 and 100 kb, containing at least one each of the following: a functional variable (V) gene segment, a functional joining (J) region segment, at least one functional constant (C) region gene segment, and--if it is a heavy chain minilocus--a functional diversity (D) region segment, such that said DNA sequence contains at least one substantial discontinuity (e.g., a deletion, usually of at least about 2 to 5 kb, preferably 10-25 kb or more, relative to the
  • a light chain minilocus transgene will be at least 25 kb in length, typically 50 to 60 kb.
  • a heavy chain transgene will typically be about 70 to 80 kb in length, preferably at least about 60 kb with two
  • the individual elements of the minilocus are preferably in the germline configuration and capable of undergoing gene rearrangement in the pre-B cell of a
  • transgenic animal so as to express functional antibody
  • a heavy chain minilocus comprising at least two C H genes and the requisite switching sequences is typ.cally c pable of undergoing isotype switching, so that functional antibody molecules of different immunoglobulin classes will be generated.
  • switching may occur in vivo in B-cells residing within the transgenic nonhuman animal, or may occur in cultured cells of the B-cell lineage which have been explanted from the
  • immunoglobulin heavy chain transgenes comprise one or more of each of the V H , D, and J H gene segments and two or more of the C H genes. At least one of each appropriate type gene segment is
  • the transgene contain at least one ⁇ gene segment and at least one other constant region gene segment, more preferably a ⁇ gene segment, and most preferably ⁇ 3 or ⁇ 1.
  • constant region gene segments may also be used such as those which encode for the production of IgD, IgA and IgE.
  • transgenes wherein the order of occurrence of heavy chain C H genes will be different from the naturally-occurring spatial order found in the germline of the species serving as the donor of the C H genes.
  • C H genes from more than one individual of a species (e.g., allogeneic C H genes) and incorporate said genes in the
  • the resultant transgenic nonhuman animal may then, in some embodiments, make antibodies of various classes including all of the allotypes represented in the species from which the transgene C H genes were obtained.
  • C H gene combinations will produce a transgenic nonhuman animal which may produce antibodies of various classes corresponding to C H genes from various
  • Transgenic nonhuman animals containing interspecies C H transgenes may serve as the source of B-cells for
  • the heavy chain J region segments in the human comprise six functional J segments and three pseudo genes clustered in a 3 kb stretch of DNA. Given its relatively compact size and the ability to isolate these segments
  • J region gene segments be used in the mini-locus construct. Since this fragment spans the region between the ⁇ and ⁇ genes, it is likely to contain all of the 3' cis-linked regulatory elements required for ⁇ expression. Furthermore, because this fragment includes the entire J region, it contains the heavy chain enhancer and the ⁇ switch region (Mills et al., Nature 306:809 (1983); Yancopoulos and Alt, Ann. Rev. Immunol. 4:339-368 (1986), which are incorporated herein by reference).
  • the human D region consists of 4 or 5 homologous 9 kb subregions, linked in tandem (Siebenlist, et al. (1981), Nature, 294, 631-635). Each subregion contains up to 10 individual D segments. Some of these segments have been mapped- and are shown in Fig. 4.
  • Two different strategies are used to generate a mini-locus D region. The first strategy involves using only those D segments located in a short contiguous stretch of DNA that includes one or two of the repeated D subregions. A candidate is a single 15 kb fragment that contains 12 individual D segments. This piece of DNA consists of 2 contiguous EcoRI fragments and has been
  • D segments should be sufficient for a primary repertoire.
  • an alternative strategy is to ligate together several non-contiguous D-segment containing fragments, to produce a smaller piece of DNA with a greater number of segments. Additional D-segment genes can be identified, for example, by the presence of characteristic flanking nonamer and heptamer sequences, supra, and by reference to the
  • At least one, and preferably more than one V gene segment is used to construct the heavy chain minilocus
  • V segments, D segments, J segments, and C genes, with or without flanking sequences can be isolated as described in PCT Publication No. WO 92/03918, published March 19, 1992.
  • a minilocus light chain transgene may be similarly constructed from the human ⁇ or ⁇ immunoglobulin locus.
  • an immunoglobulin heavy chain minilocus transgene construct e.g., of about 75 kb, encoding V, D, J and constant region sequences can be formed from a plurality of DNA fragments, with each sequence being substantially homologous to human gene sequences.
  • the sequences are operably linked to transcription regulatory sequences and are capable of undergoing rearrangement.
  • constant region sequences e.g., ⁇ and ⁇
  • switch regions switch recombination also occurs.
  • An exemplary light chain transgene construct can be formed similarly from a plurality of DNA fragments, substantially homologous to human DNA and capable of undergoing
  • transgene constructs that are intended to undergo class switching should include all of the cis-acting sequences necessary to regulate sterile transcripts.
  • Switch regions and upstream promoters and regulatory sequences are preferred cis-acting sequences that are included in transgene constructs capable of isotype switching.
  • switch regions can be linked upstream of (and adjacent to) C H genes that do not naturally occur next to the particular switch region.
  • a human ⁇ 1 switch region may be linked upstream from a human ⁇ 2 C H gene, or a murine ⁇ 1 switch may be linked to a human C H gene.
  • An alternative method for obtaining ⁇ on-classical isotype switching (e.g., (S-associated deletion) in transgenic mice involves the inclusion of the 400 bp direct repeat sequences ( ⁇ and e ⁇ ) that flank the human ⁇ gene (Yasui et al., Eur. J. Immunol. 19:1399 (1989)).
  • Heavy chain transgenes can be
  • V H is a heavy chain variable region gene segment
  • D is a heavy chain D (diversity) region gene segment
  • J H is a heavy chain J (joining) region gene segment
  • S D is a donor region segment capable of participating in a recombination event with the S a acceptor region
  • C 1 is a heavy chain constant region gene segment encoding an isotype utilized in for B cell development (e.g., ⁇ or ⁇ ) ,
  • T is a cis-acting transcriptional regulatory region
  • S A is an acceptor region segment capable of participating in a recombination event with selected S D donor region segments, such that isotype switching occurs
  • C 2 is a heavy chain constant region gene segment encoding an isotype other than ⁇ (e.g., ⁇ 1 , ⁇ 2 , ⁇ 3 , ⁇ 4 , ⁇ 1 , ⁇ 2 , ⁇ ).
  • V H , D, J H , S D , C 1 , T, S A , and C Z segments may be selected from various species, preferably mammalian species, and more preferably from human and murine germline DNA.
  • V H segments may be selected from various species, but are preferably selected from V H segments that occur naturally in the human germline, such as V H251 . Typically about 2 V H gene segments are included, preferably about 4 V H segments are included, and most preferably at least about 10 V H segments are included.
  • At least one D segment is typically included, although at least 10 D segments are preferably included, and some embodiments include more than ten D segments. Some preferred embodiments include human D segments.
  • At least one J H segment is incorporated in the transgene, although it is preferable to include about six J H segments, and some preferred embodiments include more than about six J H segments. Some preferred embodiments include human J H segments, and further preferred embodiments include six human J H segments and no nonhuman J H segments.
  • S D segments are donor regions capable of
  • S D and S A are switch. regions such as S ⁇ , s ⁇ 1 , S ⁇ 2 . S ⁇ 3 , S ⁇ 4 , S ⁇ , S ⁇ 2 , and S ⁇ .
  • the switch regions are murine or human, more preferably S D is a human or murine S and S A is a human or murine S ⁇ 1 .
  • S D and S A are preferably the 400 basepair direct repeat sequences that flank the human ⁇ gene.
  • C 1 segments are typically ⁇ or ⁇ genes, preferably a ⁇ gene, and more preferably a human or murine ⁇ gene.
  • T segments typically include S' flanking sequences that are adjacent to naturally occurring (i.e., germline) switch regions. T segments typically at least about at least 50 nucleotides in length, preferably about at least 200 nucleotides in length, and more preferably at least 500-1000 nucleotides in length. Preferably T segments are 5' flanking sequences that occur immediately upstream of human or murine switch regions in a germline configuration. It is also evident to those of skill in the art that T segments may comprise cis-acting transcriptional regulatory sequences that do not occur naturally in an animal germline (e.g., viral enhancers and promoters such as those found in SV40,
  • adenovirus and other viruses that infect eukaryotic cells.
  • C 2 segments are typically a ⁇ 1 , ⁇ 2 , ⁇ 3 , ⁇ 4 , ⁇ 1 , ⁇ 2 , or ⁇ C H gene, preferably a human C H gene of these isotypes, and more preferably a human ⁇ 1 or ⁇ 3 gene.
  • Murine ⁇ 2a and ⁇ 2b may also be used, as may downstream (i.e., switched) isotype genes form various species.
  • the total length of the transgene will be typically 150 kilo basepairs or less.
  • the transgene will be other than a native heavy chain Ig locus.
  • deletion of unnecessary regions or substitutions with corresponding regions from other species will be present.
  • transgenic nonhuman animal The occurrence of isotype switching in a transgenic nonhuman animal may be identified by any method known to those in the art. Preferred embodiments include the following, employed either singly or in combination:
  • detection of mRNA transcripts that contain a sequence homologous to at least one transgene downstream C H gene other than ⁇ and an adjacent sequence homologous to a transgene V H - D H -J H rearranged gene; such detection may be by Northern hybridization, S 1 nuclease protection assays, PCR
  • detection in DNA from B-cells of the transgenic animal or in genomic DNA from hybridoma cells, of DNA rearrangements consistent with the occurrence of isotype switching in the transgene, such detection may be accomplished by Southern blot hybridization, PCR amplification, genomic cloning, or other method; or
  • each transgenic line may represent a
  • transgenes are typically integrated into host chromosomal DNA, most usually into germline DNA and propagated by subsequent breeding of germline transgenic breeding stock animals. However, other vectors and transgenic methods known in the present art or subsequently developed may be substituted as appropriate and as desired by a
  • Endogenous Immunoglobulin Loci The expression of successfully rearranged immunoglobulin heavy and light transgenes is expected to have a dominant effect by suppressing the rearrangement of the endogenous immunoglobulin genes in the transgenic nonhuman animal.
  • Another way to generate a nonhuman that is devoid of endogenous antibodies is by mutating the endogenous immunoglobulin loci. Using embryonic stem cell technology and homologous recombination, the endogenous immunoglobulin repertoire can be readily eliminated. The following describes the functional description of the mouse immunoglobulin loci.
  • the vectors and methods disclosed, however, can be readily adapted for use in other non-human animals.
  • this technology involves the inactivation of a gene, by homologous recombination, in a pluripotent cell line that is capable of differentiating into germ cell tissue.
  • a DNA construct that contains an altered, copy of a mouse immunoglobulin gene is introduced into the nuclei of embryonic stem cells. In a portion of the cells, the introduced DNA recombines with the endogenous copy of the mouse gene,
  • the mouse ⁇ locus contributes to only 5% of the immunoglobulins, inactivation of the heavy chain and/or ⁇ -light chain loci is sufficient. There are three ways to disrupt each of these loci, deletion of the J region, deletion of the J-C intron enhancer, and disruption of constant region coding sequences by the introduction of a stop codon. The last option is the most straightforward, in terms of DNA construct design. Elimination of the ⁇ gene disrupts B-cell maturation thereby preventing class switching to any of the functional heavy chain segments. The strategy for knocking out these loci is outlined below.
  • neomycin resistance gene from the plasmid pMCIneo is inserted into the coding region of the target gene.
  • the pMCIneo insert uses a hybrid viral promoter/enhancer sequence to drive neo expression. This promoter is active in embryonic stem cells. Therefore, neo can be used as a selectable marker for integration of the knock-out construct.
  • the HSV thymidine kinase (tk) gene is added to the end of the construct as a negative selection marker against random insertion events (Zijlstra, et al., supra.).
  • a preferred strategy for disrupting the heavy chain locus is the elimination of the J region. This region is fairly compact in the mouse, spanning only 1.3 kb.
  • the heavy-chain locus is knocked out by disrupting the coding region of the ⁇ gene.
  • This approach involves the same 15 kb Kpnl fragment used in the previous approach.
  • the 1.1 kb insert from pMCIneo is inserted at a unique BamHI site in exon II, and the HSV tk gene added to the 3' Kpnl end. Double crossover events on either side of the neo insert, that eliminate the tk gene, are then selected for. These are detected from pools of selected clones by PCR amplification.
  • One of the PCR primers is derived from neo sequences and the other from mouse sequences outside of the targeting vector. The functional disruption of the mouse immunoglobulin loci is presented in the Examples.
  • an alternative method for preventing the expression of an endogenous Ig locus is suppression.
  • Suppression of endogenous Ig genes may be accomplished with antisense RNA produced from one or more integrated transgenes, by antisense oligonucleotides, and/or by administration of antisera
  • Antisense RNA transgenes can be employed to
  • Antisense polynucleotides are polynucleotides that: (1) are complementary to all or part of a reference sequence, such as a sequence of an endogenous Ig C H or C L region, and (2) which specifically hybridize to a
  • complementary target sequence such as a chromosomal gene locus- or a Ig mRNA.
  • polynucleotides may include nucleotide substitutions,
  • Complementary antisense polynucleotides include soluble antisense RNA or DNA oligonucleotides which can hybridize specifically to
  • An antisense sequence is a
  • polynucleotide sequence that is complementary to at least one immunoglobulin gene sequence of at least about 15 contiguous nucleotides in length, typically at least 20 to 30 nucleotides in length, and preferably more than about 30 nucleotides in length.
  • antisense sequences may have substitutions, additions, or deletions as compared to the complementary immunoglobulin gene sequence, so long as
  • an antisense sequence is complementary to an endogenous immunoglobulin gene sequence that encodes, or has the potential to encode after DNA
  • sense sequences corresponding to an immunoglobulin gene sequence may function to suppress expression, particularly by interfering with transcription.
  • antisense polynucleotides therefore inhibit production of the encoded polypeptide (s).
  • antisense polynucleotides that inhibit transcription and/or translation of one or more endogenous Ig loci can alter the capacity and/or specificity of a non-human animal to produce immunoglobulin chains encoded by endogenous Ig loci.
  • Antisense polynucleotides may be produced from a heterologous expression cassette in a transfectant cell or transgenic cell, such as a transgenic pluripotent
  • the antisense polynucleotides may comprise soluble oligonucleotides that are administered to the external milieu, either in culture medium in vitro or in the circulatory system or interstitial fluid in vivo. Soluble antisense polynucleotides present in the external milieu have been shown to gain access to the
  • the antisense polynucleotides comprise methylphosphonate moieties, alternatively phosphorothiolates or O-methylribonucleotides may be used, and chimeric
  • oligonucleotides may also be used (Dagle et al. (1990) Nucleic Acids Res. 18: 4751). For some applications, antisense
  • oligonucleotides may comprise polyamide nucleic acids (Nielsen et al. (1991) Science 254: 1497). For general methods
  • Antisense polynucleotides complementary to one or more sequences are employed to inhibit transcription, RNA processing, and/or translation of the cognate mRNA species andthereby effect a reduction in the amount of the respective encoded polypeptide.
  • Such antisense polynucleotides can provide a therapeutic function by inhibiting the formation of one or more endogenous Ig chains in vivo.
  • the antisense polynucleotides of this invention are selected so as to hybridize preferentially to endogenous Ig sequences at physiological conditions in vivo. Most typically, the
  • selected antisense polynucleotides will not appreciably hybridize to heterologous Ig sequences encoded by a heavy or light chain transgene of the invention (i.e., the antisense oligonucleotides will not inhibit transgene Ig expression by more than about 25 to 35 percent).
  • Partial or complete suppression of endogenous Ig chain expression can be produced by injecting mice with antisera against one or more endogenous Ig chains (Weiss et al. (1984) Proc. ⁇ atl. Acad. Sci. (U.S.A.) 81 211, which is incorporated herein by reference).
  • Antisera are selected so as to react specifically with one or more endogenous Ig chains but to have minimal or no cross-reactivity with heterologous Ig chains encoded by an Ig transgene of the invention.
  • administration of selected antisera according to a schedule as typified by that of Weiss et al. op.cit. will suppress
  • nucleic acids the term "substantial homology” indicates" that two nucleic acids, or de gnated sequences thereof, when optimally aligned and compared, are identical, with appropriate nucleotide insertions or deletions, in at least about 80% of the nucleotides, usually at least about 90% to 95%, and more preferably at least about 98 to 99.5% of the nucleotides. Alternatively, substantial homology exists when the segments will hybridize under selective hybridization conditions, to the complement of the strand.
  • the nucleic acids may be present in whole cell ⁇ , in a cell lysate, or in a partially purified or substantially pure form.
  • a nucleic acid is "isolated” or “rendered substantially pure” when purified away from other cellular components or other contaminants, e.g., other cellular nucleic acids or proteins, by standard techniques, including alkaline/SDS treatment, CsCl banding, column chromatography, agarose gel electrophoresis and others well known in the art. See, F. Ausubel, et al., ed. Current Protocols in Molecular Biology, Greene Publishing and Wiley- Interscience, New York (1987).
  • DNA sequences substantially identical to a native sequence (except for modified restriction sites and the like), from either cDNA, genomic or mixtures may be mutated, thereof in accordance with standard techniques to provide gene sequences.
  • these mutations may affect amino acid sequence as desired.
  • DNA sequences substantially identical to a native sequence (except for modified restriction sites and the like), from either cDNA, genomic or mixtures may be mutated, thereof in accordance with standard techniques to provide gene sequences.
  • these mutations may affect amino acid sequence as desired.
  • DNA sequences substantially identical to a native sequence (except for modified restriction sites and the like), from either cDNA, genomic or mixtures may be mutated, thereof in accordance with standard techniques to provide gene sequences.
  • these mutations may affect amino acid sequence as desired.
  • DNA sequences substantially identical to a native sequence (except for modified restriction sites and the like), from either cDNA, genomic or mixtures may be mutated, thereof in accordance with standard techniques to provide gene sequences.
  • these mutations may affect amino acid sequence as desired.
  • a nucleic acid is "operably linked" when it is placed into a functional relationship with another nucleic acid sequence.
  • a promoter or enhancer is operably linked to a coding sequence if it affects the
  • operably linked means that the DNA sequences being linked are contiguous and, where necessary to join two protein coding regions, contiguous and in reading frame.
  • operably linked indicates that the sequences are capable of effecting switch recombination.
  • a preferred embodiment of the invention is an animal containing at least one, typically 2-10, and sometimes 25-50 or more copies of the transgene described in Example 12 (e.g., pHC1 or pHC2) bred with an animal containing a single copy of a light chain transgene described in Examples 5, 6, 8, or 14, and the offspring bred with the J H deleted animal described in Example 10. Animals are bred to homozygosity for each of these three traits.
  • Such animals have the following genotype: a single copy (per haploid set of chromosomes) of a human heavy chain unrearranged mini-locus (described in Example 12), a single copy (per haploid set of chromosomes) of a rearranged human ⁇ light chain construct (described in Example 14), and a deletion at each endogenous mouse heavy chain locus that removes all of the functional J H segments (described in
  • Example 10 Such animals are bred with mice that are
  • B cells will be monospecific with regards to human or mouse heavy chains because both endogenous mouse heavy chain gene copies are nonfunctional by virtue of the deletion spanning the J H region introduced as described in Example 9 and 12. Furthermore, a substantial fraction of the B cells will be monospecific with regards to the human or mouse light chains because expression of the single copy of the rearranged human ⁇ light chain gene will allelically and isotypically exclude the rearrangement of the endogenous mouse ⁇ and ⁇ chain genes in a significant fraction of B-cells.
  • the transgenic mouse of the preferred embodiment will exhibit immunoglobulin production with a significant repertoire, ideally substantially similar to that of a native mouse.
  • the total immunoglobulin levels will range from about 0.1 to 10 mg/ml of serum,
  • the adult mouse ratio of serum IgG to IgM is preferably about 10:1.
  • the IgG to IgM ratio will be much lower in the
  • spleen and lymph node B cells express exclusively human IgG protein.
  • the repertoire will ideally approximate that shown in a non-transgenic mouse, usually at least about 10% as high, preferably 25 to 50% or more.
  • immunoglobulins ideally IgG
  • 10 4 to 10 6 or more will be produced, depending primarily on the number of different V, J and D regions introduced into the mouse genome.
  • These immunoglobulins will typically recognize about one-half or more of highly antigenic proteins,
  • immunoglobulins will exhibit an affinity for preselected antigens of at least about 10 7 M -1 , preferably 10 8 M -1 to 10 9 M -1 or greater.
  • transgenic animal prior to rearrangement of a transgene containing various heavy or light chain gene segments, such gene segments may be readily identified, e.g. by hybridization or DNA sequencing, as being from a species of organism other than the transgenic animal.
  • transgenic animal of the invention Although the foregoing describes a preferred embodiment of the transgenic animal of the invention, other embodiments are defined by the disclosure herein and more particularly by the transgenes described in the Examples.
  • transgenic animal Four categories of transgenic animal may be defined:
  • Transgenic animals containing an unrearranged heavy and unrearranged light immunoglobulin transgene III Transgenic animal containing rearranged heavy and an unrearranged light immunoglobulin transgene, and IV.
  • the preferred order of preference is as follows II > I > III > IV where the endogenous light chain genes (or at least the ⁇ gene) have been knocked out by homologous recombination (or other method) and I > II > III >IV where the endogenous light chain genes have not been knocked out and must be dominated by allelic exclusion.
  • Transgenic mice are derived according to Hogan, et al., "Manipulating the Mouse Embryo: A Laboratory Manual", Cold Spring Harbor Laboratory, which is incorporated herein by reference.
  • Embryonic stem cells are manipulated according to published procedures (Teratocarcinomas and embryonic stem cells: a practical approach, E.J. Robertson, ed., IRL Press, Washington, D.C., 1987; Zjilstra et al., Nature 342:435-438 (1989); and Schwartzberg et al., Science 246:799-803 (1989), each of which is incorporated herein by reference).
  • Oligonucleotides are synthesized on an Applied Bio Systems oligonucleotide synthesizer according to
  • Hybridoma cells and antibodies are manipulated according to "Antibodies: A Laboratory Manual”, Ed Harlow and David Lane, Cold Spring Harbor Laboratory (1988), which is incorporated herein by reference.
  • the isolated nuclei (or PBS washed human spermatocytes) are embedded in a low melting point agarose matrix and lysed with EDTA and proteinase ⁇ to expose high molecular weight DNA, which is then digested in the agarose with the restriction enzyme NotI as described by M. Finney in Current Protocols in Molecular Biology (F. Ausubel, et al., eds. John Wiley & Sons, Supp. 4, 1988, Section 2.5.1).
  • the NotI digested DNA is then fractionated by pulsed field gel electrophoresis as described by Anand et al.,
  • Plasmid pYACNN is prepared by digestion of pYAC-4 Neo (Cook et al., Nucleic Acids Res. 16: 11817 (1988)) with EcoRI and ligation in the presence of the oligonucleotide 5' - AAT TGC GGC CGC - 3'.
  • a 450 kb Xhol to NotI fragment that includes all of C ⁇ , the 3' enhancer, all J segments, and at least five different V segments is isolated and microinjected into the nucleus of single cell embryos as described in Example 1.
  • a 750 kb Mlul to NotI fragment that includes all of the above plus at least 20 more V segments is isolated as described in Example 1 and digested with BssHII to produce a fragment of about 400 kb.
  • the 450 kb Xhol to NotI fragment plus the approximately 400 kb Mlul to BssHII fragment have sequence overlap defined by the BssHII and Xhol restriction sites.
  • pBR322 is digested with EcoRI and StyI and ligated with the following oligonucleotides to generate pGP1 which contains a 147 base pair insert containing the restriction sites shown in Fig. 8. The general overlapping of these oligos is also shown in Fig. 9.
  • the oligonucleotides are:
  • AAA AGC CCG CTC ATT AGG CGG GCT - 3'
  • This plasmid contains a large polylinker flanked by rare cutting NotI sites for building large inserts that can be isolated from vector sequences for microinjection.
  • the plasmid is based on pBR322 which is relatively low copy compared to the pUC based plasmids (pGP1 retains the pBR322 copy number control region near the origin of replication). Low copy number reduces the potential toxicity of insert sequences.
  • pGP1 contains a strong transcription terminator sequence derived from trpA (Christie et al., Proc. Natl. Acad. Sci. USA 78:4180 (1981)) inserted between the ampicillin resistance gene and the polylinker. This further reduces the toxicity associated with certain inserts by preventing readthrough transcription coming from the
  • ampicillin promoters are ampicillin promoters.
  • Plasmid pGP2 is derived from pGP1 to introduce an additional restriction site (Sfil) in the polylinker.
  • pGP1 is digested with Mlul and Spel to cut the recognition sequences in the polylinker portion of the plasmid.
  • the following adapter oligonucleotides are ligated to the thus digested pGP1 to form pGP2.
  • a 3' pGP2 is identical to pGP1 except that it contains an additional Sfi I site located between the Mlul and Spel sites. This allows inserts to be completely excised with Sfil as well as with NotI.
  • the rat IGH 3' enhancer sequence is PCR amplified by using the following oligonucleotides: 5' CAG GAT CCA GAT ATC AGT ACC TGA AAC AGG GCT TGC 3'
  • the thus formed double stranded DNA encoding the 3' enhancer is cut with BamHI and SphI and clone into BamHI/SphI cut pGP2 to yield pRE3 (rat enhancer 3').
  • pMUM is 4 kb EcoRI/Hindlll fragment isolated from human genomic DNA library with oligonucleotide:
  • pGP1 is digested with BamHI and Bglll followed by treatment with calf intestinal alkaline phosphatase.
  • Fragments (a) and (b) from Fig. 9 are cloned in the digested pGP1.
  • a clone is then isolated which is oriented such that 5' BamHI site is destroyed by BamHI/Bgl fusion. It is identified as pMU (see Fig. 10).
  • pMU is digested with BamHI and fragment (c) from Fig. 9 is inserted.
  • the resultant plasmid pHIG1 (Fig. 10) contains an 18 kb insert encoding J and C ⁇ segments.
  • pGP1 is digested with BamHI and Hindlll is followed by treatment with calf intestinal alkaline phosphatase (Fig. 14).
  • the so treated fragment (b) of Fig. 14 and fragment (c) of Fig. 14 are cloned into the BamHI/Hindlll cut pGP1.
  • Proper orientation of fragment (c) is checked by Hindlll digestion to form pCON1 containing a 12 kb insert encoding the C ⁇ region.
  • pHIGl contains J segments, switch and ⁇ sequences in its 18 kb insert with an Sfil 3' site and a Spel 5' site in a polylinker flanked by NotI sites, will be used for rearranged VDJ segments.
  • pCON1 is identical except that it lacks the J region and contains only a 12 kb insert. The use of pCON1 in the construction of fragment containing rearranged VDJ segments will be described hereinafter.
  • An intronic sequence is a nucleotide sequence of at least 15 contiguous nucleotides that occurs in an intron of a specified gene.
  • Phage clones containing the ⁇ -1 region are identified and isolated "sing the following oligonucleotide which is specific for the third exon of ⁇ -1 (CH3).
  • a 7.7 kb Hindlll to Bglll fragment (fragment (a) in Fig. 11) is cloned into Hindlll/Bglll cut pRE3 to form pREGl.
  • the upstream 5.3 kb Hindlll fragment (fragment (b) in Fig. 11) is cloned into Hindlll digested pREGl to form pREG2. Correct orientation is confirmed by BamHI/Spel digestion.
  • the previously described plasmid pHIGl contains human J segments and the C ⁇ constant region exons.
  • pHIG1 was digested with Sfil (Fig. 10).
  • the plasmid pREG2 was also digested with Sfil to produce a 13.5 kb insert containing human C ⁇ exons and the rat 3' enhancer sequence.
  • sequences were combined to produce the plasmid pHIG3' (Fig. 12) containing the human J segments, the human C ⁇ constant region, the human C ⁇ 1 constant region and the rat 3' enhancer contained on a 31.5 kb insert.
  • a second plasmid encoding human C ⁇ and human C ⁇ 1 without J segments is constructed by digesting pCON1 with Sfil and combining that with the Sfil fragment containing the human C ⁇ region and the rat 3' enhancer by digesting pREG2 with Sfil.
  • the resultant plasmid, pCON (Fig. 12) contains a 26 kb NotI/Spel insert containing human C ⁇ , human ⁇ 1 and the rat 3' enhancer sequence.
  • Fig. 13 Phage clones from the human genomic library containing D segments are identified and isolated using probes specific for diversity region sequences (Ichihara et al., EMBO J. 7:4141-4150 (1988)). The following
  • oligonucleotides are used:
  • DXP1 5' - TGG TAT TAC TAT GGT TCG GGG AGT TAT TAT
  • DXP4 5' - GCC TGA AAT GGA GCC TCA GGG CAC AGT GGG
  • CAC GGA CAC TGT - 3' DN4 5' - GCA GGG AGG ACA TGT TTA GGA TCT GAG GCC
  • a 5.2 kb Xhol fragment (fragment (b) in Fig. 13) containing DLR1, DXP1, DXP'1, and DA1 is isolated from a phage clone identified with oligo DXP1.
  • a 3.2 kb Xbal fragment (fragment (c) in Fig. 13) containing DXP4, DA4 and DK4 is isolated from a phage clone identified with oligo DXP4.
  • This plasmid contains diversity segments cloned into the polylinker with a unique 5' Sfil site and unique 3' Spel site. The entire polylinker is flanked by NotI sites.
  • a restriction map of the unrearranged V segment is determined to identify unique restriction sites which provide upon digestion a DNA fragment having a length approximately 2 kb containing the unrearranged V segment together with 5' and 3' flanking sequences.
  • the 5' prime sequences will include promoter and other regulatory sequences whereas the 3'
  • flanking sequence provides recombination sequences necessary for V-DJ joining. This approximately 3.0 kb V segment insert is cloned into the polylinker of pGB2 to form pVH1.
  • pVHl is digested with Sfil and the resultant fragment is cloned into the Sfil site of pHIG2 to form a
  • pHIG5 contains D segments only, the resultant pHIG5' plasmid contains a single V segment together with D segments.
  • the size of the insert contained in pHIG5 is 10.6 kb plus the size of the V segment insert.
  • pHIG5 The insert from pHIG5 is excised by digestion with NotI and Spel and isolated.
  • pHIG3' which contains J, C ⁇ and C ⁇ 1 segments is digested with Spel and NotI and the 3' kb fragment containing such sequences and the rat 3' enhancer sequence is isolated. These two fragments are combined and ligated into NotI digested pGP1 to produce pHIG which contains insert encoding a V segment, nine D segments, six functional J segments, C ⁇ , C ⁇ and the rat 3' enhancer.
  • the size of this insert is approximately 43 kb plus the size of the V segment insert.
  • the insert of pHIG is approximately 43 to 45 kb when a single V segment is employed. This insert size is at or near the limit of that which may be readily cloned into plasmid vectors.
  • the following describes in vivo homologous recombination of overlapping DNA fragments which upon homologous recombination within a zygote or ES cell form a transgene containing the rat 3' enhancer sequence, the human C ⁇ , the human C ⁇ 1, human J segments, human D segments and a multiplicity of human V segments.
  • a 6.3 kb BamHI/Hindlll fragment containing human J segments (see fragment (a) in Fig. 9) is cloned into Mlul/Spel digested pHIG5' using the following adapters: 5' GAT CCA AGC AGT 3'
  • the resultant is plasmid designated pHIG5'O (overlap).
  • the insert contained in this plasmid contains human V, D and J segments. When the single V segment from pVHl is used, the size of this insert is approximately 17 kb plus 2 kb.
  • This insert is isolated and combined with the insert from pHIG3' which contains the human J, C ⁇ , ⁇ 1 and rat 3' enhancer sequences. Both inserts contain human J segments which provide for approximately 6.3 kb of overlap between the two DNA fragments. When coinjected into the mouse zygote, in vivo homologous recombination occurs generating a transgene equivalent to the insert contained in pHIG.
  • This approach provides for the addition of a multiplicity of V segments into the transgene formed in vivo.
  • a multiplicity of V segments contained on (1) isolated genomic DNA, (2) ligated DNA derived from genomic DNA, or (3) DNA encoding a synthetic V segment repertoire is cloned into pHIG2 at the Sfil site to generate pHIG5' V N .
  • the J segments fragment (a) of Fig. 9 is then cloned into pHIG5' V N and the insert isolated.
  • This insert now contains a multiplicity of V segments and J segments which overlap with the J segments contained on the insert isolated from pHIG3'.
  • mouse heavy chain enhancer is isolated on the Xbal to EcoRI 678 bp fragment (Banerji et al., Cell 33:729-740 (1983)) from phage clones using oligo: 5' GAA TGG GAG TGA GGC TCT CTC ATA CCC
  • the ⁇ construct contains at least one human V ⁇ segment, all five human J ⁇ segments, the human J-C ⁇ enhancer, human ⁇ constant region exon, and, ideally, the human 3' ⁇ enhancer (Meyer et al., EMBO J. 8:1959-1964 (1989)).
  • the ⁇ enhancer in mouse is 9 kb downstream from C ⁇ . However, it is as yet unidentified in the human.
  • the construct contains a copy of the mouse heavy chain J-C ⁇ enhancers.
  • the minilocus is constructed from four component fragments:
  • this sequence is included to induce expression of the light chain construct as early as possible in B-cell development. Because the heavy chain genes are transcribed earlier than the light chain genes, this heavy chain enhancer is presumably active at an earlier stage than the intronic ⁇ enhancer); and
  • the 16 kb fraction is isolated from the Smal digested gel and the 11 kb region is similarly isolated from the gel containing DNA digested with BamHI.
  • the 11 kb BamHI fraction is cloned into ⁇ EMBL3 (Strategene, La Jolla, California) which is digested with BamHI prior to cloning.
  • the above C ⁇ specific oligonucleotide is used to probe the ⁇ EMBL3/BamHI library to identify an 11 kb clone.
  • a 5 kb Smal fragment fragment (b) in Fig. 20) is subcloned and subsequently inserted into pKapl digested with Smal.
  • Those plasmids containing the correct orientation of J segments, C ⁇ and the E ⁇ enhancer are designated pKap2.
  • V ⁇ segments are thereafter subcloned into the Mlul site of pKap2 to yield the plasmid pKapH which encodes the human V ⁇ segments, the human J ⁇ segments, the human C ⁇ segments and the human E ⁇ enhancer.
  • This insert is excised by digesting pKapH with NotI and purified by agarose gel electrophoresis. The thus purified insert is
  • the 11 kb BamHI fragment is cloned into BamHI .
  • V ⁇ segments is inserted into the polylinker between the BamHI and Spel sites in pKAPint to form pKapHV.
  • the insert of pKapHV is excised by digestion with NotI and purified.
  • the insert from pKap2 is excised by digestion with NotI and purified.
  • Each of these fragments contain regions of homology in that the fragment from pKapHV contains a 5 kb sequence of DNA that include the J ⁇ segments which is substantially homologous to the 5 kb Smal fragment contained in the insert obtained from pKap2.
  • these inserts are capable of homologously recombining when microinjected into a mouse zygote to form a transgene encoding V ⁇ , J ⁇ and C ⁇ .
  • This example describes the cloning of immunoglobulin ⁇ light chain genes from cultured cells that express an immunoglobulin of interest.
  • Such cells may contain multiple alleles of a given immunoglobulin gene.
  • a hybridoma might contain four copies of the ⁇ light chain gene, two copies from the fusion partner cell line and two copies from the original B-cell expressing the immunoglobulin of interest. Of these four copies, only one encodes the
  • immunoglobulin of interest despite the fact that several of them may be rearranged.
  • the procedure described in this example allows for the selective cloning of the expressed copy of the ⁇ light chain.
  • RNA is then used for the isolation of polyA+ RNA.
  • single-stranded cDNA is then purified and used as template for second strand synthesis (catalyzed by the enzyme DNA
  • the double stranded cDNA is isolated and used for determining the nucleotide sequence of the 5' end of the mRNAs encoding the heavy and light chains of the expressed
  • the double stranded cDNA described in part A is denatured and used as a template for a third round of DNA synthesis using the following oligonucleotide primer: 5' - GTA CGC CAT ATC AGC TGG ATG AAG TCA TCA GAT
  • This primer contains sequences specific for the constant portion of the ⁇ light chain message (TCA TCA GAT GGC GGG AAG ATG AAG ACA GAT GGT GCA) as well as unique sequences that can be used as a primer for the PCR amplification of the newly synthesized DNA strand (GTA CGC CAT ATC AGC TGG ATG AAG).
  • the sequence is amplified by PCR using the following two oligonucleotide primers: 5' - GAG GTA CAC TGA CAT ACT GGC ATG -3'
  • the PCR amplified sequence is then purified by gel electrophoresis and used as template for dideoxy sequencing reactions using the following oligonucleotide as a primer: 5' - GAG GTA CAC TGA CAT ACT GGC ATG -3'
  • the first 42 nucleotides of sequence will then be used to synthesize a unique probe for isolating the gene from which immunoglobulin message was transcribed.
  • This synthetic 42 nucleotide segment of DNA will be referred to below as o-kappa.
  • DNA from the Ig expressing cell line is then cut with Smal and second enzyme (or BamHI or Kpnl if there is Smal site inside V segment). Any resulting non-blunted ends are treated with the enzyme T4 DNA polymerase to give blunt ended DNA molecules. Then add restriction site encoding linkers (BamHI, EcoRI or Xhol depending on what site does not exist in fragment) and cut with the corresponding linker enzyme to give DNA fragments with BamHI, EcoRI or Xhol ends. The DNA is then size fractionated by agarose gel electrophoresis, and the fraction including the DNA fragment covering the expressed V segment is cloned into lambda EMBL3 or Lambda FIX (Stratagene, La Jolla, California).
  • V segment containing clones are isolated using the unique probe o-kappa.
  • DNA is isolated from positive clones and subcloned into the polylinker of pKapl. The resulting clone is called pRKL.
  • This example describes the cloning of immunoglobulin heavy chain ⁇ genes from cultured cells of expressed and immunoglobulin of interest. The procedure described in this example allows for the selective cloning of the expressed copy of a ⁇ heavy chain gene.
  • Double-stranded cDNA is prepared and isolated as described herein before.
  • the double-stranded cDNA is
  • This primer contains sequences specific for the constant portion of the ⁇ heavy chain message (ACA GGA GAC GAG GGG GAA AAG GGT TGG GGC GGA TGC) as well as unique sequences that can be used as a primer for the PCR amplification of the newly synthesized DNA strand (GTA CGC CAT ATC AGC TGG ATG AAG).
  • the sequence is amplified by PCR using the following two oligonucleotide primers:
  • PCR amplified sequence is then purified by gel electrophoresis and used as template for dideoxy sequencing reactions using the following oligonucleotide as a primer:
  • Mlul is a rare cutting enzyme that cleaves between the J segment and mu CH1
  • Mlul is a rare cutting enzyme that cleaves between the J segment and mu CH1
  • restriction endonuclease site is identified upstream of the rearranged V segment.
  • DNA from the Ig expressing cell line is then cut with Mlul and second enzyme.
  • Mlul or Spel adapter linkers are then ligated onto the ends and cut to convert the upstream site to Mlul or Spel.
  • the DNA is then size fractionated by agarose gel electrophoresis, and the fraction including the DNA fragment covering the expressed V segment is cloned directly into the plasmid pGP1.
  • V segment containing clones are isolated using the unique probe o-mu, and the insert is subcloned into Mlul or Mlul/Spel cut plasmid pCON2. The resulting plasmid is called pRMGH.
  • a human genomic DNA phage library was screened with kappa light chain specific oligonucleotide probes and isolated clones spanning the J ⁇ -C region.
  • a 5.7 kb Clal/Xhol fragment containing J ⁇ 1 together with a 13 kb Xhol fragment containing J ⁇ 2-5 and C ⁇ into pGP1d was cloned and used to create the plasmid pKcor. This plasmid contains J ⁇ 1-5, the kappa
  • V ⁇ light chain specific oligonucleotide probes were screened with V ⁇ light chain specific oligonucleotide probes and isolated clones containing human V ⁇ segments. Functional V segments were identified by DNA sequence analysis. These clones contain TATA boxes, open reading frames encoding leader and variable peptides (including 2 cysteine residues), splice sequences, and recombination heptamer-12 bp spacer-nonamer sequences. Three of the clones were mapped and sequenced.
  • Two of the clones, 65.5 and 65.8 appear to be functional, they contain TATA boxes, open reading frames encoding leader and variable peptides (including 2 cysteine residues), splice sequences, and recombination heptamer-12 bp spacer-nonamer sequences.
  • the third clone, 65.4 appears to encode a V ⁇ I pseudogene as it contains a non-canonical recombination heptamer.
  • Vk 65-8 which encodes a Vklll family gene, was used to build a light chain minilocus construct.
  • the kappa light chain minilocus transgene pKC1 (Fig. 32) was generated by inserting a 7.5 kb Xhol/Sall fragment containing V ⁇ 65.8 into the 5' Xhol site of pKcor.
  • the transgene insert was isolated by digestion with NotI prior to injection.
  • the purified insert was microinjected into the pronuclei of fertilized (C57BL/6 x CBA) F2 mouse embryos and transferred the surviving embryos into pseudopregnant females as described by Hogan et al. (in Methods of Manipulating the Mouse Embryo, 1986, Cold Spring Harbor Laboratory, New York). Mice that developed from injected embryos were analyzed for the presence of transgene sequences by Southern blot analysis of tail DNA. Transgene copy number was estimated by band intensity relative to control standards containing known quantities of cloned DNA.
  • Serum was isolated from these animals and assayed for the presence of transgene encoded human Ig kappa protein by ELISA as described by Harlow and Lane (in Antibodies: A Laboratory Manual, 1988, Cold Spring Harbor Laboratory, New York).
  • Microtiter plate wells were coated with mouse monoclonal antibodies specific for human Ig kappa (clone 6E1, #0173 , AMAC, Inc. , Westbrook, ME) , human IgM (Clone AF6, #0285, AMAC, Inc., Westbrook, ME) and human IgG1 (clone JL512, #0280, AMAC, Inc., Westbrook, ME).
  • Serum samples were serially diluted into the wells and the presence of specific immunoglobulins detected with affinity isolated alkaline phosphatase conjugated goat anti-human Ig
  • Fig. 35 shows the results of an ELISA assay of serum from 8 mice (I.D. #676, 674, 673, 670, 666, 665, 664, and 496). The first seven of these mice developed from embryos that were injected with the pKC1 transgene insert and the eighth mouse is derived from a mouse generated by
  • the kappa light chain minilocus transgene pKC2 was generated by inserting an 8 kb Xhol/Sall fragment containing V ⁇ 65.5 into the 5' Xhol site of pKC1. The resulting
  • transgene insert which contains two V ⁇ segments, was isolated prior to microinjection by digestion with NotI.
  • This construct is identical to pKCI except that it includes 1.2 kb of additional sequence 5' of J ⁇ and is missing 4.5 kb of sequence 3' of V ⁇ 65.8. In additional it contains a 0.9 kb Xbal fragment containing the mouse heavy chain J-m intronic enhancer (Banerji et al., Cell 33:729-740 (1983)) together with a 1.4 kb Mlul Hindlll fragment containing the human heavy chain J-m intronic enhancer (Hayday et al., Nature 307:334-340 (1984)) inserted downstream. This construct tests the feasibility of initiating early rearrangement of the light chain minilocus to effect allelic and isotypic exclusion.
  • enhancers i.e., the mouse or rat 3' kappa or heavy chain enhancer (Meyer and Neuberger, EMBO J. 8:1959-1964 (1989); Petterson et al. Nature 344:165-168 (1990), which are
  • a kappa light chain expression cassette was designed to reconstruct functionally rearranged light chain genes that have been amplified by PCR from human B-cell DNA.
  • the scheme is outlined in Fig. 33.
  • PCR amplified light chain genes are cloned into the vector pK5nx that includes 3.7 kb of 5' flanking sequences isolated from the kappa light chain gene 65.5.
  • the VJ segment fused to the 5' transcriptional
  • sequences are then cloned into the unique Xhol site of the vector pK31s that includes J ⁇ 2-4, the J ⁇ intronic enhancer, C ⁇ , and 9 kb of downstream sequences.
  • the resulting plasmid contains a reconstructed functionally rearranged kappa light chain transgene that can be excised with NotI for
  • the plasmids also contain unique Sall sites at the 3' end for the insertion of additional cis- acting regulatory sequences.
  • Oligonucleotide o-131 (gga ccc aga
  • Oligonucleotide o-130 (gtg caa tea att etc gag ttt gac tac aga c) is complementary to a sequence approximately 150 bp 3' of J ⁇ 1 and includes an Xhol site. These two oligonucleotides amplify a 0.7 kb DNA
  • V ⁇ III genes joined to J ⁇ 1 segments.
  • the PCR amplified DNA was digested with Ncol and Xhol and cloned individual PCR products into the plasmid pNN03.
  • the DNA sequence of 5 clones was determined and identified two with functional VJ joints (open reading frames). Additional functionally rearranged light chain clones are collected.
  • the functionally rearranged clones can be individually cloned into light chain expression
  • Transgenic mice generated with the rearranged light chain constructs can be bred with heavy chain minilocus transgenics to produce a strain of mice that express a spectrum of fully human antibodies in which all of the diversity of the primary repertoire is contributed by the heavy chain.
  • One source of light chain diversity can be from somatic mutation. Because not all light chains will be equivalent with respect to their ability to combine with a variety of different heavy chains, different strains of mice, each containing different light chain constructs can be
  • combination can result in an increased frequency of B-cells expressing fully human antibodies, and thus it can facilitate the isolation of human Ig expressing hybridomas.
  • NotI inserts of plasmids pIGM1, pHC1, pIGG1, pKC1, and pKC2 were isolated away from vector sequences by agarose gel electrophoresis. The purified inserts were microinjected into the pronuclei of fertilized (C57BL/6 x CBA) F2 mouse embryos and transferred the surviving embryos into
  • This example describes the inactivation of the mouse endogenous kappa locus by homologous recombination in
  • ES embryonic stem cells followed by introduction of the mutated gene into the mouse germ line by injection of targeted ES cells bearing an inactivated kappa allele into early mouse embryos (blastocysts).
  • the strategy is to delete J ⁇ and C ⁇ by homologous recombination with a vector containing DNA sequences
  • the plasmid pGEM7 contains the neomycin resistance gene (neo), used for drug selection of transfected ES cells, under the transcriptional control of the mouse phosphoglycerate kinase (pgk) promoter (Xbal/TaqI fragment; Adra et al., Gene 60: 65-74 (1987)) in the cloning vector pGEM- 7Zf(+).
  • the plasmid also includes a heterologous polyadenylation site for the neo gene, derived from the 3' region of the mouse pgk gene (PvulI/Hindlll fragment; Boer et al., Biochemical Genetics. 28:299-308 (1990)). This plasmid was used as the starting point for construction of the kappa targeting vector. The first step was to insert sequences homologous to the kappa locus 3' of the neo expression
  • Fig. 20a Mouse kappa chain sequences (Fig. 20a) were isolated from a genomic phage library derived from liver DNA using oligonucleotide probes specific for the C/c locus:
  • a 1.2 kb EcoRI/SphI fragment extending 5' of the J ⁇ region was also isolated from a positive phage clone.
  • An Sphl/Xbal/Bglll/EcoRI adaptor was ligated to the SphI site of this fragment, and the resulting EcoRI fragment was ligated into EcoRI digested pNEO-K3', in the same 5' to 3' orientation as the neo gene and the downstream 3' kappa sequences, to generate pNEO-K5'3' (Fig. 20c).
  • HSV Herpes Simplex Virus
  • TK thymidine kinase gene
  • the HSV TK cassette was obtained from the plasmid pGEM7 (TK), which contains the structural sequences for the HSV TK gene bracketed by the mouse pgk promoter and polyadenylation sequences as described above for pGEM7 (KJ1).
  • telomere sequences from 5' of JK and 3' of C ⁇ was inserted into pGP1b-TK to generate the targeting vector J/C KI (Fig. 20d).
  • the putative structure of the genomic kappa locus following homologous recombination with J/C Kl is shown in Fig. 20e.
  • ES cells used were the AB-1 line grown on mitotically inactive SNL76/7 cell feeder layers (McMahon and Bradley, Cell 62:1073-1085 (1990)) essentially as described (Robertson, E.J. (1987) in Teratocarcinomas and Embryonic Stem Cells: A Practical Approach. E.J. Robertson, ed. (Oxford: IRL Press), p. 71-112).
  • Other suitable ES lines include, but are not limited to, the E14 line (Hooper et al. (1987) Nature 326: 292-295), the D3 line (Doetschman et al. (1985) J. Embrvol. Exp. Morph. 87: 27-45), and the CCE line (Robertson et al.
  • the pluripotence of any given ES cell line can vary with time in culture and the care with which it has been handled.
  • the only definitive assay for pluripotence is to determine whether the specific population of ES cells to be used for targeting can give rise to chimeras capable of germline transmission of the ES genome. For this reason, prior to gene targeting, a portion of the parental population of AB-1 cells is injected into C57B1/6J blastocysts to
  • the kappa chain inactivation vector J/C Kl was digested with NotI and electroporated into AB-1 cells by the methods described (Hasty et al., Nature. 350:243-246 (1991)). Electroporated cells were plated onto 100 mm dishes at a density of 1-2 x 10 6 cells/dish. After 24 hours, G418
  • DNA analysis was carried out by Southern blot hybridization. DNA was isolated from the clones as described (Laird et al., Nucl. Acids Res. 19:4293 (1991)) digested with Xbal and probed with the 800 bp EcoRI/Xbal fragment indicated in Fig. 20e as probe A. This probe detects a 3.7 kb Xbal fragment in the wild type locus, and a diagnostic 1.8 kb band in a locus which has homologously recombined with the
  • the AB1 ES cells are an XY cell line and a majority of these high percentage chimeras were male due to sex conversion of female embryos colonized by male ES cells.
  • Male chimeras derived from 4 of the 5 targeted clones were bred with C57BL/6J females and the offspring monitored for the presence of the dominant agouti coat color indicative of germline transmission of the ES genome.
  • mice homozygous for the mutation approximately 50 percent of the agouti offspring showed a hybridizing Bgl II band of 2.4 kb in addition to the wild-type band of 4.1 kb, demonstrating the germline transmission of the targeted kappa locus.
  • heterozygotes were bred together and the kappa genotype of the offspring determined as described above.
  • three genotypes were derived from the heterozygote matings: wild-type mice bearing two copies of a normal kappa locus, heterozygotes carrying one targeted copy of the kappa gene and one NT kappa gene, and mice homozygous for the kappa mutation. The deletion of kappa sequences from these latter mice was verified by hybridization of the Southern blots with a probe specific for J ⁇ (probe C, Fig. 20a).
  • This example describes the inactivation of the endogenous murine immunoglobulin heavy chain locus by
  • homologous recombination in embryonic stem (ES) cells The strategy is to delete the endogenous heavy chain J segments by homologous recombination with a vector containing heavy chain sequences from which the J H region has been deleted and replaced by the gene for the selectable marker neo.
  • Fig. 21a were isolated from a genomic phage library derived from the D3 ES cell line (Gossler et al., Proc. Natl. Acad. Sci. U.S.A. 83:9065-9069 (1986)) using a J H 4 specific
  • restriction fragment encompassing the mutation with the corresponding sequence from a wild-type neo clone.
  • Hindlll site in the prepared pGEM7 (KJ1) was converted to a Sall site by addition of a synthetic adaptor, and the neo expression cassette excised by digestion with Xbal/Sall. The ends of the neo fragment were then blunted by treatment with the Klenow form of DNA poll, and the neo fragment was
  • pGP1b was digested with the restriction enzyme NotI and ligated with the following oligonucleotide as an adaptor:
  • pGMT mouse immunoglobulin heavy chain targeting
  • HSV Herpes Simplex Virus
  • TK thymidine kinase
  • HSV TK gene was obtained from the plasmid pGEM7 (TK) by digestion with EcoRI and Hindlll. The TK DNA fragment was subcloned between the EcoRI and Hindlll sites of pGMT, creating the plasmid pGMT-TK (Fig. 21c).
  • genomic Xbal/Xhol fragment situated 5' of the J H region, was derived from a positive genomic phage clone by limit digestion of the DNA with Xhol, and partial digestion with Xbal.
  • this Xbal site is not present in genomic DNA, but is rather derived from phage sequences immediately flanking the cloned genomic heavy chain insert in the positive phage clone. The fragment was
  • the final step in the construction involved the excision from pUC18 J H -neo of the 2.8 kb EcoRI fragment which contained the neo gene and flanking genomic sequences 3' of J H . This fragment was blunted by Klenow polymerase and subcloned into the similarly blunted Xhol site of
  • J H KO1 contains 6.9 kb of genomic sequences flanking the J H locus, with a 2.3 kb deletion spanning the J H region into which has been inserted the neo gene.
  • Fig. 21f shows the structure of an endogenous heavy chain gene after homologous recombination with the targeting construct.
  • the heavy chain inactivation vector J H KO1 was digested with NotI and electroporated into AB-1 cells by the methods described (Hasty et al., Nature 350:243-246 (1991)). Electroporated cells were plated into 100 mm dishes at a density of 1-2 x 10 6 cells/dish. After 24 hours, G418
  • DNA analysis was carried out by Southern blot hybridization. DNA was isolated from the clones as described (Laird et al. (1991) Nucleic Acids Res. 19: 4293), digested with StuI and probed with the 500 bp EcoRI/StuI fragment designated as probe A in Fig. 2If. This probe detects a StuI fragment of 4.7 kb in the wild-type locus, whereas a 3 kb band is diagnostic of homologous recombination of endogenous sequences with the targeting vector (see Fig. 21a and f).
  • mice carrying the J H deletion were generated using a neo-specific probe (probe B, Fig. 21f) to generate mice carrying the J H deletion.
  • agouti offspring Since only one copy of the heavy chain locus was targeted in the injected ES clones, each agouti pup had a 50 percent chance of inheriting the mutated locus. Screening for the targeted gene was carried out by Southern blot analysis of stul-digested DNA from tail
  • heterozygotes were bred together and the heavy chain genotype of the offspring determined as described above.
  • three genotypes were derived from the heterozygote atings: wild-type mice bearing two copies of the normal J H locus, heterozygotes caring one targeted copy of the gene and one normal copy, and mice homozygous for the J H mutation.
  • the absence of J H sequences from these latter mice was verified by hybridization of the Southern blots of Stul-digested DNA with a probe specific for J H (probe C, Fig. 21a).
  • the plasmid pBR322 was digested with EcoRI and Styl and ligated with the following oligonucleotides: oligo-42 5'- caa gag ccc gcc taa tga gcg ggc ttt ttt ttg cat act gcg gcc get -3'
  • pGP1a The resulting plasmid, pGP1a, is designed for cloning very large DNA constructs that can be excised by the rare cutting restriction enzyme NotI. It contains a NotI restriction site downstream (relative to the ampicillin resistance gene, AmpR) of a strong transcription termination signal derived from the trpA gene (Christie et al., Proc.
  • This termination signal reduces the potential toxicity of coding sequences inserted into the NotI site by eliminating readthrough transcription from the AmpR gene.
  • this plasmid is low copy relative to the pUC plasmids because it retains the pBR322 copy number control region. The low copy number further reduces the potential toxicity of insert sequences and reduces the selection against large inserts due to DNA replication.
  • the vectors pGP1b, pGP1c, pGP1d, and pGP1f are derived from pGP1a and contain different polylinker cloning sites. The polylinker sequences are given below pGP1a
  • pGP1a was digested with NotI and ligated with the following oligonucleotides: oligo-47 5'- ggc cgc aag ctt act get gga tec tta att aat cga tag tga tct cga ggc -3'
  • oligo-48 5'- ggc cgc etc gag ate act ate gat taa tta agg ate cag cag taa get tgc -3'
  • the resulting plasmid, pGP1b contains a short polylinker region flanked by NotI sites. This facilitates the construction of large inserts that can be excised by NotI digestion.
  • oligonucleotides oligo-44 5'- etc cag gat cca gat ate agt ace tga aac agg get tgc -3'
  • oligo-45 5'- etc gag cat gca cag gac ctg gag cac aca cag cct tec -3' were used to amplify the immunoglobulin heavy chain 3'
  • pNN03 is a pUC derived plasmid that contains a polylinker with the following restriction sites, listed in order: NotI, BamHI, Ncol, Clal, EcoRV, Xbal, Sad, Xhol, SphI, PstI, Bglll, EcoRI, Smal, Kpnl, Hindlll, and NotI).
  • the resulting plasmid, pRE3 was digested with BamHI and Hindlll, and the insert containing the rat Ig heavy chain 3' enhancer cloned into BamHI/Hindlll digested pGP1b.
  • the resulting plasmid, pGPe (Fig. 22 and Table 1), contains several unique restriction sites into which sequences can be cloned and subsequently excised together with the 3' enhancer by NotI digestion.
  • a human placental genomic DNA library cloned into the phage vector XEMBL3/SP6/T7 was screened with the human heavy chain J region specific oligonucleotide: oligo-1 5'- gga ctg tgt ccc tgt gtg atg ctt ttg atg tct ggg gcc aag -3' and the phage clone ⁇ 1.3 isolated.
  • a 4 kb Xhol fragment was isolated from phage clone ⁇ 2.1 that contains sequences immediately downstream of the sequences in pJMl, including the so called ⁇ element involved in (S-associated deleteon of the ⁇ in certain IgD expressing B-cells (Yasui et al., Eur. J. Immunol. 19:1399 (1989), which is incorporated herein by reference).
  • This fragment was treated with the Klenow fragment of DNA polymerase I and ligated to Xhol cut, Klenow treated, pJMl.
  • the resulting plasmid, pJM2 (Fig. 23), had lost the internal Xhol site but retained the 3' Xhol site due to incomplete reaction by the Klenow enzyme.
  • pJM2 contains the entire human J region, the heavy chain J- ⁇ intronic enhancer, the ⁇ switch region and all of the ⁇ constant region exons, as well as the two 0.4 kb direct repeats, ⁇ and ⁇ , involved in ⁇ -associated deletion of the ⁇ gene. 3. Isolation of D region clones and construction of pDH1
  • oligonucleotide oligo-4 5'- tgg tat tac tat ggt teg ggg agt tat tat aac cac agt gtc -3' was used to screen the human placenta genomic library for D region clones. Phage clones ⁇ 4.1 and ⁇ 4.3 were isolated. A 5.5 kb Xhol fragment, that includes the D elements D ⁇ 1 , D N1 , and D M2 (Ichihara et al., EMBO J. 1:4141 (1988)), was isolated from phage clone ⁇ 4.1.
  • pDH1 contains a 10.6 kb insert that includes at least 7 D segments and can be excised with Xhol (5') and EcoRV (3'). 4. pCOR1
  • the plasmid pJM2 was digested with Asp718 (an isoschizomer of Kpnl) and the overhang filled in with the Klenow fragment of DNA polymerase I. The resulting DNA was then digested with Clal and the insert isolated. This insert was ligated to the XhoI/EcoRV insert of pDH1 and Xhol/Clal digested pGPe to generate pCORl (Fig. 24).
  • pIGM1 contains 2 functional human variable region segments, at least 8 human D segments all 6 human J H segments, the human
  • J- ⁇ enhancer the human ⁇ element, the human ⁇ switch region, all of the human ⁇ coding exons, and the human ⁇ element, together with the rat heavy chain 3' enhancer, such that all of these sequence elements can be isolated on a single
  • oligonucleotide specific for human Ig g constant region genes: oligo-29 5'- cag cag gtg cac ace caa tgc cca tga gcc cag aca ctg gac -3' was used to screen the human genomic library. Phage clones 129.4 and ⁇ 29.5 were isolated. A 4 kb Hindlll fragment of phage clone ⁇ 29.4, containing a ⁇ switch region, was used to probe a human placenta genomic DNA library cloned into the phage vector lambda FIXTM II (Stratagene, La Jolla, CA). Phage clone ⁇ Sg1.13 was isolated.
  • a 7.8 kb Hindlll fragment of phage clone ⁇ 29.5, containing the ⁇ 1 coding region was cloned into pUC18.
  • the resulting plasmid, pLT1 was digested with Xhol, Klenow treated, and religated to destroy the internal Xhol site.
  • the resulting clone, pLTlxk was digested with Hindlll and the insert isolated and cloned into pSP ⁇ 2 to generate the plasmid clone pLTlxks.
  • p ⁇ e1 contains all of the 71 constant region coding exons, together with 5 kb of downstream sequences, linked to the rat heavy chain 3' enhancer.
  • a 5.3 kb Hindlll fragment containing the 71 switch region and the first exon of the pre-switch sterile transcript (P. Sideras et al. (1989) International Immunol. 1, 631) was isolated from phage clone XS ⁇ 1.13 and cloned into pSP72 with the polylinker Xhol site adjacent to the 5' end of the insert, to generate the plasmid clone pS ⁇ 1S.
  • the Xhol/Sall insert of pS ⁇ 1S was cloned into Xhol digested p ⁇ e1 to generate the plasmid clone p ⁇ e2 (Fig. 26).
  • p ⁇ e2 contains all of the 71 constant region coding exons, and the upstream switch region and sterile transcript exons, together with 5 kb of downstream sequences, linked to the rat heavy chain 3' enhancer.
  • This clone contains a unique Xhol site at the 5' end of the insert. The entire insert, together with the Xhol site and the 3' rat enhancer can be excised from vector sequences by digestion with NotI. 4 . pHC1
  • pHC1 contains 2
  • Phage clone X49.8 was isolated and a 6.1 kb Xbal fragment containing the variable segment VH49.8 subcloned into pNN03 (such that the polylinker Clal site is downstream of VH49.8 and the polylinker Xhol site is upstream) to generate the plasmid pVH49.8.
  • An 800 bp region of this insert was sequenced, and VH49.8 found to have an open reading frame and intact splicing and recombination signals, thus indicating that the gene is functional (Table 2). 2.
  • a 4 kb Xbal genomic fragment containing the human V H IV family gene V H 4-21 (Sanz et al., EMBO J., 8:3741 (1989)), subcloned into the plasmid pUC12, was excised with Smal and Hindlll, and treated with the Klenow fragment of polymerase I. The blunt ended fragment was then cloned into Clal digested, Klenow treated, pVH49.8. The resulting plasmid, pV2, contains the human heavy chain gene VH49.8 linked upstream of VH4-21 in the same orientation, with a unique Sall site at the 3' end of the insert and a unique Xhol site at the 5' end.
  • sequences immediately upstream of, and adjacent to, the 5.3 kb ⁇ 1 switch region containing fragment in the plasmid p ⁇ e2) together with the neighboring upstream 3.1 kb Xbal fragment were isolated from the phage clone ⁇ Sg1.13 and cloned into Hindlll/Xbal digested pUC18 vector.
  • the resulting plasmid, pS ⁇ 1-5' contains a 3.8 kb insert representing sequences upstream of the initiation site of the sterile transcript found in B-cells prior to switching to the ⁇ 1 isotype (P.
  • transgene constructs to promote correct expression of the sterile transcript and the associated switch
  • the pS ⁇ 1-5' insert was excised with Smal and
  • the ligation product was digested with Sall and ligated to Sall digested pV2.
  • the resulting plasmid, pVP contains 3.8 kb of ⁇ 1 switch 5' flanking sequences linked downstream of the two human variable gene segments VH49.8 and VH4-21 (see Table 2).
  • the pVP insert is isolated by partial digestion with Sall and complete digestion with Xhol, followed by purification of the 15 kb fragment on an agarose gel. The insert is then cloned into the Xhol site of p ⁇ e2 to generate the plasmid clone pVGE1 (Fig. 27).
  • pVGEl contains two human heavy chain variable gene segments upstream of the human ⁇ 1 constant gene and associated switch region.
  • a unique Sall site between the variable and constant regions can be used to clone in D, J, and ⁇ gene segments.
  • the rat heavy chain 3' enhancer is linked to the 3' end of the ⁇ 1 gene and the entire insert is flanked by NotI sites. 5.
  • the plasmid clone pVGEl is digested with Sall and the Xhol insert of pIGMl is cloned into it.
  • the resulting clone, pHC2 contains 4 functional human variable region segments, at least 8 human D segments all 6 human J H segments, the human J-m enhancer, the human ⁇ element, the human ⁇ switch region, all of the human ⁇ coding exons, the human ⁇ element, and the human ⁇ 1 constant region, including the associated switch region and sterile transcript associated exons, together with 4 kb flanking sequences upstream of the sterile transcript initiation site.
  • These human sequences are linked to the rat heavy chain 3' enhancer, such that all of the sequence elements can be isolated on a single fragment, away from vector sequences, by digestion with NotI and
  • transgenic mice A unique Xhol site at the 5' end of the insert can be used to clone in additional human variable gene segments to further expand the recombinational diversity of this heavy chain minilocus.
  • E. Transgenic mice A unique Xhol site at the 5' end of the insert can be used to clone in additional human variable gene segments to further expand the recombinational diversity of this heavy chain minilocus.
  • the NotI inserts of plasmids pIGM1 and pHC1 were isolated from vector sequences by agarose gel electrophoresis. The purified inserts were microinjected into the pronuclei of fertilized (C57BL/6 x CBA) F2 mouse embryos and transferred the surviving embryos into pseudopregnant females as described by Hogan et al. (B. Hogan, F. Costantini, and E. Lacy, Methods of Manipulating the Mouse Embryo, 1986, Cold Spring Harbor
  • mice that developed from injected embryos were analyzed for the presence of transgene sequences by Southern blot analysis of tail DNA. Transgene copy number was estimated by band intensity relative to control standards containing known quantities of cloned DNA. At 3 to 8 weeks of age, serum was isolated from these animals and assayed for the presence of transgene encoded human IgM and IgG1 by ELISA as described by Harlow and Lane (E. Harlow and D. Lane.
  • mice were coated with mouse monoclonal antibodies specific for human IgM (clone AF6, #0285, AMAC, Inc. Westbrook, ME) and human IgG1 (clone JL512, #0280, AMAC, Inc. Westbrook, ME). Serum samples were serially diluted into the wells and the presence of specific immunoglobulins detected with affinity isolated alkaline phosphatase conjugated goat anti-human Ig (polyvalent) that had been pre-adsorbed to minimize cross-reactivity with mouse immunoglobulins. Table 3 and Fig.
  • mice 28 show the results of an ELISA assay for the presence of human IgM and IgG1 in the serum of two animals that developed from embryos injected with the transgene insert of plasmid pHC1. All of the control non- transgenic mice tested negative for expression of human IgM and IgG1 by this assay.
  • Mice from two lines containing the pIGM1 NotI insert (lines #6 and 15) express human IgM but not human IgG1.
  • mice from 6 lines that contain the pHC1 insert and found that 4 of the lines (lines #26, 38, 57 and 122) express both human IgM and human IgG1, while mice from two of the lines (lines #19 and 21) do not express detectable levels of human immunoglobulins.
  • the pHC1 transgenic mice that did not express human immunoglobulins were so-called G o mice that developed directly from microinjected embryos and may have been mosaic for the presence of the transgene.
  • Table 3 shows a correlation between the presence of integrated transgene DNA and the presence of transgene encoded immunoglobulins in the serum. Two of the animals that were found to contain the pHC1 transgene did not express detectable levels of human immunoglobulins. These were both low copy animals and may not have contained complete copies of the transgenes, or the animals may have been genetic mosaics
  • the transgene containing cells may not have populated the hematopoietic lineage.
  • the transgenes may have integrated into genomic locations that are not conducive to their expression.
  • the detection of human IgM in the serum of pIGMl transgenics, and human IgM and IgG1 in pHC1 transgenics, indicates that the transgene sequences function correctly in directing VDJ joining, transcription, and isotype switching.
  • immunoglobulin cDNA clones derived from transgenic mouse spleen mRNA were examined.
  • the overall diversity of the transgene encoded heavy chains, focusing on D and J segment usage, N region addition, CDR3 length distribution, and the frequency of joints resulting in functional mRNA molecules was examined.
  • Transcripts encoding IgM and IgG incorporating VH105 and VH251 were examined.
  • Polyadenylated RNA was isolated from an eleven week old male second generation line-57 pHC1 transgenic mouse.
  • This RNA was used to synthesize oligo-dT primed single
  • cDNA was then used as template for four individual PCR amplifications using the following four synthetic oligonucleotides as primers: VH251 specific oligo-149, eta get cga gtc caa gga gtc tgt gcc gag gtg cag ctg (g,a,t,c); VH105 specific o-150, gtt get cga gtg aaa ggt gtc cag tgt gag gtg cag ctg (g,a,t,c); human gamma1 specific oligo-151, ggc get cga gtt cca cga cac cgt cac egg ttc; and human mu specific oligo-152, cct get cga ggc age caa egg cca cgc tgc teg.
  • Reaction 1 used primers 0-149 and o-151 to amplify VH251-gammal transcripts
  • reaction 2 used o-149 and o- 152 to amplify VH251-mu transcripts
  • reaction 3 used o-150 and o-151 to amplify VH105-gammal transcripts
  • reaction 4 used o-150 and o-152 to amplify VH105-mu transcripts.
  • resulting 0.5 kb PCR products were isolated from an agarose gel; the ⁇ transcript products were more abundant than the ⁇ transcript products, consistent with the corresponding ELISA data (Fig. 34).
  • the PCR products were digested with Xhol and cloned into the plasmid pNN03. Double-stranded plasmid DNA was isolated from minipreps of nine clones from each of the four PCR amplifications and dideoxy sequencing reactions were performed. Two of the clones turned out to be deletions containing no D or J segments. These could not have been derived from normal RNA splicing products and are likely to have originated from deletions introduced during PCR
  • VH105 primer turned out not to be specific for VH105 in the reactions performed. Therefore many of the clones from reactions 3 and 4 contained VH251 transcripts.
  • Table 5 compared the distribution of J segments incorporated into pHC1 transgene encoded transcripts to J segments found in adult human PBL immunoglobulin transcripts. The distribution profiles are very similar, J4 is the dominant segment in both systems, followed by J6. J2 is the least common segment in human PBL and the transgenic animal.
  • Table 7 shows the predicted amino acid sequences of the VDJ regions from 30 clones that were analyzed from the pHC1 transgenic.
  • the translated sequences indicate that 23 of the 30 VDJ joints (77%) are in-frame with respect to the variable and J segments.
  • Table 8 compared the length of the CDR3 peptides from transcripts with in-frame VDJ joints in the pHC1
  • transgenic mouse to those in human PBL.
  • human PBL data comes from Yamada et al.
  • the profiles are similar with the transgenic profile skewed slightly toward smaller CDR3 peptides than observed from human PBL.
  • the average length of CDR3 in the transgenic mouse is 10.3 amino acids. This is substantially the same as the average size reported for authentic human CDR3 peptides by Sanz (J. Immunol. 147:1720- 1729 (1991)).
  • Two human leukocyte genomic DNA libraries cloned into the phage vector XEMBL3/SP6/T7 are screened with a 1 kb Pacl/Hindlll fragment of ⁇ 1.3 containing the human heavy chain J- ⁇ intronic enhancer.
  • Fragments containing functional VJ segments are subcloned into the plasmid vector pSP72 such that the plasmid derived Xhol site is adjacent to the 5' end of the insert seguence.
  • a subclone containing a functional VDJ segment is digested with Xhol and Pad (Pad, a rare-cutting enzyme, recognizes a site near the J-m intronic enhancer), and the insert cloned into Xhol/Pad digested pHC2 to generate a transgene construct with a
  • VDJ segment functional VDJ segment, the J- ⁇ intronic enhancer, the ⁇ switch element, the ⁇ constant region coding exons, and the ⁇ 1 constant region, including the sterile transcript associated sequences, the ⁇ 1 switch, and the coding exons.
  • transgene construct is excised with NotI and microinjected into the pronuclei of mouse embryos to generate transgenic animals as described above.
  • Plasmid vector pGP1a is digested with NotI and the following oligonucleotides ligated in: oligo-81 5'-ggc cgc ate ccg ggt etc gag gtc gac aag ctt teg agg ate cgc-3' oligo-82 5'-ggc cgc gga tec teg aaa get tgt cga cct cga gac ccg gga tgc-3'
  • the resulting plasmid, pGP1c contains a polylinker with Xmal, Xhol, Sall, Hindlll, and BamHI restriction sites flanked by NotI sites.
  • Plasmid vector pGP1a is digested with NotI and the following oligonucleotides ligated in: oligo-87 5'-ggc cgc tgt cga caa get tat cga tgg ate etc gag tgc -3' oligo-88 5'-ggc cgc act cga gga tec ate gat aag ctt gtc gac age -3'
  • the resulting plasmid, pGP1d contains a polylinker with Sall, Hindlll, Clal, BamHI, and Xhol restriction sites flanked by NotI sites.
  • a human placental genomic DNA library cloned into the phage vector ⁇ EMBL3/SP6/T7 (Clonetech Laboratories, Inc., Palo Alto, CA) was screened with the human kappa light chain J region specific oligonucleotide: oligo-36 5'- cac ctt egg cca agg gac aeg act gga gat taa acg taa gca -3' and the phage clones 136.2 and 136.5 isolated.
  • a 7.4 kb Xhol fragment that includes the J ⁇ 1 segment was isolated from
  • pCK1 a C ⁇ vector for expressing rearranged variable segments
  • the resulting clone, pCK1 can accept cloned fragments containing rearranged VJ ⁇ segments into the unique 5' Xhol site.
  • the transgene can then be excised with NotI and purified from vector sequences by gel electrophoresis.
  • the resulting transgene construct will contain the human J-C ⁇ intronic enhancer and may contain the human 3' ⁇ enhancer. 2.
  • pCK2 a C ⁇ vector with heavy chain enhancers for
  • pMHEl human heavy chain J- ⁇ intronic enhancer
  • pMHE2 BamHI/Hindlll fragment. This 2.3 kb fragment is isolated and cloned into pGP1c to generate pMHE2.
  • pMHE2 is digested with Sall and the 13 kb Xhol insert of p36.5 cloned in.
  • the resulting plasmid, pCK2 is identical to pCK1, except that the mouse and human heavy chain J- ⁇ intronic enhancers are fused to the 3' end of the transgene insert.
  • analogous constructs can be generated with different enhancers, i.e. the mouse or rat 3' kappa or heavy chain enhancer (Meyer and Neuberger, EMBO J..
  • V ⁇ specific oligonucleotide oligo-65 5'-agg ttc agt ggc agt ggg tct ggg aca gac ttc act etc ace ate age-3'
  • VJ ⁇ segment Clones that hybridized with both V and J probes are isolated and the DNA sequence of the rearranged VJ ⁇ segment determined.
  • Fragments containing functional VJ segments are subcloned into the unique Xhol sites of vectors pCK1 and pCK2 to generate
  • transgene constructs are isolated from vector sequences by digestion with NotI. Agarose gel purified insert is microinjected into mouse embryo pronuclei to generate transgenic animals.
  • Animals expressing human kappa chain are bred with heavy chain minilocus containing transgenic animals to generate mice expressing fully human antibodies.
  • VJK combinations may be capable of forming stable heavy-light chain complexes with a broad spectrum of different heavy chain VDJ combinations
  • several different light chain transgene constructs are generated, each using a different rearranged VJk clone, and transgenic mice that result from these constructs are bred with heavy chain minilocus transgene expressing mice.
  • Peripheral blood, spleen, and lymph node lymphocytes are isolated from double transgenic (both heavy and light chain constructs) animals, stained with fluorescent antibodies specific for human and mouse heavy and light chain immunoglobulins (Pharmingen, San Diego, CA) and analyzed by flow cytometry using a FACScan analyzer (Becton Dickinson, San Jose, CA).
  • Rearranged light chain transgenes constructs that result in the highest level of human heavy/light chain complexes on the surface of the highest number of B cells, and do not adversely affect the immune cell compartment (as assayed by flow cytometric
  • the 13 kb CK containing Xhol insert of p36.5 is treated with Klenow enzyme and cloned into Hindlll digested, Klenow-treated, plasmid pGPld.
  • a plasmid clone is selected such that the 5' end of the insert is adjacent to the vector derived Clal site.
  • the resulting plasmid, p36.5-ld is digested with Clal and Klenow-treated.
  • the J ⁇ l containing 7.4 kb Xhol insert of p36.2 is then Klenow-treated and cloned into the Clal, Klenow-treated p36.5-ld.
  • a clone is selected in which the p36.2 insert is in the same orientation as the p36.5 insert.
  • This clone, pJCKl (Fig. 34), contains the entire human JK region and C ⁇ , together with 7.2 kb of upstream sequences and 9 kb of downstream sequences.
  • the insert also contains the human J-C ⁇ intronic enhancer and may contain a human 3' ⁇ enhancer.
  • the insert is flanked by a unique 3' Sall site for the purpose of cloning additional 3' flanking sequences such as heavy chain or light chain enhancers.
  • a unique Xhol site is located at the 5' end of the insert for the purpose of cloning in unrearranged VK gene segments.
  • the unique Sall and Xhol sites are in turn flanked by NotI sites that are used to isolate the completed transgene construct away from vector sequences. 2. Isolation of unrearranged V ⁇ gene segments and generation of transgenic animals expressing human Ig light chain protein
  • V ⁇ specific oligonucleotide cligo-65 (discussed above), is used to probe a human placental genomic DNA library cloned into the phage vector 1EMBL3/3P6/T7 (Clonetech).
  • DNA fragments containing selected variable gene segments are cloned into the unique Xhol site of plasmid pJCK1 to generate minilocus constructs.
  • the resulting clones are digested with NotI and the inserts isolated and injected into mouse embryo pronuclei to generate transgenic animals.
  • the transgenes of these animals will undergo V to J joining in developing B-cells.
  • Animals expressing human kappa chain are bred with heavy chain minilocus containing transgenic animals to generate mice expressing fully human antibodies.
  • This Example describes the cloning of a human genomic heavy chain immunoglobulin transgene which is then introduced into the murine germline via microinjection into zygotes or integration in ES cells.
  • Nuclei are isolated from fresh human placental tissue as described by Marzluff, W.F., et al. (1985),
  • the isolated nuclei (or PBS washed human spermatocytes) are embedded in 0.5% low melting point agarose blocks and lyse with 1 mg/ml proteinase K in 500mM EDTA, 1% SDS for nuclei, or with lmg/ml proteinase K in 500mM EDTA, 1% SDS, lOmM DTT for spermatocytes at 50°C for 18 hours.
  • the proteinase K is inactivated by incubating the blocks in 40 ⁇ g/ml PMSF in TE for 30 minutes at 50°C, and then washing extensively with TE.
  • the DNA is then digested in the agarose with the restriction enzyme NotI as described by M. Finney in Current Protocols in Molecular Biology (F. Ausubel et al., eds. John Wiley & Sons, Supp. 4, 1988, e.g., Section 2.5.1).
  • the NotI digested DNA is then fractionated by pulsed field gel electrophoresis as described by Anand et al., Nuc. Acids Res. 17:3425-3433 (1989). Fractions enriched for the NotI fragment are assayed by Southern hybridization to detect one or more of the sequences encoded by this fragment. Such sequences include the heavy chain D segments, J segments, and ⁇ I constant regions together with representatives of all 6 V H families (although this fragment is identified as 670 kb fragment from HeLa cells by Berman et al. (1988), supra., we have found it to be an 830 kb fragment from human placental and sperm DNA). Those fractions containing this NotI
  • Plasmid pYACNN is prepared by digestion of pYACneo (Clontech) with EcoRI and ligation in the presence of the oligonucleotide 5' - AAT TGC GGC CGC - 3'.
  • YAC clones containing the heavy chain NotI fragment are isolated as described by Traver et al., Proc. Natl. Acad. Sci. USA. 86:5898-5902 (1989).
  • the cloned NotI insert is isolated from high molecular weight yeast DNA by pulse field gel electrophoresis as described by M. Finney, op. cit.
  • the DNA is condensed by the addition of 1 mM spermine and
  • the DNA is isolated by pulsed field gel electrophoresis and introduced into ES cells by lipofection (Gnirke et al., EMBO J. 10:1629-1634 (1991)), or the YAC is introduced into ES cells by spheroplast fusion.
  • the two fragments are coinjected into the nucleus of a mouse single cell embryo as described in Example 1.
  • An antigen of interest is used to immunize (see
  • the spleen is removed, and spleen cells used to generate hybridomas. Cells from an individual hybridoma clone that secretes
  • genomic DNA antibodies reactive with the antigen of interest are used to prepare genomic DNA.
  • a sample of the genomic DNA is digested with several different restriction enzymes that recognize unique six base pair sequences, and fractionated on an agarose gel.
  • Southern blot hybridization is used to identify two DNA fragments in the 2-10 kb range, one of which contains the single copy of the rearranged human heavy chain VDJ sequences and one of which contains the single copy of the rearranged human light chain VJ sequence. These two fragments are size fractionated on agarose gel and cloned directly into pUC18. The cloned inserts are then subcloned respectively into heavy and light chain expression cassettes that contain constant region sequences.
  • the plasmid clone p ⁇ el (Example 12) is used as a heavy chain expression cassette and rearranged VDJ sequences are cloned into the Xhol site.
  • the plasmid clone pCK1 is used as a light chain expression cassette and rearranged VJ
  • sequences are cloned into the Xhol site.
  • the resulting clones are used together to transfect SP 0 cells to produce antibodies that react with the antigen of interest (Co. et al., Proc. Natl. Acad. Sci. USA 88:2869 (1991), which is incorporated herein by reference).
  • mRNA is isolated from the cloned hybridoma cells described above, and used to synthesize cDNA.
  • the expressed human heavy and light chain VDJ and VJ sequence are then amplified by PCR and cloned (Larrick et al., Biol. Technology, 7:934-938 (1989)).
  • oligonucleotides are synthesized that encode the same polypeptides, and synthetic expression vectors generated as described by Queen et al., Proc. Natl. Acad. Sci. USA., 84: 5454-5458 (1989).
  • transgenic animals can be successfully immunized with complex antigens such as those on human red blood cells and respond with kinetics that are similar to the response kinetics observed in normal mice.
  • Blood cells generally are suitable immunogens and comprise many different types of antigens on the surface of red and white blood cells.
  • Tubes of human blood from a single donor were collected and used to immunize transgenic mice having
  • J H D functionally disrupted endogenous heavy chain loci
  • HCI human heavy chain minigene construct
  • mice are designated as line 112.
  • Blood was washed and resuspended in 50 mis Hanks' and diluted to 1x10 8 cells/ml 0.2 mis (2x10 7 cells) were then injected interperitoneally using a 28 gauge needle and 1 ce syringe. This immunization protocol was repeated approximately weekly for 6 weeks. Serum titers were monitored by taking blood from retro-orbital bleeds and collecting serum and later testing for specific antibody. A pre-immune bleed was also taken as a control. On the very last immunization, three days before these animals were sacrificed for serum and for hybridomas, a single immunization of 1 x 10 7 cells was given intravenously through the tail to enhance the production of hybridomas.
  • Mice # 2343 and 2348 have a desired phenotype: human heavy chain mini-gene transgenic on heavy chain knock-out
  • Hybridomas were generated by fusing mouse spleen cells of approximately 16 week-old transgenic mice (Table 9) that had been immunized as described (supra) to a fusion partner consisting of the non-secreting HAT-sensitive myeloma cell line, X63 Ag8.653.
  • Hybridoma clones were cultivated and hybridoma supernatants containing immunoglobulins having specific binding affinity for blood cell antigens were
  • Serum and hybridoma supernatants were tested using flow cytometry.
  • Red blood cells from the donor were washed 4X in Hanks' balanced salt solution and 50,000 cells were placed in 1.1 ml polypropylene microtubes.
  • Cells were incubated with antisera or supernatant from the hybridomas for 30 minutes on ice in staining media (lx RPMI 1640 media without phenol red or biotin (Irvine Scientific) 3% newborn calf serum, 0.1% Na azide). Controls consisted of littermate mice with other genotypes. Cells were then washed by centrifugation at 4°C in Sorvall RT600B for 5-10 minutes at 1000 rpm.
  • mice and littermate controls Serum of transgenic mice and littermate controls was incubated with either red blood cells from the donor, or white blood cells from another individual, washed and then developed with anti-human IgM FITC labeled antibody and analyzed in a flow cytometer. Results showed that serum from mice that are transgenic for the human mini-gene locus (mice 2343 and 2348) show human IgM reactivity whereas all littermate animals (2344, 2345, 2346, 2347) do not. Normal mouse serum (NS) and phosphate buffer saline (PBS) were used as negative controls. Red blood cells were ungated and white blood cells were gated to include only lymphocytes. Lines are drawn on the x and y axis to provide a reference. Flow cytometry was performed on 100 supernatants from fusion 2348. Four supernatants showed positive reactivity for blood cell antigens.
  • NS normal mouse serum
  • PBS phosphate buffer saline
  • the vector pGP1b (referred to in a previous example) is digested with Xhol and BamHI and ligated with the following oligonucleotides: 5'- gat cct cga gac cag gta cca gat ctt gtg aat teg -3'
  • This plasmid contains a polylinker that includes the following restriction sites:
  • a 0.8 kb Xbal/Bglll fragment of pVH251 (referred to in a previous example), that includes the promoter leader sequence exon, first intron, and part of the second exon of the human VH-V family immunoglobulin variable gene segment, was inserted into Xbal/Bglll digested vector pNN03 to generate the plasmid pVH251.
  • the BamHI/Hindlll fragment of pMHEl is cloned into BamHI/Hindlll cut pVhgh to generate the B-cell expression vector pBCE1.
  • This vector depicted in Fig. 36, contains unique Xhol and Asp718 cloning sites into which antisense DNA fragments can be cloned.
  • the expression of these antisense sequences is driven by the upstream heavy chain promoter- enhancer combination the downstream hGH gene sequences provide polyadenylation sequences in addition to intron sequences that promote the expression of transgene constructs.
  • Antisense transgene constructs generated from pBCEl can be separated from vector sequences by digestion with NotI.
  • PCR polymerase chain reaction
  • the resulting 0.3 kb PCR product is digested with Asp7l8 and Xhol and cloned into Asp718/XhoI digested pBCE1 to generate the antisense transgene construct pMASl.
  • the purified NotI insert of pMAS1 is microinjected into the pronuclei of half day mouse embryos-- alone or in combination with one or more other transgene constructs--to generate transgenic mice.
  • This construct expresses an RNA transcript in B-cells that hybridizes with mouse IgM mRNA, thus down-regulating the expression of mouse IgM protein.
  • Double transgenic mice containing pMAS1 and a human heavy chain transgene minilocus such as pHC1 will express the human transgene encoded Ig receptor on a higher percentage of B-cell than mice transgenic for the human heavy chain minilocus alone.
  • the ratio of human to mouse Ig receptor expressing cells is due in part to competition between the two populations for factors and cells that promoter B-cell differentiation and expansion. Because the Ig receptor plays a key role in B-cell
  • mouse Ig receptor expressing B-cells that express reduced levels of IgM on their surface (due to mouse Ig specific antisense down-regulation) during B-cell development will not compete as well as cells that express the human receptor.
  • RNA transcript in B-cells that hybridizes with mouse IgK mRNA, thus down-regulating the expression of mouse IgK protein as described above for pMAS1.
  • This example demonstrates the successful immunization and immune response in a transgenic mouse of the present invention.
  • KLH-DNP Keyhole limpet hemocyanin conjugated with greater than 400 dinitrophenyl groups per molecule (Calbiochem, La Jolla, California) (KLH-DNP) was alum precipitated according to a previously published method (Practical Immunology, L. Hudson and F.C. Hay, Blackwell Scientific (Pubs.), p. 9, 1980).
  • PBS phosphate buffered saline

Abstract

The invention relates to transgenic non-human animals capable of producing heterologous antibodies and transgenic non-human animals having inactivated endogenous immunoglobulin genes. In one aspect of the invention, endogenous immunoglobulin genes are suppresed by antisense polynucleotides and/or by antiserum directed against endogenous immunoglobulins. Heterologous antibodies are encoded by immunoglobulin genes not normally found in the genome of that species of non-human animal. In one aspect of the invention, one or more transgenes containing sequences of unrearranged heterogolous human immunoglobulin heavy chains are introduced into a non-human animal thereby forming a transgenic animal capable of functionally rearranging transgenic immunoglobulin sequences and producing a repertoire of antibodies of various isotypes encoded by human immunoglobulin genes. Such heterologous human antibodies are produced in B-cells which are thereafter immortalized, e.g., by fusing with an immortalizing cell line such as a myeloma or by manipulating such B-cells by other techniques to perpetuate a cell line capable of producing a monoclonal heterologous antibody. The invention also relates to heavy and light chain immunoglobulin transgenes for making such transgenic non-human animals as well as methods and vectors for disrupting endogenous immunoglobulin loci in the transgenic animal.

Description

Transgenic non-human animals capable of producing heterologous antibodies
TECHNICAL FIELD
The invention relates to transgenic non-human animals capable of producing heterologous antibodies,
transgenes used to produce such transgenic animals,
transgenes capable of functionally rearranging a heterologous D gene in V-D-J recombination, immortalized B-cells capable of producing heterologous antibodies, methods and transgenes for producing heterologous antibodies of multiple isotypes, methods and transgenes for inactivating or suppressing
expression of endogenous immunoglobulin loci, methods and transgenes for producing heterologous antibodies wherein a variable region sequence comprises somatic mutation as
compared to germline rearranged variable region sequences, and transgenic nonhuman animals which produce antibodies having a human primary sequence and which bind to human antigens.
BACKGROUND OF THE INVENTION
One of the major impediments facing the development of in vivo therapeutic and diagnostic applications for
monoclonal antibodies in humans is the intrinsic
immunogenicity of non-human immunoglobulins. For example, when immunocompetent human patients are administered therapeutic doses of rodent monoclonal antibodies, the patients produce antibodies against the rodent immunoglobulin sequences; these human anti-mouse antibodies (KAMA) neutralize the therapeutic antibodies and can cause acute toxicity. Hence, it is
desirable to produce human immunoglobulins that are reactive with specific human antigens that are promising therapeutic and/or diagnostic targets. However, producing human immunoglobulins that bind specifically with human antigens is problematic.
The present technology for generating monoclonal antibodies involves pre-exposing, or priming, an animal
(usually a rat or mouse) with antigen, harvesting B-cells from that animal, and generating a library of hybridoma clones. By screening a hybridoma population for. antigen binding
specificity (idiotype) and also screening for immunoglobulin class (isotype), it is possible to select hybridoma clones that secrete the desired antibody.
However, when present methods for generating
monoclonal antibodies are applied for the purpose of
generating human antibodies that have binding specificities for human antigens, obtaining B-lymphocytes which produce human immunoglobulins a serious obstacle, since humans will typically not make immune responses against self-antigens.
Hence, present methods of generating human monoclonal antibodies that are specifically reactive with human antigens are clearly insufficient. It is evident that the same limitations on generating monoclonal antibodies to authentic self antigens apply where non-human species are used as the source of B-cells for making the hybridoma.
The construction of transgenic animals harboring a functional heterologous immunoglobulin transgene are a method by which antibodies reactive with self antigens may be
produced. However, in order to obtain expression of
therapeutically useful antibodies, or hybridoma clones
producing such antibodies, the transgenic animal must produce transgenic B cells that are capable of maturing through the B lymphocyte development pathway. Such maturation requires the presence of surface IgM on the transgenic B cells, however isotypes other than IgM are desired for therapeutic uses.
Thus, there is a need for transgenes and animals harboring such transgenes that are able to undergo functional V-D-J rearrangement to generate recombinational diversity and
junctional diversity. Further, such transgenes and transgenic animals preferably include cis-acting sequences that
facilitate isotype switching from a first isotype that is required for B cell maturation to a subsequent isotype that has superior therapeutic utility.
A number of experiments have reported the use of transfected cell lines to determine the specific DNA sequences required for Ig gene rearrangement (reviewed by Lewis and Gellert (1989), Cell, 59, 585-588). Such reports have
identified putative sequences and concluded that the
accessibility of these sequences to the recombinase enzymes used for rearrangement is modulated by transcription
(Yancopoulos and Alt (1985), Cell, 40, 271-281). The
sequences for V(D)J joining are reportedly a highly conserved, near-palindromic heptamer and a less well conserved AT-rich nanomer separated by a spacer of either 12 or 23 bp (Tonegawa (1983), Nature, 302, 575-581; Hesse, et al. (1989), Genes in Dev. , 3, 1053-1061). Efficient recombination reportedly occurs only between sites containing recombination signal sequences with different length spacer regions.
Ig gene rearrangement, though studied in tissue culture cells, has not been extensively examined in transgenic mice. Only a handful of reports have been published
describing rearrangement test constructs introduced into mice [Buchini, et al. (1987), Nature. 326, 409-411 (unrearranged chicken λ transgene); Goodhart, et al. (1987) , Proc. Natl. Acad. Sci. USA, 84, 4229-4233) (unrearranged rabbit k gene); and Bruggemann, et al. (1989), Proc. Natl. Acad. Sci. USA, 86, 6709-6713 (hybrid mouse-human heavy chain)]. The results of such experiments, however, have been variable, in some cases, producing incomplete or minimal rearrangement of the
transgene.
Further, a variety of biological functions of antibody molecules are exerted by the Fc portion of molecules, such as the interaction with mast cells or basophils through Fee, and binding of complement by Fcμ or Fcγ, it further is desirable to generate a functional diversity of antibodies of a given specificity by variation of isotype.
Although transgenic animals have been generated that incorporate transgenes encoding one or more chains of a heterologous antibody, there have been no reports of hererologous transgenes that undergo successful isotype switching. Transgenic animals that cannot switch isotypes are limited to producing h-iterologous antibodies of a single isotype, and more specifically are limited to producing an isotype that is essential for B cell maturation, such as IgM and possibly IgD, which may be of limited therapeutic utility. Thus, there is a need for heterologous immunoglobulin
transgenes and transgenic animals that are capable of
switching from an isotype needed for B cell development to an isotype that has a desired characteristic for therapeutic use.
Based on the foregoing, it is clear that a need exists for methods of efficiently producing heterologous antibodies, e.g. antibodies encoded by genetic sequences of a first species that are produced in a second species. More particularly, there is a need in the art for heterologous immunoglobulin transgenes and transgenic animals that are capable of undergoing functional V-D-J gene rearrangement that incorporates all or a portion of a D gene segment which contributes to recombinational diversity. Further, there is a need in the art for transgenes and transgenic animals that can support V-D-J recombination and isotype switching so that (1) functional B cell development may occur, and (2)
therapeutically useful heterologous antibodies may be
produced. There is also a need for a source of B cells which can be used to make hybridomas that produce monoclonal
antibodies for therapeutic or diagnostic use in the particular species for which they are designed. A heterologous
immunoglobulin transgene capable of functional V-D-J
recombination and/or capable of isotype switching could
fulfill these needs.
In accordance with the foregoing object transgenic nonhuman animals are provided which are capable of producing a heterologous antibody, such as a human antibody.
Further, it is an object to provide B-cells from such transgenic animals which are capable of expressing
heterologous antibodies wherein such B-cells are immortalized to provide a source of a monoclonal antibody specific for a particular antigen. In accordance with this foregoing object, it is a further object of the invention to provide hybridoma cells that are capable of producing such heterologous monoclonal antibodies.
Still further, it is an object herein to provide heterologous unrearranged and rearranged immunoglobulin heavy and light chain transgenes useful for producing the
aforementioned non-human transgenic animals.
Still further, it is an object herein to provide methods to disrupt endogenous immunoglobulin loci in the transgenic animals.
Still further, it is an object herein to provide methods to induce heterologous antibody production in the aforementioned transgenic non-human animal.
A further object of the invention is to provide methods to generate an immunoglobulin variable region gene segment repertoire that is used to construct one or more transgenes of the invention.
The references discussed herein are provided solely for their disclosure prior to the filing date of the present application. Nothing herein is to be construed as an
admission that the inventors are not entitled to antedate such disclosure by virtue of prior invention.
SUMMARY OF THE INVENTION
Transgenic nonhuman animals are provided which are capable of producing a heterologous antibody, such as a human antibody. Such heterologous antibodies may be of various isotypes, including: IgG1, IgG2, IgG3, IgG4, IgM, IgA1, IgA2, IgAsec, IgD, of IgE. In order for such transgenic nonhuman animals to make an immune response, it is necessary for the transgenic B cells and pre-B cells to produce surface-bound immunoglobulin, particularly of the IgM (or possibly IgD) isotype, in order to effectuate B cell development and
antigen-stimulated maturation. Such expression of an IgM (or IgD) surface-bound immunoglobulin is only required during the antigen-stimulated maturation phase of B cell development, and mature B cells may produce other isorypes, although only a single switched isotype may be produced at a time.
Typically, a cell of the B-cell lineage will produce only a single isotype at a time, although cis or trans
alternative RNA splicing, such as occurs naturally with the μs (secreted μ) and μM (membrane-bound μ) forms, and the μ and δ immunoglobulin chains, may lead to the contemporaneous
expression of multiple isotypes by a single cell. Therefore, in order to produce heterologous antibodies of multiple isotypes, specifically the therapeutically useful IgG, IgA, and IgE isotypes, it is necessary that isotype switching occur. Such isotype switching may be classical class- switching or may result from one or more non-classical isotype switching mechanisms.
The invention provides heterologous immunoglobulin transgenes and transgenic nonhuman animals harboring such transgenes, wherein the transgenic animal is capable of producing heterologous antibodies of multiple isotypes by undergoing isotype switching. Classical isotype switching occurs by recombination events which involve at least one switch sequence region in the transgene. Non-classical isotype switching may occur by, for example, homologous recombination between human σμ and human Σμ sequences (δ- associated deletion). Alternative non-classical switching mechanisms, such as intertransgene and/or interchromosomal recombination, among others, may occur and effectuate isotype switching. Such transgenes and transgenic nonhuman animals produce a first immunoglobulin isotype that is necessary for antigen-stimulated B cell maturation and can switch to encode and produce one or more subsequent heterologous isotypes that have therapeutic and/or diagnostic utility. Transgenic
nonhuman animals of the invention are thus able to produce, in one embodiment, IgG, IgA, and/or IgE antibodies that are encoded by human immunoglobulin genetic sequences and which also bind specific human antigens with high affinity.
The invention also encompasses B-cells from such transgenic animals that are capable of expressing heterologous antibodies of various isotypes, wherein such B-cells are immortalized to provide a source of a monoclonal antibody specific for a particular antigen. Hybridoma cells that are derived from such B-cells can serve as one source of such heterologous monoclonal antibodies.
The invention provides heterologous unrearranged and rearranged immunoglobulin heavy and light chain transgenes capable of undergoing isotype switching in vivo in the
aforementioned non-human transgenic animals or in explanted lymphocytes of the B-cell lineage from such transgenic
animals. Such isotype switching may occur spontaneously or be induced by treatment of the transgenic animal or explanted B- lineage lymphocytes with agents that promote isotype
switching, such as T-cell-derived lymphokines (e.g., IL-4 and IFNγ).
Still further, the invention includes methods to induce heterologous antibody production in the aforementioned transgenic non-human animal, wherein such antibodies may be of various isotypes. These methods include producing an antigen- stimulated immune response in a transgenic nonhuman animal for the generation of heterologous antibodies, particularly heterologous antibodies of a switched isotype (i.e., IgG, IgA, and IgE).
This invention provides methods whereby the transgene contains sequences that effectuate isotype
switching, so that the heterologous immunoglobulins produced in the transgenic animal and monoclonal antibody clones derived from the B-cells of said animal may be of various isotypes.
This invention further provides methods that facilitate isotype switching of the transgene, so that
switching between particular isotypes may occur at much higher or lower frequencies or in different temporal orders than typically occurs in germline immunoglobulin loci. Switch regions may be grafted from various CH genes and ligated to other CH genes in a transgene construct; such grafted switch sequences will typically function independently of the
associated CH gene so that switching in the transgene
construct will typically be a function of the origin of the associated switch regions. Alternatively, or in combination with switch sequences, δ-associated deletion sequences may be linked to various CH genes to effect non-classical switching by deletion of sequences between two δ-associated deletion sequences. Thus, a transgene may be constructed so that a particular CH gene is linked to a different switch sequence and thereby is switched to more frequently than occurs when the naturally associated switch region is used.
This invention also provides methods to determine whether isotype switching of transgene sequences has occurred in a transgenic animal containing an immunoglobulin transgene.
The invention provides immunoglobulin transgene constructs and methods for producing immunoglobulin transgene constructs, some of which contain a subset of germline
immunoglobulin loci sequences (which may include deletions). The invention includes a specific method for facilitated cloning and construction of immunoglobulin transgenes,
involving a vector that employs unique Xhol and Sall
restriction sites flanked by two unique NotI sites. This method exploits the complementary termini of Xhol and Sall restrictions sites and is useful for creating large constructs by ordered concatemerization of restriction fragments in a vector.
The transgenes of the invention include a heavy chain transgene comprising DNA encoding at least one variable gene segment, one diversity gene segment, one joining gene segment and one constant region gene segment. The
immunoglobulin light chain transgene comprises DNA encoding at least one variable gene segment, one joining gene segment and one constant region gene segment. The gene segments encoding the light and heavy chain gene segments are heterologous to the transgenic non-human animal in that they are derived from, or correspond to, DNA encoding immunoglobulin heavy and light chain gene segments from a species not consisting of the transgenic non-human animal. In one aspect of the invention, the transgene is constructed such that the individual gene segments are unrearranged, i.e., not rearranged so as to encode a functional immunoglobulin light or heavy chain. Such unrearranged transgenes permit recombination of the gene segments (functional rearrangement) and expression of the resultant rearranged immunoglobulin heavy and/or light chains within the transgenic non-human animal when said animal is exposed to antigen.
In one aspect of the invention, heterologous heavy and light immunoglobulin transgenes comprise relatively large fragments of unrearranged heterologous DNA. Such fragments typically comprise a substantial portion of the C, J (and in the case of heavy chain, D) segments from a heterologous immunoglobulin locus. In addition, such fragments also comprise a substantial portion of the variable gene segments.
In one embodiment, such transgene constructs
comprise regulatory sequences, e.g. promoters, enhancers, class switch regions, recombination signals and the like, corresponding to sequences derived from the heterologous DNA. Alternatively, such regulatory sequences may be incorporated into the transgene from the same or a related species of the non-human animal used in the invention. For example, human immunoglobulin gene segments may be combined in a transgene with a rodent immunoglobulin enhancer sequence for use in a transgenic mouse.
In a method of the invention, a transgenic non-human animal containing germline unrearranged light and heavy immunoglobulin transgenes - that undergo VDJ joining during D-cell differentiation - is contacted with an antigen to induce production of a heterologous antibody in a secondary repertoire B-cell.
Also included in the invention are vectors and methods to disrupt the endogenous immunoglobulin loci in the non-human animal to be used in the invention. Such vectors and methods utilize a transgene, preferably positive-negative selection vector, which is constructed such that it targets the functional disruption of a class of gene segments encoding a heavy and/or light immunoglobulin chain endogenous to the non-human animal used in the invention. Such endogenous gene segments include diversity, joining and constant region gene segments. In this aspect of the invention, the positive-negative selection vector is contacted, with at least one embryonic stem cell of a non-human animal after which cells are selected wherein the positive-negative selection vector has integrated into the genome of the non-human animal by way of homologous recombination. After transplantation, the resultant transgenic non-human animal is substantially incapable of mounting an immunoglobulin-mediated immune response as a result of homologous integration of the vector into chromosomal DNA. Such immune deficient non-human animals may thereafter be used for study of immune deficiencies or used as the recipient of heterologous immunoglobulin heavy and light chain transgenes.
The invention also provides vectors, methods, and compositions useful for suppressing the expression of one or more species of immunoglobulin chain(s), without disrupting an endogenous immunoglobulin locus. Such methods are useful for suppressing expression of one or more endogenous
immunoglobulin chains while permitting the expression of one or more transgene-encoded immunoglobulin chains. Unlike genetic disruption of an endogenous immunoglobulin chain locus, suppression of immunoglobulin chain expression does not require the time-consuming breeding that is needed to
establish transgenic animals homozygous for a disrupted endogenous Ig locus. An additional advantage of suppression as compared to engognous Ig gene disruption is that, in certain embodiments, chain suppression is reversible within an individual animal. For example, Ig chain suppression may be accomplished with: (1) transgenes encoding and expressing antisense RNA that specifically hybridizes to an endogenous Ig chain gene sequence, (2) antisense oligonucleotides that specifically hybridize to. an endogenous Ig chain gene
sequence, and (3) immunoglobulins that bind specifically to an endogenous Ig chain polypeptide.
The references discussed herein are provided solely for their disclosure prior to the filing date of the present application. Nothing herein is to be construed as an
admission that the inventors are not entitled to antedate such disclosure by virtue of prior invention. BRIEF DESCRIPTION OF THE FIGURES
Fig. 1 depicts the complementarity determining regions CDR1, CDR2 and CDR3 and framework regions FR1, FR2, FR3 and FR4 in unrearranged genomic DNA and mRNA expressed from a rearranged immunoglobulin heavy chain gene,
Fig. 2 depicts the human λ chain locus,
Fig. 3 depicts. the human κ chain locus,
Fig. 4 depicts the human heavy chain locus, Fig. 5 depicts a transgene construct containing a rearranged IgM gene ligated to a 25 kb fragment that contains human γ3 and γ1 constant regions followed by a 700 bp fragment containing the rat chain 3' enhancer sequence.
Fig. 6 is a restriction map of the human κ chain locus depicting the fragments to be used to form a light chain transgene by way of in vivo homologous recombination.
Fig. 7 depicts the construction of pGP1.
Fig. 8 depicts the construction of the polylinker contained in pGP1.
Fig. 9 depicts the fragments used to construct a human heavy chain transgene of the invention.
Fig. 10 depicts the construction of pHIG1 and pCON1. Fig. 11 depicts the human Cγl fragments which are inserted into pRE3 (rat enhancer 3') to form pREG2.
Fig. 12 depicts the construction of pHIG3' and PCON.
Fig. 13 depicts the fragment containing human D region segments used in construction of the transgenes of the invention.
Fig. 14 depicts the construction of pHIG2 (D segment. containing plasmid).
Fig. 15 depicts the fragments covering the human Jκ and human Cκ gene segments used in constructing a transgene of the invention.
Fig. 16 depicts the structure of pEμ.
Fig. 17 depicts the construction of pKapH.
Figs. 18A through 18D depict the construction of a positive-negative selection vector for functionally disrupting the endogenous heavy chain immunoglobulin locus of mouse. Figs. 19A through 19C depict the construction of a positive-negative selection vector for functionally disrupting the endogenous immunoglobulin light-chain loci in mouse.
Figs. 20 a through e depict the structure of a kappa light chain targeting vector.
Figs. 21 a through f depict the structure of a mouse heavy chain targeting vector.
Fig. 22 depicts the map of vector pGPe.
Fig. 23 depicts the structure of vector pJM2.
Fig. 24 depicts the structure of vector pCOR1.
Fig. 25 depicts the transgene constructs for pIGM1, pHC1 and pHC2.
Fig. 26 depicts the structure of pγe2.
Fig. 27 depicts the structure of pVGE1.
Fig. 28 depicts the assay results of human Ig expression in a pHC1 transgenic mouse.
Fig,. 29 depicts the structure of pJCK1.
Fig. 30 depicts the construction of a synthetic heavy chain variable region.
Fig. 31 is a schematic representation of the'heavy chain minilocus constructs pIGM1, pHC1, and pHC2.
Fig. 32 is a schematic representation of the heavy chain minilocus construct pIGG1 and the κ light chain
minilocus construct pKC1, pKVe1, and pKC2.
Fig. 33 depicts a scheme to reconstruct functionally rearranged light chain genes.
Fig. 34 depicts serum ELISA results
Fig. 35 depicts the results of an ELISA assay of serum from 8 transgenic mice.
Fig. 36 is a schematic representation of plasmid pBCE1.
Fig. 37 depicts the immune response of transgenic mice of the present invention against KLH-DNP, by measuring IgG and IgM levels specific for KLH-DNP (37A), KLH (37B) and BSA-DNP (37C).
Fig. 38 shows ELISA data demonstrating the presence of antibodies that bind human carcinoembryonic antigen (CEA) and comprise human μ chains; each panel shows reciprocal serial dilutions from pooled serum samples obtained from mice on the indicated day following immunization.
Fig. 39 shows ELISA data demonstrating the presence of antibodies that bind human carcinoembryonic antigen (CEA) and comprise human γ chains; each panel shows reciprocal serial dilutions from pooled serum samples obtained from mice on the indicated day following immunization.
Fig. 40 shows aligned variable region sequences of 23 randomly-chosen cDNAs generated from mRNA obtained from lymphoid tissue of HCI transgenic mice immunized, with human carcinoembryonic antigen (CEA) as compared to the germline transgene sequence (top line); on each line nucleotide changes relative to germline sequence are shown above the alteration in deduced amino acid sequence (if any); the regions
corresponding to heavy chain CDR1, CDR2, and CDR3 are
indicated. Non-germline encoded nucleotides are shown in capital letters. Germline VH251 and JH are shown in lower case letters. Deduced amino acid changes are given beneath
nucleotide sequences using th conventional single-letter notation.
Fig. 41 shows the data from Fig. 40 in histogram format; deduced amino acid residue position is shown as the ordinate (left is the amino-terminal direction, right is in the direction towards the carboxy-terminus) and frequency of sequence variation is shown as the abscissa.
Fig..42 show the nucleotide sequence of a human DNA fragment, designated vk65.3, containing a Vκ gene segment; the deduced amino acid sequences of the Vκ coding regions are also shown; splicing and recombination signal sequences
(heptamer/nonamer) are shown boxed.
Fig. 43 show the nucleotide sequence of a human DNA fragment, designated vk65.5, containing a Vκ gene segment; the deduced amino acid sequences of the Vκ coding regions are also shown; splicing and recombination signal sequences
(heptamer/nonamer) are shown boxed.
Fig. 44 show the nucleotide sequence of a human DNA fragment, designated vk65.8, containing a Vκ gene segment; the deduced amino acid sequences of the Vκ coding regions are also shown; splicing and recombination signal sequences
(hεptamer/nonamer) are shewn boxed.
Fig. 45 show the nucleotide sequence of a human DNA fragment, designated vk65.15, containing a Vκ gene segment; the deduced amino acid sequences of the Vκ coding regions are also shown; splicing and recombination signal sequences
(heptamer/nonamer) are shown boxed.
Fig. 46 shows formation of a light chain minilocus by homologous recombination between two overlapping fragments which were co-injected.
Table 1 depicts the sequence of vector pGPe.
Table 2 depicts the sequence of gene VH4 .8.
Table 3 depicts the detection of human IgM and IgG in the serum of transgenic mice of this invention.
Table 4 depicts sequences of VDJ joints.
Table 5 depicts the distribution of J segments incorporated into pHC1 transgene encoded transcripts to J segments found in adult human peripheral blood lymphocytes (PBL).
Table 6 depicts the distribution of D segments incorporated into pHC1 transgene encoded transcripts to D segments found in adult human peripheral blood lymphocytes (PBL).
Table 7 depicts the length of the CDR3 peptides from transcripts with in-frame VDJ joints in the pHC1 transgenic mouse and in human PBL.
Table 8 depicts the predicted amino acid sequences of the VDJ regions from 30 clones analyzed from a pHC1
transgenic.
Table 9 shows transgenic mice of line 112 that were used in the indicated experiments; (+) indicates the presence of the respective transgene, (++) indicates that the animal is homozygous for the JHD knockout transgene. DETAILED DESCRIPTION
As has been discussed supra, it is desirable to produce human immunoglobulins that are reactive with specific human antigens that are promising therapeutic and/or diagnostic targets. However, producing human immunoglobulins that bind specifically with human antigens is problematic.
First, the immunized animal that serves as the source of B cells must make an immune response against the presented antigen. In order for an animal to make an immune response, the antigen presented must be foreign and the animal must not be tolerant to the antigen. Thus, for example, if it is desired to produce a human monoclonal antibody with an idiotype that binds to a human protein, self-tolerance will prevent an immunized human from making a substantial immune response to the human protein, since the only epitopes of the antigen that may be immunogenic will be those that result from polymorphism of the protein within the human population
(allogeneic epitopes).
Second, if the animal that serves as the source of
B-cells for forming a hybridoma (a human in the illustrative given example) does make an immune response against an
authentic self antigen, a severe autoimmune disease may result in the animal. Where humans would be used as a source of B- cells for a hybridoma, such autoimmunization would be
considered unethical by contemporary standards.
One methodology that can be used to obtain human antibodies that are specifically reactive with human antigens is the production of a transgenic mouse harboring the human immunoglobulin transgene constructs of this invention.
Briefly, transgenes containing all or portions of the human immunoglobulin heavy and light chain loci, or transgenes containing synthetic "miniloci" (described infra, and in
PCT/US91/06185 filed August 28, 1991) which comprise essential functional elements of the human heavy and light chain loci, are employed to produce a transgenic nonhuman animal. Such a transgenic nonhuman animal will have the capacity to produce immunoglobulin chains that are encoded by human immunoglobulin genes, and additionally will be capable of making an immune response against human antigens. Thus, such transgenic animals can serve as a source of immune sera reactive with specified human antigens, and B-cells from such transgenic animals can be fused with myeloma cells to produce hybridomas that secrete monoclonal antibodies that are encoded by human, immunoglobulin genes and which are specifically reactive with human antigens.
The production of transgenic mice containing various forms of immunoglobulin genes has been reported previously. Rearranged mouse immunoglobulin heavy or light chain genes have been used to produce transgenic mice. In addition, functionally rearranged human Ig genes including the μ or γ1 constant region have been expressed in transgenic mice.
However, experiments in which the transgene comprises
unrearranged (V-D-J or V-J not rearranged) immunoglobulin genes have been variable, in some cases, producing incomplete or minimal rearrangement of the transgene. However, there are no published examples of either rearranged or unrearranged immunoglobulin transgenes which undergo successful isotype switching between CH genes within a transgene.
Definitions
As used herein, the term "antibody" refers to a glycoprotein comprising at least two light polypeptide chains and two heavy polypeptide chains. Each of the heavy and light polypeptide chains contains a variable region (generally the amino terminal portion of the polypeptide chain) which
contains a binding domain which interacts with antigen. Each of the heavy and light polypeptide chains also comprises a constant region of the polypeptide chains (generally the carboxyl terminal portion) which may mediate the binding of the immunoglobulin to host tissues or factors including
various cells of the immune system, some phagocytic cells and the first component (C1q) of the classical complement system.
As used herein, a "heterologous antibody" is defined in relation to the transgenic non-human organism producing such an antibody. It is defined as an antibody having an amino acid sequence or an encoding DNA sequence corresponding to that found in an organism not consisting of the transgenic non-human animal.
As used herein, a "heterohybrid antibody" refers to an antibody having a light and heavy chains of different organismal origins. For example, an antibody having a human heavy chain associated with a murine light chain is a
heterohybrid antibody.
As used herein, "isotype" refers to the antibody class (e.g., IgM or IgG1) that is encoded by heavy chain constant region genes.
As used herein, "isotype switching" refers to the phenomenon by which the class, or isotype, of an antibody changes from one Ig class to one of the other Ig classes.
As used herein, "nonswitched isotype" refers to the isotypic class of heavy chain that is produced when no isotype switching has taken place; the CH gene encoding the
nonswitched isotype is typically the first CH gene immediately downstream from the functionally rearranged VDJ gene.
As used herein, the term "switch sequence" refers to those DNA sequences responsible for switch recombination. A "switch donor" sequence, typically a μ switch region, will be 5' (i.e., upstream) of the construct region to be deleted during the switch recombination. The "switch acceptor" region will be between the construct region to be deleted and the replacement constant region (e.g., γ, ε, etc.). As there is no specific site where recombination always occurs, the final gene sequence will typically not be predictable from the construct.
As used herein, "glycosylation pattern" is defined as the pattern of carbohydrate units that are covalently attached to a protein, more specifically to an immunoglobulin protein. A glycosylation pattern of a heterologous antibody can be characterized as being substantially similar to
glycosylation patterns which occur naturally on antibodies produced by the species of the nonhuman transgenic animal, when one of ordinary skill in the art- would recognize the glycosylation pattern of the heterologous antibody as being more similar to said pattern of glycosylation in the species of the nonhuman transgenic animal than to the species from which the CH genes of the transgene were derived.
As used herein, "specific binding" refers to the property of the antibody: (1) to bind to a predetermined antigen with an affinity of at least 1 x 107 M-1, and (2) to preferentially bind to the predetermined antigen with an affinity that is at least twc-fold greater than its affinity for binding to a non-specific antigen (e.g., BSA, casein) other than the predetermined antigen.
The term "naturally-occurring" as used herein as applied to an object refers to the fact that an object can be found in nature. For example, a polypeptide or polynucleotide sequence that is present in an organism (including viruses) that can be isolated from a source in nature and which has not been intentionally modified by man in the laboratory is naturally-occurring.
The term "rearranged" as used herein refers to a configuration of a heavy chain or light chain immunoglobulin locus wherein a V segment is positioned immediately adjacent to a D-J or J segment in a conformation encoding essentially a complete VH or VL domain, respectively. A rearranged
immunoglobulin gene locus can be identified by comparison to germline DNA; a rearranged locus will have at least one recombined heptamer/nonamer homology element.
The term "unrearranged" or "germline configuration" as used herein in reference to a V segment refers to the configuration wherein the V segment is not recombined so as to be immediately adjacent to a D or J segment.
Transgenic Nonhuman Animals Capable
of Producing Heterologous Antibodies
The design of a transgenic non-human animal that responds to foreign antigen stimulation with a heterologous antibody repertoire, requires that the heterologous
immunoglobulin transgenes contained within the transgenic animal function correctly throughout the pathway of B-cell development. In a preferred embodiment, correct function of a heterologous heavy chain transgene includes isotype switching. Accordingly, the transgenes of the invention are constructed so as to produce isotype switching and one or more of the following: (1) high level and cell-type specific expression, (2) functional gene rearrangement, (3) activation of and response to allelic exclusion, (4) expression of a sufficient primary repertoire, (5) signal transduction, (6) somatic hypermutation, and (7) domination of the transgene antibody locus during the immune response.
As will be apparent from the following disclosure, not all of the foregoing criteria need be met. For example, in those embodiments wherein the endogenous immunoglobulin loci of the transgenic animal are functionally disrupted, the transgene need not activate allelic exclusion. Further, in those embodiments wherein the transgene comprises a
functionally rearranged heavy and/or light chain
immunoglobulin gene, the second criteria of functional gene rearrangement is unnecessary, at least for that transgene which is already rearranged. For background on molecular immunology, see, Fundamental Immunology, 2nd edition (1989), Paul William E., ed. Raven Press, N.Y., which is incorporated herein by reference.
In one aspect of the invention, transgenic non-human animals are provided that contain rearranged, unrearranged or a combination of rearranged and unrearranged heterologous immunoglobulin heavy and light chain transgenes in the
germline of the transgenic animal. Each of the heavy chain transgenes comprises at least one CH gene. In addition, the heavy chain transgene may contain functional isotype switch sequences, which are capable of supporting isotype switching of a heterologous transgene encoding multiple CH genes in B- cells of the transgenic animal. Such switch sequences may be those which occur naturally in the germline immunoglobulin locus from the species that serves as the source of the transgene CH genes, or such switch sequences may be derived from those which occur in the species that is to receive the transgene construct (the transgeneic animal). For example, a human transgene construct that is used to produce a transgenic mouse may produce a higher frequency of isotype switching events if it incorporates switch sequences similar to those that occur naturally in the mouse heavy chain locus, as presumably the mouse switch sequences are optimized to
function with the mouse switch recombinase enzyme system, whereas the human switch sequences are not. Switch sequences made be isolated and cloned by conventional cloning methods, or may be synthesized de novo from overlapping synthetic oligonucleotides designed on the basis of published sequence information relating to immunoglobulin switch region sequences (Mills et al., Nucl. Acids Res. 18:7305-7316 (1991);
Sideras et al., Intl. Immunol. 1:631-642 (1989), which are incorporated herein by reference).
For each of the foregoing transgenic animals, functionally rearranged heterologous heavy and light chain immunoglobulin transgenes are found in a significant fraction of the B-cells of the transgenic animal (at least 10 percent).
The transgenes of the invention include a heavy chain transgene comprising DNA encoding at least one variable gene segment, one diversity gene segment, one joining gene segment and at least one constant region gene segment. The immunoglobulin light chain transgene comprises DNA encoding at least one variable gene segment, one joining gene segment and at least one constant region gene segment. The gene segments encoding the light and heavy chain gene segments are
heterologous to the transgenic non-human animal in that they are derived from, or correspond to, DNA encoding
immunoglobulin heavy and light chain gene segments from a species not consisting of the transgenic non-human animal. In one aspect of the invention, the transgene is constructed such that the individual gene segments are unrearranged, i.e., not rearranged so as to encode a functional immunoglobulin light or heavy chain. Such unrearranged transgenes support
recombination of the V, D, and J gene segments (functional rearrangement) and preferably support incorporation of all ora portion of a D region gene segment in the resultant
rearranged immunoglobulin heavy chain within the transgenic non-human animal when exposed to antigen.
In an alternate embodiment, the transgenes comprise an unrearranged "mini-locus". Such transgenes typically
comprise a substantial portion of the C, D, and J segments as well as a subset of the V gene segments. In such transgene constructs, the various regulatory sequences, e.g. promoters, enhancers, class switch regions, splice-donor and splice- acceptor sequences for RNA processing, recombination signals and the like, comprise corresponding sequences derived from the heterologous DNA. Such regulatory sequences may be incorporated into the transgene from the same or a related species of the non-human animal used in the invention. For example, human immunoglobulin gene segments may be combined in a transgene with a rodent immunoglobulin enhancer sequence for use in a transgenic mouse. Alternatively, synthetic regulatory sequences may be incorporated into the transgene, wherein such synthetic regulatory sequences are not homologous to a
functional DNA sequence that is known to occur naturally in the genomes of mammals. Synthetic regulatory sequences are designed according to consensus rules, such as, for example, those specifying the permissible sequences of a splice- acceptor site or a promoter/enhancer motif.
The invention also includes transgenic animals containing germ line cells having a heavy and light transgene wherein one of the said transgenes contains rearranged gene segments with the other containing unrearranged gene segments. In the preferred embodiments, the rearranged transgene is a light chain immunoglobulin transgene and the unrearranged transgene is a heavy chain immunoglobulin transgene. The Structure and Generation of Antibodies
The basic structure of all immunoglobulins is based upon a unit consisting of two light polypeptide chains and two heavy polypeptide chains. Each light chain comprises two regions known as the variable light chain region and the constant light chain region. Similarly, the immunoglobulin heavy chain comprises two regions designated the variable heavy chain region and the constant heavy chain region.
The constant region for the heavy or light chain is encoded by genomic sequences referred to as heavy or light constant region gerie (CH) segments. The use of a particular heavy chain gene segment defines the class of immunoglobulin. For example, in humans, the μ constant region gene segments define the IgM class of antibody whereas the use of a γ, γ2, γ3 or γ4 constant region gene segment defines the IgG class of antibodies as well as the IgG subclasses IgG1 through IgG4. Similarly, the use of a α1 or α2 constant region gene segment defines the IgA class of antibodies as well as the subclasses IgA1 and IgA2. The δ and e constant region gene segments define the IgD and IgE antibodv classes, respectively.
The variable regions of the heavy and light
immunoglobulin chains together contain the antigen binding domain of the antibody. Because of the need for diversity in this region of the antibody to permit binding to a wide range of antigens, the DNA encoding the initial or primary
repertoire variable region comprises a number of different DNA segments derived from families of specific variable region gene segments. In the case of the light chain variable region, such families comprise variable (V) gene segments and joining (J) gene segments. Thus, the initial variable region of the light chain is encoded by one V gene segment and one J gene segment each selected from the family of V and J gene segments contained in the genomic DNA of the organism. In the case of the heavy chain variable region, the DNA encoding the initial or primary repertoire variable region of the heavy chain comprises one heavy chain V gene segment, one heavy chain diversity (D) gene segment and one J gene segment, each selected from the appropriate V, D and J families of
immunoglobulin gene segments in genomic DNA.
In order to increase the diversity of sequences that contribute to forming antibody binding sites, it is preferable that a heavy chain transgene include cis-acting sequences that support functional V-D-J rearrangement that can incorporate all or part of a D region gene sequence in a rearranged V-D-J gene sequence. Typically, at least about 1 percent of
expressed transgene-encoded heavy chains (or mRNAsJ include recognizable D region sequences in the V region. Preferably, at least about 10 percent of transgene-encoded V regions include recognizable D region sequences, more preferably at least about 30 percent, and most preferably more than 50 percent include recognizable D region sequences. A recognizable D region sequence is generally at least about eight consecutive nucleotides corresponding to a sequence present in a D region gene segment of a heavy chain transgene and/or the amino acid sequence encoded by such D region nucleotide sequence. For example, if a transgene includes the D region gene DHQ52, a transgene-encoded mRNA containing the sequence 5'-TAACTGGG-3' located in the V region between a V gene segment sequence and a J gene segment
sequence is recognizable as containing a D region sequence, specifically a DHQ52 sequence. Similarly, for example, if a transgene includes the D region gene DHQ52, a transgeneencoded heavy chain polypeptide containing the amino acid sequence -DAF- located in the V region between a V gene segment amino acid sequence and a J gene segment amino acid sequence is recognizable as containing a D region sequence, specifically a DHQ52 sequence.
However, because of somatic mutation and N-region addition, some D region sequences may be recognizable but may not correspond identically to a consecutive D region sequence in the transgene. For example, a nucleotide sequence 5'-
CTAAXTGGGG-3', where X is A, T, or G, and which is located in a heavy chain V region and flanked by a V region gene sequence and a J region gene sequence, can be recognized as
corresponding to the DHQ52 sequence 5'-CTAACTGGG-3'.
Similarly, for example, the polypeptide sequences -DAFDI-, -DYFDY-, or -GAFDI- located in a V region and flanked on the amino-terminal side by an amino acid sequence encoded by a transgene V gene sequence and flanked on the carboxyterminal side by an amino acid sequence encoded by a transgene J gene sequence is recognizable as a D region sequence.
Therefore, because somatic mutation and N-region addition can produce mutations in sequences derived from a transgene D region, the following definition is provided as a guide for determining the presence of a recognizable D region sequence. An amino acid sequence or nucleotide sequence is recognizable as a D region sequence if: (1) the sequence is located in a V region and is flanked on one side by a V gene sequence (nucleotide sequence or deduced amino acid sequence) and on the other side by a J gene sequence (nucleotide sequence or deduced amino acid sequence) and (2) the sequence is substantially identical or substantially similar to a known D gene sequence (nucleotide sequence or encoded amino acid sequence).
The term "substantial identity" as used herein denotes a characteristic of a polypeptide sequence or nucleic acid sequence, wherein the polypeptide sequence has at least 50 percent sequence identity compared to a reference sequence, and the nucleic acid sequence has at least 70 percent sequence identity compared to a reference sequence. The percentage of sequence identity is calculated excluding small deletions or additions which total less than 35 percent of the reference sequence. The reference sequence may be a subset of a larger sequence, such as an entire D gene; however, the reference sequence is at least 8 nucleotides long in the case of
polynucleotides, and at least 3 amino residues long in the case of a polypeptide. Typically, the reference sequence is at least 8 to 12 nucleotides or at least 3 to 4 amino acids, and preferably the reference sequence is 12 to 15 nucleotides or more, or at least 5 amino acids.
The term "substantial similarity" denotes a characteristic of an polypeptide sequence, wherein the
polypeptide sequence has at least 80 percent similarity to a reference sequence. The percentage of sequence similarity is calculated by scoring identical amino acids or positional conservative amino acid substitutions as similar. A
positional conservative amino acid substitution is one that can result from a single nucleotide substitution; a first amino acid is replaced by a second amino acid where a codon for the first amino acid and a codon for the second amino acid can differ by a single nucleotide substitution. Thus, for example, the sequence -Lys-Glu-Arg-Val- is substantially similar to the sequence -Asn-Asp-Ser-Val-, since the codon sequence -AAA-GAA-AGA-GUU- can be mutated to -AAC-GAC-AGC-GUU- by introducing only 3 substitution mutations, single
nucleotide substitutions in three of the four original codons. The reference sequence may be a subset of a larger sequence. such as an entire D gene; however, the reference sequence is at least 4 amino residues long. Typically, the reference sequence is at least 5 amino acids, and preferably the
reference sequence is 6 amino acids or more.
The Primary Repertoire
The process for generating DNA encoding the heavy and light chain immunoglobulin genes occurs primarily in developing B-cells. Prior to the joining of various
immunoglobulin gene segments, the V, D, J and constant (C) gene segments are found, for the most part, in clusters of V, D, J and C gene segments in the precursors of primary
repertoire B-cells. Generally, all of the gene segments for a heavy or light chain are located in relatively close proximity on a single chromosome. Such genomic DNA prior to
recombination of the various immunoglobulin gene segments is referred to herein as "unrearranged" genomic DNA. During B-cell differentiation, one of each of the appropriate family members of the V, D, J (or only V and J in the case of light chain genes) gene segments are recombined to form functionally rearranged heavy and light immunoglobulin genes. Such
functional rearrangement is of the variable region segments to form DNA encoding a functional variable region. This gene segment rearrangement process appears to be sequential.
First, heavy chain D-to-J joints are made, followed by heavy chain V-to-DJ joints and light chain V-to-J joints. The DNA encoding this initial form of a functional variable region in a light and/or heavy chain is referred to as "functionally rearranged DNA" or "rearranged DNA". In the case of the heavy chain, such DNA. is referred to as "rearranged heavy chain DNA" and in the case of the light chain, such DNA is referred to as "rearranged light chain DNA". Similar language is used to describe the functional rearrangement of the transgenes of the invention.
The recombination of variable region gene segments to form functional heavy and light chain variable regions is mediated by recombination signal sequences (RSS's) that flank recombinationally competent V, D and J segments. RSS's necessary and sufficient to direct recombination, comprise a dyad-symmetric heptamer, an AT-rich nonamer and an intervening spacer region of either 12 or 23 base pairs. These signals are conserved among the different loci and species that carry out D-J (or V-J) recombination and are functionally
interchangeable. See Oettinger, et al. (1990), Science, 248, 1517-1523 and references cited therein. The heptamer
comprises the sequence CACAGTG or its analogue followed by a spacer of unconserved sequence and then a nonamer having the sequence ACAAAAACC or its analogue. These sequences are found on the J, or downstream side, of each V and D gene segment. Immediately preceding the germline D and J segments are again two recombination signal sequences, first the nonamer and then the heptamer again separated by an unconserved sequence. The heptameric and nonameric sequences following a VL, VH or D segment are complementary to those preceding the JL, D or JH segments with which they recombine. The spacers between the heptameric and nonameric sequences are either 12 base pairs long or between 22 and 24 base pairs long.
In addition to the rearrangement of V, D and J segments, further diversity is generated in the primary repertoire of immunoglobulin heavy and light chain by way of variable recombination between the V and J segments in the light chain and between the D and J segments of the heavy chain. Such variable recombination is generated by variation in the exact place at which such segments are joined. Such variation in the light chain typically occurs within the last codon of the V gene segment and the first codon of the J segment. Similar imprecision in joining occurs on the heavy chain chromosome between the D and JH segments and may extend over as many as 10 nucleotides. Furthermore, several
nucleotides may be inserted between the D and JH and between the VH and D gene segments which are not encoded by genomic DNA. The addition of these nucleotides is known as N-region diversity.
After VJ and/or VDJ rearrangement, transcription of the rearranged variable region and one or more constant region gene segments located downstream from the rearranged variable region produces a primary RNA transcript which upon
appropriate RNA splicing results in an mRNA which encodes a full length heavy or light immunoglobulin chain. Such heavy and light chains include a leader signal sequence to effect secretion through and/or insertion of the immunoglobulin into the transmembrane region of the B-cell. The DNA encoding this signal sequence is contained within the first exon of the V segment used to form the variable region of the heavy or light immunoglobulin chain. Appropriate regulatory sequences are also present in the mRNA to control translation of the mRNA to produce the encoded heavy and light immunoglobulin
polypeptides which upon proper association with each other form an antibody molecule.
The net effect of such rearrangements in the
variable region gene segments and the variable recombination which may occur during such joining, is the production of a primary antibody repertoire. Generally, each B-cell which has differentiated to this stage, produces a single primary repertoire antibody. During this differentiation process, cellular events occur which suppress the functional
rearrangement of gene segments other than those contained within the functionally rearranged Ig gene. The process by which diploid B-cells maintain such mono-specificity is termed allelic exclusion.
The Secondary Repertoire
B-cell clones expressing immunoglobulins from within the set of sequences comprising the primary repertoire are immediately available to respond to foreign antigens. Because of the limited diversity generated by simple VJ and VDJ joining, the antibodies produced by the so-called primary response are of relatively low affinity. Two different types of B-cells make up this initial response: precursors of primary antibody-forming cells and precursors of secondary repertoire B-cells (Linton et al., Cell 59:1049-1059 (1989)). The first type of B-cell matures into IgM-secreting plasma cells in response to certain antigens. The other B-cells respond to initial exposure to antigen by entering a T-cell dependent maturation pathway.
During the T-cell dependent maturation of antigen stimulated B-cell clones, the structure of the antibody molecule on the cell surface changes in two ways: the constant region switches to a non-IgM subtype and the sequence of the variable region can be modified by multiple single amino acid substitutions to produce a higher affinity antibody molecule.
As previously indicated, each variable region of a heavy or light Ig chain contains an antigen binding domain. It has been determined by amino acid and nucleic acid
sequencing that somatic mutation during the secondary response occurs throughout the V region including the three
complementary determining regions (CDRl, CDR2 and CDR3) also referred to as hypervariable regions 1, 2 and 3 (Kabat et al. Sequences of Proteins of Immunological Interest (1991) U.S. Department of Health and Human Services, Washington, DC, incorporated herein by reference. The CDR1 and CDR2 are located within the variable gene segment whereas the CDR3 is largely the result of recombination between V and J gene segments or V, D and J gene segments. Those portions of the variable region which do not consist of CDR1, 2 or 3 are commonly referred to as framework regions designated FR1, FR2, FR3 and FR4. See Fig. 1. During hypermutation, the
rearranged DNA is mutated to give rise to new clones with altered Ig molecules. Those clones with higher affinities for the foreign antigen are selectively expanded by helper
T-cells, giving rise to affinity maturation of the expressed antibody. Clonal selection typically results in expression of clones containing new mutation within the CDR1, 2 and/or 3 regions. However, mutations outside these regions also occur which influence the specificity and affinity of the antiggn binding domain. Transgenic Non-Human Animals Capable
of Producing Heterologous Antibody
Transgenic non-human animals in one aspect of the invention are produced by introducing at least one of the immunoglobulin transgenes of the invention (discussed
hereinafter) into a zygote or early embryo of a non-human animal. The non-human animals which are used in the invention generally comprise any mammal which is capable of rearranging immunoglobulin gene segments to produce a primary antibody response. Such nonhuman transgenic animals may include, for example, transgenic pigs, transgenic rats, transgenic rabbits, transgenic cattle, and other transgenic animal species, particularly mammalian species, known in the art. A
particularly preferred non-human animal is the mouse or other members of the rodent family.
However, the invention is not limited to the use of mice. Rather, any non-human mammal which is capable of mounting a primary and secondary antibody response may be used. Such animals include non-human primates, such as chimpanzee, bovine, ovine, and porcine species, other members of the rodent family, e.g. rat, as well as rabbit and guinea pig. Particular preferred animals are mouse, rat, rabbit and guinea pig, most preferably mouse.
In one embodiment of the invention, various gene segments from the human genome are used in heavy and light chain transgenes in an unrearranged form. In this embodiment, such transgenes are introduced into mice. The unrearranged gene segments of the light and/or heavy chain transgene have DNA sequences unique to the human species which are
distinguishable from the endogenous immunoglobulin gene segments in the mouse genome. They may be readily detected in unrearranged form in the germ line and somatic cells not consisting of B-cells and in rearranged form in B-cells.
In an alternate embodiment of the invention, the transgenes comprise rearranged heavy and/or light
immunoglobulin transgenes. Specific segments of such
transgenes corresponding to functionally rearranged VDJ or VJ segments, contain immunoglobulin DNA sequences which are also clearly distinguishable from the endogenous immunoglobulin gene segments in the mouse.
Such differences in DNA sequence are also reflected in the amino acid sequence encoded by such human
immunoglobulin transgenes as compared to those encoded by mouse B-cells. Thus, human immunoglobulin amino acid
sequences may be detected in the transgenic non-human animals of the invention with antibodies specific for immunoglobulin epitopes encoded by human immunoglobulin gene segments.
Transgenic B-cells containing unrearranged transgenes from human or other species functionally recombine the appropriate gene segments to form functionally rearranged light and heavy chain variable regions. It will be readily apparent that the antibody encoded by such rearranged
transgenes has a DNA and/or amino acid sequence which iε heterologous to that normally encountered in the nonhuman animal used to practice the indention.
Unrearranged Transgenes
As used herein, an "unrearranged immunoglobulin heavy chain transgene" comprises DNA encoding at least one variable gene segment, one diversity gene segment, one joining gene segment and one constant region gene segment. Each of the gene segments of said heavy chain transgene are derived from, or has a sequence corresponding to, DNA encoding
immunoglobulin heavy chain gene segments from a species not consisting of the non-human animal into which said transgene is introduced. Similarly, as used herein, an "unrearranged immunoglobulin light chain transgene" comprises DNA encoding at least one variable gene segment, one joining gene segment and at least one constant region gene segment wherein each gene segment of said light chain tr; nsgene is derived from, or has a sequence corresponding to, DNA encoding immunoglobulin light chain gene segments from a species not consisting of the non-human animal into which said light chain transgene is introduced.
Such heavy and light chain transgenes in this aspect of the invention contain the above-identified gene segments in an unrearranged form. Thus, interposed between the V, D and J segments in the heavy chain transgene and between the V and J segments on the light chain transgene are appropriate
recombination signal sequences (RSS's). In addition, such transgenes also include appropriate RNA splicing signals to join a constant region gene segment with the VJ or VDJ
rearranged variable region.
In order to facilitate isotype switching within a heavy chain transgene containing more than one C region gene segment, e.g. Cμ and Cγ1 from the human genome, as explained below "switch regions" are incorporated upstream from each of the constant region gene segments and downstream from the variable region gene segments to permit recombination between such constant regions to allow for immunoglobulin class switching, e.g. from IgM to IgG. Such heavy and light
immunoglobulin transgenes also contain transcription control sequences including promoter regions situated upstream from the variable region gene segments which typically contain TATA motifs. A promoter region can be defined approximately as a DNA sequence that, when operably linked to a downstream sequence, can produce transcription of the downstream
sequence. Promoters may require the presence of additional linked cis-acting sequences in order to produce efficient transcription. In addition, other sequences that participate in the transcription of sterile transcripts are preferably included. Examples of sequences that participate in
expression of sterile transcripts can be found in the
published literature, including Rothman et al., Intl. Immunol. 2:621-627 (1990); Reid et al., Proc. Natl. Acad. Sci. USA 86-840-844 (1989); Stavnezer et al., Proc. Natl. Acad. Sci. USA 85:7704-7708 (1988); and Mills et al., Nucl. Acids Res. 18:7305-7316 (1991), each of which is incorporated herein by reference. These sequences typically include about at least 50 bp immediately upstream of a switch region, preferably about at least 200 bp upstream of a switch region; and more preferably about at least 200-1000 bp or more upstream of a switch region. Suitable sequences occur immediately upstream of the human Sγ1, Sγ2, Sγ3, Sγ4, Sα1, Sα2, and Sε switch regions, although the sequences immediately upstream of the human Sγ1, and Sγ3 switch regions are preferable. In
particular, interferon (IFN) inducible transcriptional regulatory elements, such as IFN-inducible enhancers, are preferably included immediately upstream of transgene switch sequences.
In addition to promoters, other regulatory sequences which function primarily in B-lineage cells are used. Thus, for example, a light chain enhancer sequence situated
preferably between the J and constant region gene segments on the light chain transgene is used to enhance transgene
expression, thereby facilitating allelic exclusion. In the case of the heavy chain transgene, regulatory enhancers and also employed. Such regulatory sequences are used to maximize the transcription and translation of the transgene so as to induce allelic exclusion and to provide relatively high levels of transgene expression.
Although the foregoing promoter and enhancer
regulatory control sequences have been generically described, such regulatory sequences may be heterologous to the nonhuman animal being derived from the genomic DNA from which the heterologous transgene immunoglobulin gene segments are obtained. Alternately, such regulatory gene segments are derived from the corresponding regulatory sequences in the genome of the non-human animal, or closely related species, which contains the heavy and light transgene.
In the preferred embodiments, gene segments are derived from human beings. The transgenic non-human animals harboring such heavy and light transgenes are capable of mounting an Ig-mediated immune response to a specific antigen administered to such an animal. B-cells are produced within such an animal which are capable of producing heterologous human antibody. After immortalization, and the selection for an appropriate monoclonal antibody (Mab), e.g. a hybridoma, a source of therapeutic human monoclonal antibody is provided. Such human Mabs have significantly reduced immunogenicity when therapeutically administered to humans. Although the preferred embodiments disclose the construction of heavy and light transgenes containing human gene segments, the invention is not so limited. In this regard, it is to be understood that the teachings described herein may be readily adapted to utilize immunoglobulin gene segments from a species other than human beings. For example, in addition to the therapeutic treatment of humans with the antibodies of the invention, therapeutic antibodies encoded by appropriate gene segments may be utilized to generate
monoclonal antibodies for use in the veterinary sciences.
Rearranged Transgenes
In an alternative embodiment, transgenic nonhuman animals contain functionally at least one rearranged
heterologous heavy chain immunoglobulin transgene in the germline of the transgenic animal. Such animals contain primary repertoire B-cells that express such rearranged heavy transgenes. Such B-cells preferably are capable of undergoing somatic mutation when contacted with an antigen to form a heterologous antibody having high affinity and specificity for the antigen. Said rearranged transgenes will contain at least two CH genes and the associated sequences required for isotype switching.
The invention also includes transgenic animals containing germ line cells having heavy and light transgenes wherein one of the said transgenes contains rearranged gene segments with the other containing unrearranged gene segments. In such animals, the heavy chain transgenes shall have at least two CH genes and the associated sequences required for isotype switching.
The invention further includes methods for generating a synthetic variable region gene segment repertoire to be used in the transgenes of the invention. The method comprises generating a population of immunoglobulin V segment DNAs wherein each of the V segment DNAs encodes an
immunoglobulin V segment and contains at each end a cleavage recognition site of a restriction endonuclease. The
population of immunoglobulin V segment DNAs is thereafter concatenated to form the synthetic immunoglobulin V segment repertoire. Such synthetic variable region heavy chain transgenes shall have at least two CH genes and the associated sequences required for isotype switching.
Isotype Switching
In the development of a B lymphocyte, the cell initially produces IgM with a binding specificity determined by the productively rearranged VH and VL regions.
Subsequently, each B cell and its progeny cells synthesize antibodies with the same L and H chain V regions, but they may switch the isotype of the H chain.
The use of μ or δ constant regions is largely determined by alternate splicing, permitting IgM and IgD to be coexpressed in a single cell. The other heavy chain isotypes (γ, α, and ε) are only expressed natively after a gene
rearrangement event deletes the Cμ and Cδ exons. This gene rearrangement process, termed isotype switching, typically occurs by recombination between so called switch segments located immediately upstream of each heavy chain gene (except S) . The individual switch segments are between 2 and 10 kb in length, and consist primarily of short repeated sequences.
The exact point of recombination differs for individual class switching events. Investigations which have used solution hybridization kinetics or Southern blotting with cDNA-derived CH probes have confirmed that switching can be associated with loss of CH sequences from the cell.
The switch (S) region of the μ gene, Sμ, is located about 1 to 2 kb 5' to the coding sequence and is composed of numerous tandem repeats of sequences of the form
(GAGCT)n(GGGGT), where n is usually 2 to 5 but can range as high as 17. (See T. Nikaido et al. Nature 292:845-848 (1981))
Similar internally repetitive switch sequences spanning several kilobases have been found 5' of the other CH genes. The Sα region has been sequenced and found to consist of tandemly repeated 80-bp homology units, whereas Sγ2a, Sγ2b, and Sγ3 all contain repeated 49-bp homology units very similar to each other. (See. P. Szurek et al., J. Immunol 135:620-626 (1985) and T. Nikaido et al., J. Biol. Chem. 257:7322-7329 (1982), which are incorporated herein by reference.) All the sequenced S regions include numerous occurrences of the pentamers GAGCT and GGGGT that are the basic repeated elements of the S gene (T. Nikaido et al., J. Biol. Chem. 257:7322- 7329 (1982) which is incorporated herein by reference); in the other S regions these pentamers are not precisely tandemly repeated as in Sμ, but instead are embedded in larger repeat units. The Sγ1 region has an additional higher-order
structure: two direct repeat sequences flank each of two clusters of 49-bp tandem repeats. (See M. R. Mowatt et al., J. Immunol. 136:2674-2683 (1986), which is incorporated herein by reference).
Switch regions of human H chain genes have been found to be very similar to their mouse homologs. Indeed, similarity between pairs of human and mouse clones 5' to the CH genes has been found to be confined to the S regions, a fact that confirms the biological significance of these regions.
A switch recombination between μ and a genes produces a composite Sμ-Sα sequence. Typically, there is no specific site, either in S„ or in any other S region, where the recombination always occurs.
Generally, unlike the enzymatic machinery of V-J recombination, the switch machinery can apparently accommodate different alignments of the repeated homologous regions of germline S precursors and then join the sequences at different positions within the alignment. (See. T. H. Rabbits et al., Nucleic Acids Res. 9:4509-4524 (1981) and J. Ravetch et al., Proc. Natl. Acad. Sci. USA 77:6734-6738 (1980), which are incorporated herein by reference.)
The exact details of the mechanism(s) of selective activation of switching to a particular isotype are unknown. Although exogenous influences such as lymphokines and
cytokines might upregulate isotype-specific recombinases, it is also possible that the same enzymatic machinery catalyzes switches to all isotypes and that specificity lies in
targeting this machinery to specific switch regions. The T-cell-derived lymphokines IL-4 and IFNγ have been shown to specifically promote the expression of certain isotypes: IL-4 decreases IgM, IgG2a, IgG2b, and IgG3
expression and increases IgE and IgG1 expression; while IFNγ selectively stimulates IgG2a expression and antagonizes the IL-4-induced increase in IgE and IgG1 expression (Coffman et al., J. Immunol. 136:949-954 (1986) and Snapper et al.,
Science 236:944-947 (1987), which are incorporated herein by reference). A combination of IL-4 and IL-5 promotes IgA expression (Coffman et al., J. Immunol. 139:3685-3690 (1987), which is incorporated herein by reference).
Most of the experiments implicating T-cell effects on switching have not ruled out the possibility that the observed increase in cells with particular switch
recombinations might reflect selection of preswitched or precommitted cells; but the most likely explanation is that the lymphokines actually promote switch recombination.
Induction of class switching appears to be associated with sterile transcripts that initiate upstream of the switch segments (Lutzker et al., Mol. Cell. Biol. 8: 1849 (1988); Stavnezer et al., Proc. Natl. Acad. Sci. USA 85:7704 (1988); Esser and Radbruch, EMBO J. 8:483 (1989); Berton et al., Proc. Natl. Acad. Sci. USA 86:2829 (1989); Rothman et al., Int. Immunol. 2 : 621 (1990), each of which is incorporated by reference). For example, the observed induction of the γI sterile transcript by IL-4 and inhibition by IFN-γ correlates with the observation that IL-4 promotes class switching to γ1 in B-cells in culture, while IFN-γ inhibits γ1 expression. Therefore, the inclusion of regulatory sequences that affect the transcription of sterile transcripts may also affect the rate of isotype switching. For example, increasing the transcription of a particular sterile transcript typically can be expected to enhance the frequency of isotype switch
recombination involving adjacent switch sequences.
For these reasons, it is preferable that transgenes incorporate transcriptional regulatory sequences within about 1-2 kb upstream of each switch region that is to be utilized for isotype switching. These transcriptional regulatory sequences preferably include a promoter and an enhancer element, and more preferably include the 5' flanking (i.e., upstream) region that is naturally associated (i.e., occurs in germline configuration) with a switch region. This 5'
flanking region is typically about at least 50 nucleotides in length, preferably about at least 200 nucleotides in length, and more preferably at least 500-1000 nucleotides.
Although a 5' flanking sequence from one switch region can be operably linked to a different switch region for transgene construction (e.g., the 5' flanking sequence from the human Sγ1 switch can be grafted immediately upstream of the Sα1 switch), in some embodiments it is preferred that each switch region incorporated in the transgene construct have the 5' flanking region that occurs immediately upstream in the naturally occurring germline configuration.
The Transgenic Primary Repertoire
A. The Human Immunoglobulin Loci
An important requirement for transgene function is the generation of a primary antibody repertoire that is diverse enough to trigger a secondary immune response for a wide range of antigens. The rearranged heavy chain gene
consists of a signal peptide exon, a variable region exon and a tandem array of multi-domain constant region regions, each of which is encoded by several exons. Each of the constant region genes encode the constant portion of a different class of immunoglobulins. During B-cell development, V region proximal constant regions are deleted leading to the
expression of new heavy chain classes. For each heavy chain . class, alternative patterns of RNA splicing give rise to both transmembrane and secreted immunoglobulins.
The human heavy chain locus consists of
approximately 200 V gene segments spanning 2 Mb, approximately 30 D gene segments spanning about 40 kb, six J segments
clustered within a 3 kb span, and nine constant region gene segments spread out over approximately 300 kb. The entire locus spans approximately 2.5 Mb of the distal portion of the long arm of chromosome 14. B. Gene Fragment Transgenes
1. Heavy Chain Transgene
In a preferred embodiment, immunoglobulin heavy and light chain transgenes comprise unrearranged genomic DNA from humans. In the case of the heavy chain, a preferred transgene comprises a NotI fragment having a length between 670 to 830 kb. The length of this fragment is ambiguous because the 3' restriction site has not been accurately mapped. It is known, however, to reside between the αl and
Figure imgf000040_0001
gene segments. This fragment contains members of all six of the known VH families, the D and J gene segments, as well as the μ, δ, γ3 , γ1 and α1 constant regions (Berman et al., EMBO J. 7:727-738 (1988), which is incorporated herein by reference). A transgenic mouse line containing this transgene correctly expresses a heavy chain class required for B-cell development (IgM) and at least one switched heavy chain class (IgG1), in conjunction with a sufficiently large repertoire of variable regions to trigger a secondary response for most antigens. 2. Light Chain Transgene
A genomic fragment containing all of the necessary gene segments and regulatory sequences from a human light chain locus may be similarly constructed. Such transgenes are constructed as described in the Examples.
C. Transgenes Generated Intracellularly
by In Vivo Recombination
It is not necessary to isolate the all or part of the heavy chain locus on a single DNA fragment. Thus, for example, the 670-830 kb NotI fragment from the human
immunoglobulin heavy chain locus may be formed in vivo in the non-human animal during transgenesis. Such in vivo transgene construction is produced by introducing two or more
overlapping DNA fragments into an embryonic nucleus of the non-human animal. The overlapping portions of the DNA
fragments have DNA sequences which are substantially
homologous. Upon exposure to the recombinases contained within the embryonic nucleus, the overlapping DNA fragments homologously recombined in proper orientation to form the 670-830 kb NotI heavy chain fragment.
In vivo transgene construction can be used to form any number of immunoglobulin transgenes which because of their size are otherwise difficult, or impossible, to make or manipulate by present technology. Thus, in vivo transgene construction is useful to generate immunoglobulin transgenes which are larger than DNA fragments which may be manipulated by YAC vectors (Murray and Szostak, Nature 305: 189-193
(1983)). Such in vivo transgene construction may be used to introduce into a non-human animal substantially the entire immunoglobulin loci from a species not consisting of the transgenic non-human animal.
In addition to forming genomic immunoglobulin transgenes, in vivo homologous recombination may also be utilized to form "mini-locus" transgenes as described in the Examples.
In the preferred embodiments utilizing in vivo
transgene construction, each overlapping DNA fragment
preferably has an overlapping substantially homologous DNA sequence between the end portion of one DNA fragment and the end portion of a second DNA fragment. Such overlapping
portions of the DNA fragments preferably comprise about 500 bp to about 2000 bp, most preferably 1.0 kb to 2.0 kb.
Homologous recombination of overlapping DNA fragments to form transgenes in vivo is further described in commonly assigned PCT Publication No. WO 92/03917 entitled "Homologous
Recombination in Mammalian Cells" published March 19, 1992. D. Minilocus Transgenes
As used herein, the term "immunoglobulin minilocus" refers to a DNA sequence (which may be within a longer
sequence), usually of less than about 150 kb, typically between about 25 and 100 kb, containing at least one each of the following: a functional variable (V) gene segment, a functional joining (J) region segment, at least one functional constant (C) region gene segment, and--if it is a heavy chain minilocus--a functional diversity (D) region segment, such that said DNA sequence contains at least one substantial discontinuity (e.g., a deletion, usually of at least about 2 to 5 kb, preferably 10-25 kb or more, relative to the
homologous genomic DNA sequence). A light chain minilocus transgene will be at least 25 kb in length, typically 50 to 60 kb. A heavy chain transgene will typically be about 70 to 80 kb in length, preferably at least about 60 kb with two
constant regions operably linked to switch regions.
Furthermore, the individual elements of the minilocus are preferably in the germline configuration and capable of undergoing gene rearrangement in the pre-B cell of a
transgenic animal so as to express functional antibody
molecules with diverse antigen specificities encoded entirely by the elements of the minilocus. Further, a heavy chain minilocus comprising at least two CH genes and the requisite switching sequences is typ.cally c pable of undergoing isotype switching, so that functional antibody molecules of different immunoglobulin classes will be generated. Such isotype
switching may occur in vivo in B-cells residing within the transgenic nonhuman animal, or may occur in cultured cells of the B-cell lineage which have been explanted from the
transgenic nonhuman animal.
In an alternate preferred embodiment, immunoglobulin heavy chain transgenes comprise one or more of each of the VH, D, and JH gene segments and two or more of the CH genes. At least one of each appropriate type gene segment is
incorporated into the minilocus transgene. With regard to the CH segments for the heavy chain transgene, it is preferred that the transgene contain at least one μ gene segment and at least one other constant region gene segment, more preferably a γ gene segment, and most preferably γ3 or γ1. This
preference is to allow for class switching between IgM and IgG forms of the encoded immunoglobulin and the production of a secretable form of high affinity non-IgM immunuglobulin.
Other constant region gene segments may also be used such as those which encode for the production of IgD, IgA and IgE.
Those skilled in the art will also construct transgenes wherein the order of occurrence of heavy chain CH genes will be different from the naturally-occurring spatial order found in the germline of the species serving as the donor of the CH genes.
Additionally, those skilled in the art can select CH genes from more than one individual of a species (e.g., allogeneic CH genes) and incorporate said genes in the
transgene as supernumerary CH genes capable of undergoing isotype switching; the resultant transgenic nonhuman animal may then, in some embodiments, make antibodies of various classes including all of the allotypes represented in the species from which the transgene CH genes were obtained.
Still further, those skilled in the art can select CH genes from different species to incorporate into the transgene. Functional switch sequences are included with each CH gene, although the switch sequences used are not
necessarily those which occur naturally adjacent to the CH gene. Interspecies CH gene combinations will produce a transgenic nonhuman animal which may produce antibodies of various classes corresponding to CH genes from various
species. Transgenic nonhuman animals containing interspecies CH transgenes may serve as the source of B-cells for
constructing hybridomas to produce monoclonals for veterinary uses.
The heavy chain J region segments in the human comprise six functional J segments and three pseudo genes clustered in a 3 kb stretch of DNA. Given its relatively compact size and the ability to isolate these segments
together with the μ gene and the 5' portion of the δ gene on a single 23 kb SFil/Spel fragment (Sado et al., Biochem.
Biophys. Res. Comm. 154:264271 (1988), which is incorporated herein by reference), it is preferred that all of the J region gene segments be used in the mini-locus construct. Since this fragment spans the region between the μ and δ genes, it is likely to contain all of the 3' cis-linked regulatory elements required for μ expression. Furthermore, because this fragment includes the entire J region, it contains the heavy chain enhancer and the μ switch region (Mills et al., Nature 306:809 (1983); Yancopoulos and Alt, Ann. Rev. Immunol. 4:339-368 (1986), which are incorporated herein by reference). It also contains the transcription start sites which trigger VDJ joining to form primary repertoire B-cells (Yancopoulos and Alt, Cell 40:271-281 (1985), which is incorporated herein by reference). Alternatively, a 36 kb BssHII/Spell fragment, which includes part on the D region, may be used in place of the 23 kb Sfil/Spell fragment. The use of such a fragment increases the amount of 5' flanking sequence to facilitate efficient D-to-J joining.
The human D region consists of 4 or 5 homologous 9 kb subregions, linked in tandem (Siebenlist, et al. (1981), Nature, 294, 631-635). Each subregion contains up to 10 individual D segments. Some of these segments have been mapped- and are shown in Fig. 4. Two different strategies are used to generate a mini-locus D region. The first strategy involves using only those D segments located in a short contiguous stretch of DNA that includes one or two of the repeated D subregions. A candidate is a single 15 kb fragment that contains 12 individual D segments. This piece of DNA consists of 2 contiguous EcoRI fragments and has been
completely sequenced (Ichihara, et al. (1988), EMBO J., 7, 4141-4150). Twelve D segments should be sufficient for a primary repertoire. However, given the dispersed nature of the D region, an alternative strategy is to ligate together several non-contiguous D-segment containing fragments, to produce a smaller piece of DNA with a greater number of segments. Additional D-segment genes can be identified, for example, by the presence of characteristic flanking nonamer and heptamer sequences, supra, and by reference to the
literature.
At least one, and preferably more than one V gene segment is used to construct the heavy chain minilocus
transgene. Rearranged or unrearranged V segments with or without flanking sequences can be isolated as described PCT Publication No. WO 92/03918, published March 19, 1992,
entitled "Transgenic Non-Human Animals Capable of Producing Heterologous Antibodies." Rearranged or unrearranged V segments, D segments, J segments, and C genes, with or without flanking sequences, can be isolated as described in PCT Publication No. WO 92/03918, published March 19, 1992.
A minilocus light chain transgene may be similarly constructed from the human λ or κ immunoglobulin locus.
Thus, for example, an immunoglobulin heavy chain minilocus transgene construct, e.g., of about 75 kb, encoding V, D, J and constant region sequences can be formed from a plurality of DNA fragments, with each sequence being substantially homologous to human gene sequences. Preferably, the sequences are operably linked to transcription regulatory sequences and are capable of undergoing rearrangement. With two or more appropriately placed constant region sequences (e.g., μ and γ) and switch regions, switch recombination also occurs. An exemplary light chain transgene construct can be formed similarly from a plurality of DNA fragments, substantially homologous to human DNA and capable of undergoing
rearrangement.
E. Transgene Constructs Capable of Isotype Switching
Ideally, transgene constructs that are intended to undergo class switching should include all of the cis-acting sequences necessary to regulate sterile transcripts.
Naturally occurring switch regions and upstream promoters and regulatory sequences (e.g., IFN-inducible elements) are preferred cis-acting sequences that are included in transgene constructs capable of isotype switching. About at least 50 basepairs, preferably about at least 200 basepairs, and more preferably at least 500 to 1000 basepairs or more of sequence immediately upstream of a switch region, preferably a human γ1 switch region, should be operably linked to a switch sequence, preferably a human γ1 switch sequence. Further, switch regions can be linked upstream of (and adjacent to) CH genes that do not naturally occur next to the particular switch region. For example, but not for limitation, a human γ1 switch region may be linked upstream from a human α2 CH gene, or a murine γ1 switch may be linked to a human CH gene. An alternative method for obtaining ήon-classical isotype switching (e.g., (S-associated deletion) in transgenic mice involves the inclusion of the 400 bp direct repeat sequences (σμ and eμ) that flank the human μ gene (Yasui et al., Eur. J. Immunol. 19:1399 (1989)). Homologous
recombination between these two sequences deletes the μ gene in IgD-only B-cells. Heavy chain transgenes can be
represented by the following formulaic description: (VH)x-(D)y-(JH)z-(SD)m-(C1)n-[(T)-(SA)p-(C2)]q where:
VH is a heavy chain variable region gene segment,
D is a heavy chain D (diversity) region gene segment, JH is a heavy chain J (joining) region gene segment,
SD is a donor region segment capable of participating in a recombination event with the Sa acceptor region
segments such that isotype switching occurs,
C1 is a heavy chain constant region gene segment encoding an isotype utilized in for B cell development (e.g., μ or δ) ,
T is a cis-acting transcriptional regulatory region
segment containing at least a promoter,
SA is an acceptor region segment capable of participating in a recombination event with selected SD donor region segments, such that isotype switching occurs, C2 is a heavy chain constant region gene segment encoding an isotype other than μ (e.g., γ1, γ2, γ3, γ4, α1, α2, ε).
x, y, z, m, n, p, and q are integers, x is 1-100, n is 0-10, y is 1-50, p is 1-10, z is 1-50, q is 0-50, m is 0-10. Typically, when he transgene is capable of isotype switching, q must be at least 1, m is at least 1, n is at least 1, and m is greater than or equal to n. VH, D, JH, SD, C1, T, SA, and CZ segments may be selected from various species, preferably mammalian species, and more preferably from human and murine germline DNA.
VH segments may be selected from various species, but are preferably selected from VH segments that occur naturally in the human germline, such as VH251. Typically about 2 VH gene segments are included, preferably about 4 VH segments are included, and most preferably at least about 10 VH segments are included.
At least one D segment is typically included, although at least 10 D segments are preferably included, and some embodiments include more than ten D segments. Some preferred embodiments include human D segments.
Typically at least one JH segment is incorporated in the transgene, although it is preferable to include about six JH segments, and some preferred embodiments include more than about six JH segments. Some preferred embodiments include human JH segments, and further preferred embodiments include six human JH segments and no nonhuman JH segments.
SD segments are donor regions capable of
participating in recombination events with the SA segment of the transgene. For classical isotype switching, SD and SA are switch. regions such as Sμ, sγ1, Sγ2. Sγ3, Sγ4, Sα, Sα2, and Sε. Preferably the switch regions are murine or human, more preferably SD is a human or murine S and SA is a human or murine Sγ1. For nonclassical isotype switching (5-associated deletion) , SD and SA are preferably the 400 basepair direct repeat sequences that flank the human μ gene.
C1 segments are typically μ or δ genes, preferably a μ gene, and more preferably a human or murine μ gene.
T segments typically include S' flanking sequences that are adjacent to naturally occurring (i.e., germline) switch regions. T segments typically at least about at least 50 nucleotides in length, preferably about at least 200 nucleotides in length, and more preferably at least 500-1000 nucleotides in length. Preferably T segments are 5' flanking sequences that occur immediately upstream of human or murine switch regions in a germline configuration. It is also evident to those of skill in the art that T segments may comprise cis-acting transcriptional regulatory sequences that do not occur naturally in an animal germline (e.g., viral enhancers and promoters such as those found in SV40,
adenovirus, and other viruses that infect eukaryotic cells).
C2 segments are typically a γ1 , γ2 , γ3 , γ4, α1, α2, or ε CH gene, preferably a human CH gene of these isotypes, and more preferably a humanγ1 or γ3 gene. Murine γ2a and γ2b may also be used, as may downstream (i.e., switched) isotype genes form various species. Where the heavy chain transgene
contains an immunoglobulin heavy chain minilocus, the total length of the transgene will be typically 150 kilo basepairs or less.
In general, the transgene will be other than a native heavy chain Ig locus. Thus, for example, deletion of unnecessary regions or substitutions with corresponding regions from other species will be present.
F. Methods for Determining Functional
Isotype Switching in Ig Transgenes
The occurrence of isotype switching in a transgenic nonhuman animal may be identified by any method known to those in the art. Preferred embodiments include the following, employed either singly or in combination:
1. detection of mRNA transcripts that contain a sequence homologous to at least one transgene downstream CH gene other than δ and an adjacent sequence homologous to a transgene VH- DH-JH rearranged gene; such detection may be by Northern hybridization, S1 nuclease protection assays, PCR
amplification, cDNA cloning, or other methods;
2. detection in the serum of the transgenic animal, or in supernatants of cultures of hybridoma cells made from B-cells of the transgenic animal, of immunoglobulin proteins encoded by downstream CH genes, where such proteins can also be shown by immunochemical methods to comprise a functional variable region;
3. detection, in DNA from B-cells of the transgenic animal or in genomic DNA from hybridoma cells, of DNA rearrangements consistent with the occurrence of isotype switching in the transgene, such detection may be accomplished by Southern blot hybridization, PCR amplification, genomic cloning, or other method; or
4. identification of other indicia of isotype switching, such as production of sterile transcripts, production of characteristic enzymes involved in switching (e.g., "switch recombinase"), or other manifestations that may be detected, measured, or observed by contemporary techniques.
Because each transgenic line may represent a
different site of integration of the transgene, and a
potentially different tandem array of transgene inserts, and because each different configuration of transgene and flanking DNA sequences can affect gene expression, it is preferable to identify and use lines of mice that express high levels of human immunoglobulins, particularly of the IgG isotype, and contain the least number of copies of the transgene. Single copy transgenics minimize the potential problem of incomplete allelic expression. Transgenes are typically integrated into host chromosomal DNA, most usually into germline DNA and propagated by subsequent breeding of germline transgenic breeding stock animals. However, other vectors and transgenic methods known in the present art or subsequently developed may be substituted as appropriate and as desired by a
practitioner.
G. Functional Disruption of
Endogenous Immunoglobulin Loci The expression of successfully rearranged immunoglobulin heavy and light transgenes is expected to have a dominant effect by suppressing the rearrangement of the endogenous immunoglobulin genes in the transgenic nonhuman animal. However, another way to generate a nonhuman that is devoid of endogenous antibodies is by mutating the endogenous immunoglobulin loci. Using embryonic stem cell technology and homologous recombination, the endogenous immunoglobulin repertoire can be readily eliminated. The following describes the functional description of the mouse immunoglobulin loci. The vectors and methods disclosed, however, can be readily adapted for use in other non-human animals.
Briefly, this technology involves the inactivation of a gene, by homologous recombination, in a pluripotent cell line that is capable of differentiating into germ cell tissue. A DNA construct that contains an altered, copy of a mouse immunoglobulin gene is introduced into the nuclei of embryonic stem cells. In a portion of the cells, the introduced DNA recombines with the endogenous copy of the mouse gene,
replacing it with the altered copy. Cells containing the newly engineered genetic lesion are injected into a host mouse embryo, which is reimplanted into a recipient female. Some of these embryos develop into chimeric mice that possess germ cells entirely derived from the mutant cell line. Therefore, by breeding the chimeric mice it is possible to obtain a new line of mice containing the introduced genetic lesion
(reviewed by Capecchi (1989), Science, 244, 1288-1292).
Because the mouse λ locus contributes to only 5% of the immunoglobulins, inactivation of the heavy chain and/or κ-light chain loci is sufficient. There are three ways to disrupt each of these loci, deletion of the J region, deletion of the J-C intron enhancer, and disruption of constant region coding sequences by the introduction of a stop codon. The last option is the most straightforward, in terms of DNA construct design. Elimination of the μ gene disrupts B-cell maturation thereby preventing class switching to any of the functional heavy chain segments. The strategy for knocking out these loci is outlined below.
To disrupt the mouse μ andκ genes, targeting
vectors are used based on the design employed by Jaenisch and co-wόrkers (Zijlstra, et al. (1989), Nature, 342, 435-438) for the successful disruption of the mouse β2-microglobulin gene. The neomycin resistance gene (neo), from the plasmid pMCIneo is inserted into the coding region of the target gene. The pMCIneo insert uses a hybrid viral promoter/enhancer sequence to drive neo expression. This promoter is active in embryonic stem cells. Therefore, neo can be used as a selectable marker for integration of the knock-out construct. The HSV thymidine kinase (tk) gene is added to the end of the construct as a negative selection marker against random insertion events (Zijlstra, et al., supra.).
A preferred strategy for disrupting the heavy chain locus is the elimination of the J region. This region is fairly compact in the mouse, spanning only 1.3 kb. To
construct a gene targeting vector, a 15 kb Kpnl fragment containing all of the secreted A constant region exons from mouse genomic library is isolated. The 1.3 kb J region is replaced with the 1.1 kb insert from pMCIneo. The HSV tk gene is then added to the 5' end of the Kpnl fragment. Correct integration of this construct, via homologous recombination, will result in the replacement of the mouse JH region with the neo gene. Recombinants are screened by PCR, using a primer based on the neo gene and a primer homologous to mouse
sequences 5' of the Kpnl site in the D region.
Alternatively, the heavy-chain locus is knocked out by disrupting the coding region of the μ gene. This approach involves the same 15 kb Kpnl fragment used in the previous approach. The 1.1 kb insert from pMCIneo is inserted at a unique BamHI site in exon II, and the HSV tk gene added to the 3' Kpnl end. Double crossover events on either side of the neo insert, that eliminate the tk gene, are then selected for. These are detected from pools of selected clones by PCR amplification. One of the PCR primers is derived from neo sequences and the other from mouse sequences outside of the targeting vector. The functional disruption of the mouse immunoglobulin loci is presented in the Examples.
G. Suppressing Expression of
Endogenous Immunoglobulin Loci
In addition to functional disruption of endogenous
Ig loci, an alternative method for preventing the expression of an endogenous Ig locus is suppression. Suppression of endogenous Ig genes may be accomplished with antisense RNA produced from one or more integrated transgenes, by antisense oligonucleotides, and/or by administration of antisera
specific for one or more endogenous Ig chains. Antisense Polynucleotides
Antisense RNA transgenes can be employed to
partially or totally knock-out expression of specific genes (Pepin et al. (1991) Nature 355: 725; Helene., C. and Toulme, J. (1990) Biochimica Biophys. Acta 1049: 99; Stout, J. and Caskey, T. (1990) Somat. Cell Mol. Genet. 16: 369; Munir et al. (1990) Somat. Cell Mol. Genet. 16: 383, each of which is incorporated herein by reference).
"Antisense polynucleotides" are polynucleotides that: (1) are complementary to all or part of a reference sequence, such as a sequence of an endogenous Ig CH or CL region, and (2) which specifically hybridize to a
complementary target sequence, such as a chromosomal gene locus- or a Ig mRNA. Such complementary antisense
polynucleotides may include nucleotide substitutions,
additions, deletions, or transpositions, so long as specific hybridization to the relevant target sequence is retained as a functional property of the polynucleotide. Complementary antisense polynucleotides include soluble antisense RNA or DNA oligonucleotides which can hybridize specifically to
individual mRNA species and prevent transcription and/or RNA processing of the mRNA species and/or translation of the encoded polypeptide (Ching et al., Proc. Natl. Acad. Sci.
U.S.A. 86:10006-10010 (1989); Broder et al., Ann. Int. Med. 113: 604-618 (1990); Loreau et al., FEBS Letters 274:53-56
(1990); Holcenberg et al., WO91/11535; WO91/09865; WO91/04753; W090/13641; and EP 386563, each of which is incorporated herein by reference). An antisense sequence is a
polynucleotide sequence that is complementary to at least one immunoglobulin gene sequence of at least about 15 contiguous nucleotides in length, typically at least 20 to 30 nucleotides in length, and preferably more than about 30 nucleotides in length. However, in some embodiments, antisense sequences may have substitutions, additions, or deletions as compared to the complementary immunoglobulin gene sequence, so long as
specific hybridization is retained as a property of the antisense polynucleotide. Generally, an antisense sequence is complementary to an endogenous immunoglobulin gene sequence that encodes, or has the potential to encode after DNA
rearrangement, an immunoglobulin chain. In some cases, sense sequences corresponding to an immunoglobulin gene sequence may function to suppress expression, particularly by interfering with transcription.
The antisense polynucleotides therefore inhibit production of the encoded polypeptide (s). In this regard, antisense polynucleotides that inhibit transcription and/or translation of one or more endogenous Ig loci can alter the capacity and/or specificity of a non-human animal to produce immunoglobulin chains encoded by endogenous Ig loci.
Antisense polynucleotides may be produced from a heterologous expression cassette in a transfectant cell or transgenic cell, such as a transgenic pluripotent
hematopoietic stem cell used to reconstitute all or part of the hematopoietic stem cell population of an individual, or a transgenic nonhuman animal. Alternatively, the antisense polynucleotides may comprise soluble oligonucleotides that are administered to the external milieu, either in culture medium in vitro or in the circulatory system or interstitial fluid in vivo. Soluble antisense polynucleotides present in the external milieu have been shown to gain access to the
cytoplasm and inhibit translation of specific mRNA species. In some embodiments the antisense polynucleotides comprise methylphosphonate moieties, alternatively phosphorothiolates or O-methylribonucleotides may be used, and chimeric
oligonucleotides may also be used (Dagle et al. (1990) Nucleic Acids Res. 18: 4751). For some applications, antisense
oligonucleotides may comprise polyamide nucleic acids (Nielsen et al. (1991) Science 254: 1497). For general methods
relating to antisense polynucleotides, see Antisense RNA and DNA, (1988), D.A. Melton, Ed., Cold Spring Harbor Laboratory, Cold Spring Harbor, NY).
Antisense polynucleotides complementary to one or more sequences are employed to inhibit transcription, RNA processing, and/or translation of the cognate mRNA species andthereby effect a reduction in the amount of the respective encoded polypeptide. Such antisense polynucleotides can provide a therapeutic function by inhibiting the formation of one or more endogenous Ig chains in vivo.
Whether as soluble antisense oligonucleotides or as antisense RNA transcribed from an antisense transgene, the antisense polynucleotides of this invention are selected so as to hybridize preferentially to endogenous Ig sequences at physiological conditions in vivo. Most typically, the
selected antisense polynucleotides will not appreciably hybridize to heterologous Ig sequences encoded by a heavy or light chain transgene of the invention (i.e., the antisense oligonucleotides will not inhibit transgene Ig expression by more than about 25 to 35 percent).
Antiserum Suppression
Partial or complete suppression of endogenous Ig chain expression can be produced by injecting mice with antisera against one or more endogenous Ig chains (Weiss et al. (1984) Proc. Νatl. Acad. Sci. (U.S.A.) 81 211, which is incorporated herein by reference). Antisera are selected so as to react specifically with one or more endogenous Ig chains but to have minimal or no cross-reactivity with heterologous Ig chains encoded by an Ig transgene of the invention. Thus, administration of selected antisera according to a schedule as typified by that of Weiss et al. op.cit. will suppress
endogenous Ig chain expression but permits expression of heterologous Ig chain(s) encoded by a transgene of the present invention.
Nucleic Acids
The nucleic acids, the term "substantial homology" indicates" that two nucleic acids, or de gnated sequences thereof, when optimally aligned and compared, are identical, with appropriate nucleotide insertions or deletions, in at least about 80% of the nucleotides, usually at least about 90% to 95%, and more preferably at least about 98 to 99.5% of the nucleotides. Alternatively, substantial homology exists when the segments will hybridize under selective hybridization conditions, to the complement of the strand. The nucleic acids may be present in whole cellε, in a cell lysate, or in a partially purified or substantially pure form. A nucleic acid is "isolated" or "rendered substantially pure" when purified away from other cellular components or other contaminants, e.g., other cellular nucleic acids or proteins, by standard techniques, including alkaline/SDS treatment, CsCl banding, column chromatography, agarose gel electrophoresis and others well known in the art. See, F. Ausubel, et al., ed. Current Protocols in Molecular Biology, Greene Publishing and Wiley- Interscience, New York (1987).
The nucleic acid compositions of the present
invention, while often in a native sequence (except for modified restriction sites and the like), from either cDNA, genomic or mixtures may be mutated, thereof in accordance with standard techniques to provide gene sequences. For coding sequences, these mutations, may affect amino acid sequence as desired. In particular, DNA sequences substantially
homologous to or derived from native V, D, J, constant, switches and other such sequences described herein are
contemplated (where "derived" indicates that a sequence is identical or modified from another sequence).
A nucleic acid is "operably linked" when it is placed into a functional relationship with another nucleic acid sequence. For instance, a promoter or enhancer is operably linked to a coding sequence if it affects the
transcription of the sequence. With respect to transcription regulatory sequences, operably linked means that the DNA sequences being linked are contiguous and, where necessary to join two protein coding regions, contiguous and in reading frame. For switch sequences, operably linked indicates that the sequences are capable of effecting switch recombination.
Specific Preferred Embodiments
A preferred embodiment of the invention is an animal containing at least one, typically 2-10, and sometimes 25-50 or more copies of the transgene described in Example 12 (e.g., pHC1 or pHC2) bred with an animal containing a single copy of a light chain transgene described in Examples 5, 6, 8, or 14, and the offspring bred with the JH deleted animal described in Example 10. Animals are bred to homozygosity for each of these three traits. Such animals have the following genotype: a single copy (per haploid set of chromosomes) of a human heavy chain unrearranged mini-locus (described in Example 12), a single copy (per haploid set of chromosomes) of a rearranged humanκ light chain construct (described in Example 14), and a deletion at each endogenous mouse heavy chain locus that removes all of the functional JH segments (described in
Example 10). Such animals are bred with mice that are
homozygous for the deletion of the JH segments (Examples 10) to produce offspring that are homozygous for the JH deletion and hemizygous for the human heavy and light chain constructs. The resultant animals are injected with antigens and used for production of human monoclonal antibodies against these antigens.
B cells isolated from such an animal are
monospecific with regard to the human heavy and light chains because they contain only a single copy of each gene.
Furthermore, they will be monospecific with regards to human or mouse heavy chains because both endogenous mouse heavy chain gene copies are nonfunctional by virtue of the deletion spanning the JH region introduced as described in Example 9 and 12. Furthermore, a substantial fraction of the B cells will be monospecific with regards to the human or mouse light chains because expression of the single copy of the rearranged humanκ light chain gene will allelically and isotypically exclude the rearrangement of the endogenous mouse κ and λ chain genes in a significant fraction of B-cells.
The transgenic mouse of the preferred embodiment will exhibit immunoglobulin production with a significant repertoire, ideally substantially similar to that of a native mouse. Thus, for example, in embodiments where the endogenous Ig genes have been inactivated, the total immunoglobulin levels will range from about 0.1 to 10 mg/ml of serum,
preferably 0.5 to 5 mg/ml, ideally at least about 1.0 mg/ml. When a transgene capable of effecting a switch to IgG from IgM has been introduced into the transgenic mouse, the adult mouse ratio of serum IgG to IgM is preferably about 10:1. Of course, the IgG to IgM ratio will be much lower in the
immature mouse. In general, greater than about 10%,
preferably 40 to 80% of the spleen and lymph node B cells express exclusively human IgG protein.
The repertoire will ideally approximate that shown in a non-transgenic mouse, usually at least about 10% as high, preferably 25 to 50% or more. Generally, at least about a thousand different immunoglobulins (ideally IgG), preferably 104 to 106 or more, will be produced, depending primarily on the number of different V, J and D regions introduced into the mouse genome. These immunoglobulins will typically recognize about one-half or more of highly antigenic proteins,
including, but not limited to: pigeon cytochrome C, chicken lysozyme, pokeweed mitogen, bovine serum albumin, keyhole limpit hemocyanin, influenza hemagglutinin, staphylococcus protein A, sperm whale myoglobin, influenza neuraminidase, and lambda repressor protein. Some of the immunoglobulins will exhibit an affinity for preselected antigens of at least about 107M-1, preferably 108M-1 to 109M-1 or greater.
Thus, prior to rearrangement of a transgene containing various heavy or light chain gene segments, such gene segments may be readily identified, e.g. by hybridization or DNA sequencing, as being from a species of organism other than the transgenic animal.
Although the foregoing describes a preferred embodiment of the transgenic animal of the invention, other embodiments are defined by the disclosure herein and more particularly by the transgenes described in the Examples.
Four categories of transgenic animal may be defined:
I. Transgenic animals containing an unrearranged heavy and rearranged light immunoglobulin transgene.
II. Transgenic animals containing an unrearranged heavy and unrearranged light immunoglobulin transgene III. Transgenic animal containing rearranged heavy and an unrearranged light immunoglobulin transgene, and IV. Transgenic animals containing rearranged heavy and rearranged light immunoglobulin transgenes. Of these categories of transgenic animal, the preferred order of preference is as follows II > I > III > IV where the endogenous light chain genes (or at least the κ gene) have been knocked out by homologous recombination (or other method) and I > II > III >IV where the endogenous light chain genes have not been knocked out and must be dominated by allelic exclusion.
EXPERIMENTAL EXAMPLES METHODS AND MATERIALS
Transgenic mice are derived according to Hogan, et al., "Manipulating the Mouse Embryo: A Laboratory Manual", Cold Spring Harbor Laboratory, which is incorporated herein by reference.
Embryonic stem cells are manipulated according to published procedures (Teratocarcinomas and embryonic stem cells: a practical approach, E.J. Robertson, ed., IRL Press, Washington, D.C., 1987; Zjilstra et al., Nature 342:435-438 (1989); and Schwartzberg et al., Science 246:799-803 (1989), each of which is incorporated herein by reference).
DNA cloning procedures are carried out according to J. Sambrook, et al. in Molecular Cloning: A Laboratory
Manual, 2d ed., 1989, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., which is incorporated herein by reference.
Oligonucleotides are synthesized on an Applied Bio Systems oligonucleotide synthesizer according to
specifications provided by the manufacturer.
Hybridoma cells and antibodies are manipulated according to "Antibodies: A Laboratory Manual", Ed Harlow and David Lane, Cold Spring Harbor Laboratory (1988), which is incorporated herein by reference.
EXAMPLE 1
Genomic Heavy Chain Human Ig Transgene
This Example describes the cloning and
microinjection of a human genomic heavy chain immunoglobulin transgene which is microinjected into a murine zygote. Nuclei are isolated from fresh human placental tissue as described by Marzluff et al., "Transcription and Translation: A Practical Approach", B.D. Hammes and
S.J. Higgins, eds., pp. 89-129, IRL Press, Oxford (1985)).
The isolated nuclei (or PBS washed human spermatocytes) are embedded in a low melting point agarose matrix and lysed with EDTA and proteinaseκ to expose high molecular weight DNA, which is then digested in the agarose with the restriction enzyme NotI as described by M. Finney in Current Protocols in Molecular Biology (F. Ausubel, et al., eds. John Wiley & Sons, Supp. 4, 1988, Section 2.5.1).
The NotI digested DNA is then fractionated by pulsed field gel electrophoresis as described by Anand et al.,
Nucl. Acids Res. 17:3425-3433 (1989). Fractions enriched for the NotI fragment are assayed by Southern hybridization to detect one or more of the sequences encoded by this fragment. Such sequences include the heavy chain D segments, J segments, μ and γ1 constant regions together with representatives of all 6 VH families (although this fragment is identified as 670 kb fragment from HeLa cells by Berman et al. (1988), supra., we have found it to be as 830 kb fragment from human placental an sperm DNA). Those fractions containing this NotI fragment (see Fig; 4) are pooled and cloned into the NotI site of the vector pYACNN in Yeast cells. Plasmid pYACNN is prepared by digestion of pYAC-4 Neo (Cook et al., Nucleic Acids Res. 16: 11817 (1988)) with EcoRI and ligation in the presence of the oligonucleotide 5' - AAT TGC GGC CGC - 3'.
YAC clones containing the heavy chain NotI fragment are isolated as described by Brownstein et al., Science
244:1348-1351 (1989), and Green et al., Proc. Natl. Acad. Sci. USA 87:1213-1217 (1990), which are incorporated herein by reference. The cloned NotI insert is isolated from high molecular weight yeast DNA by pulse field gel electrophoresis as described by M. Finney, op cit. The DNA is condensed by the addition of 1 mM spermine and microinjected directly into the nucleus of single cell embryos previously described. EXAMPLE 2
Genomicκ Light Chain Human Ig Transgene
Formed by In Vivo Homologous Recombination
A map of the humanκ light chain has been described in Lorenz et al., Nucl. Acids Res. 15:9667-9677 (1987), which is incorporated herein by reference.
A 450 kb Xhol to NotI fragment that includes all of Cκ, the 3' enhancer, all J segments, and at least five different V segments is isolated and microinjected into the nucleus of single cell embryos as described in Example 1.
EXAMPLE 3
Genomicκ Light Chain Human Ig Transgene
Formed by In Vivo Homologous Recombination
A 750 kb Mlul to NotI fragment that includes all of the above plus at least 20 more V segments is isolated as described in Example 1 and digested with BssHII to produce a fragment of about 400 kb.
The 450 kb Xhol to NotI fragment plus the approximately 400 kb Mlul to BssHII fragment have sequence overlap defined by the BssHII and Xhol restriction sites.
Homologous recombination of these two fragments upon
microinjection of a mouse zygote results in a transgene containing at least an additional 15-20 V segments over that found in the 450 kb XhoI/NotI fragment (Example 2). EXAMPLE 4
Construction of Heavy Chain Mini-Locus A. Construction of pGP1 and pGP2
pBR322 is digested with EcoRI and StyI and ligated with the following oligonucleotides to generate pGP1 which contains a 147 base pair insert containing the restriction sites shown in Fig. 8. The general overlapping of these oligos is also shown in Fig. 9. The oligonucleotides are:
oligo-1 5' - CTT GAG CCC GCC TAA TGA GCG GGC TTT
TTT TTG CAT ACT GCG GCC - 3'
oligo-2 5' - GCA ATG GCC TGG ATC CAT GGC GCG CTA
GCA TCG ATA TCT AGA GCT CGA GCA -3'
oligo-3 5' - TGC AGA TCT GAA TTC CCG GGT ACC AAG
CTT ACG CGT ACT AGT GCG GCC GCT -3'
oligo-4 5' - AAT TAG CGG CCG CAC TAG TAC GCG TAA
GCT TGG TAC CCG GGA ATT - 3'
oligo-5 5' - CAG ATC TGC ATG CTC GAG CTC TAG ATA
TCG ATG CTA GCG CGC CAT GGA TCC - 3'
oligo-6 5' - AGG CCA TTG CGG CCG CAG TAT GCA AAA
AAA AGC CCG CTC ATT AGG CGG GCT - 3'
This plasmid contains a large polylinker flanked by rare cutting NotI sites for building large inserts that can be isolated from vector sequences for microinjection. The plasmid is based on pBR322 which is relatively low copy compared to the pUC based plasmids (pGP1 retains the pBR322 copy number control region near the origin of replication). Low copy number reduces the potential toxicity of insert sequences. In addition, pGP1 contains a strong transcription terminator sequence derived from trpA (Christie et al., Proc. Natl. Acad. Sci. USA 78:4180 (1981)) inserted between the ampicillin resistance gene and the polylinker. This further reduces the toxicity associated with certain inserts by preventing readthrough transcription coming from the
ampicillin promoters.
Plasmid pGP2 is derived from pGP1 to introduce an additional restriction site (Sfil) in the polylinker. pGP1 is digested with Mlul and Spel to cut the recognition sequences in the polylinker portion of the plasmid.
The following adapter oligonucleotides are ligated to the thus digested pGP1 to form pGP2.
5' CGC GTG GCC GCA ATG GCC A 3'
5' CTA GTG GCC ATT GCG GCC A 3' pGP2 is identical to pGP1 except that it contains an additional Sfi I site located between the Mlul and Spel sites. This allows inserts to be completely excised with Sfil as well as with NotI.
B. Construction of pRE3 (rat enhancer 3')
An enhancer sequence located downstream of the rat constant region is included in the heavy chain constructs.
Tue heavy chain region 3' enhancer described by Petterson et al., Nature 344:165-168 (1990), which is
incorporated herein by reference) is isolated and cloned. The rat IGH 3' enhancer sequence is PCR amplified by using the following oligonucleotides: 5' CAG GAT CCA GAT ATC AGT ACC TGA AAC AGG GCT TGC 3'
5' GAG CAT GCA CAG GAC CTG GAG CAC ACA CAG CCT TCC 3'
The thus formed double stranded DNA encoding the 3' enhancer is cut with BamHI and SphI and clone into BamHI/SphI cut pGP2 to yield pRE3 (rat enhancer 3').
C. Cloning of Human J-μ Region
A substantial portion of this region is cloned by combining two or more fragments isolated from phage lambda inserts. See Fig. 9.
A 6.3 kb BamHI/Hindlll fragment that includes all human J segments (Matsuda et al., EMBO J.. 2:1047-1051 (1988); Ravetech et al.m Cell, 27:583-591 (1981), which are
incorporated herein by reference) is isolated from human genomic DNA library using the oligonucleotide GGA CTG TGT CCC TGT GTG ATG CTT TTG ATG TCT GGG GCC AAG.
An adjacent If kb Hindlll/Bamll fragment that
contains enhancer, switch and constant region coding exons (Yasui et al., Eur. J. Immunol. 19:1399-1403 (1989)) is
similarly isolated using the oligonucleotide:
CAC CAA GTT GAC CTG CCT GGT CAC AGA CCT GAC CAC CTA TGA
An adjacent 3' 1.5 kb BamHI fragment is similarly isolated using clone pMUM insert as probe. (pMUM is 4 kb EcoRI/Hindlll fragment isolated from human genomic DNA library with oligonucleotide:
CCT GTG GAC CAC CGC CTC CAC CTT CAT CGT CCT CTT CCT CCT
mu membrane exon 1) and cloned into pUC19.
pGP1 is digested with BamHI and Bglll followed by treatment with calf intestinal alkaline phosphatase.
Fragments (a) and (b) from Fig. 9 are cloned in the digested pGP1. A clone is then isolated which is oriented such that 5' BamHI site is destroyed by BamHI/Bgl fusion. It is identified as pMU (see Fig. 10). pMU is digested with BamHI and fragment (c) from Fig. 9 is inserted. The
orientation is checked with Hindlll digest. The resultant plasmid pHIG1 (Fig. 10) contains an 18 kb insert encoding J and Cμ segments.
D. Cloning of Cμ Region
pGP1 is digested with BamHI and Hindlll is followed by treatment with calf intestinal alkaline phosphatase (Fig. 14). The so treated fragment (b) of Fig. 14 and fragment (c) of Fig. 14 are cloned into the BamHI/Hindlll cut pGP1. Proper orientation of fragment (c) is checked by Hindlll digestion to form pCON1 containing a 12 kb insert encoding the Cμ region.
Whereas pHIGl contains J segments, switch and μ sequences in its 18 kb insert with an Sfil 3' site and a Spel 5' site in a polylinker flanked by NotI sites, will be used for rearranged VDJ segments. pCON1 is identical except that it lacks the J region and contains only a 12 kb insert. The use of pCON1 in the construction of fragment containing rearranged VDJ segments will be described hereinafter.
E. Cloning of γ-1 Constant Region (pREG2)
The cloning of the human γ-1 region is depicted in Fig. 16.
Yamamura et al., Proc. Natl. Acad. Sci. USA
83:2152-2156 (1986) reported the expression of membrane bound human γ-1 from a transgene construct that had been partially deleted on integration. Their results indicate that the 3' BamHI site delineates a sequence that includes the
transmembrane rearranged and switched copy of the gamma gene with a V-C intron of less than 5kb. Therefore, in the unrearranged, unswitched gene, the entire switch region is included in a sequence beginning less than 5 kb from the 5' end of the first γ-1 constant exon. Therefore it is included in the 5' 5.3 kb Hindlll fragment (Ellison et al., Nucleic Acids Res. 10:4071-4079 (1982), which is incorporated herein by reference). Takahashi et al., Cell 29: 671-679 (1982), which is incorporated herein by reference, also reports that this fragment contains the switch sequence, and this fragment together with the 7.7 kb Hindlll to BamHI fragment must include all of the sequences we need for the transgene
construct. An intronic sequence is a nucleotide sequence of at least 15 contiguous nucleotides that occurs in an intron of a specified gene.
Phage clones containing the γ-1 region are identified and isolated "sing the following oligonucleotide which is specific for the third exon of γ-1 (CH3).
5' TGA GCC ACG AAG ACC CTG AGG
TCA AGT TCA ACT GGT ACG TGG 3'
A 7.7 kb Hindlll to Bglll fragment (fragment (a) in Fig. 11) is cloned into Hindlll/Bglll cut pRE3 to form pREGl. The upstream 5.3 kb Hindlll fragment (fragment (b) in Fig. 11) is cloned into Hindlll digested pREGl to form pREG2. Correct orientation is confirmed by BamHI/Spel digestion. F. Combining Cγ and Cμ
The previously described plasmid pHIGl contains human J segments and the Cμ constant region exons. To provide a transgene containing the Cμ constant region gene segments, pHIG1 was digested with Sfil (Fig. 10). The plasmid pREG2 was also digested with Sfil to produce a 13.5 kb insert containing human Cγ exons and the rat 3' enhancer sequence. These sequences were combined to produce the plasmid pHIG3' (Fig. 12) containing the human J segments, the human Cμ constant region, the human Cγ1 constant region and the rat 3' enhancer contained on a 31.5 kb insert.
A second plasmid encoding human Cμ and human Cγ1 without J segments is constructed by digesting pCON1 with Sfil and combining that with the Sfil fragment containing the human Cγ region and the rat 3' enhancer by digesting pREG2 with Sfil. The resultant plasmid, pCON (Fig. 12) contains a 26 kb NotI/Spel insert containing human Cμ, human γ1 and the rat 3' enhancer sequence.
G. Cloning of D Segment
The strategy for cloning the human D segments is depicted in Fig. 13. Phage clones from the human genomic library containing D segments are identified and isolated using probes specific for diversity region sequences (Ichihara et al., EMBO J. 7:4141-4150 (1988)). The following
oligonucleotides are used:
DXP1: 5' - TGG TAT TAC TAT GGT TCG GGG AGT TAT TAT
AAC CAC AGT GTC - 3'
DXP4: 5' - GCC TGA AAT GGA GCC TCA GGG CAC AGT GGG
CAC GGA CAC TGT - 3' DN4: 5' - GCA GGG AGG ACA TGT TTA GGA TCT GAG GCC
GCA CCT GAC ACC - 3'
A 5.2 kb Xhol fragment (fragment (b) in Fig. 13) containing DLR1, DXP1, DXP'1, and DA1 is isolated from a phage clone identified with oligo DXP1.
A 3.2 kb Xbal fragment (fragment (c) in Fig. 13) containing DXP4, DA4 and DK4 is isolated from a phage clone identified with oligo DXP4.
Fragments (b), (c) and (d) from Fig. 13 are combined and cloned into the Xbal/Xhol site of pGP1 to form pHIG2 which contains a 10.6 kb insert.
This cloning is performed sequentially. First, the 5.2 kb fragment (b) in Fig. 13 and the 2.2 kb fragment (d) of Fig. 13 are treated with calf intestinal alkaline phosphatase and cloned into pGP1 digested with Xhol and Xbal. The resultant clones are screened with the 5.2 and 2.2 kb insert. Half of those clones testing positive with the 5.2 and 2.2 kb inserts have the 5.2 kb insert in the proper orientation as determined by BamHI digestion. The 3.2 kb Xbal fragment from Fig. 13 is then cloned into this intermediate plasmid
containing fragments (b) and (d) to form pHIG2. This plasmid contains diversity segments cloned into the polylinker with a unique 5' Sfil site and unique 3' Spel site. The entire polylinker is flanked by NotI sites.
H. Construction of Heavy Chain Minilocus
The following describes the construction of a human heavy chain mini-locus which contain one or more V segments.
An unrearranged V segment corresponding to that identified as the V segment contained in the hybridoma of Newkirk et al., J. Clin. Invest, 81:1511-1518 (1988), which is incorporated herein by reference, is isolated using the following oligonucleotide:
5' - GAT CCT GGT TTA GTT AAA GAG GAT TTT
ATT CAC CCC TGT GTC - 3'
A restriction map of the unrearranged V segment is determined to identify unique restriction sites which provide upon digestion a DNA fragment having a length approximately 2 kb containing the unrearranged V segment together with 5' and 3' flanking sequences. The 5' prime sequences will include promoter and other regulatory sequences whereas the 3'
flanking sequence provides recombination sequences necessary for V-DJ joining. This approximately 3.0 kb V segment insert is cloned into the polylinker of pGB2 to form pVH1.
pVHl is digested with Sfil and the resultant fragment is cloned into the Sfil site of pHIG2 to form a
PHIG5 ' . Since pHIG2 contains D segments only, the resultant pHIG5' plasmid contains a single V segment together with D segments. The size of the insert contained in pHIG5 is 10.6 kb plus the size of the V segment insert.
The insert from pHIG5 is excised by digestion with NotI and Spel and isolated. pHIG3' which contains J, Cμ and Cγ1 segments is digested with Spel and NotI and the 3' kb fragment containing such sequences and the rat 3' enhancer sequence is isolated. These two fragments are combined and ligated into NotI digested pGP1 to produce pHIG which contains insert encoding a V segment, nine D segments, six functional J segments, Cμ, Cγ and the rat 3' enhancer. The size of this insert is approximately 43 kb plus the size of the V segment insert.
I. Construction of Heavy Chain Minilocus
by Homologous Recombination
As indicated in the previous section, the insert of pHIG is approximately 43 to 45 kb when a single V segment is employed. This insert size is at or near the limit of that which may be readily cloned into plasmid vectors. In order to provide for the use of a greater number of V segments, the following describes in vivo homologous recombination of overlapping DNA fragments which upon homologous recombination within a zygote or ES cell form a transgene containing the rat 3' enhancer sequence, the human Cμ, the human Cγ1, human J segments, human D segments and a multiplicity of human V segments.
A 6.3 kb BamHI/Hindlll fragment containing human J segments (see fragment (a) in Fig. 9) is cloned into Mlul/Spel digested pHIG5' using the following adapters: 5' GAT CCA AGC AGT 3'
5' CTA GAC TGC TTG 3'
5' CGC GTC GAA CTA 3'
5' AGC TTA GTT CGA 3'
The resultant is plasmid designated pHIG5'O (overlap). The insert contained in this plasmid contains human V, D and J segments. When the single V segment from pVHl is used, the size of this insert is approximately 17 kb plus 2 kb. This insert is isolated and combined with the insert from pHIG3' which contains the human J, Cμ, γ1 and rat 3' enhancer sequences. Both inserts contain human J segments which provide for approximately 6.3 kb of overlap between the two DNA fragments. When coinjected into the mouse zygote, in vivo homologous recombination occurs generating a transgene equivalent to the insert contained in pHIG.
This approach provides for the addition of a multiplicity of V segments into the transgene formed in vivo. For example, instead of incorporating a single V segment into pHIG5', a multiplicity of V segments contained on (1) isolated genomic DNA, (2) ligated DNA derived from genomic DNA, or (3) DNA encoding a synthetic V segment repertoire is cloned into pHIG2 at the Sfil site to generate pHIG5' VN. The J segments fragment (a) of Fig. 9 is then cloned into pHIG5' VN and the insert isolated. This insert now contains a multiplicity of V segments and J segments which overlap with the J segments contained on the insert isolated from pHIG3'. When
cointroduced into the nucleus of a mouse zygote, homologous recombination occurs to generate in vivo the transgene
encoding multiple V segments and multiple J segments, multiple D segments, the Cμ region, the Cγ1 region (all from human) and the rat 3' enhancer sequence.
EXAMPLE 5
Construction of Light Chain Minilocus
A. Construction of pEul
The construction of pEμl is depicted in Fig. 16.
The mouse heavy chain enhancer is isolated on the Xbal to EcoRI 678 bp fragment (Banerji et al., Cell 33:729-740 (1983)) from phage clones using oligo: 5' GAA TGG GAG TGA GGC TCT CTC ATA CCC
TAT TCA GAA CTG ACT 3' This Eμ fragment is cloned into EcoRV/Xbal digested pGP1 by blunt end filling in EcoRI site. The resultant plasmid is designated pEmul. B. Construction Of κ Light chain Minilocus
Theκ construct contains at least one human Vκ segment, all five human Jκ segments, the human J-Cκ enhancer, humanκ constant region exon, and, ideally, the human 3' κ enhancer (Meyer et al., EMBO J. 8:1959-1964 (1989)). The κ enhancer in mouse is 9 kb downstream from Cκ. However, it is as yet unidentified in the human. In addition, the construct contains a copy of the mouse heavy chain J-Cμ enhancers.
The minilocus is constructed from four component fragments:
(a) A 16 kb Smal fragment that contains the human
Cκ exon and the 3' human enhancer by analogy with the mouse locus;
(b) A 5' adjacent 5 kb Smal fragment, which
contains all five J segments;
(c) The mouse heavy chain intronic enhancer
isolated from pEμl (this sequence is included to induce expression of the light chain construct as early as possible in B-cell development. Because the heavy chain genes are transcribed earlier than the light chain genes, this heavy chain enhancer is presumably active at an earlier stage than the intronic κ enhancer); and
(d) A fragment containing one or more V segments. The preparation of this construct is as follows. Human placental DNA is digested with Smal and fractionated on agarose gel by electrophoresis. Similarly, human placental DNA is digested with BamHI and fractionated by
electrophoresis. The 16 kb fraction is isolated from the Smal digested gel and the 11 kb region is similarly isolated from the gel containing DNA digested with BamHI.
The 16 kb Smal fraction is cloned into Lambda FIX II
(Stratagene, La Jolla, California) which has been digested with Xhol, treated with klenow fragment DNA polymerase to fill in the Xhol restriction digest product. Ligation of the 16 kb Smal fraction destroys the Smal sites and lases Xhol sites intact.
The 11 kb BamHI fraction is cloned into λ EMBL3 (Strategene, La Jolla, California) which is digested with BamHI prior to cloning.
Clones from each library were probed with the CK specific oligo: 5' GAA CTG TGG CTG CAC CAT CTG TCT
TCA TCT TCC CGC CAT CTG 3'
A 16 kb Xhol insert that was subcloned into the Xhol cut pEμl so that Cκ is adjacent to the Smal site. The
resultant plasmid was designated pKapl.
The above Cκ specific oligonucleotide is used to probe the λ EMBL3/BamHI library to identify an 11 kb clone. A 5 kb Smal fragment (fragment (b) in Fig. 20) is subcloned and subsequently inserted into pKapl digested with Smal. Those plasmids containing the correct orientation of J segments, Cκ and the Eμ enhancer are designated pKap2.
One or more Vκ segments are thereafter subcloned into the Mlul site of pKap2 to yield the plasmid pKapH which encodes the human Vκ segments, the human Jκ segments, the human Cκ segments and the human Eμ enhancer. This insert is excised by digesting pKapH with NotI and purified by agarose gel electrophoresis. The thus purified insert is
microinjected into the pronucleus of a mouse zygote as
previously described. C. Construction of κ Light Chain Minilocus by
In Vivo Homologous Recombination
The 11 kb BamHI fragment is cloned into BamHI .
digested pGP1 such that the 3' end is toward the Sfil site. The resultant plasmid is designated pKAPint. One or more Vκ segments is inserted into the polylinker between the BamHI and Spel sites in pKAPint to form pKapHV. The insert of pKapHV is excised by digestion with NotI and purified. The insert from pKap2 is excised by digestion with NotI and purified. Each of these fragments contain regions of homology in that the fragment from pKapHV contains a 5 kb sequence of DNA that include the Jκ segments which is substantially homologous to the 5 kb Smal fragment contained in the insert obtained from pKap2. As such, these inserts are capable of homologously recombining when microinjected into a mouse zygote to form a transgene encoding Vκ, Jκ and Cκ.
EXAMPLE 6
Isolation of Genomic Clones
Corresponding to Rearranged and Expressed Copies of Immunoglobulin κ Light Chain Genes
This example describes the cloning of immunoglobulinκ light chain genes from cultured cells that express an immunoglobulin of interest. Such cells may contain multiple alleles of a given immunoglobulin gene. For example, a hybridoma might contain four copies of theκ light chain gene, two copies from the fusion partner cell line and two copies from the original B-cell expressing the immunoglobulin of interest. Of these four copies, only one encodes the
immunoglobulin of interest, despite the fact that several of them may be rearranged. The procedure described in this example allows for the selective cloning of the expressed copy of theκ light chain.
A. Double Stranded cDNA
Cells from human hybridoma, or lymphoma, or other cell line that synthesizes either cell surface or secreted or both forms of IgM with aκ light chain are used for the isolation of polyA+ RNA. The RNA is then used for the
synthesis of oligo dT primed cDNA using the enzyme reverse transcriptase. The single. stranded cDNA is then isolated and G residues are added to the 3' end using the enzyme
polynucleotide terminal transferase. The Gtailed
single-stranded cDNA is then purified and used as template for second strand synthesis (catalyzed by the enzyme DNA
polymerase) using the following oligonucleotide as a primer: 5' - GAG GTA CAC TGA CAT ACT GGC ATG CCC
CCC CCC CCC - 3' The double stranded cDNA is isolated and used for determining the nucleotide sequence of the 5' end of the mRNAs encoding the heavy and light chains of the expressed
immunoglobulin molecule. Genomic clones of these expressed genes are then isolated. The procedure for cloning the expressed light chain gene is outlined in part B below.
B. Light Chain
The double stranded cDNA described in part A is denatured and used as a template for a third round of DNA synthesis using the following oligonucleotide primer: 5' - GTA CGC CAT ATC AGC TGG ATG AAG TCA TCA GAT
GGC GGG AAG ATG AAG-.ACA GAT GGT GCA - 3'
This primer contains sequences specific for the constant portion of theκ light chain message (TCA TCA GAT GGC GGG AAG ATG AAG ACA GAT GGT GCA) as well as unique sequences that can be used as a primer for the PCR amplification of the newly synthesized DNA strand (GTA CGC CAT ATC AGC TGG ATG AAG). The sequence is amplified by PCR using the following two oligonucleotide primers: 5' - GAG GTA CAC TGA CAT ACT GGC ATG -3'
5' - GTA CGC CAT ATC AGC TGG ATG AAG -3'
The PCR amplified sequence is then purified by gel electrophoresis and used as template for dideoxy sequencing reactions using the following oligonucleotide as a primer: 5' - GAG GTA CAC TGA CAT ACT GGC ATG -3' The first 42 nucleotides of sequence will then be used to synthesize a unique probe for isolating the gene from which immunoglobulin message was transcribed. This synthetic 42 nucleotide segment of DNA will be referred to below as o-kappa.
A Southern blot of DNA, isolated from the Ig expressing cell line and digested individually and in pairwise combinations with several different restriction endonucleases including Smal, is then probed with the 32-P labelled unique oligonucleotide o-kappa. A unique restriction endonuclease site is identified upstream of the rearranged V segment.
DNA from the Ig expressing cell line is then cut with Smal and second enzyme (or BamHI or Kpnl if there is Smal site inside V segment). Any resulting non-blunted ends are treated with the enzyme T4 DNA polymerase to give blunt ended DNA molecules. Then add restriction site encoding linkers (BamHI, EcoRI or Xhol depending on what site does not exist in fragment) and cut with the corresponding linker enzyme to give DNA fragments with BamHI, EcoRI or Xhol ends. The DNA is then size fractionated by agarose gel electrophoresis, and the fraction including the DNA fragment covering the expressed V segment is cloned into lambda EMBL3 or Lambda FIX (Stratagene, La Jolla, California). V segment containing clones are isolated using the unique probe o-kappa. DNA is isolated from positive clones and subcloned into the polylinker of pKapl. The resulting clone is called pRKL. EXAMPLE 7
Isolation of Genomic Clones
Corresponding to Rearranged Expressed Copies of Immunoglobulin Heavy Chain μ Genes
This example describes the cloning of immunoglobulin heavy chain μ genes from cultured cells of expressed and immunoglobulin of interest. The procedure described in this example allows for the selective cloning of the expressed copy of a μ heavy chain gene.
Double-stranded cDNA is prepared and isolated as described herein before. The double-stranded cDNA is
denatured and used as a template for a third round of DNA synthesis using the following oligonucleotide primer:
5' - GTA CGC CAT ATC AGC TGG ATG AAG ACA GGA GAC
GAG GGG GAA AAG GGT TGG GGC GGA TGC - 3'
This primer contains sequences specific for the constant portion of the μ heavy chain message (ACA GGA GAC GAG GGG GAA AAG GGT TGG GGC GGA TGC) as well as unique sequences that can be used as a primer for the PCR amplification of the newly synthesized DNA strand (GTA CGC CAT ATC AGC TGG ATG AAG). The sequence is amplified by PCR using the following two oligonucleotide primers:
5' - GAG GTA CAC TGA CAT ACT GGC ATG - 3' 5' - GTA CTC CAT ATC AGC TGG ATG AAG - 3'
The PCR amplified sequence is then purified by gel electrophoresis and used as template for dideoxy sequencing reactions using the following oligonucleotide as a primer:
5' - GAG GTA CAC TGA CAT ACT GGC ATG - 3' The first 42 nucleotides of sequence are then used to synthesize a unique probe for isolating the gene from which immunoglobulin message was transcribed. This synthetic 42 nucleotide segment of DNA will be referred to below as o-mu.
A Southern blot of DNA, isolated from the Ig
expressing cell line and digested individually and in pairwise combinations with several different restriction endonucleases including Mlul (Mlul is a rare cutting enzyme that cleaves between the J segment and mu CH1), is then probed with the 32-P labelled unique oligonucleotide o-mu. A unique
restriction endonuclease site is identified upstream of the rearranged V segment.
DNA from the Ig expressing cell line is then cut with Mlul and second enzyme. Mlul or Spel adapter linkers are then ligated onto the ends and cut to convert the upstream site to Mlul or Spel. The DNA is then size fractionated by agarose gel electrophoresis, and the fraction including the DNA fragment covering the expressed V segment is cloned directly into the plasmid pGP1. V segment containing clones are isolated using the unique probe o-mu, and the insert is subcloned into Mlul or Mlul/Spel cut plasmid pCON2. The resulting plasmid is called pRMGH. EXAMPLE 8
Construction of Human κ Miniloci Transgenes
Light Chain Minilocus
A human genomic DNA phage library was screened with kappa light chain specific oligonucleotide probes and isolated clones spanning the Jκ-C region. A 5.7 kb Clal/Xhol fragment containing Jκ1 together with a 13 kb Xhol fragment containing Jκ2-5 and Cκ into pGP1d was cloned and used to create the plasmid pKcor. This plasmid contains Jκ1-5, the kappa
intronic enhancer and Cκ together with 4.5 kb of 5' and 9 kb of 3' flanking sequences. It also has a unique 5' Xhol site for cloning Vκ segments and a unique 3' Sall site for
inserting additional cis-acting regulatory sequences. V kappa genes
A human genomic DNA phage library was screened with Vκ light chain specific oligonucleotide probes and isolated clones containing human Vκ segments. Functional V segments were identified by DNA sequence analysis. These clones contain TATA boxes, open reading frames encoding leader and variable peptides (including 2 cysteine residues), splice sequences, and recombination heptamer-12 bp spacer-nonamer sequences. Three of the clones were mapped and sequenced.
Two of the clones, 65.5 and 65.8 appear to be functional, they contain TATA boxes, open reading frames encoding leader and variable peptides (including 2 cysteine residues), splice sequences, and recombination heptamer-12 bp spacer-nonamer sequences. The third clone, 65.4, appears to encode a VκI pseudogene as it contains a non-canonical recombination heptamer.
One of the functional clones, Vk 65-8, which encodes a Vklll family gene, was used to build a light chain minilocus construct. pKCI
The kappa light chain minilocus transgene pKC1 (Fig. 32) was generated by inserting a 7.5 kb Xhol/Sall fragment containing Vκ 65.8 into the 5' Xhol site of pKcor. The transgene insert was isolated by digestion with NotI prior to injection.
The purified insert was microinjected into the pronuclei of fertilized (C57BL/6 x CBA) F2 mouse embryos and transferred the surviving embryos into pseudopregnant females as described by Hogan et al. (in Methods of Manipulating the Mouse Embryo, 1986, Cold Spring Harbor Laboratory, New York). Mice that developed from injected embryos were analyzed for the presence of transgene sequences by Southern blot analysis of tail DNA. Transgene copy number was estimated by band intensity relative to control standards containing known quantities of cloned DNA. Serum was isolated from these animals and assayed for the presence of transgene encoded human Ig kappa protein by ELISA as described by Harlow and Lane (in Antibodies: A Laboratory Manual, 1988, Cold Spring Harbor Laboratory, New York). Microtiter plate wells were coated with mouse monoclonal antibodies specific for human Ig kappa (clone 6E1, #0173 , AMAC, Inc. , Westbrook, ME) , human IgM (Clone AF6, #0285, AMAC, Inc., Westbrook, ME) and human IgG1 (clone JL512, #0280, AMAC, Inc., Westbrook, ME). Serum samples were serially diluted into the wells and the presence of specific immunoglobulins detected with affinity isolated alkaline phosphatase conjugated goat anti-human Ig
(polyvalent) that had been pre-adsorbed to minimize cross- reactivity with mouse immunoglobulins.
Fig. 35 shows the results of an ELISA assay of serum from 8 mice (I.D. #676, 674, 673, 670, 666, 665, 664, and 496). The first seven of these mice developed from embryos that were injected with the pKC1 transgene insert and the eighth mouse is derived from a mouse generated by
microinjection of the pHC1 transgera (described previously). Two of the seven mice from KC1 injected embryos (I.D.#'s 666 and 664) did not contain the transgene insert as assayed by DAN Southern blot analysis, and five of the mice (I.D.#'s 676, 674, 673, 670, and 665) contained the transgene. Al! but one of the KC1 transgene positive animals express detectable levels of human Ig kappa protein, and the single non- expressing animal appears to be a genetic mosaic on the basis of DNA Southern blot analysis. The pHC1 positive transgenic mouse expresses human IgM and IgG1 but not Ig kappa,
demonstrating the specificity of the reagents used in the assay. pKC2
The kappa light chain minilocus transgene pKC2 was generated by inserting an 8 kb Xhol/Sall fragment containing Vκ 65.5 into the 5' Xhol site of pKC1. The resulting
transgene insert, which contains two Vκ segments, was isolated prior to microinjection by digestion with NotI.
pKVe2
This construct is identical to pKCI except that it includes 1.2 kb of additional sequence 5' of Jκ and is missing 4.5 kb of sequence 3' of Vκ 65.8. In additional it contains a 0.9 kb Xbal fragment containing the mouse heavy chain J-m intronic enhancer (Banerji et al., Cell 33:729-740 (1983)) together with a 1.4 kb Mlul Hindlll fragment containing the human heavy chain J-m intronic enhancer (Hayday et al., Nature 307:334-340 (1984)) inserted downstream. This construct tests the feasibility of initiating early rearrangement of the light chain minilocus to effect allelic and isotypic exclusion.
Analogous constructs can be generated with different
enhancers, i.e., the mouse or rat 3' kappa or heavy chain enhancer (Meyer and Neuberger, EMBO J. 8:1959-1964 (1989); Petterson et al. Nature 344:165-168 (1990), which are
incorporated herein by reference).
Rearranged Light Chain Transgenes
A kappa light chain expression cassette was designed to reconstruct functionally rearranged light chain genes that have been amplified by PCR from human B-cell DNA. The scheme is outlined in Fig. 33. PCR amplified light chain genes are cloned into the vector pK5nx that includes 3.7 kb of 5' flanking sequences isolated from the kappa light chain gene 65.5. The VJ segment fused to the 5' transcriptional
sequences are then cloned into the unique Xhol site of the vector pK31s that includes Jκ2-4, the Jκ intronic enhancer, Cκ, and 9 kb of downstream sequences. The resulting plasmid contains a reconstructed functionally rearranged kappa light chain transgene that can be excised with NotI for
microinjection into embryos. The plasmids also contain unique Sall sites at the 3' end for the insertion of additional cis- acting regulatory sequences.
Two synthetic oligonucleotides (o-130, o-131) were used J amplify rearranged kappa light chain genes from human spleen genomic DNA. Oligonucleotide o-131 (gga ccc aga
(g,c)gg aac cat gga a(g,a) (g,a,t,c)) is complementary to the 5' region of V^III family light chain genes and overlaps the first ATC of the leader sequence. Oligonucleotide o-130 (gtg caa tea att etc gag ttt gac tac aga c) is complementary to a sequence approximately 150 bp 3' of Jκ1 and includes an Xhol site. These two oligonucleotides amplify a 0.7 kb DNA
fragment from human spleen DNA corresponding to rearranged
VκIII genes joined to Jκ1 segments. The PCR amplified DNA was digested with Ncol and Xhol and cloned individual PCR products into the plasmid pNN03. The DNA sequence of 5 clones was determined and identified two with functional VJ joints (open reading frames). Additional functionally rearranged light chain clones are collected. The functionally rearranged clones can be individually cloned into light chain expression
cassette described above (Fig. 33). Transgenic mice generated with the rearranged light chain constructs can be bred with heavy chain minilocus transgenics to produce a strain of mice that express a spectrum of fully human antibodies in which all of the diversity of the primary repertoire is contributed by the heavy chain. One source of light chain diversity can be from somatic mutation. Because not all light chains will be equivalent with respect to their ability to combine with a variety of different heavy chains, different strains of mice, each containing different light chain constructs can be
generated and tested. The advantage of this scheme, as opposed to the use of unrearranged light chain miniloci, is the increased light chain allelic and isotypic exclusion that comes from having the light chain ready to pair with a heavy chain as soon as heavy chain VDJ joining occurs. This
combination can result in an increased frequency of B-cells expressing fully human antibodies, and thus it can facilitate the isolation of human Ig expressing hybridomas.
NotI inserts of plasmids pIGM1, pHC1, pIGG1, pKC1, and pKC2 were isolated away from vector sequences by agarose gel electrophoresis. The purified inserts were microinjected into the pronuclei of fertilized (C57BL/6 x CBA) F2 mouse embryos and transferred the surviving embryos into
pseudopregnant females as described by Hogan et al. (Hogan et al., Methods of Manipulating the Mouse Embryo. Cold Spring Harbor Laboratory, New York (1986)).
EXAMPLE 9
Inactivation of the Mouse Kappa Light Chain Gene by Homologous Recombination
This example describes the inactivation of the mouse endogenous kappa locus by homologous recombination in
embryonic stem (ES) cells followed by introduction of the mutated gene into the mouse germ line by injection of targeted ES cells bearing an inactivated kappa allele into early mouse embryos (blastocysts).
The strategy is to delete Jκ and Cκ by homologous recombination with a vector containing DNA sequences
homologous to the mouse kappa locus in which a 4.5 kb segment of the locus, spanning the Jκ gene and Cκ segments, is deleted and replaced by the selectable marker neo.
Construction of the kappa targeting vector
The plasmid pGEM7 (KJ1) contains the neomycin resistance gene (neo), used for drug selection of transfected ES cells, under the transcriptional control of the mouse phosphoglycerate kinase (pgk) promoter (Xbal/TaqI fragment; Adra et al., Gene 60: 65-74 (1987)) in the cloning vector pGEM- 7Zf(+). The plasmid also includes a heterologous polyadenylation site for the neo gene, derived from the 3' region of the mouse pgk gene (PvulI/Hindlll fragment; Boer et al., Biochemical Genetics. 28:299-308 (1990)). This plasmid was used as the starting point for construction of the kappa targeting vector. The first step was to insert sequences homologous to the kappa locus 3' of the neo expression
cassette.
Mouse kappa chain sequences (Fig. 20a) were isolated from a genomic phage library derived from liver DNA using oligonucleotide probes specific for the C/c locus:
5'- GGC TGA TGC TGC ACC AAC TGT ATC CAT CTT CCC ACC ATC CAG -3'
and for the Jκ5 gene segment:
5'- CTC ACG TTC GGT GCT GGG ACC AAG CTG GAG CTG AAA CGT AAG - 3'.
An 8 kb Bglll/SacI fragment extending 3' of the mouse Cκ segment was isolated from a positive phage clone in two pieces, as a 1.2 kb Bglll/SacI fragment and a 6.8 kb SacI fragment, and subcloned into Bglll/SacI digested pGEM7 (KJ1) to generate the plasmid pNEO-K3' (Fig. 20b).
A 1.2 kb EcoRI/SphI fragment extending 5' of the Jκ region was also isolated from a positive phage clone. An Sphl/Xbal/Bglll/EcoRI adaptor was ligated to the SphI site of this fragment, and the resulting EcoRI fragment was ligated into EcoRI digested pNEO-K3', in the same 5' to 3' orientation as the neo gene and the downstream 3' kappa sequences, to generate pNEO-K5'3' (Fig. 20c).
The Herpes Simplex Virus (HSV) thymidine kinase (TK) gene was then included in the construct in order to allow for enrichment of ES clones bearing homologous recombinants, as described by Mansour et al., Nature 336:348-352 (1988), which is incorporated herein by reference. The HSV TK cassette was obtained from the plasmid pGEM7 (TK), which contains the structural sequences for the HSV TK gene bracketed by the mouse pgk promoter and polyadenylation sequences as described above for pGEM7 (KJ1). The EcoRI site of pGEM7 (TK) was modified to a BamHI site and the TK cassette was then excised as a BamHI/Hindlll fragment and subcloned into pGFlb to generate pGP1b-TK. This plasmid was linearized at the Xhol site and the Xhol fragment from pNEO-K5'3', containing the neo gene flanked by genomic sequences from 5' of JK and 3' of Cκ, was inserted into pGP1b-TK to generate the targeting vector J/C KI (Fig. 20d). The putative structure of the genomic kappa locus following homologous recombination with J/C Kl is shown in Fig. 20e.
Generation and analysis of ES cells with targeted inactivation of a kappa allele
The ES cells used were the AB-1 line grown on mitotically inactive SNL76/7 cell feeder layers (McMahon and Bradley, Cell 62:1073-1085 (1990)) essentially as described (Robertson, E.J. (1987) in Teratocarcinomas and Embryonic Stem Cells: A Practical Approach. E.J. Robertson, ed. (Oxford: IRL Press), p. 71-112). Other suitable ES lines include, but are not limited to, the E14 line (Hooper et al. (1987) Nature 326: 292-295), the D3 line (Doetschman et al. (1985) J. Embrvol. Exp. Morph. 87: 27-45), and the CCE line (Robertson et al.
(1986) Nature 323: 445-448). The success of generating a mouse line from ES cells bearing a specific targeted mutation depends on the pluripotence of the ES cells (i.e., their ability, once injected into a host blastocyst, to participate in embryogenesis and contribute to the germ cells of the resulting animal).
The pluripotence of any given ES cell line can vary with time in culture and the care with which it has been handled. The only definitive assay for pluripotence is to determine whether the specific population of ES cells to be used for targeting can give rise to chimeras capable of germline transmission of the ES genome. For this reason, prior to gene targeting, a portion of the parental population of AB-1 cells is injected into C57B1/6J blastocysts to
ascertain whether the cells are capable of generating chimeric mice with extensive ES cell contribution and whether the majority of these chimeras can transmit the ES genome to progeny.
The kappa chain inactivation vector J/C Kl was digested with NotI and electroporated into AB-1 cells by the methods described (Hasty et al., Nature. 350:243-246 (1991)). Electroporated cells were plated onto 100 mm dishes at a density of 1-2 x 106 cells/dish. After 24 hours, G418
(200μg/ml of active component) and FIAU (0.5μM) were added to the medium, and drug-resistant clones were allowed to develop over 10-11 days. Clones were picked, trypsinized, divided into two portions, and further expanded. Half of the cells derived from each clone were then frozen and the other half analyzed for homologous recombination between vector and target sequences.
DNA analysis was carried out by Southern blot hybridization. DNA was isolated from the clones as described (Laird et al., Nucl. Acids Res. 19:4293 (1991)) digested with Xbal and probed with the 800 bp EcoRI/Xbal fragment indicated in Fig. 20e as probe A. This probe detects a 3.7 kb Xbal fragment in the wild type locus, and a diagnostic 1.8 kb band in a locus which has homologously recombined with the
targeting vector (see Fig. 20a and e). Of 901 G418 and FIAU resistant clones screened by Southern blot analysis, 7 displayed the 1.8 kb Xbal band indicative of a homologous recombination into one of the kappa genes. These 7 clones were further digested with the enzymes Bglll, Sad, and PstI to verify that the vector integrated homologously into one of the kappa genes. When probed with the diagnostic 800 bp
EcoRI/Xbal fragment (probe A), Bglll, Sad, and PstI digests of wild type DNA produce fragments of 4.1, 5.4, and 7 kb, respectively, whereas the presence of a targeted kappa allele would be indicated by fragments of 2.4,
Figure imgf000082_0001
, and 5.7 kb, respectively (see Fig. 20a and e). All positive clones detected by the Xbal digest showed the expected Bglll, Sad, and PstI restriction fragments diagnostic of a homologous recombination at the kappa light chain. In addition. Southern blot analysis of an Nsil digest of the targeted clones using a neo specific probe (probe B, Fig. 20e) generated only the predicted fragment of 4.2 kb, demonstrating that the clones each contained only a single copy of the targeting vector.
Generation of mice bearing the inactivated kappa chain
Five of the targeted ES clones described in the previous section were thawed and injected into C57B1/6J blastocysts as described (Bradley, A. (1987) in
Teratocarcinomas and Embryonic Stem Cells: A Practical
Approach. E.J. Robertson, ed. (Oxford: IRL Press), p. 113-151) and transferred into the uteri of pseudopregnant females to generate chimeric mice resulting from a mixture of cells derived from the input ES cells and the host blastocyst. The extent of ES cell contribution to the chimeras can be visually estimated by the amount of agouti coat coloration, derived from the ES cell line, on the black C57B1/6J background.
Approximately half of the offspring resulting from blastocyst injection of the targeted clones were chimeric (i.e., showed agouti as well as black pigmentation) and of these, the majority showed extensive (70 percent or greater) ES cell contribution to coat pigmentation. The AB1 ES cells are an XY cell line and a majority of these high percentage chimeras were male due to sex conversion of female embryos colonized by male ES cells. Male chimeras derived from 4 of the 5 targeted clones were bred with C57BL/6J females and the offspring monitored for the presence of the dominant agouti coat color indicative of germline transmission of the ES genome.
Chimeras from two of these clones consistently generated agouti offspring. Since only one copy of the kappa locus was targeted in the injected ES clones, each agouti pup had a 50 percent chance of inheriting the mutated locus. Screening for the targeted gene was carried out by Southern blot analysis of Bgl II-digested DNA from tail biopsies, using the probe utilized in identifying targeted ES clones (probe A, Fig.
20e). As expected, approximately 50 percent of the agouti offspring showed a hybridizing Bgl II band of 2.4 kb in addition to the wild-type band of 4.1 kb, demonstrating the germline transmission of the targeted kappa locus. In order to generate mice homozygous for the mutation, heterozygotes were bred together and the kappa genotype of the offspring determined as described above. As expected, three genotypes were derived from the heterozygote matings: wild-type mice bearing two copies of a normal kappa locus, heterozygotes carrying one targeted copy of the kappa gene and one NT kappa gene, and mice homozygous for the kappa mutation. The deletion of kappa sequences from these latter mice was verified by hybridization of the Southern blots with a probe specific for Jκ (probe C, Fig. 20a). Whereas
hybridization of the Jκ probe was observed to DNA samples from heterozygous and wild-type siblings, no hybridizing signal was present in the homozygotes, attesting to the generation of a novel mouse strain in which both copies of the kappa locus have been inactivated by deletion as a result of targeted mutation.
EXAMPLE 10
Inactivation of the Mouse Heavy Chain Gene by Homologous
Recombination
This example describes the inactivation of the endogenous murine immunoglobulin heavy chain locus by
homologous recombination in embryonic stem (ES) cells. The strategy is to delete the endogenous heavy chain J segments by homologous recombination with a vector containing heavy chain sequences from which the JH region has been deleted and replaced by the gene for the selectable marker neo.
Construction of a heavy chain targeting vector
Mouse heavy chain sequences containing the JH region
(Fig. 21a) were isolated from a genomic phage library derived from the D3 ES cell line (Gossler et al., Proc. Natl. Acad. Sci. U.S.A. 83:9065-9069 (1986)) using a JH4 specific
oligonucleotide probe:
5'- ACT ATG CTA TGG ACT ACT GGG GTC AAG GAA CCT CAG TCA CCG -3' A 3.5 kb genomic SacI/StuI fragment, spanning the JH region, was isolated from a positive phage clone and subcloned into Sacl/Smal digested pUC18. The resulting plasmid was designated pUC18 JH. The neomycin resistance gene (neo), used for drug selection of transfected ES cells, was derived from a repaired version of the plasmid pGEM7 (KJ1). A report in the literature (Yenofsky et al. (1990) Proc. Natl. Acad. Sci.
(U.S.A. ) 87: 3435-3439) documents a point mutation the neo coding sequences of several commonly used expression vectors, including the construct pMCIneo (Thomas and Cappechi (1987) Cell 51: 503-512) which served as the source of the neo gene used in pGEM7 (KJ1). This mutation reduces the activity of the neo gene product and was repaired by replacing a
restriction fragment encompassing the mutation with the corresponding sequence from a wild-type neo clone. The
Hindlll site in the prepared pGEM7 (KJ1) was converted to a Sall site by addition of a synthetic adaptor, and the neo expression cassette excised by digestion with Xbal/Sall. The ends of the neo fragment were then blunted by treatment with the Klenow form of DNA poll, and the neo fragment was
subcloned into the Nael site of pUC18 JH, generating the plasmid pUC18 JH-neo (Fig. 21b) .
Further construction of the targeting vector was carried out in a derivative of the plasmid pGP1b. pGP1b was digested with the restriction enzyme NotI and ligated with the following oligonucleotide as an adaptor:
5'- GGC CGC TCG ACG ATA GCC TCG AGG CTA TAA ATC TAG AAG AAT
TCC AGC AAA GCT TTG GC -3'
The resulting plasmid, called pGMT, was used to build the mouse immunoglobulin heavy chain targeting
construct.
The Herpes Simplex Virus (HSV) thymidine kinase (TK) gene was included in the construct in order to allow for enrichment of ES clones bearing homologous recombinants, as described by Mansour et al. (Nature 336, 348-352 (1988)). The
HSV TK gene was obtained from the plasmid pGEM7 (TK) by digestion with EcoRI and Hindlll. The TK DNA fragment was subcloned between the EcoRI and Hindlll sites of pGMT, creating the plasmid pGMT-TK (Fig. 21c).
To provide an extensive region of homology to the target sequence, a 5.9 kb genomic Xbal/Xhol fragment, situated 5' of the JH region, was derived from a positive genomic phage clone by limit digestion of the DNA with Xhol, and partial digestion with Xbal. As noted in Fig. 21a, this Xbal site is not present in genomic DNA, but is rather derived from phage sequences immediately flanking the cloned genomic heavy chain insert in the positive phage clone. The fragment was
subcloned into Xbal/Xhol digested pGMT-TK, to generate the plasmid pGMT-TK-JH5' (Fig. 21d).
The final step in the construction involved the excision from pUC18 JH-neo of the 2.8 kb EcoRI fragment which contained the neo gene and flanking genomic sequences 3' of JH. This fragment was blunted by Klenow polymerase and subcloned into the similarly blunted Xhol site of
pGMT-TK-JH5'. The resulting construct, JHKO1 (Fig. 21e), contains 6.9 kb of genomic sequences flanking the JH locus, with a 2.3 kb deletion spanning the JH region into which has been inserted the neo gene. Fig. 21f shows the structure of an endogenous heavy chain gene after homologous recombination with the targeting construct.
EXAMPLE 11
Generation and analysis of targeted ES cells
AB-1 ES cells (McMahon and Bradley, Cell
62:1073-1085 (1990)) were grown on mitotically inactive
SNL76/7 cell feeder layers essentially as described
(Robertson, E.J. (1987) Teratocarcinomas and Embryonic Stem Cells: A Practical Approach. E.J. Robertson, ed. (Oxford: IRL Press), pp. 71-112). As described in the previous example, prior to electroporation of ES cells with the targeting
construct JHKO1, the pluripotency of the ES cells was
determined by generation of AB-1 derived chimeras which were shown capable of germline transmission of the ES genome. The heavy chain inactivation vector JHKO1 was digested with NotI and electroporated into AB-1 cells by the methods described (Hasty et al., Nature 350:243-246 (1991)). Electroporated cells were plated into 100 mm dishes at a density of 1-2 x 106 cells/dish. After 24 hours, G418
(200mg/ml of active component) and FIAU (0.5mM) were added to the medium, and drug-resistant clones were allowed to develop over 8-10 days. Clones were picked, trypsinized, divided into two portions, and further expanded. Half of the cells derived from each clone were then frozen and the other half analyzed for homologous recombination between vector and target
sequences.
DNA analysis was carried out by Southern blot hybridization. DNA was isolated from the clones as described (Laird et al. (1991) Nucleic Acids Res. 19: 4293), digested with StuI and probed with the 500 bp EcoRI/StuI fragment designated as probe A in Fig. 2If. This probe detects a StuI fragment of 4.7 kb in the wild-type locus, whereas a 3 kb band is diagnostic of homologous recombination of endogenous sequences with the targeting vector (see Fig. 21a and f). Of 525 G418 and FIAU doubly-resistant clones screened by Southern blot hybridization, 12 were found to contain the 3 kb fragment diagnostic of recombination with the targeting vector. That these clones represent the expected targeted events at the JH locus (as shown in Fig. 21f) was confirmed by further
digestion with Hindlll, Spel and Hpal. Hybridization of probe A (see Fig. 21f) to Southern blots of Hindlll, Spel, and Hpal digested DNA produces bands of 2.3 kb, >10 kb, and >10kb, respectively, for the wild-type locus (see Fig. 21a), whereas bands of 5.3 kb, 3.8 kb, and 1.9 kb, respectively, are
expected for the targeted heavy chain locus (see Fig 21f). All 12 positive clones detected by the StuI digest showed the predicted Hindlll, Spel, and Hpal bands diagnostic of a targeted JH gene. In addition, Southern blot analysis of a StuI digest of all 12 clones using a neo-specific probe (probe B, Fig. 21f) generated only the predicted fragment of 3 kb, demonstrating that the clones each contained only a single copy of the targeting vector. Generation of mice carrying the JH deletion
Three of the targeted ES clones described in the previous section were thawed and injected into C57BL/6J blastocysts as described (Bradley, A. (1987) in
Teratocarcinomas and Embryonic Stem Cells: A Practical
Approach, E.J. Robertson, ed. (Oxford: IRL Press), p.113-151) and transferred into the uteri of pseudopregnant females. The extent of ES cell contribution to the chimera was visually estimated from the amount of agouti coat coloration, derived from the ES cell line, on the black C57BL/6J background. Half of the offspring resulting from blastocyst injection of two of the targeted clones were chimeric (i.e., showed agouti as well as black pigmentation); the third targeted clone did not generate any chimeric animals. The majority of the chimeras showed significant (approximately 50 percent or greater) ES cell contribution to coat pigmentation. Since the AB-1 ES cells are an XY cell line, most of the chimeras were male, due to sex conversion of female embryos colonized by male ES cells. Males chimeras were bred with C57BL/6J females and the offspring monitored for the presence of the dominant agouti coat color indicative of germline transmission of the ES genome. Chimeras from both of the clones consistently
generated agouti offspring. Since only one copy of the heavy chain locus was targeted in the injected ES clones, each agouti pup had a 50 percent chance of inheriting the mutated locus. Screening for the targeted gene was carried out by Southern blot analysis of stul-digested DNA from tail
biopsies, using the probe utilized in identifying targeted ES clones (probe A, Fig. 21f). As expected, approximately 50 percent of the agouti offspring showed a hybridizing StuI band of approximately 3 kb in addition to the wild-type band of 4.7 kb, demonstrating germline transmission of the targeted CH gene segment.
In order to generate mice homozygous for the mutation, heterozygotes were bred together and the heavy chain genotype of the offspring determined as described above. As expected, three genotypes were derived from the heterozygote atings: wild-type mice bearing two copies of the normal JH locus, heterozygotes caring one targeted copy of the gene and one normal copy, and mice homozygous for the JH mutation. The absence of JH sequences from these latter mice was verified by hybridization of the Southern blots of Stul-digested DNA with a probe specific for JH (probe C, Fig. 21a). Whereas
hybridization of the JH probe to a 4.7 kb fragment in DNA samples from heterozygous and wild-type siblings was observed, no signal was present in samples from the JH-mutant
homozygotes, attesting to the generation of a novel mouse strain in which both copies of the heavy chain gene have been mutated by deletion of the JH sequences.
EXAMPLE 12
Heavy Chain Minilocus Transgene
A. Construction of plasmid vectors for cloning large DNA sequences
1. pGPla
The plasmid pBR322 was digested with EcoRI and Styl and ligated with the following oligonucleotides: oligo-42 5'- caa gag ccc gcc taa tga gcg ggc ttt ttt ttg cat act gcg gcc get -3'
oligo-43 5'- aat tag egg ccg cag tat gca aaa aaa age ccg etc att agg egg get -3'
The resulting plasmid, pGP1a, is designed for cloning very large DNA constructs that can be excised by the rare cutting restriction enzyme NotI. It contains a NotI restriction site downstream (relative to the ampicillin resistance gene, AmpR) of a strong transcription termination signal derived from the trpA gene (Christie et al., Proc.
Natl. Acad. Sci. USA 78:4180 (1981)). This termination signal reduces the potential toxicity of coding sequences inserted into the NotI site by eliminating readthrough transcription from the AmpR gene. In addition, this plasmid is low copy relative to the pUC plasmids because it retains the pBR322 copy number control region. The low copy number further reduces the potential toxicity of insert sequences and reduces the selection against large inserts due to DNA replication. The vectors pGP1b, pGP1c, pGP1d, and pGP1f are derived from pGP1a and contain different polylinker cloning sites. The polylinker sequences are given below pGP1a
NotI
GCGGCCGC
pGP1b
NotI Xhol Clal BamHI Hindlll NotI GCggccgcctcgagatcactatcgattaattaaggatccagcagtaagcttgcGGCCGC pGIlc
NotI Smal Xhol Sall Hindlll BamHI SacII NotI
GCggccgcatcccgggtctcgaggtcgacaagctttcgaggatccgcGGCCGC pGP1d
NotI Sall Hindlll Clal BamHI Xhol NotI
GCggccgctgtcgacaagcttatcgatggatcctcgagtgcGGCCGC pGP1f
NotI Sall Hindlll EcoRI Clal Kpnl BamHI Xhol NotI GCggccgctgtcgacaagcttcgaattcagatcgatgtggtacctggatcctcgagtgcGGCC Each of these plasmids can be used for the construction of large transgene inserts that are excisable with NotI so that the transgene DNA can be purified away from vector sequences prior to microinjection. 2. pGP1b
pGP1a was digested with NotI and ligated with the following oligonucleotides: oligo-47 5'- ggc cgc aag ctt act get gga tec tta att aat cga tag tga tct cga ggc -3'
oligo-48 5'- ggc cgc etc gag ate act ate gat taa tta agg ate cag cag taa get tgc -3' The resulting plasmid, pGP1b, contains a short polylinker region flanked by NotI sites. This facilitates the construction of large inserts that can be excised by NotI digestion.
3. pGPe
The following oligonucleotides: oligo-44 5'- etc cag gat cca gat ate agt ace tga aac agg get tgc -3'
oligo-45 5'- etc gag cat gca cag gac ctg gag cac aca cag cct tec -3' were used to amplify the immunoglobulin heavy chain 3'
enhancer (S. Petterson, et al., Nature 344:165-168 (1990)) from rat liver DNA by the polymerase chain reaction technique.
The amplified product was digested with BamHI and SphI and cloned into BamHI/SphI digested pNN03 (pNN03 is a pUC derived plasmid that contains a polylinker with the following restriction sites, listed in order: NotI, BamHI, Ncol, Clal, EcoRV, Xbal, Sad, Xhol, SphI, PstI, Bglll, EcoRI, Smal, Kpnl, Hindlll, and NotI). The resulting plasmid, pRE3, was digested with BamHI and Hindlll, and the insert containing the rat Ig heavy chain 3' enhancer cloned into BamHI/Hindlll digested pGP1b. The resulting plasmid, pGPe (Fig. 22 and Table 1), contains several unique restriction sites into which sequences can be cloned and subsequently excised together with the 3' enhancer by NotI digestion.
Figure imgf000092_0001
B. Construction of IgM expressing minilocus transgene. pIGM1
1. Isolation of J-μ constant region clones and construction of pJM1
A human placental genomic DNA library cloned into the phage vector XEMBL3/SP6/T7 (Clonetech Laboratories, Inc., Palo Alto, CA) was screened with the human heavy chain J region specific oligonucleotide: oligo-1 5'- gga ctg tgt ccc tgt gtg atg ctt ttg atg tct ggg gcc aag -3' and the phage clone λ1.3 isolated. A 6 kb Hindlll/Kpnl fragment from this clone, containing all six J segments as well as D segment DHQ52 and the heavy chain J-μ intronic enhancer, was isolated. The same library was screened with the human μ specific oligonucleotide: oligo-2 5'- cac caa gtt gac ctg cct ggt cac aga cct gac cac eta tga -3' and the phage clone λ2.1 isolated. A 10.5 kb Hindlll/Xhol fragment, containing the μ switch region and all of the μ constant region exons, was isolated from this clone. These two fragments were ligated together with Kpnl/Xhol digested pNN03 to obtain the plasmid pJMl. 2. pJM2
A 4 kb Xhol fragment was isolated from phage clone λ2.1 that contains sequences immediately downstream of the sequences in pJMl, including the so called ∑μ element involved in (S-associated deleteon of the μ in certain IgD expressing B-cells (Yasui et al., Eur. J. Immunol. 19:1399 (1989), which is incorporated herein by reference). This fragment was treated with the Klenow fragment of DNA polymerase I and ligated to Xhol cut, Klenow treated, pJMl. The resulting plasmid, pJM2 (Fig. 23), had lost the internal Xhol site but retained the 3' Xhol site due to incomplete reaction by the Klenow enzyme. pJM2 contains the entire human J region, the heavy chain J-μ intronic enhancer, the μ switch region and all of the μ constant region exons, as well as the two 0.4 kb direct repeats, σμ and ∑μ, involved in δ-associated deletion of the μ gene. 3. Isolation of D region clones and construction of pDH1
The following human D region specific
oligonucleotide: oligo-4 5'- tgg tat tac tat ggt teg ggg agt tat tat aac cac agt gtc -3' was used to screen the human placenta genomic library for D region clones. Phage clones λ4.1 and λ4.3 were isolated. A 5.5 kb Xhol fragment, that includes the D elements Dκ1, DN1, and DM2 (Ichihara et al., EMBO J. 1:4141 (1988)), was isolated from phage clone λ4.1. An adjacent upstream 5.2 kb Xhol fragment, that includes the D elements DLR1, DXP1, DXP'1, and DA1, was isolated from phage clone λ4.3. Each of these D region Xhol fragments were cloned into the Sall site of the plasmid vector pSP72 (Promega, Madison, WI) so as to destroy the Xhol site linking the two sequences. The upstream
fragment was then excised with Xhol and Smal, and the
downstream fragment with EcoRV and Xhol. The resulting isolated fragments were ligated together with Sall digested pSP72 to give the plasmid pDH1. pDH1 contains a 10.6 kb insert that includes at least 7 D segments and can be excised with Xhol (5') and EcoRV (3'). 4. pCOR1
The plasmid pJM2 was digested with Asp718 (an isoschizomer of Kpnl) and the overhang filled in with the Klenow fragment of DNA polymerase I. The resulting DNA was then digested with Clal and the insert isolated. This insert was ligated to the XhoI/EcoRV insert of pDH1 and Xhol/Clal digested pGPe to generate pCORl (Fig. 24).
5. pVH251
A 10.3 kb genomic Hindlll fragment containing the two human heavy chain variable region segments VH251 and VH105 (Humphries et al., Nature 331:446 (1988), which is
incorporated herein by reference) was subcloned into pSP72 to give the plasmid pVH251. 6. plGM1
The plasmid pCOR1 was partially digested with Xhol and the isolated Xhol/Sail insert of pVH251 cloned into the upstream Xhol site to generate the plasmid pIGMl (Fig. 25). pIGM1 contains 2 functional human variable region segments, at least 8 human D segments all 6 human JH segments, the human
J-μ enhancer, the human σμ element, the human μ switch region, all of the human μ coding exons, and the human ∑μ element, together with the rat heavy chain 3' enhancer, such that all of these sequence elements can be isolated on a single
fragment, away from vector sequences, by digestion with NotI and microinjected into mouse embryo pronuclei to generate transgenic animals.
C. Construction of IgM and IgG expressing minilocus
transgene, pHC1
1. Isolation of γ constant region clones
The following oligonucleotide, specific for human Ig g constant region genes: oligo-29 5'- cag cag gtg cac ace caa tgc cca tga gcc cag aca ctg gac -3' was used to screen the human genomic library. Phage clones 129.4 and λ29.5 were isolated. A 4 kb Hindlll fragment of phage clone λ29.4, containing a γ switch region, was used to probe a human placenta genomic DNA library cloned into the phage vector lambda FIX™ II (Stratagene, La Jolla, CA). Phage clone λSg1.13 was isolated. To determine the subclass of the different γ clones, dideoxy sequencing reactions were carrred out using subclones of each of the three phage clones as templates and the following oligonucleotide as a primer: oligo-67 5'- tga gcc cag aca ctg gac -3' Phage clones λ29.5 and λSγ1.13 were both determined to be of the γ1 subclass.
2. pγe1
A 7.8 kb Hindlll fragment of phage clone λ29.5, containing the γ1 coding region was cloned into pUC18. The resulting plasmid, pLT1, was digested with Xhol, Klenow treated, and religated to destroy the internal Xhol site. The resulting clone, pLTlxk, was digested with Hindlll and the insert isolated and cloned into pSPγ2 to generate the plasmid clone pLTlxks. Digestion of pLT1xks at a polylinker Xhol site and a human sequence derived BamHI site generates a 7.6 kb fragment containing the γ1 constant region coding exons. This 7.6 kb XhoI/BamHI fragment was cloned together with an
adjacent downstream 4.5 kb BamHI fragment from phage clone λ29.5 into XhoI/BamHI digested pGPe to generate the plasmid clone pγe1. pγe1 contains all of the 71 constant region coding exons, together with 5 kb of downstream sequences, linked to the rat heavy chain 3' enhancer.
3. pγe2
A 5.3 kb Hindlll fragment containing the 71 switch region and the first exon of the pre-switch sterile transcript (P. Sideras et al. (1989) International Immunol. 1, 631) was isolated from phage clone XSγ1.13 and cloned into pSP72 with the polylinker Xhol site adjacent to the 5' end of the insert, to generate the plasmid clone pSγ1S. The Xhol/Sall insert of pSγ1S was cloned into Xhol digested pγe1 to generate the plasmid clone pγe2 (Fig. 26). pγe2 contains all of the 71 constant region coding exons, and the upstream switch region and sterile transcript exons, together with 5 kb of downstream sequences, linked to the rat heavy chain 3' enhancer. This clone contains a unique Xhol site at the 5' end of the insert. The entire insert, together with the Xhol site and the 3' rat enhancer can be excised from vector sequences by digestion with NotI. 4 . pHC1
The plasmid pIGMl was digested with Xhol and the 43 kb insert isolated and cloned into Xhol digested pge2 to generate the plasmid pHC1 (Fig. 25). pHC1 contains 2
functional human variable region segments, at least 8 human D segments all 6 human JH segments, the human J-μ enhancer, the human σμ element, the human μ switch region, all of the human μ coding exons, the human ∑μ element, and the human γl constant region, including the associated switch region and sterile transcript associated exons, together with the rat heavy chain 3' enhancer, such that all of these sequence elements can be isolated on a single fragment, away from vector sequences, by digestion with NotI and microinjected into mouse embryo pronuclei to generate transgenic animals.
D. Construction of IgM and IgG expressing minilocus
transgene, pHC2
1. Isolation of human heavy chain V region gene VH49.8
The human placental genomic DNA library lambda, FIX™ II, Stratagene, La Jolla, CA) was screened with the following human VH1 family specific oligonucleotide: oligo-49 5'- gtt aaa gag gat ttt att cac ccc tgt gtc etc tec aca ggt gtc -3'
Phage clone X49.8 was isolated and a 6.1 kb Xbal fragment containing the variable segment VH49.8 subcloned into pNN03 (such that the polylinker Clal site is downstream of VH49.8 and the polylinker Xhol site is upstream) to generate the plasmid pVH49.8. An 800 bp region of this insert was sequenced, and VH49.8 found to have an open reading frame and intact splicing and recombination signals, thus indicating that the gene is functional (Table 2).
Figure imgf000098_0001
2. pV2
A 4 kb Xbal genomic fragment containing the human VHIV family gene VH4-21 (Sanz et al., EMBO J., 8:3741 (1989)), subcloned into the plasmid pUC12, was excised with Smal and Hindlll, and treated with the Klenow fragment of polymerase I. The blunt ended fragment was then cloned into Clal digested, Klenow treated, pVH49.8. The resulting plasmid, pV2, contains the human heavy chain gene VH49.8 linked upstream of VH4-21 in the same orientation, with a unique Sall site at the 3' end of the insert and a unique Xhol site at the 5' end.
3. pS-γ1-5'
A 0.7 kb Xbal/Hindlll fragment (representing
sequences immediately upstream of, and adjacent to, the 5.3 kb γ1 switch region containing fragment in the plasmid pγe2) together with the neighboring upstream 3.1 kb Xbal fragment were isolated from the phage clone λSg1.13 and cloned into Hindlll/Xbal digested pUC18 vector. The resulting plasmid, pSγ1-5', contains a 3.8 kb insert representing sequences upstream of the initiation site of the sterile transcript found in B-cells prior to switching to the γ1 isotype (P.
Sideras et al., International Immunol. 1:631 (1989)). Because the transcript is implicated in the initiation of isotype switching, and upstream cis-acting sequences are often
important for transcription regulation, these sequences are included in transgene constructs to promote correct expression of the sterile transcript and the associated switch
recombination. 4. pVGE1
The pSγ1-5' insert was excised with Smal and
Hindlll, treated with Klenow enzyme, and ligated with the following oligonucleotide linker: 5'- ccg gtc gac egg -3'
The ligation product was digested with Sall and ligated to Sall digested pV2. The resulting plasmid, pVP, contains 3.8 kb of γ1 switch 5' flanking sequences linked downstream of the two human variable gene segments VH49.8 and VH4-21 (see Table 2). The pVP insert is isolated by partial digestion with Sall and complete digestion with Xhol, followed by purification of the 15 kb fragment on an agarose gel. The insert is then cloned into the Xhol site of pγe2 to generate the plasmid clone pVGE1 (Fig. 27). pVGEl contains two human heavy chain variable gene segments upstream of the human γ1 constant gene and associated switch region. A unique Sall site between the variable and constant regions can be used to clone in D, J, and μ gene segments. The rat heavy chain 3' enhancer is linked to the 3' end of the γ1 gene and the entire insert is flanked by NotI sites. 5. pHC2
The plasmid clone pVGEl is digested with Sall and the Xhol insert of pIGMl is cloned into it. The resulting clone, pHC2 (Fig. 25), contains 4 functional human variable region segments, at least 8 human D segments all 6 human JH segments, the human J-m enhancer, the human σμ element, the human μ switch region, all of the human μ coding exons, the human∑μ element, and the human γ1 constant region, including the associated switch region and sterile transcript associated exons, together with 4 kb flanking sequences upstream of the sterile transcript initiation site. These human sequences are linked to the rat heavy chain 3' enhancer, such that all of the sequence elements can be isolated on a single fragment, away from vector sequences, by digestion with NotI and
microinjected into mouse embryo pronuclei to generate
transgenic animals. A unique Xhol site at the 5' end of the insert can be used to clone in additional human variable gene segments to further expand the recombinational diversity of this heavy chain minilocus. E. Transgenic mice
The NotI inserts of plasmids pIGM1 and pHC1 were isolated from vector sequences by agarose gel electrophoresis. The purified inserts were microinjected into the pronuclei of fertilized (C57BL/6 x CBA) F2 mouse embryos and transferred the surviving embryos into pseudopregnant females as described by Hogan et al. (B. Hogan, F. Costantini, and E. Lacy, Methods of Manipulating the Mouse Embryo, 1986, Cold Spring Harbor
Laboratory, New York). Mice that developed from injected embryos were analyzed for the presence of transgene sequences by Southern blot analysis of tail DNA. Transgene copy number was estimated by band intensity relative to control standards containing known quantities of cloned DNA. At 3 to 8 weeks of age, serum was isolated from these animals and assayed for the presence of transgene encoded human IgM and IgG1 by ELISA as described by Harlow and Lane (E. Harlow and D. Lane.
Antibodies: A Laboratory Manual, 1988, Cold Spring Harbor Laboratory, New York). Microtiter plate wells were coated with mouse monoclonal antibodies specific for human IgM (clone AF6, #0285, AMAC, Inc. Westbrook, ME) and human IgG1 (clone JL512, #0280, AMAC, Inc. Westbrook, ME). Serum samples were serially diluted into the wells and the presence of specific immunoglobulins detected with affinity isolated alkaline phosphatase conjugated goat anti-human Ig (polyvalent) that had been pre-adsorbed to minimize cross-reactivity with mouse immunoglobulins. Table 3 and Fig. 28 show the results of an ELISA assay for the presence of human IgM and IgG1 in the serum of two animals that developed from embryos injected with the transgene insert of plasmid pHC1. All of the control non- transgenic mice tested negative for expression of human IgM and IgG1 by this assay. Mice from two lines containing the pIGM1 NotI insert (lines #6 and 15) express human IgM but not human IgG1. We tested mice from 6 lines that contain the pHC1 insert and found that 4 of the lines (lines #26, 38, 57 and 122) express both human IgM and human IgG1, while mice from two of the lines (lines #19 and 21) do not express detectable levels of human immunoglobulins. The pHC1 transgenic mice that did not express human immunoglobulins were so-called Go mice that developed directly from microinjected embryos and may have been mosaic for the presence of the transgene.
Southern blot analysis indicates that many of these mice contain one or fewer copies of the transgene per cell. The detection of human IgM in the serum of pIGMl transgenics, and human IgM and IgG1 in pHC1 transgenics, provides evidence that the transgene sequences functicn correctly in directing VDJ joining, transcription, and isotyp switching. One of the animals (#18) was negative for the transgene by Southern blot analysis, and showed no detectable levels of human IgM or IgG1. The second animal (#38) contained approximately 5 copies of the transgene, as assayed by Southern blotting, and showed detectable leve's of both human IgM and IgG1. The results of ELISA assays for 11 animals that developed from transgene injected embryos is summarized in the table below (Table 3).
TABLE 3
Detection of human IgM and IgG1 in the serum of transgenic animals by ELISA assay
Figure imgf000102_0001
Table 3 shows a correlation between the presence of integrated transgene DNA and the presence of transgene encoded immunoglobulins in the serum. Two of the animals that were found to contain the pHC1 transgene did not express detectable levels of human immunoglobulins. These were both low copy animals and may not have contained complete copies of the transgenes, or the animals may have been genetic mosaics
(indicated by the <1 copy per cell estimated for animal #21), and the transgene containing cells may not have populated the hematopoietic lineage. Alternatively, the transgenes may have integrated into genomic locations that are not conducive to their expression. The detection of human IgM in the serum of pIGMl transgenics, and human IgM and IgG1 in pHC1 transgenics, indicates that the transgene sequences function correctly in directing VDJ joining, transcription, and isotype switching.
F. cDNA clones
To assess the functionality of the pHC1 transgene in VDJ joining and class switching, as well the participation of the transgene encoded human B-cell receptor in B-cell
development and allelic exclusion, the structure of
immunoglobulin cDNA clones derived from transgenic mouse spleen mRNA were examined. The overall diversity of the transgene encoded heavy chains, focusing on D and J segment usage, N region addition, CDR3 length distribution, and the frequency of joints resulting in functional mRNA molecules was examined. Transcripts encoding IgM and IgG incorporating VH105 and VH251 were examined.
Polyadenylated RNA was isolated from an eleven week old male second generation line-57 pHC1 transgenic mouse.
This RNA was used to synthesize oligo-dT primed single
stranded cDNA. The resulting cDNA was then used as template for four individual PCR amplifications using the following four synthetic oligonucleotides as primers: VH251 specific oligo-149, eta get cga gtc caa gga gtc tgt gcc gag gtg cag ctg (g,a,t,c); VH105 specific o-150, gtt get cga gtg aaa ggt gtc cag tgt gag gtg cag ctg (g,a,t,c); human gamma1 specific oligo-151, ggc get cga gtt cca cga cac cgt cac egg ttc; and human mu specific oligo-152, cct get cga ggc age caa egg cca cgc tgc teg. Reaction 1 used primers 0-149 and o-151 to amplify VH251-gammal transcripts, reaction 2 used o-149 and o- 152 to amplify VH251-mu transcripts, reaction 3 used o-150 and o-151 to amplify VH105-gammal transcripts, and reaction 4 used o-150 and o-152 to amplify VH105-mu transcripts. The
resulting 0.5 kb PCR products were isolated from an agarose gel; the μ transcript products were more abundant than the γ transcript products, consistent with the corresponding ELISA data (Fig. 34). The PCR products were digested with Xhol and cloned into the plasmid pNN03. Double-stranded plasmid DNA was isolated from minipreps of nine clones from each of the four PCR amplifications and dideoxy sequencing reactions were performed. Two of the clones turned out to be deletions containing no D or J segments. These could not have been derived from normal RNA splicing products and are likely to have originated from deletions introduced during PCR
amplification. One of the DNA samples turned out to be a mixture of two individual clones, and three additional clones did not produce readable DNA sequence (presumably because the DNA samples were not clean enough). The DNA sequences of the VDJ joints from the remaining 30 clones are compiled in Table 4. Each of the sequences are unique, indicating that no single pathway of gene rearrangement, or single clone of transgene expressing B-cells is dominant. The fact that no two sequences are alike is also an indication of the large diversity of immunoglobulins that can be expressed from a compact minilocus containing only 2 V segments, 10 D segments, and 6 J segments. Both of the V segments, all six of the J segments, and 7 of the 10 D segments that are included in the transgene are used in VDJ joints. In addition, both constant region genes (mu and gammal) are incorporated into
transcripts. The VH105 primer turned out not to be specific for VH105 in the reactions performed. Therefore many of the clones from reactions 3 and 4 contained VH251 transcripts.
Additionally, clones isolated from ligated reaction 3 PCR product turned out to encode IgM rather than IgG; however this may reflect contamination with PCR product from reaction 4 as the DNA was isolated on the same gel. An analogous
experiment, in which immunoglobulin heavy chain sequences were amplified from adult human peripheral blood lymphocytes (PBL), and the DNA sequence of the VDJ joints determined, was
recently reported by Yamada et al. (J . Exp. Med. 173 :395-407 (1991), which is incorporated herein by reference). We compared the data from human PBL with our data from the pHC1 transgenic mouse.
Figure imgf000106_0001
G. J segment choice
Table 5 compared the distribution of J segments incorporated into pHC1 transgene encoded transcripts to J segments found in adult human PBL immunoglobulin transcripts. The distribution profiles are very similar, J4 is the dominant segment in both systems, followed by J6. J2 is the least common segment in human PBL and the transgenic animal.
Figure imgf000107_0001
H. D segment choice
49% (40 of 82) of the clones analyzed by Yamada et al. incorporated D segments that are included in the pHC1 transgene. An additional 11 clones contained seguences that were not assigned by the authors to any of the known D
segments. Two of these 11 unassigned clones appear to be derived from an inversion of the DIR2 segments which is included in the pHC1 construct. This mechanism, which was predicted by Ichihara et al. (EMBO J. 7:4141 (1988)) and observed by Sanz (J. Immunol. 147:1720-1729 (1991)), was not considered by Yamada et al. (J. Exp. Med. 173:395-407 (1991)). Table 5 is a comparison of the D segment distribution for the pHC1 transgenic mouse and that observed for human PBL
transcripts by Yamada et al. The data of Yamada et al. was recompiled to include DIR2 use, and to exclude D segments that are not in the pHC1 transgene. Table 6 demonstrates that the distribution of D segment incorporation is very similar in the transgenic mouse and in human PBL. The two dominant human D segments, DXP'1 and DN1, are also found with high frequency in the transgenic mouse. The most dramatic dissimilarity between the two distributions is the high frequency of DHQ52 in the transgenic mouse as compared to the human. The high frequency of DHQ52 is reminiscent of the D segment distribution in the human fetal liver. Sanz has observed that 14% of the heavy chain transcripts contained DHQ52 sequences. If D segments not found in pHC1 are excluded from the analysis, 31% of the fetal transcripts analyzed by Sanz contain DHQ52. This is comparable to the 27% that we observe in the pHC1 transgenic mouse.
Figure imgf000108_0001
I. Functionality of VDJ joints
Table 7 shows the predicted amino acid sequences of the VDJ regions from 30 clones that were analyzed from the pHC1 transgenic. The translated sequences indicate that 23 of the 30 VDJ joints (77%) are in-frame with respect to the variable and J segments.
Figure imgf000109_0001
J. CDR3 length distribution
Table 8 compared the length of the CDR3 peptides from transcripts with in-frame VDJ joints in the pHC1
transgenic mouse to those in human PBL. Again the human PBL data comes from Yamada et al. The profiles are similar with the transgenic profile skewed slightly toward smaller CDR3 peptides than observed from human PBL. The average length of CDR3 in the transgenic mouse is 10.3 amino acids. This is substantially the same as the average size reported for authentic human CDR3 peptides by Sanz (J. Immunol. 147:1720- 1729 (1991)).
Figure imgf000110_0001
EXAMPLE 13
Rearranged Heavy Chain Transgenes
A. Isolation of Rearranged Human Heavy Chain VDJ segments.
Two human leukocyte genomic DNA libraries cloned into the phage vector XEMBL3/SP6/T7 (Clonetech Laboratories, Inc., Palo Alto, CA) are screened with a 1 kb Pacl/Hindlll fragment of λ1.3 containing the human heavy chain J-μ intronic enhancer. Positive clones are tested for hybridization with a mixture of the following VH specific oligonucleotides: oligo-7 5'-tea gtg aag gtt tec tgc aag gca tct gga tac ace ttc acc-3' oligo-8 5'-tec ctg aga etc tec tgt gca gcc tct gga ttc ace ttc acc'3' Clones that hybridized with both V and J-μ probes are isolated and the DNA seguence of the rearranged VDJ segment determined. B. Construction of rearranged human heavy chain transgenes
Fragments containing functional VJ segments (open reading frame and splice signals) are subcloned into the plasmid vector pSP72 such that the plasmid derived Xhol site is adjacent to the 5' end of the insert seguence. A subclone containing a functional VDJ segment is digested with Xhol and Pad (Pad, a rare-cutting enzyme, recognizes a site near the J-m intronic enhancer), and the insert cloned into Xhol/Pad digested pHC2 to generate a transgene construct with a
functional VDJ segment, the J-μ intronic enhancer, the μ switch element, the μ constant region coding exons, and the γ1 constant region, including the sterile transcript associated sequences, the γ1 switch, and the coding exons. This
transgene construct is excised with NotI and microinjected into the pronuclei of mouse embryos to generate transgenic animals as described above.
EXAMPLE 14
Light Chain Transgenes
A. Construction of Plasmid vectors
1. Plasmid vector pGP1c
Plasmid vector pGP1a is digested with NotI and the following oligonucleotides ligated in: oligo-81 5'-ggc cgc ate ccg ggt etc gag gtc gac aag ctt teg agg ate cgc-3' oligo-82 5'-ggc cgc gga tec teg aaa get tgt cga cct cga gac ccg gga tgc-3' The resulting plasmid, pGP1c, contains a polylinker with Xmal, Xhol, Sall, Hindlll, and BamHI restriction sites flanked by NotI sites. 2. Plasmid vector pGP1d
Plasmid vector pGP1a is digested with NotI and the following oligonucleotides ligated in: oligo-87 5'-ggc cgc tgt cga caa get tat cga tgg ate etc gag tgc -3' oligo-88 5'-ggc cgc act cga gga tec ate gat aag ctt gtc gac age -3'
The resulting plasmid, pGP1d, contains a polylinker with Sall, Hindlll, Clal, BamHI, and Xhol restriction sites flanked by NotI sites. B. Isolation of Jκ and Cκ clones
A human placental genomic DNA library cloned into the phage vector λEMBL3/SP6/T7 (Clonetech Laboratories, Inc., Palo Alto, CA) was screened with the human kappa light chain J region specific oligonucleotide: oligo-36 5'- cac ctt egg cca agg gac aeg act gga gat taa acg taa gca -3' and the phage clones 136.2 and 136.5 isolated. A 7.4 kb Xhol fragment that includes the Jκ1 segment was isolated from
136.2 and subcloned into the plasmid pNN03 to generate the plasmid ulone p36.2. A neighboring 13 kb Xhol fragment that includes Jk segments 2 through 5 together with the Cκ gene segment was isolated from phage clone 136.5 and subcloned into the plasmid pNN03 to generate the plasmid clone p36.5.
Together these two clones span the region beginning 7.2 kb upstream of Jκ1 and ending 9 kb downstream of Cκ.
C. Construction of rearranged light chain transgenes
1. pCK1, a Cκ vector for expressing rearranged variable segments
The 13 kb Xhol insert of plasmid clone p36.5 containing the Cκ gene, together with 9 kb of downstream sequences, is cloned into the Sall site of plasmid vector pGP1c with the 5' end of the insert adjacent to the plasmid Xhol site. The resulting clone, pCK1 can accept cloned fragments containing rearranged VJκ segments into the unique 5' Xhol site. The transgene can then be excised with NotI and purified from vector sequences by gel electrophoresis. The resulting transgene construct will contain the human J-Cκ intronic enhancer and may contain the human 3' κ enhancer. 2. pCK2, a Cκ vector with heavy chain enhancers for
expressing rearranged variable segments
A 0.9 kb Xbal fragment of mouse genomic DNA containing the mouse heavy chain J-μ intronic enhancer (J. Banerji et al., Cell 33:729-740 (1983)) was subcloned into pUC18 to generate the plasmid pJH22 . 1. This plasmid was linearized with SphI and the ends filled in with Klenow enzyme. The Klenow treated DNA was then digested with Hindlll and a 1.4 kb Mlul/Hindlll fragment of phage clone λ1.3
(previous example), containing the human heavy chain J-μ intronic enhancer (Hayday et al., Nature 307:334-340 (1984)), to it. The resulting plasmid, pMHEl, consists of the mouse and human heavy chain J-μ intronic enhancers ligated together into pUC18 such that they are excised on a single
BamHI/Hindlll fragment. This 2.3 kb fragment is isolated and cloned into pGP1c to generate pMHE2. pMHE2 is digested with Sall and the 13 kb Xhol insert of p36.5 cloned in. The resulting plasmid, pCK2, is identical to pCK1, except that the mouse and human heavy chain J-μ intronic enhancers are fused to the 3' end of the transgene insert. To modulate expression of the final transgene, analogous constructs can be generated with different enhancers, i.e. the mouse or rat 3' kappa or heavy chain enhancer (Meyer and Neuberger, EMBO J..
8:1959-1964 (1989); Petterson et al., Nature. 344:165-168 (1990)). 3. Isolation of rearranged kappa light chain variable segments
Two human leukocyte genomic DNA libraries cloned into the phage vector λEMBL3/SP6/T7 (Clonetech Laboratories, Inc., Palo Alto, CA) were screened with the human kappa light chain J region containing 3.5 kb Xhol/Smal fragment of p36.5. Positive clones were tested for hybridization with the
following Vκ specific oligonucleotide: oligo-65 5'-agg ttc agt ggc agt ggg tct ggg aca gac ttc act etc ace ate age-3'
Clones that hybridized with both V and J probes are isolated and the DNA sequence of the rearranged VJκ segment determined.
4. Generation of transgenic mice containing rearranged human light chain constructs.
Fragments containing functional VJ segments (open reading frame and splice signals) are subcloned into the unique Xhol sites of vectors pCK1 and pCK2 to generate
rearranged kappa light chain transgenes. The transgene constructs are isolated from vector sequences by digestion with NotI. Agarose gel purified insert is microinjected into mouse embryo pronuclei to generate transgenic animals.
Animals expressing human kappa chain are bred with heavy chain minilocus containing transgenic animals to generate mice expressing fully human antibodies.
Because not all VJK combinations may be capable of forming stable heavy-light chain complexes with a broad spectrum of different heavy chain VDJ combinations, several different light chain transgene constructs are generated, each using a different rearranged VJk clone, and transgenic mice that result from these constructs are bred with heavy chain minilocus transgene expressing mice. Peripheral blood, spleen, and lymph node lymphocytes are isolated from double transgenic (both heavy and light chain constructs) animals, stained with fluorescent antibodies specific for human and mouse heavy and light chain immunoglobulins (Pharmingen, San Diego, CA) and analyzed by flow cytometry using a FACScan analyzer (Becton Dickinson, San Jose, CA). Rearranged light chain transgenes constructs that result in the highest level of human heavy/light chain complexes on the surface of the highest number of B cells, and do not adversely affect the immune cell compartment (as assayed by flow cytometric
analysis with B and T cell subset specific antibodies), are selected for the generation of human monoclonal antibodies. D. Construction of unrearranged light chain minilocus
transgenes
1. pJCKl, a JK, Cκ containing vector for constructing
minilocus transgenes
The 13 kb CK containing Xhol insert of p36.5 is treated with Klenow enzyme and cloned into Hindlll digested, Klenow-treated, plasmid pGPld. A plasmid clone is selected such that the 5' end of the insert is adjacent to the vector derived Clal site. The resulting plasmid, p36.5-ld, is digested with Clal and Klenow-treated. The Jκl containing 7.4 kb Xhol insert of p36.2 is then Klenow-treated and cloned into the Clal, Klenow-treated p36.5-ld. A clone is selected in which the p36.2 insert is in the same orientation as the p36.5 insert. This clone, pJCKl (Fig. 34), contains the entire human JK region and Cκ, together with 7.2 kb of upstream sequences and 9 kb of downstream sequences. The insert also contains the human J-Cκ intronic enhancer and may contain a human 3' κ enhancer. The insert is flanked by a unique 3' Sall site for the purpose of cloning additional 3' flanking sequences such as heavy chain or light chain enhancers. A unique Xhol site is located at the 5' end of the insert for the purpose of cloning in unrearranged VK gene segments. The unique Sall and Xhol sites are in turn flanked by NotI sites that are used to isolate the completed transgene construct away from vector sequences. 2. Isolation of unrearranged Vκ gene segments and generation of transgenic animals expressing human Ig light chain protein
The Vκ specific oligonucleotide, cligo-65 (discussed above), is used to probe a human placental genomic DNA library cloned into the phage vector 1EMBL3/3P6/T7 (Clonetech
Laboratories, Inc., Palo Alto, CA). Variable gene segments from the resulting clones are sequenced, and clones that appear functional are selected. Criteria for judging
functionality include: open reading frames, intact splice acceptor and doncr sequences, and intact recombination
sequence. DNA fragments containing selected variable gene segments are cloned into the unique Xhol site of plasmid pJCK1 to generate minilocus constructs. The resulting clones are digested with NotI and the inserts isolated and injected into mouse embryo pronuclei to generate transgenic animals. The transgenes of these animals will undergo V to J joining in developing B-cells. Animals expressing human kappa chain are bred with heavy chain minilocus containing transgenic animals to generate mice expressing fully human antibodies.
EXAMPLE 15
Genomic Heavy Chain Human Ig Transgene
This Example describes the cloning of a human genomic heavy chain immunoglobulin transgene which is then introduced into the murine germline via microinjection into zygotes or integration in ES cells.
Nuclei are isolated from fresh human placental tissue as described by Marzluff, W.F., et al. (1985),
Transcription and Translation: A Practical Approach, B.D.
Hammes and S.J. Higgins, eds., pp. 89-129, IRL Press, Oxford). The isolated nuclei (or PBS washed human spermatocytes) are embedded in 0.5% low melting point agarose blocks and lyse with 1 mg/ml proteinase K in 500mM EDTA, 1% SDS for nuclei, or with lmg/ml proteinase K in 500mM EDTA, 1% SDS, lOmM DTT for spermatocytes at 50°C for 18 hours. The proteinase K is inactivated by incubating the blocks in 40μg/ml PMSF in TE for 30 minutes at 50°C, and then washing extensively with TE. The DNA is then digested in the agarose with the restriction enzyme NotI as described by M. Finney in Current Protocols in Molecular Biology (F. Ausubel et al., eds. John Wiley & Sons, Supp. 4, 1988, e.g., Section 2.5.1).
The NotI digested DNA is then fractionated by pulsed field gel electrophoresis as described by Anand et al., Nuc. Acids Res. 17:3425-3433 (1989). Fractions enriched for the NotI fragment are assayed by Southern hybridization to detect one or more of the sequences encoded by this fragment. Such sequences include the heavy chain D segments, J segments, and γI constant regions together with representatives of all 6 VH families (although this fragment is identified as 670 kb fragment from HeLa cells by Berman et al. (1988), supra., we have found it to be an 830 kb fragment from human placental and sperm DNA). Those fractions containing this NotI
fragment are ligated into the NotI cloning site of the vector pYACNN as described (McCormick et al., Technique 2: 65-71
(1990)). Plasmid pYACNN is prepared by digestion of pYACneo (Clontech) with EcoRI and ligation in the presence of the oligonucleotide 5' - AAT TGC GGC CGC - 3'.
YAC clones containing the heavy chain NotI fragment are isolated as described by Traver et al., Proc. Natl. Acad. Sci. USA. 86:5898-5902 (1989). The cloned NotI insert is isolated from high molecular weight yeast DNA by pulse field gel electrophoresis as described by M. Finney, op. cit. The DNA is condensed by the addition of 1 mM spermine and
microinjected directly into the nucleus of single cell embryos previously described. Alternatively, the DNA is isolated by pulsed field gel electrophoresis and introduced into ES cells by lipofection (Gnirke et al., EMBO J. 10:1629-1634 (1991)), or the YAC is introduced into ES cells by spheroplast fusion.
EXAMPLE 16
Discontinuous Genomic Heavy Chain Ig Transgene
An 85 kb Spel fragment of human genomic DNA,
containing VH6, D segments, J segments, the μ constant region and part of the γ constant region, has been isolated by YAC cloning essentially as described in Example 1. A YAC carrying a fragment from the germline variable region, such as a 570 kb NotI fragment upstream of the 830 kb NotI fragment
Figure imgf000118_0001
described above containing multiple copies of V1 through V5 is isolated as described. (Berman et al. (198P), supra, detected two 570 kb NotI fragments, each containing multiple V
segments.) The two fragments are coinjected into the nucleus of a mouse single cell embryo as described in Example 1.
Typically, coinjection of two different DNA fragments result in the integration of both fragments at the same insertion site within the chromosome. Therefore, approximately 50% of the resulting transgenic animals that contain at least one copy of each of the two fragments will have the V segment fragment inserted upstream of the constant region containing fragment. Of these animals, about 50% will carry out V to DJ joining by DNA inversion and about 50% by deletion, depending on the orientation of the 570 kb NotI fragment relative to the position of the 85 kb Spel fragment. DNA is isolated from resultant transgenic animals and those animals found to be containing both transgenes by Southern blot hybridization (specifically, those animals containing both multiple human V segments and human constant region genes) are tested for their ability to express human
immunoglobulin molecules in accordance with standard
techniques. EXAMPLE 17
Identification of functionally rearranged variable region sequences in transgenic B cells
An antigen of interest is used to immunize (see
Harlow and Lane, Antibodies: A Laboratory Manual. Cold Spring Harbor, New York (1988)) a mouse with the following genetic traits: homozygosity at the endogenous having chain locus for a deletion of JH (Examples 10); hemizygous for a single copy of unrearranged human heavy chain minilocus transgene
(examples 5 and 14); and hemizygous for a single copy of a rearranged human kappa light chain transgene (Examples 6 and 14).
Following the schedule of immunization, the spleen is removed, and spleen cells used to generate hybridomas. Cells from an individual hybridoma clone that secretes
antibodies reactive with the antigen of interest are used to prepare genomic DNA. A sample of the genomic DNA is digested with several different restriction enzymes that recognize unique six base pair sequences, and fractionated on an agarose gel. Southern blot hybridization is used to identify two DNA fragments in the 2-10 kb range, one of which contains the single copy of the rearranged human heavy chain VDJ sequences and one of which contains the single copy of the rearranged human light chain VJ sequence. These two fragments are size fractionated on agarose gel and cloned directly into pUC18. The cloned inserts are then subcloned respectively into heavy and light chain expression cassettes that contain constant region sequences.
The plasmid clone pγel (Example 12) is used as a heavy chain expression cassette and rearranged VDJ sequences are cloned into the Xhol site. The plasmid clone pCK1 is used as a light chain expression cassette and rearranged VJ
sequences are cloned into the Xhol site. The resulting clones are used together to transfect SP0 cells to produce antibodies that react with the antigen of interest (Co. et al., Proc. Natl. Acad. Sci. USA 88:2869 (1991), which is incorporated herein by reference).
Alternatively, mRNA is isolated from the cloned hybridoma cells described above, and used to synthesize cDNA. The expressed human heavy and light chain VDJ and VJ sequence are then amplified by PCR and cloned (Larrick et al., Biol. Technology, 7:934-938 (1989)). After the nucleotide sequence of these clones has been determined, oligonucleotides are synthesized that encode the same polypeptides, and synthetic expression vectors generated as described by Queen et al., Proc. Natl. Acad. Sci. USA., 84: 5454-5458 (1989).
Immunization of Transgenic Animals with Complex Antigens
The following experiment demonstrates that transgenic animals can be successfully immunized with complex antigens such as those on human red blood cells and respond with kinetics that are similar to the response kinetics observed in normal mice.
Blood cells generally are suitable immunogens and comprise many different types of antigens on the surface of red and white blood cells.
Immunization with human blood
Tubes of human blood from a single donor were collected and used to immunize transgenic mice having
functionally disrupted endogenous heavy chain loci (JHD) and harboring a human heavy chain minigene construct (HCI); these mice are designated as line 112. Blood was washed and resuspended in 50 mis Hanks' and diluted to 1x108 cells/ml 0.2 mis (2x107 cells) were then injected interperitoneally using a 28 gauge needle and 1 ce syringe. This immunization protocol was repeated approximately weekly for 6 weeks. Serum titers were monitored by taking blood from retro-orbital bleeds and collecting serum and later testing for specific antibody. A pre-immune bleed was also taken as a control. On the very last immunization, three days before these animals were sacrificed for serum and for hybridomas, a single immunization of 1 x 107 cells was given intravenously through the tail to enhance the production of hybridomas.
Figure imgf000120_0001
Mice # 2343 and 2348 have a desired phenotype: human heavy chain mini-gene transgenic on heavy chain knock-out
background. Generation of Hybridomas
Hybridomas were generated by fusing mouse spleen cells of approximately 16 week-old transgenic mice (Table 9) that had been immunized as described (supra) to a fusion partner consisting of the non-secreting HAT-sensitive myeloma cell line, X63 Ag8.653. Hybridoma clones were cultivated and hybridoma supernatants containing immunoglobulins having specific binding affinity for blood cell antigens were
identified, for example, by flow cytometry. Flow cytometry
Serum and hybridoma supernatants were tested using flow cytometry. Red blood cells from the donor were washed 4X in Hanks' balanced salt solution and 50,000 cells were placed in 1.1 ml polypropylene microtubes. Cells were incubated with antisera or supernatant from the hybridomas for 30 minutes on ice in staining media (lx RPMI 1640 media without phenol red or biotin (Irvine Scientific) 3% newborn calf serum, 0.1% Na azide). Controls consisted of littermate mice with other genotypes. Cells were then washed by centrifugation at 4°C in Sorvall RT600B for 5-10 minutes at 1000 rpm. Cells were washed two times and then antibody detected on the cell surface with a fluorescent developing reagent. Two monoclonal reagents were used to test. One was a FITC-labeled mouse anti-human μ heavy chain antibody (Pharmagen, San Diego, CA) and the other was a PE-labeled rat anti-mouse kappa light chain (Becton-Dickenson, San Jose, CA). Both of these
reagents gave similar results. Whole blood (red blood cells and white blood cells) and white blood cells alone were used as target cells. Both sets gave positive results.
Serum of transgenic mice and littermate controls was incubated with either red blood cells from the donor, or white blood cells from another individual, washed and then developed with anti-human IgM FITC labeled antibody and analyzed in a flow cytometer. Results showed that serum from mice that are transgenic for the human mini-gene locus (mice 2343 and 2348) show human IgM reactivity whereas all littermate animals (2344, 2345, 2346, 2347) do not. Normal mouse serum (NS) and phosphate buffer saline (PBS) were used as negative controls. Red blood cells were ungated and white blood cells were gated to include only lymphocytes. Lines are drawn on the x and y axis to provide a reference. Flow cytometry was performed on 100 supernatants from fusion 2348. Four supernatants showed positive reactivity for blood cell antigens.
EXAMPLE 18
Reduction of Endogenous Mouse Immunoglobulin Expression by Antisense RNA
A. Vector for Expression of Antisense Ig Sequences
1. Construction of the cloning vector pGP1h
The vector pGP1b (referred to in a previous example) is digested with Xhol and BamHI and ligated with the following oligonucleotides: 5'- gat cct cga gac cag gta cca gat ctt gtg aat teg -3'
5'- teg acg aat tea caa gat ctg gta cct ggt etc gag -3' to generate the plasmid pGP1h. This plasmid contains a polylinker that includes the following restriction sites:
NotI, EcoRI, Bglll, Asp718, Xhol, BamHI, Hindlll, NotI.
Construction of pBCEl.
A 0.8 kb Xbal/Bglll fragment of pVH251 (referred to in a previous example), that includes the promoter leader sequence exon, first intron, and part of the second exon of the human VH-V family immunoglobulin variable gene segment, was inserted into Xbal/Bglll digested vector pNN03 to generate the plasmid pVH251.
The 2.2 kb BamHI/EcoRI DNA fragment that includes the coding exons of the human growth hormone gene (hGH;
Seeburg, (1982) DNA 1:239-249) is cloned into Bglll/EcoRI digested pGHlh. The resulting plasmid is digested with BamHI and the BamHI/Bglll of pVH251N is inserted in the same
orientation as the bGH gene to generate the plasmid pVhgh.
A 0.9 kb Xbal fragment of mouse genomic DNA containing the mouse heavy chain J-μ intronic enhancer
(Banerji et al., (1983) Cell 33:729-740) was subcloned into pUClδ to generate the plasmid pJH22.1. This plasmid was linearized with SphI and the ends filled in with klenow enzyme. The klenow treated DNA was then digested with Hindlll and a 1.4 kb Mlul (klenow) /Hindlll fragment of phage clone λ1.3 (previous example), containing the human heavy chain J-μ intronic enhancer (Hayday et al., (1984) Nature 307:334-340), to it. The resulting plasmid, pMHE1, consists of the mouse and human heavy chain J-μ intron enhancers ligated together into pUC18 such that they can be excised on a single
BamHI/Hindlll fragment.
The BamHI/Hindlll fragment of pMHEl is cloned into BamHI/Hindlll cut pVhgh to generate the B-cell expression vector pBCE1. This vector, depicted in Fig. 36, contains unique Xhol and Asp718 cloning sites into which antisense DNA fragments can be cloned. The expression of these antisense sequences is driven by the upstream heavy chain promoter- enhancer combination the downstream hGH gene sequences provide polyadenylation sequences in addition to intron sequences that promote the expression of transgene constructs. Antisense transgene constructs generated from pBCEl can be separated from vector sequences by digestion with NotI.
B. An IgM antisense transgene construct.
The following two oligonucleotides: 5'- cgc ggt ace gag agt cag tec ttc cca aat gtc -3'
5'- cgc etc gag aca get gga atg ggc aca tgc aga -3' are used as primers for the amplification of mouse IgM
constant region sequences by polymerase chain reaction (PCR) using mouse spleen cDNA as a substrate. The resulting 0.3 kb PCR product is digested with Asp7l8 and Xhol and cloned into Asp718/XhoI digested pBCE1 to generate the antisense transgene construct pMASl. The purified NotI insert of pMAS1 is microinjected into the pronuclei of half day mouse embryos-- alone or in combination with one or more other transgene constructs--to generate transgenic mice. This construct expresses an RNA transcript in B-cells that hybridizes with mouse IgM mRNA, thus down-regulating the expression of mouse IgM protein. Double transgenic mice containing pMAS1 and a human heavy chain transgene minilocus such as pHC1 (generated either by coinjection of both constructs or by breeding of singly transgenic mice) will express the human transgene encoded Ig receptor on a higher percentage of B-cell than mice transgenic for the human heavy chain minilocus alone. The ratio of human to mouse Ig receptor expressing cells is due in part to competition between the two populations for factors and cells that promoter B-cell differentiation and expansion. Because the Ig receptor plays a key role in B-cell
development, mouse Ig receptor expressing B-cells that express reduced levels of IgM on their surface (due to mouse Ig specific antisense down-regulation) during B-cell development will not compete as well as cells that express the human receptor.
C. An IgKappa antisense transgene construct.
The following two oligonucleotides:
5'- cgc ggt ace get gat get gca cca act gta tec -3'
5'- cgc etc gag eta aca etc att cct gtt gaa get -3' are used as primers for the amplification of mouse IgKappa constant region sequences by polymerase chain reaction (PCR) using mouse spleen cDNA as a substrate. The resulting 0.3 kb PCR product is digested with Asp718 and Xhol and cloned into Asp718/XhoI digested pBCE1 to generate the antisense transgene construct pKAS1. The purified NotI insert of pKAS1 is
microinjected into the pronuclei of half day mouse embryos-- alone or in combination with one or more other transgene constructs--to generate transgenic mice. This construct expresses an RNA transcript in B-cells that hybridizes with mouse IgK mRNA, thus down-regulating the expression of mouse IgK protein as described above for pMAS1.
EXAMPLE 19
This example demonstrates the successful immunization and immune response in a transgenic mouse of the present invention.
Immunization of Mice
Keyhole limpet hemocyanin conjugated with greater than 400 dinitrophenyl groups per molecule (Calbiochem, La Jolla, California) (KLH-DNP) was alum precipitated according to a previously published method (Practical Immunology, L. Hudson and F.C. Hay, Blackwell Scientific (Pubs.), p. 9, 1980). Four hundred μg of alum precipitated KLH-DNP along with 100 μg dimethyldioctadecyl Ammonium Bromide in 100 μL of phosphate buffered saline (PBS) was injected intraperitoneally into each mouse. Serum samples were collected six days later by retro-orbital sinus bleeding.
Analysis of Human Antibody Reactivity in Serum
Antibody reactivity and specificity were assessed using an indirect enzyme-linked immunosorbent assay (ELISA). Several target antigens were tested to analyze antibody induction by the immunogen. Keyhole limpet hemocyanin
(Calbiochem) was used to identify reactivity against the protein component, bovine serum albumin-DNP for reactivity against the hapten and/or modified amino groups, and KLH-DNP for reactivity against the total immunogen. Human antibody binding to antigen was detected by enzyme conjugates specific for IgM and IgG sub-classes with no cross reactivity to mouse immunoglobulin. Briefly, PVC microtiter plates were coated with antigen drying overnight at 37°C of 5 μg/mL protein in PBS. Serum samples diluted in PBS, 5% chicken serum, 0.5% Tween-20 were incubated in the wells for 1 hour at room temperature, followed by anti-human IgG Fc and IgG F(ab')- horseradish peroxidase or anti-human IgM Fc-horseradish peroxidase in the same diluent. After 1 hour at room temperature enzyme activity was assessed by addition of ABTS substrate (Sigma, St. Louis, Missouri) and read after 30 minutes at 415-490 nm. Human Heavy Chain Participation in Immune Response in
Transgenic Mice
Figure 37 illustrates the response of three mouse iittermates to immunization with KLH-DNP. Mouse number 1296 carried the human IgM and IgG unrearranged transgene and was homozygous for mouse Ig heavy chain knockout. Mouse number
1299 carried the transgene on a non-knockout background, while mouse 1301 inherited neither of these sets of genes. Mouse 1297, another littermate, carried the human transgene and was hemizygous with respect to mouse heavy chain knockout. It was included as a non-immunized control.
The results demonstrate that both human IgG and IgM responses were developed to the hapten in the context of conjugation to protein. Human IgM also developed to the KLH molecule, but no significant levels of human IgG were present at this time point. In pre-immunization serum samples from the same mice, titers of human antibodies to the same target antigens were insignificant.
EXAMPLE 20
This example demonstrates the successful
immunization with a human antigen and immune response in a transgenic mouse of the present invention, and provides data demonstrating that nonrandom somatic mutation occurs in the variable region sequences of the human transgene. Demonstration of antibody responses comprising human
immunoglobulin heavy chains against a human glycoprotein antigen
Transgenic mice used for the experiment were homozygous for functionally disrupted murine immunoglobulin heavy chain loci produced by introduction of a transgene at the joining (J) region (supra) resulting in the absence of functional endogenous (murine) heavy chain production. The transgenic mice also harbored at least one complete
unrearranged human heavy chain mini-locus transgene, (HCl, supra), which included a single functional VH gene (VH251), human μ constant region gene, and human γ1 constant region gene. Transgenic mice shown to express human immunoglobulin transgene products (supra) were selected for immunization with a human antigen to demonstrate the capacity of the transgenic mice to make an immune response against a human antigen immunization. Three mice of the HCl-26 line and three mice of the HCl-57 line (supra) were injected with human antigen.
One hundred μg of purified human carcinoembryonic antigen (CEA) insolubilized on alum was injected in complete Freund's adjuvant on Day 0, followed by further weekly
injections of alum-precipitated CEA in incomplete Freund's adjuvant on Days 7, 14, 21, and 28. Serum samples were collected by retro-orbital bleeding on each day prior to injection of CEA. Equal volumes of serum were pooled from each of the three mice in each group for analysis.
Titres of human μ chain-containing immunoglobulin and human γ chain-containing immunoglobulin which bound to human CEA immobilized on microtitre wells were determined by ELISA assay. Results of the ELISA assays for human μ chain- containing immunoglobulins and human γ chain-containing immmunoglbulins are shown in Figs. 38 and 39, respectively. Significant human μ chain Ig titres were detected for both lines by Day 7 and were observed to rise until about Day 21. For human γ chain Ig, significant titres were delayed, being evident first for line HCl-57 at Day 14, and later for line HCl-26 at Day 21. Titres for human γ chain Ig continued to show an increase over time during the course of the
experiment. The observed human μ chain Ig response, followed by a plateau, combined with a later geveloping γ chain
response which continues to rise is characteristic of the pattern seen with affinity maturation. Analysis of Day 21 samples showed lack of reactivity to an unrelated antigen, keyhole limpet hemocyanin (KLC), indicating that the antibody response was directed against CEA in a specific manner. These data indicate that animals transgenic for human unrearranged immunoglobulin gene loci: (1) can respond to a human antigen (e.g., the human glycoprotein, CEA), (2) can undergo isotype switching ("class switching) as
exemplified by the observed μ to γ class switch, and (3) exhibit characteristics of affinity maturation in their humoral immune responses. In general, these data indicate: (1) the human Ig transgenic mice have the ability to induce heterologous antibody production in response to a defined antigen, (2) the capacity of a single transgene heavy chain variable region to respond to a defined antigen, (3) response kinetics over a time period typical of primary and secondary response development, (4) class switching of a transgeneencoded humoral immune response from IgM to IgG, and (5) the capacity of transgenic animal to produce human-sequence antibodies against a human antigen.
Demonstration of somatic mutation in a human heavy chain transgene minilocus.
Line HCl-57 transgenic mice, containing multiple copies of the HCl transgene, were bred with immunoglobulin heavy chain deletion mice to obtain mice that contain the HCl transgene and contain disruptions at both alleles of the endogenous mouse heavy chain (supra). These mice express human mu and gammal heavy chains together with mouse kappa and lambda light chains (supra). One of these mice was
hyperimmunized against human carcinoembryonic antigen by repeated intraperitoneal injections over the course of 1.5 months. This mouse was sacrificed and lymphoid cells isolated from the spleen, inguinal and mesenteric lymph nodes, and peyers patches. The cells were combined and total RNA
isolated. First strand cDNA was synthesized from the RNA and used as a template for PCR amplification with the following 2 oligonucleotide primers:
149 5' -eta get cga gtc caa gga gtc tgt gcc gag gtg cag ctg
(g/a/t/c)-3'
151 5' -ggc get cga gtt cca cga cac cgt cac egg ttc-3' These primers specifically amplify VH251/gammal cDNA sequences. The amplified sequences were digested with Xhol and cloned into the vector pNN03. DNA sequence from the inserts of 23 random clones is shown in Fig. 40; sequence variations from germline sequence are indicated, dots indicate sequence is identical to germline. Comparison of the cDNA sequences with the germline sequence of the VH251 transgene reveals that 3 of the clones are completely unmutated, while the other 20 clones contain somatic mutations. One of the 3 non-mutated sequences is derived from an out-of-frame VDJ joint. Observed somatic mutations at specific positions of occur at similar frequencies and in similar distribution patterns to those observed in human lymphocytes (Cai et al. (1992) J. Exp. Med. 176: 1073, incorporated herein by
reference). The overall frequency of somatic mutations is approximately 1%; however, the frequency goes up to about 5% within CDRl, indicating selection for amino acid changes that affect antigen binding. This demonstrates antigen driven affinity maturation of the human heavy chain sequences.
EXAMPLE 21
This example demonstrates the successful formation of a transgene by co-introduction of two separate
polynucleotides which recombine to form a complete human light chain minilocus transgene.
Generation of an unrearranged light chain minilocus transgene by co-injection of two overlapping DNA fragments
1. Isolation of unrearranged functional Vκ gene segments
vk65.3, vk65.5, vk65.8 and vk65.15
The Vκ specific oligonucleotide, oligo-65 (5'-agg ttc agt ggc agt ggg tct ggg aca gac ttc act etc ace ate agc- 3'), was used to probe a human placental genomic DNA library cloned into the phage vector XEMBL3/SP6/T7 (Clonetech
Laboratories, Inc., Palo Alto, CA). DNA fragments containing Vκ segments from positive phage clones were subcloned into plasmid vectors. Variable gene segments from the resulting clones are sequenced, and clones that appear functional were selected. Criteria for judging functionality include: open reading frames, intact splice acceptor and donor sequences, and intact recombination sequence. DNA sequences of 4 functional Vκ gene segments (vk65.3, vk65.5, vk65.8, and vk65.15) from 4 different plasmid clones isolated by this procedure are shown in Figs. 41-44. The four plasmid clones, p65.3f, p65.5gl, p65.8, and p65.15f, are described- below.
(1 a) p65.3f
A 3 kb Xba fragment of phage clone λ65.3 was subcloned into pUC19 so that the vector derived Sall site was proximal to the 3' end of the insert and the vector derived BamHI site 5'. The 3 kb BamHI/Sall insert of this clone was subcloned into pGP1f to generate p65.3f.
(1 b) p65.5gl
A 6.8 kb EcoRI fragment of phage clone λ65.5 was subcloned into pGP1f so that the vector derived Xhol site is proximal to the 5' end of the insert and the vector derived Sall site 3'. The resulting plasmid is designated p65.5gl.
(1 c) p65.8
A 6.5 kb Hindlll fragment of phage clone λ65.8 was cloned into pSP72 to generate p65.8.
(1 d) p65.15f
A 10 kb EcoRI fragment of phage clone λ65.16 was subcloned into pUC18 to generate the plasmid p65.15.3. The Vκ gene segment within the plasmid insert was mapped to a 4.6 kb EcoRI/Hindlll subfragment, which was cloned into pGP1f. The resulting clone, p65.15f, has unique Xhol and Sall sites located at the respective 5' and 3' ends of the insert.
2. pKV4
The Xhol/Sall insert of p65.8 was cloned into the
Xhol site of p65.15f to generate the plasmid pKV2. The
Xhol/Sall insert of p65.5gl was cloned into the Xhol site of pKV2 to generate pKV3. The Xhol/Sall insert of pKV3 was cloned into the Xhol site of p65.3f to generate the plasmid pKV4. This plasmid contains a single 21 kb Xhol/Sall insert that includes 4 functional Vκ gene segments. The entire insert can also be excised with NotI.
3. pKClB
(3 a) pKcor
Two Xhol fragments derived from human genomic DNA phage λ clones were subcloned into plasmid vectors. The first, a 13 kb Jκ2-Jκ5/Cκ containing fragment, was treated with Klenow enzyme and cloned into Hindlll digested, Klenow
treated, plasmid pGP1d. A plasmid clone (pK-31) was selected such that the 5' end of the insert is adjacent to the vector derived Clal site. The second Xhol fragment, a 7.4 kb piece of DNA containing Jκ1 was cloned into XhoI/Sall-digested pSP72, such that the 3' insert Xhol site was destroyed by ligation to the vector Sall site. The resulting clone, p36.2s, includes an insert derived Clal site 4.5 kb upstream of Jκ1 and a polylinker derived Clal site downstream in place of the naturally occurring Xhol site between Jκ1 and Jκ2. This clone was digested with Clal to release a 4.7 kb fragment which was cloned into Clal digested pK-31 in the correct 5' to 3' orientation to generate a plasmid containing all 5 human Jκ segments, the human intronic enhancer human Cκ, 4.5 kb of 5' flanking sequence, and 9 kb of 3' flanking sequence. This plasmid, pKcor, includes unique flanking Xhol and Sall sites on the respective 5' and 3' sides of the insert.
(3 b) pKcorB
A 4 kb BamHI fragment containing the human 3' kappa enhancer (Judde, J.-G. arid Max, E.E. (1992) Mol. Cell. Biol. 12: 5206, incorporated herein by reference) was cloned into pGP1f such that the 5' end is proximal to the vector Xhol site. The resulting plasmid, p24Bf, was cut with Xhol and the 17.7 kb Xhol/Sall fragment of pKcor cloned into it in the same orientation as the enhancer fragment. The resulting plasmid, pKcorB, includes unique Xhol and Sall sites at the 5' and 3' ends of the insert respectively. (3 c) pKClB
The Xhol/Sall insert of pKcorB was cloned into the Sall site of p65.3f to generate the light-chain minilocus- transgene plasmid pKClB. This plasmid includes a single functional human Vκ segment, all 5 human Jκ segments, the human intronic enhancer, human Cκ, and the human 3' kappa enhancer. The entire 25 kb insert can be isolated by NotI digestion.
4. Co4
Th two NotI inserts from plasmids pKV4 and pKClB were mixed at a concentration of 2.5 μg/ml each in
microinjection buffer, and co-injected into the pronuclei of half day mouse embryos as described in previous examples.
Resulting transgenic animals contain transgene inserts
(designated Co4, product of the recombination shown in Fig. 45) in which the two fragments co-integrated. The 3' 3 kb of the pKV4 insert and the 5'3 kb of the pKClB insert are
identical. Some of the integration events will represent homologous recombinations between the two fragments over the 3 kb of shared sequence. The Co4 locus will direct the
expression of a repertoire of human sequence light chains in a transgenic mouse.
The foregoing description of the preferred embodiments of the present invention has been presented for purposes of illustration and description. They are not
intended to be exhaustive or to limit the invention to the precise form disclosed, and many modifications and variations are possible in light of the above teaching.
All publications and patent applications herein are incorporated by reference to the same extent as if each
individual publication or patent application was specifically and individually indicated to be incorporated by reference.
Although the present invention has been described in some detail by way of illustration for purposes of clarity of understanding, it will be apparent that certain changes and modifications may be practiced within the scope of the claims.

Claims

WHAT IS CLAIMED IS: TRANSGENE CLAIMS
1. An isolated immunoglobulin heavy chain
transgene that is expressed in B cells of a transgenic nonhuman animal containing at least one integrated copy of a polynucleotide comprising a DNA sequence of the formula:
(VH)x-(D)y-(JH)z-(SD)m-(C1)n-[(T)-(SA)p-(C2)]q wherein x, y, z, m, n, p, and q are integers and x is 2-100, n is 1-10, y is 2-50, p is 1-10, z is 1-50, g is 0-50, and m is 0-10.
2. A transgene of Claim 1, wherein said polynucleotide comprises at least one heterologous D gene segment that can be incorporated into a functionally
rearranged V-D-J sequence.
3. A transgene of Claim 2, wherein said heterologous D gene segment contains at least one human D gene.
4. A transgene of Claim 1, wherein said polynucleotide comprises a human μ CH gene segment and a human γ1 CH gene segment.
5. A transgene of Claim 1, wherein q is at least 1, m is at least 1, n is at least 1, and said polynucleotide comprises at least about 50 basepairs of a segment immediately upstream of a germline switch sequence.
6. A transgene of Claim 5, wherein said polynucleotide comprises about 200 basepairs of sequence immediately upstream of a human germline γ1 switch sequence.
7. A transgene of Claim 1, wherein a SD segment is a γ1 switch sequence.
8. A transgene of Claim 1, wherein said
polynucleotide comprises about 200 basepairs naturally upstream of a human germline γ1 switch sequence, and wherein said 200 basepairs are operably linked to a human γ1 switch sequence.
9. A transgene of Claim 1, wherein said D region comprises only heterologous D genes.
10. A transgene of Claim 9, wherein said D region comprises only human D genes.
11. A transgene of Claim 1, wherein the polynucleotide is functionally rearranged in vivo to produce a rearranged V-D-J gene segment that contains a recognizable D region gene sequence.
12. A transgenic non-human animal comprising a transgene of Claim 1 in the germline of said non-human animal.
13. A transgenic non-human animal of Claim 12, wherein said transgene is rearranged.
14. A transgenic non-human animal of Claim 12, wherein said transgene is unrearranged.
15. A transgenic non-human animal of Claim 14, wherein said B cells produce a heterologous antibody.
16. A transgenic non-human animal of Claim 15, wherein said B cells produce a population of heterologous antibodies of more than one isotype.
17. A transgenic non-human animal of Claim 12, wherein said transgene encodes VH, D, JH, and CH regions.
18. A transgenic non-human animal of Claim 12 wherein said non-human animal is a rodent.
19. A transgenic non-human animal of Claim 12, wherein serum of said animal comprises human antibodies that contain a recognizable D region gene sequence.
20. A transgenic non-human animal of Claim 12, wherein at least one lymphocyte of said animal contains a mRNA encoding a heterologous immunoglobulin chain.
21. A transgenic animal of claim 20, wherein said mRNA contains a recognizable D region gene sequence.
22. A transgenic animal of claim 21, wherein said mRNA contains a functionally rearranged V-D-J sequence.
23. A transgenic animal of claim 12, wherein said transgenic animal comprises heterologous antibodies which comprise a human-sequence μ chain and which specifically bind to an antigen.
24. A transgenic animal of claim 23, further comprising heterologous antibodies which comprise a human- sequence γ chain and which specifically bind to an antigen.
25. A transgenic animal of claim 24, wherein the antigen is a human antigen.
26. A transgenic animal of claim 25, wherein the human antigen is CEA or a human blood cell antigen.
27. A transgenic nonhuman animal comprising a human transgene of claim 1, wherein the transgenic animal further comprises lymphoid tissue containing a population of mRNA species having somatic mutations clustered in CDR regions of a variable region encoded by an immunoglobulin transgene.
28. A transgenic nonhuman animal of claim 27, wherein the immunoglobulin transgene is a heavy chain
minilocus corresponding to HCI.
29. A hybridoma comprising a transgenic non-human B cell fused with a second cell capable of immortalizing said B cell, wherein said hybridoma produces a monoclonal antibody heterologous to said non-human animal.
30. A hybridoma of Claim 29 wherein the heterologous antibody comprises a human heavy chain containing a recognizable D region gene sequence.
31. A hybridoma of Claim 29 wherein said B cell is of murine origin.
32. A hybridoma of Claim 29 wherein the monoclonal antibody binds to a human antigen with an affinity of at least 1 x 107 M-1.
33. A human monoclonal antibody produced by a hybridoma of Claim 29.
34. A transgenic non-human animal having serum comprising detectable heterologous antibodies and having at least one suppressed endogenous immunoglobulin locus.
35. A method for producing heterologous
immunoglobulins from a transgenic nonhuman animal, the animal having a genome comprising germline copies of at least one transgene of Claim 1, the method comprising:
suppressing an endogenous immunoglobulin locus;
contacting the animal with a preselected antigen; and
collecting said heterologous immunoglobulins.
36. A method according to Claim 35, wherein suppression is produced by an antisense polynucleotide.
37. A method according to Claim 36, wherein the antisense polynucleotide is transcribed from an integrated antisense transgene.
38. A method according to Claim 37, wherein
transcription of the antisense transgene produces a transcript containing an antisense sequence linked to a second sequence.
39. A method according to Claim 37, wherein the non-human animal has a genome comprising germline copies of at least one light chain immunoglobulin transgene.
40. A method for suppressing at least one endogenous immunoglobulin locus in a transgenic non-human animal, comprising the steps of:
introducing an antisense transgene into a non-human animal to produce a non-human transgenic animal bearing an antisense transgene;
transcribing antisense RNA from the antisense transgene in vivo;
hybridizing the antisense RNA to a polynucleotide containing an endogenous immunoglobulin sequence; and
inhibiting expression of an endogenous
immunoglobulin chain.
41. A method of Claim 40, wherein said antisense transgene contains a nucleotide sequence that is homologous to an endogenous kappa chain immunoglobulin gene sequence.
42. A method of Claim 40, wherein said antisense transgene contains a nucleotide sequence that is homologous to an endogenous heavy chain immunoglobulin gene sequence.
43. A method for inactivating an endogenous
immunoglobulin gene, comprising the steps of:
integrating a targeting vector into an endogenous immunoglobulin gene; and
selecting for a cell bearing an integrated targeting vector.
44. A method according to Claim 43, further
comprising the step of: generating a line of non-human animals bearing a copy of the integrated targeting vector.
45. A method according to Claim 43, wherein the endogenous immunoglobulin gene is a light chain gene.
46. A method according to Claim 45, wherein the light chain gene is a kappa light chain gene.
47. A method according to Claim 43, wherein the endogenous immunoglobulin gene is a heavy chain gene.
48. A method according to Claim 43, wherein the endogenous immunoglobulin gene is a murine immunoglobulin gene.
49. A method according to Claim 44, wherein the non-human animal is a mouse.
50. A method according to Claim 49, wherein the mouse further comprises a human immunoglobulin transgene of Claim 1.
51. A method according to Claim 43, wherein the cell is an embryonic stem cell.
52. A method of generating a non-human animal having an inactivated endogenous immunoglobulin gene, comprising the steps of:
breeding a line of non-human animals generated by the method of Claim.44; and
identifying individual non-human animal offspring that are homozygous for an inactivated immunoglobulin gene.
53. A method according to Claim 52, wherein said offspring have an inactivated heavy chain immunoglobulin gene and at least one inactivated light chain immunoglobulin gene.
54. A transgenic non-human animal with an
inactivated endogenous immunoglobulin gene.
55. A non-human animal of Claim 54, wherein an integrated targeting vector is present in germline DNA.
56. A non-human animal of Claim 55, wherein a light chain immunoglobulin gene is inactivated.
57. A non-human animal of Claim 55, wherein a heavy chain immunoglobulin gene is inactivated.
58. A non-human animal of Claim 55 which is
homozygous for at least one inactivated endogenous
immunoglobulin gene.
59. An antisense transgene comprising a nucleotide sequence that is complementary to a polynucleotide seguence that is substantially identical to an immunoglobulin gene sequence.
60. A transgenic non-human animal bearing an antisense transgene of Claim 59.
61. A transgenic non-human animal comprising a functionally disrupted endogenous heavy chain locus and a heterologous immunoglobulin heavy chain transgene, wherein said animal makes an antibody response following immunization with an antigen.
62. A transgenic noh-human animal of Claim 61, wherein said functionally disrupted endogenous heavy chain locus is a JH region homologous recombination knockout, said heterologous immunoglobulin heavy chain transgene is the HCI human minigene transgene, and said antigen is a human antigen.
63. A transgenic non-human animal of Claim 61, wherein the antibody response comprises a population of antibodies which comprise human μ chain-containing
immunoglobulins and human γ chain-containing immunoglobulins.
64. A transgenic non-human animal of Claim 63, wherein the heterologous antibodies comprise a population of heterologous immunoglobulins which comprise somatic mutation in the variable regions which cluster in the CDRs.
65. A transgenic non-human animal of Claim 63, wherein the antigen is selected from the group consisting of human blood cell surface antigens, KLH, and human CEA.
PCT/US1992/010983 1990-08-29 1992-12-17 Transgenic non-human animals capable of producing heterologous antibodies WO1993012227A1 (en)

Priority Applications (8)

Application Number Priority Date Filing Date Title
JP5511191A JPH07503132A (en) 1991-12-17 1992-12-17 Transgenic non-human animals capable of producing xenoantibodies
EP93901143A EP0746609A4 (en) 1991-12-17 1992-12-17 Transgenic non-human animals capable of producing heterologous antibodies
CA002124967A CA2124967C (en) 1991-12-17 1992-12-17 Transgenic non-human animals capable of producing heterologous antibodies
US08/053,131 US5661016A (en) 1990-08-29 1993-04-26 Transgenic non-human animals capable of producing heterologous antibodies of various isotypes
US08/544,404 US5770429A (en) 1990-08-29 1995-10-10 Transgenic non-human animals capable of producing heterologous antibodies
US11/009,840 US20060015949A1 (en) 1990-08-29 2004-12-10 Transgenic non-human animals for producing heterologous and chimeric antibodies
US11/009,873 US7501552B2 (en) 1991-08-28 2004-12-10 Transgenic non-human animals for producing chimeric antibodies
US11/009,769 US20060026703A1 (en) 1990-08-29 2004-12-10 Transgenic non-human animals for producing heterologous and chimeric antibodies

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US07/810,279 US5569825A (en) 1990-08-29 1991-12-17 Transgenic non-human animals capable of producing heterologous antibodies of various isotypes
US07/810,279 1991-12-17
US07/853,408 1992-03-18
US07/853,408 US5789650A (en) 1990-08-29 1992-03-18 Transgenic non-human animals for producing heterologous antibodies
US90406892A 1992-06-23 1992-06-23
US07/904,068 1992-06-23

Publications (1)

Publication Number Publication Date
WO1993012227A1 true WO1993012227A1 (en) 1993-06-24

Family

ID=27420056

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1992/010983 WO1993012227A1 (en) 1990-08-29 1992-12-17 Transgenic non-human animals capable of producing heterologous antibodies

Country Status (5)

Country Link
EP (1) EP0746609A4 (en)
JP (2) JPH07503132A (en)
AU (1) AU3328493A (en)
CA (1) CA2124967C (en)
WO (1) WO1993012227A1 (en)

Cited By (755)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0754225A1 (en) * 1993-04-26 1997-01-22 GenPharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
EP0817835A1 (en) * 1995-03-29 1998-01-14 Xenotech Incorporated Production of antibodies using cre-mediated site-specific recombination
EP0822830A1 (en) * 1995-04-27 1998-02-11 Abgenix, Inc. Human antibodies derived from immunized xenomice
EP0854917A1 (en) * 1995-10-10 1998-07-29 GenPharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
US5827690A (en) * 1993-12-20 1998-10-27 Genzyme Transgenics Corporatiion Transgenic production of antibodies in milk
US5843678A (en) * 1997-04-16 1998-12-01 Amgen Inc. Osteoprotegerin binding proteins
WO1998054348A1 (en) * 1997-05-31 1998-12-03 Babraham Institute Telomere-associated chromosome fragmentation
US5885574A (en) * 1994-07-26 1999-03-23 Amgen Inc. Antibodies which activate an erythropoietin receptor
WO1999015696A1 (en) * 1997-09-19 1999-04-01 Medelys Laboratories Inc. Method and kit for early diagnosis of autoimmunity and lymphoma in central nervous system
EP0921189A1 (en) * 1997-11-14 1999-06-09 Sankyo Company Limited Transgenic animal allergy models and methods for their use
US5912176A (en) * 1996-06-03 1999-06-15 United Biomedical, Inc. Antibodies against a host cell antigen complex for pre and post exposure protection from infection by HIV
US6046310A (en) * 1996-03-13 2000-04-04 Protein Design Labs., Inc. FAS ligand fusion proteins and their uses
JP2000342279A (en) * 1999-03-30 2000-12-12 Japan Tobacco Inc Production of monoclonal antibody
WO2001018200A1 (en) 1999-09-06 2001-03-15 Chugai Seiyaku Kabushiki Kaisha Tsg-like gene
EP1167537A1 (en) * 1999-03-30 2002-01-02 Japan Tobacco Inc. Process for producing monoclonal antibody
JP2003501103A (en) * 1999-06-10 2003-01-14 アブジェニックス インク. Transgenic animals for producing specific isotypes of human antibodies via non-homologous switch regions
WO2003068259A1 (en) 2002-02-14 2003-08-21 Chugai Seiyaku Kabushiki Kaisha Antibody-containing solution pharmaceuticals
WO2003097082A2 (en) 2002-05-17 2003-11-27 Protein Design Labs Treatment of crohn's disease or psoriasis using anti-inteferon gamma antibodies
US6682736B1 (en) 1998-12-23 2004-01-27 Abgenix, Inc. Human monoclonal antibodies to CTLA-4
WO2004019966A1 (en) 2002-08-27 2004-03-11 Chugai Seiyaku Kabushiki Kaisha Method of stabilizing protein solution preparation
WO2004022739A1 (en) 2002-09-04 2004-03-18 Chugai Seiyaku Kabushiki Kaisha Antibody against n-terminal peptide or c-terminal peptide of gpc3 solubilized in blood
US6713610B1 (en) 1990-01-12 2004-03-30 Raju Kucherlapati Human antibodies derived from immunized xenomice
WO2004035747A2 (en) 2002-10-16 2004-04-29 Amgen Inc. Human anti-ifn-ϝ neutralizing antibodies as selective ifn-ϝ pathway inhibitors
WO2004050683A2 (en) 2002-12-02 2004-06-17 Abgenix, Inc. Antibodies directed to tumor necrosis factor and uses thereof
WO2004084823A2 (en) 2003-03-19 2004-10-07 Abgenix, Inc. Antibodies against t cell immunoglobulin domain and mucin domain 1 (tim-1) antigen and uses thereof
US6855808B2 (en) 1995-02-20 2005-02-15 Sankyo Co., Ltd. Proteins and methods for producing the proteins
WO2005014818A1 (en) 2003-08-08 2005-02-17 Perseus Proteomics Inc. Gene overexpressed in cancer
WO2005017155A1 (en) 2003-06-18 2005-02-24 Chugai Seiyaku Kabushiki Kaisha Fucose transporter
WO2005016111A2 (en) 2003-08-08 2005-02-24 Abgenix, Inc. Antibodies directed to parathyroid hormone (pth) and uses thereof
US6864235B1 (en) 1999-04-01 2005-03-08 Eva A. Turley Compositions and methods for treating cellular response to injury and other proliferating cell disorders regulated by hyaladherin and hyaluronans
WO2005035754A1 (en) 2003-10-14 2005-04-21 Chugai Seiyaku Kabushiki Kaisha Double specific antibodies substituting for functional protein
US6894149B2 (en) 2001-10-11 2005-05-17 Protein Design Labs, Inc. Anti-HLA-DA antibodies and the methods of using thereof
WO2005054467A1 (en) 2003-12-03 2005-06-16 Chugai Seiyaku Kabushiki Kaisha EXPRESSION SYSTEM WITH THE USE OF MAMMALIAN β-ACTIN PROMOTER
WO2005056605A1 (en) 2003-12-12 2005-06-23 Chugai Seiyaku Kabushiki Kaisha Modified antibodies recognizing trimer receptor or higher
US6911429B2 (en) 1999-04-01 2005-06-28 Transition Therapeutics Inc. Compositions and methods for treating cellular response to injury and other proliferating cell disorders regulated by hyaladherin and hyaluronans
WO2005092926A2 (en) 2004-03-19 2005-10-06 Amgen Inc. Reducing the risk of human and anti-human antibodies through v gene manipulation
EP1151010B1 (en) * 1999-02-05 2005-10-26 Therapeutic Human Polyclonals, Inc. Human polyclonal antibodies from transgenic nonhuman animals
AU2003227322B2 (en) * 1995-04-27 2005-12-01 Amgen Fremont Inc. Human Antibodies Derived From Immunized Xenomice
EP1604997A1 (en) * 1999-02-05 2005-12-14 Therapeutic Human Polyclonals, Inc. Human polyclonal antibodies from transgenic nonhuman animals
WO2006002177A2 (en) 2004-06-21 2006-01-05 Medarex, Inc. Interferon alpha receptor 1 antibodies and their uses
US6984720B1 (en) 1999-08-24 2006-01-10 Medarex, Inc. Human CTLA-4 antibodies
WO2006003179A2 (en) 2004-07-01 2006-01-12 Novo Nordisk A/S Antibodies binding to receptors kir2dl1, -2, 3 but not kir2ds4 and their therapeutic use
JP2006077030A (en) * 1997-12-02 2006-03-23 Neuralab Ltd Preventing and treating agent of amyloidogenic disease
WO2006055638A2 (en) 2004-11-17 2006-05-26 Abgenix, Inc. Fully human monoclonal antibodies to il-13
US7064244B2 (en) 1996-12-03 2006-06-20 Abgenix, Inc. Transgenic mammals having human Ig loci including plural VH and VK regions and antibodies produced therefrom
WO2006068975A2 (en) 2004-12-20 2006-06-29 Abgenix, Inc. Binding proteins specific for human matriptase
US7097834B1 (en) 1997-04-16 2006-08-29 Amgen Inc. Osteoprotegerin binding proteins
US7109003B2 (en) 1998-12-23 2006-09-19 Abgenix, Inc. Methods for expressing and recovering human monoclonal antibodies to CTLA-4
WO2006132363A1 (en) 2005-06-10 2006-12-14 Chugai Seiyaku Kabushiki Kaisha Stabilizer for protein preparation comprising meglumine and use thereof
US7192718B2 (en) 1997-04-15 2007-03-20 Sankyo Co. Ltd Methods for identifying a substance which specifically binds to an osteoclastogenesis inhibitory factor-binding molecule and modulates the biological activity thereof
WO2007038637A2 (en) 2005-09-26 2007-04-05 Medarex, Inc. Human monoclonal antibodies to cd70
WO2007043641A1 (en) 2005-10-14 2007-04-19 Fukuoka University Inhibitor of transplanted islet dysfunction in islet transplantation
WO2007046489A1 (en) 2005-10-21 2007-04-26 Chugai Seiyaku Kabushiki Kaisha Therapeutic agent for heart disease
WO2007055378A1 (en) 2005-11-14 2007-05-18 Cell Signals Inc. Method for treatment or prevention of disease associated with functional disorder of regulatory t cell
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
WO2007058194A1 (en) 2005-11-15 2007-05-24 National Hospital Organization Inhibitor of cytotoxic t cell induction
WO2007061029A1 (en) 2005-11-25 2007-05-31 Keio University Therapeutic agent for prostate cancer
WO2007067992A2 (en) 2005-12-08 2007-06-14 Medarex, Inc. Human monoclonal antibodies to fucosyl-gm1 and methods for using anti-fucosyl-gm1
WO2007074880A1 (en) 2005-12-28 2007-07-05 Chugai Seiyaku Kabushiki Kaisha Antibody-containing stabilizing preparation
WO2007077028A2 (en) 2005-12-30 2007-07-12 U3 Pharma Ag Antibodies directed to her-3 and uses thereof
WO2007084672A2 (en) 2006-01-17 2007-07-26 Medarex, Inc. Monoclonal antibodies against cd30 lacking in fucosyl and xylosyl residues
WO2007086490A1 (en) 2006-01-27 2007-08-02 Keio University Remedy for disease associated with choroidal angiogenesis
WO2007114325A1 (en) 2006-03-31 2007-10-11 Chugai Seiyaku Kabushiki Kaisha Antibody modification method for purifying bispecific antibody
WO2007114319A1 (en) 2006-03-31 2007-10-11 Chugai Seiyaku Kabushiki Kaisha Method for control of blood kinetics of antibody
WO2007117410A2 (en) 2006-03-31 2007-10-18 Medarex, Inc. Transgenic animals expressing chimeric antibodies for use in preparing human antibodies
WO2007116962A1 (en) 2006-04-07 2007-10-18 Osaka University Muscle regeneration promoter
WO2007119796A1 (en) 2006-04-14 2007-10-25 Medical And Biological Laboratories Co., Ltd. Mutant polypeptide having effector function
EP1854481A2 (en) 1996-04-23 2007-11-14 Chugai Seiyaku Kabushiki Kaisha Cerebral stroke/cerebral edema preventive or remedy containing IL-8 binding inhibitor as active ingredient
WO2007129457A1 (en) 2006-04-25 2007-11-15 The University Of Tokyo Therapeutic agents for alzheimer's disease and cancer
WO2007142325A1 (en) 2006-06-08 2007-12-13 Chugai Seiyaku Kabushiki Kaisha Preventive or remedy for inflammatory disease
WO2007145227A1 (en) 2006-06-14 2007-12-21 Chugai Seiyaku Kabushiki Kaisha Hematopoietic stem cell proliferation promoter
EP1878746A2 (en) 1997-05-05 2008-01-16 Amgen Fremont Inc. Human monoclonal antibodies to epidermal growth factor receptor
WO2008007755A1 (en) 2006-07-13 2008-01-17 Chugai Seiyaku Kabushiki Kaisha Cell death inducer
WO2008010556A1 (en) 2006-07-21 2008-01-24 Chugai Seiyaku Kabushiki Kaisha Remedy for renal disease
WO2008030611A2 (en) 2006-09-05 2008-03-13 Medarex, Inc. Antibodies to bone morphogenic proteins and receptors therefor and methods for their use
WO2008032833A1 (en) 2006-09-14 2008-03-20 Medical & Biological Laboratories Co., Ltd. Antibody having enhanced adcc activity and method for production thereof
WO2008042024A2 (en) 2006-06-01 2008-04-10 Elan Pharmaceuticals, Inc. Neuroactive fragments of app
WO2008047914A1 (en) 2006-10-20 2008-04-24 Forerunner Pharma Research Co., Ltd. Cancer therapeutic agent comprising anti-hb-egf antibody as active ingredient
WO2008047925A1 (en) 2006-10-20 2008-04-24 Forerunner Pharma Research Co., Ltd. Pharmaceutical composition comprising anti-hb-egf antibody as active ingredient
WO2008047723A1 (en) 2006-10-12 2008-04-24 Forerunner Pharma Research Co., Ltd. Diagnosis and treatment of cancer using anti-ereg antibody
EP1916020A2 (en) 1997-08-15 2008-04-30 Chugai Seiyaku Kabushiki Kaisha Preventives and/or remedies for systemic lupus erythematosus containing anti-IL-6 receptor antibody as the active ingredient
EP1916303A1 (en) 2000-11-30 2008-04-30 Medarex, Inc. Nucleic acids encoding rearranged human immunoglobulin sequences from transgenic transchromosomal mice
WO2008070569A2 (en) 2006-12-01 2008-06-12 Medarex, Inc. Human antibodies that bind cd22 and uses thereof
WO2008072723A1 (en) 2006-12-14 2008-06-19 Forerunner Pharma Research Co., Ltd. ANTI-Claudin-3 MONOCLONAL ANTIBODY, AND TREATMENT AND DIAGNOSIS OF CANCER USING THE SAME
WO2008074004A2 (en) 2006-12-14 2008-06-19 Medarex, Inc. Human antibodies that bind cd70 and uses thereof
WO2008076560A2 (en) 2006-11-15 2008-06-26 Medarex, Inc. Human monoclonal antibodies to btla and methods of use
WO2008081942A1 (en) 2007-01-05 2008-07-10 The University Of Tokyo Diagnosis and treatment of cancer by using anti-prg-3 antibody
WO2008090901A1 (en) 2007-01-23 2008-07-31 Shinshu University Chronic rejection inhibitor
US7411050B2 (en) 1996-12-23 2008-08-12 Immunex Corporation Monoclonal blocking antibody to human RANKL
WO2008099920A1 (en) 2007-02-15 2008-08-21 Kyushu University, National University Corporation Therapeutic agent for interstitial pulmonary disease comprising anti-hmgb-1 antibody
WO2008105560A1 (en) 2007-02-27 2008-09-04 Forerunner Pharma Research Co., Ltd. Pharmaceutical composition comprising anti-grp78 antibody as active ingredient
WO2008111597A1 (en) 2007-03-12 2008-09-18 Chugai Seiyaku Kabushiki Kaisha Remedy for chemotherapy-resistant cancer containing hla class i-recognizing antibody as the active ingredient and use of the same
EP1972638A1 (en) 2001-04-02 2008-09-24 Chugai Seiyaku Kabushiki Kaisha Remedies for juvenile chronic arthritis and related diseases
EP1972639A2 (en) 2007-03-07 2008-09-24 Cell Signaling Technology, Inc. Reagents for the detection of protein phosphorylation in carcinoma signaling pathways
EP1975184A2 (en) 2007-03-26 2008-10-01 Albrecht Moritz Serine or threonine phosphorylation sites
US7435871B2 (en) 2001-11-30 2008-10-14 Amgen Fremont Inc. Transgenic animals bearing human Igλ light chain genes
EP1983002A2 (en) 2007-04-19 2008-10-22 Peter Hornbeck Tyrosine phosphorylation sites and antibodies specific for them
EP1983003A2 (en) 2007-04-19 2008-10-22 Peter Hornbeck Tyrosine phosphorylation sites and antibodies specific for them
EP1987842A1 (en) 2000-04-28 2008-11-05 Chugai Seiyaku Kabushiki Kaisha Cell proliferation inhibitor
US7452535B2 (en) 2002-04-12 2008-11-18 Medarex, Inc. Methods of treatment using CTLA-4 antibodies
WO2008153926A2 (en) 2007-06-05 2008-12-18 Yale University Inhibitors of receptor tyrosine kinases and methods of use thereof
WO2009001840A1 (en) 2007-06-25 2008-12-31 Forerunner Pharma Research Co., Ltd. Anti-prominin-1 antibody having adcc activity or cdc activity
EP2011514A1 (en) 1997-03-21 2009-01-07 Chugai Seiyaku Kabushiki Kaisha A preventive or therapeutic agent for sensitized T cell-mediated diseases comprising IL-6 antagonist as an active ingredient
WO2009014263A1 (en) 2007-07-26 2009-01-29 Osaka University Agent for treatment of ophthalmia containing interleukin-6 receptor inhibitor as active ingredient
EP2026073A1 (en) 2000-04-29 2009-02-18 University Of Iowa Research Foundation Diagnostics and therapeutics for macular degeneration-related disorders
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
US7501552B2 (en) 1991-08-28 2009-03-10 Medarex, Inc. Transgenic non-human animals for producing chimeric antibodies
WO2009032845A2 (en) 2007-09-04 2009-03-12 Compugen, Ltd. Polypeptides and polynucleotides, and uses thereof as a drug target for producing drugs and biologics
WO2009041062A1 (en) 2007-09-28 2009-04-02 Chugai Seiyaku Kabushiki Kaisha Anti-glypican-3 antibody having improved kinetics in plasma
WO2009040134A1 (en) 2007-09-26 2009-04-02 U3 Pharma Gmbh Heparin-binding epidermal growth factor-like growth factor antigen binding proteins
EP2045267A2 (en) 2000-12-06 2009-04-08 Elan Pharma International Limited Humanized antibodies that recognize beta amyloid peptide
WO2009044774A1 (en) 2007-10-02 2009-04-09 Chugai Seiyaku Kabushiki Kaisha Remedy for graft-versus-host disease comprising interleukin-6 receptor inhibitor as the active ingredient
WO2009052439A2 (en) 2007-10-17 2009-04-23 Elan Pharma International Limited Immunotherapy regimes dependent on apoe status
WO2009051108A1 (en) 2007-10-15 2009-04-23 Chugai Seiyaku Kabushiki Kaisha Method for production of antibody
WO2009054435A1 (en) 2007-10-24 2009-04-30 Otsuka Chemical Co., Ltd. Polypeptide having enhanced effector function
WO2009054863A2 (en) 2006-12-13 2009-04-30 Medarex, Inc. Human antibodies that bind cd19 and uses thereof
WO2009054873A2 (en) 2007-08-02 2009-04-30 Novimmune S.A. Anti-rantes antibodies and methods of use thereof
WO2009063970A1 (en) 2007-11-14 2009-05-22 Forerunner Pharma Research Co., Ltd. Diagnosis and treatment of cancer using anti-gpr49 antibody
EP2062917A2 (en) 2004-04-09 2009-05-27 Abbott Laboratories Antibodies to erythropoietin receptor and uses thereof
EP2062920A2 (en) 2007-11-21 2009-05-27 Peter Hornbeck Protein phosphorylation by basophilic serine/threonine kinases in insulin signalling pathways
WO2009072604A1 (en) 2007-12-05 2009-06-11 Chugai Seiyaku Kabushiki Kaisha Anti-nr10 antibody and use thereof
WO2009072598A1 (en) 2007-12-05 2009-06-11 Chugai Seiyaku Kabushiki Kaisha Therapeutic agent for pruritus
WO2009075344A1 (en) 2007-12-12 2009-06-18 Japan As Represented By Director General Of Agency Of National Cancer Center Therapeutic agent for mll leukemia and moz leukemia of which molecular target is m-csf receptor, and use thereof
WO2009078799A1 (en) 2007-12-17 2009-06-25 Marfl Ab New vaccine for the treatment of mycobacterium related disorders
WO2009084659A1 (en) 2007-12-27 2009-07-09 Chugai Seiyaku Kabushiki Kaisha Solution preparation containing antibody at high concentration
WO2009086539A2 (en) 2007-12-28 2009-07-09 Elan Pharmaceuticals, Inc. Treatment and prophylaxis of amyloidosis
US7560534B2 (en) 2000-05-08 2009-07-14 Celldex Research Corporation Molecular conjugates comprising human monoclonal antibodies to dendritic cells
WO2009087978A1 (en) 2008-01-11 2009-07-16 The University Of Tokyo Anti-cldn6 antibody
FR2926438A1 (en) * 2008-01-22 2009-07-24 Univ Limoges NON-HUMAN TRANSGENIC MAMMAL FOR THE CONSTANT REGION OF THE HEAVY CHAIN OF HUMAN CLASS G IMMUNOGLOBULINS AND ITS APPLICATIONS
EP2087908A1 (en) 2001-06-26 2009-08-12 Amgen, Inc. Antibodies to opgl
WO2009122667A1 (en) 2008-04-04 2009-10-08 中外製薬株式会社 Therapeutic for hepatic cancer
WO2009125825A1 (en) 2008-04-11 2009-10-15 中外製薬株式会社 Antigen-binding molecule capable of binding to two or more antigen molecules repeatedly
US7605238B2 (en) 1999-08-24 2009-10-20 Medarex, Inc. Human CTLA-4 antibodies and their uses
EP2110434A1 (en) 2002-02-25 2009-10-21 Genentech, Inc. Type-1 cytokine receptor GLM-R
EP2112166A2 (en) 1998-12-23 2009-10-28 Pfizer Inc. Human monoclonal antibodies to CTLA-4
EP2123679A2 (en) 2007-05-01 2009-11-25 Peter Hornbeck Tyrosine phosphorylation sites
US7625549B2 (en) 2004-03-19 2009-12-01 Amgen Fremont Inc. Determining the risk of human anti-human antibodies in transgenic mice
WO2009148928A1 (en) 2008-05-29 2009-12-10 Galaxy Biotech, Llc Monoclonal antibodies to basic fibroblast growth factor
WO2009148148A1 (en) 2008-06-05 2009-12-10 国立がんセンター総長が代表する日本国 Neuroinvasion inhibitor
WO2009147781A1 (en) 2008-06-02 2009-12-10 国立大学法人東京大学 Antitumor agent
WO2009154025A1 (en) 2008-06-20 2009-12-23 国立大学法人岡山大学 ANTIBODY AGAINST OXIDIZED LDL/β2GPI COMPLEX AND USE OF THE SAME
WO2010015608A1 (en) 2008-08-05 2010-02-11 Novartis Ag Compositions and methods for antibodies targeting complement protein c5
EP2161336A1 (en) 2005-05-09 2010-03-10 ONO Pharmaceutical Co., Ltd. Human monoclonal antibodies to programmed death 1(PD-1) and methods for treating cancer using anti-PD-1 antibodies alone or in combination with other immunotherapeutics
US7709215B2 (en) 2007-06-01 2010-05-04 Cytonics Corporation Method for diagnosing and treating acute joint injury
WO2010054265A2 (en) 2008-11-07 2010-05-14 Galaxy Biotech, Llc. Monoclonal antibodies to fibroblast growth factor receptor 2
WO2010054403A1 (en) 2008-11-10 2010-05-14 Alexion Pharmaceuticals, Inc. Methods and compositions for treating complement-associated disorders
US7718776B2 (en) 2002-04-05 2010-05-18 Amgen Inc. Human anti-OPGL neutralizing antibodies as selective OPGL pathway inhibitors
US7722873B2 (en) 1996-10-10 2010-05-25 Genpharm International, Inc. Heterologous antibodies which bind human CD4
WO2010064697A1 (en) 2008-12-05 2010-06-10 中外製薬株式会社 Anti-nr10 antibody, and use thereof
WO2010067308A2 (en) 2008-12-08 2010-06-17 Compugen Ltd. Polypeptides and polynucleotides, and uses thereof as a drug target for producing drugs and biologics
WO2010070263A1 (en) * 2008-12-18 2010-06-24 Erasmus University Medical Center Rotterdam Non-human transgenic animals expressing humanised antibodies and use therof
WO2010074049A1 (en) 2008-12-22 2010-07-01 株式会社 未来創薬研究所 Anti-hs6st2 antibody and use thereof
WO2010073694A1 (en) 2008-12-25 2010-07-01 国立大学法人東京大学 Diagnosis of treatment of cancer using anti-tm4sf20 antibody
WO2010074192A1 (en) 2008-12-26 2010-07-01 国立大学法人東京大学 Diagnosis and treatment of cancer using anti-lgr7 antibody
WO2010072740A2 (en) 2008-12-23 2010-07-01 Astrazeneca Ab TARGETED BINDING AGENTS DIRECTED TO α5β1 AND USES THEREOF
EP2204191A1 (en) 2004-04-15 2010-07-07 Galaxy Biotech, LLC Monoclonal antibodies to hepatocyte growth factor
EP2208783A1 (en) 2004-12-22 2010-07-21 Chugai Seiyaku Kabushiki Kaisha Method of producing an antibody using a cell in which the function of fucose transporter is inhibited
WO2010085590A1 (en) 2009-01-23 2010-07-29 Biosynexus Incorporated Opsonic and protective antibodies specific for lipoteichoic acid gram positive bacteria
EP2213685A1 (en) 2002-09-06 2010-08-04 Amgen Inc. Therapeutic anti-IL-1R1 monoclonal antibody
EP2216046A2 (en) 2004-07-09 2010-08-11 Chugai Seiyaku Kabushiki Kaisha Anti-glypican 3 antibody
WO2010093928A2 (en) 2009-02-12 2010-08-19 Cell Signaling Technology, Inc. Mutant ros expression in human cancer
EP2221064A1 (en) 2003-03-12 2010-08-25 Janssen Alzheimer Immunotherapy Humanized antibodies that recognize beta amyloid peptide
WO2010102175A1 (en) 2009-03-05 2010-09-10 Medarex, Inc. Fully human antibodies specific to cadm1
US7807789B2 (en) 2004-12-21 2010-10-05 Cell Signaling Technology, Inc. Reagents for the detection of protein phosphorylation in EGFR-signaling pathways
EP2236519A1 (en) 2007-09-18 2010-10-06 Amgen, Inc Human GM-CSF antigen binding proteins
WO2010112458A1 (en) 2009-03-31 2010-10-07 Novartis Ag Composition and methods of use for therapeutic antibodies specific for the il-12 receptore betal subunit
WO2010117325A1 (en) 2009-04-08 2010-10-14 Olle Hernell New methods for treatment of inflammatory diseases
WO2010119991A2 (en) 2009-04-17 2010-10-21 Takeda Pharmaceutical Company Limited Novel method of treating cancer
WO2010119691A1 (en) 2009-04-16 2010-10-21 国立大学法人東京大学 Diagnosis and treatment of cancer using anti-tmprss11e antibody
WO2010125003A1 (en) 2009-04-27 2010-11-04 Novartis Ag Compositions and methods for increasing muscle growth
WO2010126137A1 (en) 2009-05-01 2010-11-04 国立大学法人 東京大学 Anti-cadherin antibody
WO2010128398A1 (en) 2009-05-04 2010-11-11 Pangenetics 110 B.V. Antibodies against nerve growth factor (ngf) with enhanced in vivo stability
WO2010128407A2 (en) 2009-05-05 2010-11-11 Novimmune S.A. Anti-il-17f antibodies and methods of use thereof
WO2010137654A1 (en) 2009-05-29 2010-12-02 株式会社未来創薬研究所 Pharmaceutical composition containing antagonist of egf family ligand as component
EP2261230A1 (en) 2002-09-11 2010-12-15 Chugai Seiyaku Kabushiki Kaisha Protein purification method
EP2264186A1 (en) 2002-09-30 2010-12-22 Oncotherapy Science, Inc. Method for diagnosing non-small cell lung cancers
WO2010151632A1 (en) 2009-06-25 2010-12-29 Bristol-Myers Squibb Company Protein purifacation by caprylic acid (octanoic acid ) precipitation
US7863029B2 (en) 2004-06-04 2011-01-04 Fox Chase Cancer Center Alternate morpheeins of allosteric proteins as a target for the development of bioactive molecules
US7868140B2 (en) 2005-07-18 2011-01-11 Amgen Inc. Human anti-B7RP1 neutralizing antibodies
EP2272539A2 (en) 2004-10-19 2011-01-12 Elan Pharmaceuticals, Inc. Truncated fragments of alpha-synuclein in lewy body disease
US7875705B2 (en) 2007-05-28 2011-01-25 The University Of Tokyo Tumor diagnostic agent used in PET comprising anti-ROBO1 antibody
WO2011013786A1 (en) 2009-07-31 2011-02-03 Maeda Shin Cancer metastasis inhibitor
WO2011014438A1 (en) 2009-07-31 2011-02-03 N.V. Organon Fully human antibodies to btla
WO2011017294A1 (en) 2009-08-07 2011-02-10 Schering Corporation Human anti-rankl antibodies
US7888480B2 (en) 2005-08-31 2011-02-15 Cell Signaling Technology, Inc. Reagents for the detection of protein phosphorylation in leukemia signaling pathways
EP2284194A1 (en) 2004-12-21 2011-02-16 AstraZeneca AB Antibodies directed to angiopoietin-2 and uses thereof
WO2011021146A1 (en) 2009-08-20 2011-02-24 Pfizer Inc. Osteopontin antibodies
WO2011021381A1 (en) 2009-08-17 2011-02-24 株式会社未来創薬研究所 Pharmaceutical composition containing anti-hb-egf antibody as active ingredient
EP2289944A2 (en) 2003-10-10 2011-03-02 Chugai Seiyaku Kabushiki Kaisha Bispecific antibody substituting for functional proteins
EP2298816A2 (en) 1995-12-05 2011-03-23 Amgen Inc. Apoptosis induced by an antibody against Her2
EP2301576A1 (en) 2004-03-29 2011-03-30 Abbott Biotherapeutics Corp. Therapeutic use of Anti-CS1 Antibodies
WO2011037983A1 (en) 2009-09-23 2011-03-31 Medarex, Inc. Cation exchange chromatography
WO2011037160A1 (en) 2009-09-24 2011-03-31 中外製薬株式会社 Antibody capable of recognizing hla class i
EP2305302A2 (en) 1999-05-28 2011-04-06 Janssen Alzheimer Immunotherapy Prevention and treatment of amyloidogenic disease
WO2011045080A2 (en) 2009-10-16 2011-04-21 Biorealites S.A.S. Monoclonal antibodies to progastrin and their uses
US7931897B2 (en) 2001-02-07 2011-04-26 Chugai Seiyaku Kabushiki Kaisha Therapeutic agent for hematopoietic tumors
EP2314627A2 (en) 2002-06-28 2011-04-27 The Government of the U.S.A. as represented by The Secretary of the dept. of Health & Human Services Method for the treatment of multiple sclerosis
WO2011049758A1 (en) 2009-10-09 2011-04-28 Amgen Inc. Human anti-ngf neutralizing antibodies as selective ngf pathway inhibitors
US7935790B2 (en) 2004-10-04 2011-05-03 Cell Singaling Technology, Inc. Reagents for the detection of protein phosphorylation in T-cell receptor signaling pathways
EP2316921A1 (en) 2002-05-24 2011-05-04 Schering Corporation Neutralizing human anti-IGFR antibody
EP2316848A1 (en) 1999-09-21 2011-05-04 Chugai Seiyaku Kabushiki Kaisha Transporter genes OATP-B, C, D and E
US7939636B2 (en) 2006-08-11 2011-05-10 Cell Signaling Technology, Inc. Reagents for the detection of protein phosphorylation in c-Src signaling pathways
WO2011057188A1 (en) 2009-11-06 2011-05-12 Idexx Laboratories, Inc. Canine anti-cd20 antibodies
WO2011057250A1 (en) 2009-11-09 2011-05-12 Alexion Pharmaceuticals, Inc. Reagents and methods for detecting pnh type ii white blood cells and their identification as risk factors for thrombotic disorders
EP2322549A1 (en) 2003-06-04 2011-05-18 Fibrogen, Inc. Connective tissue growth factor antibodies
WO2011060206A2 (en) 2009-11-13 2011-05-19 U3 Pharma Gmbh Material and methods for treating or preventing her-3 associated diseases
WO2011062926A2 (en) 2009-11-17 2011-05-26 Medarex, Inc. Methods for enhanced protein production
WO2011067711A2 (en) 2009-12-01 2011-06-09 Compugen Ltd Novel heparanase splice variant
US7973134B2 (en) 2004-07-07 2011-07-05 Cell Signaling Technology, Inc. Reagents for the detection of protein phosphorylation in anaplastic large cell lymphoma signaling pathways
EP2341067A1 (en) 2003-07-18 2011-07-06 Amgen, Inc Specific binding agents to hepatocyte growth factor
US7977462B2 (en) 2007-04-19 2011-07-12 Cell Signaling Technology, Inc. Tyrosine phosphorylation sites
EP2343384A2 (en) 2004-03-23 2011-07-13 Oncotherapy Science, Inc. Method for diagnosing non-small cell lung cancer
WO2011085343A1 (en) 2010-01-11 2011-07-14 Alexion Pharmaceuticals, Inc Biomarkers of immunomodulatory effects in humans treated with anti-cd200 antibodies
WO2011090088A1 (en) 2010-01-20 2011-07-28 中外製薬株式会社 Solution preparation containing stabilized antibody
EP2351483A1 (en) 1999-10-01 2011-08-03 Chugai Seiyaku Kabushiki Kaisha Prevention and treatment of blood coagulation-related diseases
WO2011092989A1 (en) 2010-01-29 2011-08-04 東レ株式会社 Polylactic acid-based resin sheet
WO2011093097A1 (en) 2010-01-29 2011-08-04 株式会社未来創薬研究所 Anti-dll3 antibody
WO2011097511A1 (en) 2010-02-05 2011-08-11 The United States Of America, As Represented By The Secretary Department Of Health & Human Services REGULATORY B CELLS (tBREGS) AND THEIR USE
EP2357202A1 (en) 2006-04-10 2011-08-17 AstraZeneca AB Targeted binding agents directed to Upar and uses thereof
WO2011099524A1 (en) 2010-02-10 2011-08-18 富士フイルムRiファーマ株式会社 Radioactive metal-labeled anti-cadherin antibody
US8003097B2 (en) 2007-04-18 2011-08-23 Janssen Alzheimer Immunotherapy Treatment of cerebral amyloid angiopathy
WO2011103490A2 (en) 2010-02-18 2011-08-25 The Regents Of The University Of California INTEGRIN αVβ8 NEUTRALIZING ANTIBODY
EP2361933A2 (en) 2005-01-26 2011-08-31 Amgen Fremont Inc. Antibodies against interleukin-1 beta
WO2011105573A1 (en) 2010-02-26 2011-09-01 株式会社未来創薬研究所 Anti-icam3 antibody and use thereof
WO2011109398A2 (en) 2010-03-02 2011-09-09 President And Fellows Of Harvard College Methods and compositions for treatment of angelman syndrome and autism spectrum disorders
WO2011108714A1 (en) 2010-03-04 2011-09-09 中外製薬株式会社 Antibody constant region variant
EP2364719A1 (en) 1999-06-01 2011-09-14 Janssen Alzheimer Immunotherapy Prevention and treatment of amyloidogenic diseases
EP2366715A2 (en) 2005-11-14 2011-09-21 Amgen Inc. Rankl Antibody-PTH/PTHRP Chimeric Molecules
WO2011116090A1 (en) 2010-03-17 2011-09-22 Abbott Research B.V. Anti-nerve growth factor (ngf) antibody compositions
EP2368577A2 (en) 2003-04-28 2011-09-28 Chugai Seiyaku Kabushiki Kaisha Methods for treating interleukin-6 related diseases
US8030457B2 (en) 2007-08-23 2011-10-04 Amgen, Inc. Antigen binding proteins to proprotein convertase subtilisin kexin type 9 (PCSK9)
EP2371865A2 (en) 2006-04-07 2011-10-05 Warner Chilcott Company, LLC Antibodies that bind human protein tyrosine phosphatase beta (HPTP-ß) and uses thereof
EP2371391A1 (en) 2003-05-08 2011-10-05 Abbott Biotherapeutics Corp. Therapeutic use of anti-CS1 antibodies
WO2011122011A2 (en) 2010-03-30 2011-10-06 Chugai Seiyaku Kabushiki Kaisha Antibodies with modified affinity to fcrn that promote antigen clearance
EP2383295A1 (en) 2003-12-10 2011-11-02 Medarex, Inc. IP-10 antibodies and their uses
WO2011137395A1 (en) 2010-04-30 2011-11-03 Rother Russell P Anti-c5a antibodies and methods for using the antibodies
EP2384767A1 (en) 2005-03-24 2011-11-09 Millennium Pharmaceuticals, Inc. Antibodies that bind OV064 and methods of use therefor
WO2011140254A1 (en) 2010-05-04 2011-11-10 Adimab, Llc Antibodies against epidermal growth factor receptor (egfr) and uses thereof
WO2011138392A1 (en) 2010-05-06 2011-11-10 Novartis Ag Compositions and methods of use for therapeutic low density lipoprotein -related protein 6 (lrp6) antibodies
WO2011138391A1 (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
WO2011140151A1 (en) 2010-05-04 2011-11-10 Dyax Corp. Antibodies against epidermal growth factor receptor (egfr)
WO2011149046A1 (en) 2010-05-28 2011-12-01 独立行政法人国立がん研究センター Therapeutic agent for pancreatic cancer
WO2011149051A1 (en) 2010-05-28 2011-12-01 中外製薬株式会社 Antitumor t cell response enhancer
US8071323B2 (en) 2006-04-07 2011-12-06 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Human monoclonal antibodies that bind human insulin like growth factors and their use
EP2402372A2 (en) 2005-10-18 2012-01-04 Medella Therapeutics Ltd Therapeutic agent
WO2012003338A1 (en) 2010-07-01 2012-01-05 Takeda Pharmaceutical Company Limited COMBINATION OF A cMET INHIBITOR AND AN ANTIBODY TO HGF AND/OR cMET
EP2404616A2 (en) 2005-12-13 2012-01-11 AstraZeneca AB Binding proteins specific for insulin-like growth factors and uses thereof
WO2012009442A2 (en) 2010-07-14 2012-01-19 Merck Sharp & Dohme Corp. Anti-addl monoclonal antibody and uses thereof
WO2012018404A2 (en) 2010-08-06 2012-02-09 U3 Pharma Gmbh Use of her3 binding agents in prostate treatment
WO2012019132A2 (en) 2010-08-06 2012-02-09 Cell Signaling Technology, Inc. Anaplastic lymphoma kinase in kidney cancer
EP2418220A2 (en) 2003-12-10 2012-02-15 Medarex, Inc. Interferon alpha antibodies and their uses
EP2420513A1 (en) 2006-08-03 2012-02-22 MedImmune Limited Targeted binding agents directed to PDGFR-alpha and uses thereof
WO2012022814A1 (en) 2010-08-20 2012-02-23 Novartis Ag Antibodies for epidermal growth factor receptor 3 (her3)
AU2008202860B2 (en) * 1995-04-27 2012-03-01 Amgen Fremont Inc. Human Antibodies Derived From Immunized Xenomice
US8128928B2 (en) 2002-03-12 2012-03-06 Wyeth Llc Humanized antibodies that recognize beta amyloid peptide
WO2012037534A1 (en) 2010-09-17 2012-03-22 Baxter International Inc. Stabilization of immunoglobulins through aqueous formulation with histidine at weak acidic to neutral ph
WO2012035518A1 (en) 2010-09-17 2012-03-22 Compugen Ltd. Compositions and methods for treatment of drug resistant multiple myeloma
EP2434285A1 (en) 2010-09-22 2012-03-28 IMBA-Institut für Molekulare Biotechnologie GmbH Breast cancer diagnostics
EP2433644A1 (en) 2010-09-22 2012-03-28 IMBA-Institut für Molekulare Biotechnologie GmbH Breast cancer therapeutics
WO2012040617A2 (en) 2010-09-23 2012-03-29 Neogenix Oncology, Inc. Colon and pancreas cancer peptidomimetics
US8153410B2 (en) 2003-07-07 2012-04-10 Fox Chase Cancer Center Alternate morpheein forms of allosteric proteins as a target for the development of bioactive molecules
WO2012045703A1 (en) 2010-10-05 2012-04-12 Novartis Ag Anti-il12rbeta1 antibodies and their use in treating autoimmune and inflammatory disorders
WO2012057328A1 (en) 2010-10-29 2012-05-03 株式会社ペルセウスプロテオミクス Anti-cdh3 antibody having high internalizing capability
WO2012061120A1 (en) 2010-10-25 2012-05-10 Regents Of The University Of Minnesota Therapeutic composition for treatment of glioblastoma
WO2012067176A1 (en) 2010-11-17 2012-05-24 中外製薬株式会社 Multi-specific antigen-binding molecule having alternative function to function of blood coagulation factor viii
WO2012066293A1 (en) 2010-11-17 2012-05-24 Biotecnol Inc Therapeutic agent
EP2457586A1 (en) 2003-06-27 2012-05-30 Amgen Fremont Inc. Antibodies directed to the deletion mutants of epidermal growth factor receptor and uses thereof
WO2012069466A1 (en) 2010-11-24 2012-05-31 Novartis Ag Multispecific molecules
US8193328B2 (en) 2005-09-08 2012-06-05 Philadelphia Health & Education Corporation Identification of modulators of serine protease inhibitor Kazal and their use as anti-cancer and anti-viral agents
WO2012073985A1 (en) 2010-11-30 2012-06-07 中外製薬株式会社 Cytotoxicity-inducing therapeutic agent
WO2012073992A1 (en) 2010-11-30 2012-06-07 中外製薬株式会社 Antigen-binding molecule capable of binding to plurality of antigen molecules repeatedly
EP2463305A1 (en) 2006-01-12 2012-06-13 Alexion Pharmaceuticals, Inc. Antibodies to OX-2/CD200 and uses thereof
EP2465872A2 (en) 2004-10-25 2012-06-20 Merck Sharp & Dohme Corporation Anti-ADDL Antibodies and uses thereof
EP2476706A2 (en) 2005-12-12 2012-07-18 Bayer HealthCare LLC Anti-MN antibodies and methods of using same
EP2476704A2 (en) 2006-04-01 2012-07-18 Galaxy Biotech, LLC Humanized monoclonal antibodies to heptocyte growth factor
WO2012102679A1 (en) 2011-01-24 2012-08-02 National University Of Singapore Pathogenic mycobacteria-derived mannose-capped lipoarabinomannan antigen binding proteins
US8236318B2 (en) 2007-11-07 2012-08-07 Celldex Therapeutics Inc. Antibodies that bind human dendritic and epithelial cell 205 (DEC-205)
WO2012106634A1 (en) 2011-02-03 2012-08-09 Alexion Pharmaceuticals, Inc. Use of an anti-cd200 antibody for prolonging the survival of allografts
EP2486941A1 (en) 2006-10-02 2012-08-15 Medarex, Inc. Human antibodies that bind CXCR4 and uses thereof
WO2012115241A1 (en) 2011-02-25 2012-08-30 中外製薬株式会社 Fcγriib-specific fc antibody
WO2012122484A1 (en) 2011-03-09 2012-09-13 Roberto Polakiewicz Methods and reagents for creating monoclonal antibodies
EP2500073A1 (en) 2011-03-17 2012-09-19 ChromaCon AG Method for identification and purification of multi-specific polypeptides
WO2012133782A1 (en) 2011-03-30 2012-10-04 中外製薬株式会社 Retention of antigen-binding molecules in blood plasma and method for modifying immunogenicity
EP2508608A1 (en) 2003-06-09 2012-10-10 Alnylam Pharmaceuticals Inc. Method of treating neurodegenerative disease
WO2012138997A1 (en) 2011-04-07 2012-10-11 Amgen Inc. Novel egfr binding proteins
US8287863B2 (en) 1999-08-23 2012-10-16 Chugai Seiyaku Kabushiki Kaisha Method for treating myeloma utilizing an expression enhancer for HM1.24 antigen
WO2012140627A1 (en) 2011-04-15 2012-10-18 Compugen Ltd. Polypeptides and polynucleotides, and uses thereof for treatment of immune related disorders and cancer
WO2012144208A1 (en) 2011-04-18 2012-10-26 国立大学法人東京大学 Diagnosis and treatment of cancer using anti-itm2a antibody
WO2012154999A1 (en) 2011-05-10 2012-11-15 Amgen Inc. Methods of treating or preventing cholesterol related disorders
EP2527456A1 (en) 2004-10-22 2012-11-28 Revivicor Inc. Transgenic porcines lacking endogenous immunoglobulin light chain
WO2012162373A1 (en) 2011-05-23 2012-11-29 Cell Signaling Technology, Inc. Ros kinase in lung cancer
WO2012160448A2 (en) 2011-05-25 2012-11-29 Innate Pharma, S.A. Anti-kir antibodies for the treatment of inflammatory disorders
EP2530090A2 (en) 2006-10-19 2012-12-05 CSL Limited Anti-IL-13R alpha 1 antibodies and their uses thereof
EP2532677A1 (en) 2005-10-21 2012-12-12 Novartis AG Human antibodies against il13 and therapeutic uses
WO2012174446A1 (en) 2011-06-17 2012-12-20 President And Fellows Of Harvard College Frizzled 2 as a target for therapeutic antibodies in the treatment of cancer
WO2012172495A1 (en) 2011-06-14 2012-12-20 Novartis Ag Compositions and methods for antibodies targeting tem8
WO2013002362A1 (en) 2011-06-30 2013-01-03 中外製薬株式会社 Heterodimerized polypeptide
WO2013006437A1 (en) 2011-07-01 2013-01-10 Novartis Ag Method for treating metabolic disorders
WO2013006547A2 (en) 2011-07-05 2013-01-10 Merrimack Pharmaceuticals, Inc. Antibodies against epidermal growth factor receptor (egfr) and uses thereof
EP2548576A1 (en) 2006-08-14 2013-01-23 Forerunner Pharma Research Co., Ltd. Diagnosis of cancer using anti-desmoglein-3 antibodies
WO2013012022A1 (en) 2011-07-19 2013-01-24 中外製薬株式会社 Stable protein-containing preparation containing argininamide or analogous compound thereof
WO2013010955A1 (en) 2011-07-15 2013-01-24 Morphosys Ag Antibodies that are cross-reactive for macrophage migration inhibitory factor (mif) and d-dopachrome tautomerase (d-dt)
WO2013012855A1 (en) 2011-07-18 2013-01-24 Amgen Inc. Apelin antigen-binding proteins and uses thereof
WO2013017656A1 (en) 2011-08-02 2013-02-07 Medizinische Universität Wien Antagonists of ribonucleases for treating obesity
WO2013017691A1 (en) 2011-08-04 2013-02-07 Medizinische Universität Innsbruck Cahgt1p inhibitors for use in the treatment of candidiasis
US8372412B2 (en) 2005-12-23 2013-02-12 Rapid Biosensor Systems Limited Bioassay and peptides for use therein
WO2013027802A1 (en) 2011-08-23 2013-02-28 中外製薬株式会社 Novel anti-ddr1 antibody having anti-tumor activity
EP2567709A2 (en) 2007-11-02 2013-03-13 Novartis AG Molecules and methods for modulating low-density-lipoprotein receptor-related protein 6 (LRP6)
WO2013035824A1 (en) 2011-09-07 2013-03-14 ファーマロジカルズ・リサーチ プライベート リミテッド Cancer stem cell isolation
US8398975B2 (en) 2006-08-03 2013-03-19 Medimmune Limited Antibodies directed to αVβ6 and uses thereof
WO2013046704A2 (en) 2011-09-30 2013-04-04 Chugai Seiyaku Kabushiki Kaisha THERAPEUTIC ANTIGEN-BINDING MOLECULE WITH A FcRn-BINDING DOMAIN THAT PROMOTES ANTIGEN CLEARANCE
WO2013047729A1 (en) 2011-09-30 2013-04-04 中外製薬株式会社 Antigen-binding molecule inducing immune response to target antigen
WO2013046722A1 (en) 2011-09-30 2013-04-04 中外製薬株式会社 Ion concentration-dependent binding molecule library
WO2013047748A1 (en) 2011-09-30 2013-04-04 中外製薬株式会社 Antigen-binding molecule promoting disappearance of antigens having plurality of biological activities
WO2013047752A1 (en) 2011-09-30 2013-04-04 中外製薬株式会社 Antigen-binding molecule for promoting loss of antigens
WO2013051294A1 (en) 2011-10-05 2013-04-11 中外製薬株式会社 Antigen-binding molecule for promoting clearance from plasma of antigen comprising saccharide chain receptor-binding domain
WO2013054307A2 (en) 2011-10-14 2013-04-18 Novartis Ag Antibodies and methods for wnt pathway-related diseases
WO2013056233A1 (en) 2011-10-13 2013-04-18 Aerpio Therapeutics, Inc. Treatment of ocular disease
EP2586796A1 (en) 2007-10-12 2013-05-01 Novartis AG Compositions and methods for use for antibodies against sclerostin
WO2013063496A1 (en) 2011-10-28 2013-05-02 Millennium Pharmaceuticals, Inc. Biomarkers of response to nae inhibitors
WO2013062083A1 (en) 2011-10-28 2013-05-02 ファーマロジカルズ・リサーチ プライベート リミテッド Cancer stem cell-specific molecule
WO2013067060A1 (en) 2011-11-01 2013-05-10 Bionomics, Inc. Anti-gpr49 antibodies
WO2013065708A1 (en) 2011-10-31 2013-05-10 中外製薬株式会社 Antigen-binding molecule having regulated conjugation between heavy-chain and light-chain
WO2013067054A1 (en) 2011-11-01 2013-05-10 Bionomics, Inc. Antibodies and methods of treating cancer
WO2013067055A1 (en) 2011-11-01 2013-05-10 Bionomics, Inc. Methods of blocking cancer stem cell growth
WO2013067057A1 (en) 2011-11-01 2013-05-10 Bionomics, Inc. Anti-gpr49 antibodies
WO2013071142A1 (en) 2011-11-11 2013-05-16 Millennium Pharmaceuticals, Inc. Biomarkers of response to proteasome inhibitors
WO2013071163A2 (en) 2011-11-11 2013-05-16 Millennium Pharamaceuticals, Inc. Biomarkers of response to proteasome inhibitors
EP2594587A1 (en) 2011-11-16 2013-05-22 AdrenoMed AG Anti-Adrenomedullin (ADM) antibody or anti-ADM antibody fragment or anti-ADM non-Ig scaffold for reducing the risk of mortality in a patient having a chronic or acute disease or acute condition
EP2594588A1 (en) 2011-11-16 2013-05-22 AdrenoMed AG Anti-Adrenomedullin (ADM) antibody or anti-ADM antibody fragment or an anti-ADM non-Ig scaffold for use in therapy
WO2013072514A1 (en) 2011-11-16 2013-05-23 Adrenomed Ag Anti-adrenomedullin (adm) antibody or anti-adm antibody fragment or anti-adm non-ig scaffold for regulating the fluid balance in a patient having a chronic or acute disease
WO2013075048A1 (en) 2011-11-16 2013-05-23 Amgen Inc. Methods of treating epidermal growth factor deletion mutant viii related disorders
WO2013072513A1 (en) 2011-11-16 2013-05-23 Adrenomed Ag Anti-adrenomedullin (adm) antibody or anti-adm antibody fragment or anti-adm non-ig scaffold for use in therapy of an acute disease or acute condition of a patient for stabilizing the circulation
WO2013072509A1 (en) 2011-11-16 2013-05-23 Adrenomed Ag Adrenomedullin assays and methods for determining mature adrenomedullin
WO2013072511A1 (en) 2011-11-16 2013-05-23 Adrenomed Ag Anti-adrenomedullin (adm) antibody or anti-adm antibody fragment or anti-adm non-ig scaffold for prevention or reduction of organ dysfunction or organ failure in a patient having a chronic or acute disease or acute condition
WO2013081143A1 (en) 2011-11-30 2013-06-06 中外製薬株式会社 Drug containing carrier into cell for forming immune complex
WO2013084148A2 (en) 2011-12-05 2013-06-13 Novartis Ag Antibodies for epidermal growth factor receptor 3 (her3) directed to domain ii of her3
WO2013084147A2 (en) 2011-12-05 2013-06-13 Novartis Ag Antibodies for epidermal growth factor receptor 3 (her3)
WO2013093762A1 (en) 2011-12-21 2013-06-27 Novartis Ag Compositions and methods for antibodies targeting factor p
WO2013100120A1 (en) 2011-12-28 2013-07-04 中外製薬株式会社 Humanized anti-epiregulin antibody, and cancer therapeutic agent comprising said antibody as active ingredient
WO2013118858A1 (en) 2012-02-09 2013-08-15 中外製薬株式会社 Modified fc region of antibody
EP2628750A2 (en) 2006-09-08 2013-08-21 The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc. Targeted identification of immunogenic peptides
WO2013125667A1 (en) 2012-02-24 2013-08-29 中外製薬株式会社 ANTIGEN-BINDING MOLECULE FOR PROMOTING DISAPPEARANCE OF ANTIGEN VIA FcγRIIB
EP2641612A1 (en) 2008-02-05 2013-09-25 Bristol-Myers Squibb Company Alpha 5 - beta 1 antibodies and their uses
WO2013147153A1 (en) 2012-03-29 2013-10-03 株式会社未来創薬研究所 Anti-lamp5 antibody and utilization thereof
EP2647388A1 (en) 2007-02-16 2013-10-09 Merrimack Pharmaceuticals, Inc. Antibodies Against ERBB3 and Uses Thereof
WO2013150623A1 (en) 2012-04-04 2013-10-10 株式会社ペルセウスプロテオミクス Conjugate of anti-cdh3 (p-cadherin) antibody and drug
WO2013158859A1 (en) 2012-04-18 2013-10-24 Cell Signaling Technology, Inc. Egfr and ros1 in cancer
WO2013166448A1 (en) 2012-05-03 2013-11-07 Amgen Inc. Stable formulations containing anti-pcsk9 antibodies
WO2013173496A2 (en) 2012-05-18 2013-11-21 Seattle Genetics, Inc. Cd33 antibodies and use of same to treat cancer
WO2013180201A1 (en) 2012-05-30 2013-12-05 中外製薬株式会社 Antigen-binding molecule for eliminating aggregated antigens
WO2013180200A1 (en) 2012-05-30 2013-12-05 中外製薬株式会社 Target-tissue-specific antigen-binding molecule
WO2013188448A2 (en) 2012-06-11 2013-12-19 Amgen Inc. Dual receptor antagonistic antigen-binding proteins and uses thereof
WO2013187495A1 (en) 2012-06-14 2013-12-19 中外製薬株式会社 ANTIGEN-BINDING MOLECULE CONTAINING MODIFIED Fc REGION
US8613920B2 (en) 2007-07-27 2013-12-24 Janssen Alzheimer Immunotherapy Treatment of amyloidogenic diseases
WO2014008218A1 (en) 2012-07-02 2014-01-09 Bristol-Myers Squibb Company Optimization of antibodies that bind lymphocyte activation gene-3 (lag-3), and uses thereof
WO2014007402A1 (en) 2012-07-06 2014-01-09 京都府公立大学法人 Differentiation marker for and differentiation control for ocular cells
EP2692737A1 (en) 2007-04-20 2014-02-05 Biotie Therapies Corp. Fully human anti-vap-1 monoclonal antibodies
WO2014030728A1 (en) 2012-08-24 2014-02-27 中外製薬株式会社 Fcγriib-specific fc region variant
WO2014030750A1 (en) 2012-08-24 2014-02-27 中外製薬株式会社 MOUSE FcγRII-SPECIFIC Fc ANTIBODY
WO2014037899A2 (en) 2012-09-07 2014-03-13 Novartis Ag Il-18 binding molecules
WO2014039983A1 (en) 2012-09-07 2014-03-13 The Trustees Of Dartmouth College Vista modulators for diagnosis and treatment of cancer
WO2014050926A1 (en) 2012-09-28 2014-04-03 中外製薬株式会社 Method for evaluating blood coagulation reaction
WO2014051022A1 (en) 2012-09-27 2014-04-03 中外製薬株式会社 Fgfr3 fusion gene and pharmaceutical drug targeting same
WO2014055543A2 (en) 2012-10-01 2014-04-10 Millennium Pharmaceuticals, Inc. Biomarkers and methods to predict response to inhibitors and uses thereof
WO2014074905A1 (en) 2012-11-08 2014-05-15 Eleven Biotherapeutics, Inc. Il-6 antagonists and uses thereof
WO2014073641A1 (en) 2012-11-08 2014-05-15 国立大学法人 宮崎大学 Antibody capable of specifically recognizing transferrin receptor
WO2014084859A1 (en) 2012-11-30 2014-06-05 Novartis Ag Molecules and methods for modulating tmem16a activities
WO2014089111A1 (en) 2012-12-05 2014-06-12 Novartis Ag Compositions and methods for antibodies targeting epo
WO2014099997A1 (en) 2012-12-18 2014-06-26 Novartis Ag Compositions and methods that utilize a peptide tag that binds to hyaluronan
WO2014104165A1 (en) 2012-12-27 2014-07-03 中外製薬株式会社 Heterodimerized polypeptide
US8784810B2 (en) 2006-04-18 2014-07-22 Janssen Alzheimer Immunotherapy Treatment of amyloidogenic diseases
WO2014114801A1 (en) 2013-01-25 2014-07-31 Amgen Inc. Antibodies targeting cdh19 for melanoma
WO2014114800A1 (en) 2013-01-25 2014-07-31 Amgen Research (Munich) Gmbh Antibody constructs for cdh19 and cd3
WO2014122613A1 (en) 2013-02-08 2014-08-14 Novartis Ag Anti-il-17a antibodies and their use in treating autoimmune and inflammatory disorders
WO2014123580A1 (en) 2013-02-06 2014-08-14 Inhibrx Llc Non-platelet depleting and non-red blood cell depleting cd47 antibodies and methods of use thereof
EP2769993A1 (en) 2007-12-14 2014-08-27 Novo Nordisk A/S Antibodies against human NKG2D and uses thereof
WO2014140368A1 (en) 2013-03-15 2014-09-18 Amgen Research (Munich) Gmbh Antibody constructs for influenza m2 and cd3
WO2014141189A1 (en) 2013-03-14 2014-09-18 Erasmus University Medical Center Transgenic non-human mammal for antibody production
WO2014140358A1 (en) 2013-03-15 2014-09-18 Amgen Research (Munich) Gmbh Single chain binding molecules comprising n-terminal abp
WO2014147153A1 (en) 2013-03-20 2014-09-25 Sphingotec Gmbh Adrenomedullin to guide therapy of blood pressure decline
WO2014159239A2 (en) 2013-03-14 2014-10-02 Novartis Ag Antibodies against notch 3
WO2014163101A1 (en) 2013-04-02 2014-10-09 中外製薬株式会社 Fc region variant
WO2014165271A2 (en) 2013-03-13 2014-10-09 Neotope Biosciences Limited Tau immunotherapy
US8883157B1 (en) 2013-12-17 2014-11-11 Kymab Limited Targeting rare human PCSK9 variants for cholesterol treatment
WO2014185550A1 (en) 2013-05-16 2014-11-20 Kyoto University Method for determining prognosis of cancer
WO2014200018A1 (en) 2013-06-11 2014-12-18 独立行政法人 国立精神・神経医療研究センター Method for predicting post-therapy prognosis of relapsing-remitting multiple sclerosis (rrms) patient, and method for determining applicability of novel therapy
US8916165B2 (en) 2004-12-15 2014-12-23 Janssen Alzheimer Immunotherapy Humanized Aβ antibodies for use in improving cognition
WO2014205300A2 (en) 2013-06-21 2014-12-24 Novartis Ag Lectin-like oxidized ldl receptor1 antibodies and methods of use
WO2014208482A1 (en) 2013-06-24 2014-12-31 中外製薬株式会社 Therapeutic agent comprising humanized anti-epiregulin antibody as active ingredient for non-small-cell lung carcinoma excluding adenocarcinoma
WO2014209384A1 (en) 2013-06-28 2014-12-31 Amgen Inc. Methods for treating homozygous familial hypercholesterolema
EP2824183A1 (en) 2005-04-08 2015-01-14 Chugai Seiyaku Kabushiki Kaisha Antibody substituting for function of blood coagulation factor VIII
WO2015009740A2 (en) 2013-07-15 2015-01-22 Cell Signaling Technology, Inc. Anti-mucin 1 binding agents and uses thereof
US8945560B1 (en) 2014-07-15 2015-02-03 Kymab Limited Method of treating rheumatoid arthritis using antibody to IL6R
WO2015022658A2 (en) 2013-08-14 2015-02-19 Novartis Ag Methods of treating sporadic inclusion body myositis
US8980273B1 (en) 2014-07-15 2015-03-17 Kymab Limited Method of treating atopic dermatitis or asthma using antibody to IL4RA
US8986691B1 (en) 2014-07-15 2015-03-24 Kymab Limited Method of treating atopic dermatitis or asthma using antibody to IL4RA
US8986694B1 (en) 2014-07-15 2015-03-24 Kymab Limited Targeting human nav1.7 variants for treatment of pain
WO2015041310A1 (en) 2013-09-20 2015-03-26 中外製薬株式会社 Treatment for hemorrhagic diseases by anti-protein-c antibody
US8992927B1 (en) 2014-07-15 2015-03-31 Kymab Limited Targeting human NAV1.7 variants for treatment of pain
EP2853544A1 (en) 2007-11-15 2015-04-01 Chugai Seiyaku Kabushiki Kaisha Monoclonal antibody capable of binding to anexelekto, and use thereof
WO2015046467A1 (en) 2013-09-27 2015-04-02 中外製薬株式会社 Method for producing polypeptide heteromultimer
US8999341B1 (en) 2014-07-15 2015-04-07 Kymab Limited Targeting rare human PCSK9 variants for cholesterol treatment
US9017678B1 (en) 2014-07-15 2015-04-28 Kymab Limited Method of treating rheumatoid arthritis using antibody to IL6R
EP2865754A1 (en) 1999-06-14 2015-04-29 BP Corporation North America Inc. Synthetic ligation reassembly in directed evolution
WO2015068847A1 (en) 2013-11-11 2015-05-14 中外製薬株式会社 Antigen-binding molecule containing modified antibody variable region
US9034332B1 (en) 2014-07-15 2015-05-19 Kymab Limited Precision medicine by targeting rare human PCSK9 variants for cholesterol treatment
US9045541B2 (en) 2012-02-06 2015-06-02 Inhibrx Llc CD47 antibodies and methods of use thereof
US9045548B1 (en) 2014-07-15 2015-06-02 Kymab Limited Precision Medicine by targeting rare human PCSK9 variants for cholesterol treatment
US9045545B1 (en) 2014-07-15 2015-06-02 Kymab Limited Precision medicine by targeting PD-L1 variants for treatment of cancer
US9051363B2 (en) 1997-12-02 2015-06-09 Janssen Sciences Ireland Uc Humanized antibodies that recognize beta amyloid peptide
US9051378B1 (en) 2014-07-15 2015-06-09 Kymab Limited Targeting rare human PCSK9 variants for cholesterol treatment
WO2015083764A1 (en) 2013-12-04 2015-06-11 中外製薬株式会社 Antigen-binding molecules, the antigen-binding activity of which varies according to the concentration of compounds, and libraries of said molecules
US9062116B2 (en) 2009-04-23 2015-06-23 Infinity Pharmaceuticals, Inc. Anti-fatty acid amide hydrolase-2 antibodies and uses thereof
US9062111B2 (en) 2005-12-07 2015-06-23 Medarex, L.L.C. CTLA-4 antibody dosage escalation regimens
US9062105B1 (en) 2014-07-15 2015-06-23 Kymab Limited Precision Medicine by targeting VEGF-A variants for treatment of retinopathy
US9067998B1 (en) 2014-07-15 2015-06-30 Kymab Limited Targeting PD-1 variants for treatment of cancer
US9067981B1 (en) 2008-10-30 2015-06-30 Janssen Sciences Ireland Uc Hybrid amyloid-beta antibodies
WO2015099165A1 (en) 2013-12-27 2015-07-02 中外製薬株式会社 Method for purifying antibody having low isoelectric point
WO2015099127A1 (en) 2013-12-27 2015-07-02 中外製薬株式会社 Fgfr gatekeeper mutant gene and drug targeting same
WO2015105888A1 (en) 2014-01-07 2015-07-16 Bioatla, Llc Proteins targeting orthologs
EP2905030A1 (en) 2008-08-11 2015-08-12 E. R. Squibb & Sons, L.L.C. Human antibodies that bind lymphocyte activation gene-3 (LAG-3) and uses thereof
EP2907873A1 (en) 2009-07-17 2015-08-19 Bioatla LLC Simulataneous, integrated selection and evolution of antibody/protein performance and expression in production hosts
WO2015121383A1 (en) 2014-02-12 2015-08-20 Michael Uhlin Bispecific antibodies for use in stem cell transplantation
EP2918605A1 (en) 2007-11-12 2015-09-16 U3 Pharma GmbH Axl antibodies
WO2015136471A1 (en) 2014-03-12 2015-09-17 Prothena Biosciences Limited Anti-laminin4 antibodies specific for lg1-3
WO2015136469A1 (en) 2014-03-12 2015-09-17 Prothena Biosciences Limited Anti-mcam antibodies and associated methods of use
WO2015136472A1 (en) 2014-03-12 2015-09-17 Prothena Biosciences Limited Anti-laminin4 antibodies specific for lg4-5
WO2015136470A1 (en) 2014-03-12 2015-09-17 Prothena Biosciences Limited Anti-mcam antibodies and associated methods of use
US9139648B1 (en) 2014-07-15 2015-09-22 Kymab Limited Precision medicine by targeting human NAV1.9 variants for treatment of pain
US9150660B1 (en) 2014-07-15 2015-10-06 Kymab Limited Precision Medicine by targeting human NAV1.8 variants for treatment of pain
US9150658B2 (en) 2008-12-09 2015-10-06 Genmab A/S Human antibodies against tissue factor and methods of use thereof
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
EP2927244A1 (en) 2008-09-19 2015-10-07 MedImmune, LLC Antibodies directed to DLL4 and uses thereof
US9168314B2 (en) 2010-06-15 2015-10-27 Genmab A/S Human antibody drug conjugates against tissue factor
WO2015162590A1 (en) 2014-04-24 2015-10-29 Novartis Ag Methods of improving or accelerating physical recovery after surgery for hip fracture
WO2015187835A2 (en) 2014-06-06 2015-12-10 Bristol-Myers Squibb Company Antibodies against glucocorticoid-induced tumor necrosis factor receptor (gitr) and uses thereof
WO2015195917A1 (en) 2014-06-18 2015-12-23 Mersana Therapeutics, Inc. Monoclonal antibodies against her2 epitope and methods of use thereof
WO2015198243A2 (en) 2014-06-25 2015-12-30 Novartis Ag Compositions and methods for long acting proteins
WO2015198240A2 (en) 2014-06-25 2015-12-30 Novartis Ag Compositions and methods for long acting proteins
WO2015198217A2 (en) 2013-02-08 2015-12-30 Novartis Ag Compositions and methods for long-acting antibodies targeting il-17
US9243064B2 (en) 2003-01-31 2016-01-26 Celldex Therapeutics Inc. Antibody vaccine conjugates and uses therefor
WO2016016442A1 (en) 2014-08-01 2016-02-04 INSERM (Institut National de la Santé et de la Recherche Médicale) An anti-cd45rc antibody for use as drug
WO2016016859A1 (en) 2014-07-31 2016-02-04 Amgen Research (Munich) Gmbh Optimized cross-species specific bispecific single chain antibody constructs
WO2016016412A1 (en) 2014-07-31 2016-02-04 Amgen Research (Munich) Gmbh Antibody constructs for cdh19 and cd3
WO2016016415A1 (en) 2014-07-31 2016-02-04 Amgen Research (Munich) Gmbh Bispecific single chain antibody construct with enhanced tissue distribution
EP2982379A1 (en) 2005-07-01 2016-02-10 E. R. Squibb & Sons, L.L.C. Human monoclonal antibodies to programmed death ligand 1 (pd-l1)
WO2016020882A2 (en) 2014-08-07 2016-02-11 Novartis Ag Angiopoetin-like 4 (angptl4) antibodies and methods of use
WO2016020880A2 (en) 2014-08-07 2016-02-11 Novartis Ag Angiopoietin-like 4 antibodies and methods of use
US9259459B2 (en) 2003-01-31 2016-02-16 Celldex Therapeutics Inc. Antibody vaccine conjugates and uses therefor
WO2016039796A2 (en) 2014-09-12 2016-03-17 Regeneron Pharmaceuticals, Inc. Treatment of fibrodysplasia ossificans progressiva
US9289511B2 (en) 2007-06-21 2016-03-22 The Boeing Company Bioconjugated nanoparticles
US9290489B2 (en) 2012-07-06 2016-03-22 Duke University Activation of TRPV4 ion channel by physical stimuli and critical role for TRPV4 in organ-specific inflammation and itch
WO2016044227A1 (en) 2014-09-15 2016-03-24 Abvitro, Inc. High-throughput nucleotide library sequencing
US9309315B2 (en) 2005-08-18 2016-04-12 Genmab A/S Therapy with CD4 binding peptides and radiation
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
WO2016073401A1 (en) 2014-11-03 2016-05-12 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
US9346873B2 (en) 2008-09-30 2016-05-24 Ablexis, Llc Non-human mammals for the production of chimeric antibodies
EP3023502A1 (en) 2008-04-10 2016-05-25 Cell Signaling Technology, Inc. Compositions and methods for detecting egfr mutations in cancer
WO2016081748A2 (en) 2014-11-21 2016-05-26 Bristol-Myers Squibb Company Antibodies against cd73 and uses thereof
WO2016098079A2 (en) 2014-12-19 2016-06-23 Novartis Ag Compositions and methods for antibodies targeting bmp6
EP3050963A1 (en) 2005-03-31 2016-08-03 Chugai Seiyaku Kabushiki Kaisha Process for production of polypeptide by regulation of assembly
WO2016120811A1 (en) 2015-01-28 2016-08-04 Prothena Biosciences Limited Anti-transthyretin antibodies
WO2016120810A1 (en) 2015-01-28 2016-08-04 Prothena Biosciences Limited Anti-transthyretin antibodies
WO2016120809A1 (en) 2015-01-28 2016-08-04 Prothena Biosciences Limited Anti-transthyretin antibodies
WO2016125495A1 (en) 2015-02-05 2016-08-11 Chugai Seiyaku Kabushiki Kaisha Antibodies comprising an ion concentration dependent antigen-binding domain, fc region variants, il-8-binding antibodies, and uses therof
US9428574B2 (en) 2011-06-30 2016-08-30 Compugen Ltd. Polypeptides and uses thereof for treatment of autoimmune disorders and infection
US9434782B2 (en) 2009-07-08 2016-09-06 Kymab Limited Animal models and therapeutic molecules
WO2016149088A1 (en) 2015-03-13 2016-09-22 Bristol-Myers Squibb Company Use of alkaline washes during chromatography to remove impurities
EP3072963A1 (en) 2007-10-18 2016-09-28 Cell Signaling Technology, Inc. Translocation and mutant ros kinase in human non-small cell lung carcinoma
WO2016153983A1 (en) 2015-03-20 2016-09-29 Bristol-Myers Squibb Company Use of dextran for protein purification
WO2016153978A1 (en) 2015-03-20 2016-09-29 Bristol-Myers Squibb Company Use of dextran to enhance protein purification by affinity chromatography
WO2016166360A1 (en) 2015-04-17 2016-10-20 Bayer Pharma Aktiengesellschaft Bispecific antibody constructs for cdh3 and cd3
WO2016166014A1 (en) 2015-04-17 2016-10-20 F. Hoffmann-La Roche Ag Combination therapy with coagulation factors and multispecific antibodies
WO2016172726A1 (en) 2015-04-24 2016-10-27 The Regents Of The University Of California Modulators of ror1-ror2 binding
WO2016176341A1 (en) 2015-04-29 2016-11-03 Regeneron Pharmaceuticals, Inc. Treatment of fibrodysplasia ossificans progressiva
US9504236B2 (en) 2009-07-08 2016-11-29 Kymab Limited Animal models and therapeutic molecules
WO2016193872A2 (en) 2015-06-05 2016-12-08 Novartis Ag Antibodies targeting bone morphogenetic protein 9 (bmp9) and methods therefor
WO2016196228A1 (en) 2015-05-29 2016-12-08 Bristol-Myers Squibb Company Antibodies against ox40 and uses thereof
US9535076B2 (en) 2002-09-12 2017-01-03 The Regents Of The University Of California Methods and compositions for eliciting an amyloid-selective immune response
WO2017004016A1 (en) 2015-06-29 2017-01-05 The Rockefeller University Antibodies to cd40 with enhanced agonist activity
US9546214B2 (en) 2014-04-04 2017-01-17 Bionomics, Inc. Humanized antibodies that bind LGR5
EP3127921A1 (en) 2007-09-26 2017-02-08 Chugai Seiyaku Kabushiki Kaisha Method of modifying isoelectric point of antibody via amino acid substition in cdr
WO2017021370A1 (en) 2015-07-31 2017-02-09 Amgen Research (Munich) Gmbh Bispecific antibody constructs binding egfrviii and cd3
WO2017021356A1 (en) 2015-07-31 2017-02-09 Amgen Research (Munich) Gmbh Bispecific antibody constructs binding mesothelin and cd3
WO2017021893A1 (en) 2015-08-03 2017-02-09 Novartis Ag Methods of treating fgf21-associated disorders
WO2017021362A1 (en) 2015-07-31 2017-02-09 Amgen Research (Munich) Gmbh Antibody constructs for flt3 and cd3
WO2017021354A1 (en) 2015-07-31 2017-02-09 Amgen Research (Munich) Gmbh Antibody constructs for cd70 and cd3
WO2017021349A1 (en) 2015-07-31 2017-02-09 Amgen Research (Munich) Gmbh Bispecific antibody constructs binding dll3 and cd3
US9580491B2 (en) 2010-03-31 2017-02-28 Ablexis, Llc Genetic engineering of non-human animals for the production of chimeric antibodies
US9585374B2 (en) 2004-10-22 2017-03-07 Revivicor, Inc. Ungulates with genetically modified immune systems
WO2017042701A1 (en) 2015-09-09 2017-03-16 Novartis Ag Thymic stromal lymphopoietin (tslp)-binding antibodies and methods of using the antibodies
WO2017046774A2 (en) 2015-09-16 2017-03-23 Prothena Biosciences Limited Use of anti-mcam antibodies for treatment or prophylaxis of giant cell arteritis, polymyalgia rheumatica or takayasu's arteritis
WO2017046994A1 (en) 2015-09-18 2017-03-23 Chugai Seiyaku Kabushiki Kaisha Il-8-binding antibodies and uses thereof
WO2017046776A2 (en) 2015-09-16 2017-03-23 Prothena Biosciences Limited Use of anti-mcam antibodies for treatment or prophylaxis of giant cell arteritis, polymyalgia rheumatica or takayasu's arteritis
WO2017053905A1 (en) 2015-09-24 2017-03-30 Abvitro Llc Affinity-oligonucleotide conjugates and uses thereof
WO2017053902A1 (en) 2015-09-25 2017-03-30 Abvitro Llc High throughput process for t cell receptor target identification of natively-paired t cell receptor sequences
US9617336B2 (en) 2012-02-01 2017-04-11 Compugen Ltd C10RF32 antibodies, and uses thereof for treatment of cancer
EP3156420A1 (en) 2010-12-06 2017-04-19 Seattle Genetics, Inc. Humanized antibodies to liv-1 and use of same to treat cancer
WO2017075484A2 (en) 2015-10-30 2017-05-04 Galaxy Biotech, Llc Highly potent antibodies binding to death receptor 4 and death receptor 5
EP3165540A1 (en) 2010-04-13 2017-05-10 Celldex Therapeutics, Inc. Antibodies that bind human cd27 and uses thereof
WO2017086419A1 (en) 2015-11-18 2017-05-26 中外製薬株式会社 Method for enhancing humoral immune response
WO2017087678A2 (en) 2015-11-19 2017-05-26 Bristol-Myers Squibb Company Antibodies against glucocorticoid-induced tumor necrosis factor receptor (gitr) and uses thereof
WO2017086367A1 (en) 2015-11-18 2017-05-26 中外製薬株式会社 Combination therapy using t cell redirection antigen binding molecule against cell having immunosuppressing function
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
WO2017095875A1 (en) 2015-11-30 2017-06-08 Bristol-Myers Squibb Company Anti human ip-10 antibodies and their uses
WO2017103895A1 (en) 2015-12-18 2017-06-22 Novartis Ag Antibodies targeting cd32b and methods of use thereof
WO2017110980A1 (en) 2015-12-25 2017-06-29 中外製薬株式会社 Antibody having enhanced activity, and method for modifying same
WO2017125897A1 (en) 2016-01-21 2017-07-27 Novartis Ag Multispecific molecules targeting cll-1
US9738701B2 (en) 2003-05-30 2017-08-22 Merus N.V. Method for selecting a single cell expressing a heterogeneous combination of antibodies
WO2017151176A1 (en) 2016-03-04 2017-09-08 The Rockefeller University Antibodies to cd40 with enhanced agonist activity
WO2017149513A1 (en) 2016-03-03 2017-09-08 Prothena Biosciences Limited Anti-mcam antibodies and associated methods of use
WO2017152085A1 (en) 2016-03-04 2017-09-08 Bristol-Myers Squibb Company Combination therapy with anti-cd73 antibodies
US9758805B2 (en) 2012-04-20 2017-09-12 Merus N.V. Methods and means for the production of Ig-like molecules
WO2017153953A1 (en) 2016-03-09 2017-09-14 Prothena Biosciences Limited Use of anti-mcam antibodies for treatment or prophylaxis of granulomatous lung diseases
WO2017153955A1 (en) 2016-03-09 2017-09-14 Prothena Biosciences Limited Use of anti-mcam antibodies for treatment or prophylaxis of granulomatous lung diseases
WO2017159287A1 (en) 2016-03-14 2017-09-21 中外製薬株式会社 Cell injury inducing therapeutic drug for use in cancer therapy
WO2017160754A1 (en) 2016-03-15 2017-09-21 Mersana Therapeutics,Inc. Napi2b-targeted antibody-drug conjugates and methods of use thereof
US9783618B2 (en) 2013-05-01 2017-10-10 Kymab Limited Manipulation of immunoglobulin gene diversity and multi-antibody therapeutics
US9783593B2 (en) 2013-05-02 2017-10-10 Kymab Limited Antibodies, variable domains and chains tailored for human use
WO2017177200A1 (en) 2016-04-07 2017-10-12 Duke University Small molecule dual-inhibitors of trpv4 and trpa1 for sanitizing and anesthetizing
US9788534B2 (en) 2013-03-18 2017-10-17 Kymab Limited Animal models and therapeutic molecules
WO2017182561A1 (en) 2016-04-21 2017-10-26 Sphingotec Therapeutics Gmbh Methods for determining dpp3 and therapeutic methods
WO2017182427A1 (en) 2016-04-19 2017-10-26 Amgen Research (Munich) Gmbh Administration of a bispecific construct binding to cd33 and cd3 for use in a method for the treatment of myeloid leukemia
US9803017B2 (en) 2013-07-05 2017-10-31 University Of Washington Through Its Center For Commercialization Soluble MIC neutralizing monoclonal antibody for treating cancer
WO2017191560A1 (en) 2016-05-02 2017-11-09 Prothena Biosciences Limited Antibodies recognizing tau
WO2017191561A1 (en) 2016-05-02 2017-11-09 Prothena Biosciences Limited Antibodies recognizing tau
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
US9816088B2 (en) 2013-03-15 2017-11-14 Abvitro Llc Single cell bar-coding for antibody discovery
WO2017196663A1 (en) 2016-05-09 2017-11-16 Bristol-Myers Squibb Company Tl1a antibodies and uses thereof
US9829494B2 (en) 2005-12-01 2017-11-28 Adrenomed Ag Methods of treatment using ADM antibodies
WO2017208210A1 (en) 2016-06-03 2017-12-07 Prothena Biosciences Limited Anti-mcam antibodies and associated methods of use
US9840553B2 (en) 2014-06-28 2017-12-12 Kodiak Sciences Inc. Dual PDGF/VEGF antagonists
EP3255144A1 (en) 2007-08-10 2017-12-13 E. R. Squibb & Sons, L.L.C. Recombineering construct for preparing transgenic mice capable of producing human immunoglobulin
WO2017214167A1 (en) 2016-06-06 2017-12-14 City Of Hope Baff-r targeted chimeric antigen receptor-modified t-cells and uses thereof
WO2017214170A2 (en) 2016-06-06 2017-12-14 City Of Hope Baff-r antibodies and uses thereof
WO2017216724A1 (en) 2016-06-15 2017-12-21 Novartis Ag Methods for treating disease using inhibitors of bone morphogenetic protein 6 (bmp6)
WO2018002081A1 (en) 2016-06-27 2018-01-04 Aicuris Anti-Infective Cures Gmbh Hcmv entry inhibitors
WO2018007923A2 (en) 2016-07-02 2018-01-11 Prothena Biosciences Limited Anti-transthyretin antibodies
WO2018007922A2 (en) 2016-07-02 2018-01-11 Prothena Biosciences Limited Anti-transthyretin antibodies
WO2018007588A1 (en) 2016-07-08 2018-01-11 Sphingotec Gmbh Adrenomedullin for assessing congestion in a subject with acute heart failure
WO2018007924A2 (en) 2016-07-02 2018-01-11 Prothena Biosciences Limited Anti-transthyretin antibodies
WO2018013818A2 (en) 2016-07-14 2018-01-18 Bristol-Myers Squibb Company Antibodies against tim3 and uses thereof
EP2786657B1 (en) 2001-02-16 2018-02-07 Regeneron Pharmaceuticals, Inc. A method of producing an antibody comprising a human variable region and a rodent constant region.
EP3279215A1 (en) 2009-11-24 2018-02-07 MedImmune Limited Targeted binding agents against b7-h1
WO2018031726A1 (en) 2016-08-12 2018-02-15 Bristol-Myers Squibb Company Methods of purifying proteins
WO2018029586A1 (en) 2016-08-07 2018-02-15 Novartis Ag Mrna-mediated immunization methods
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
WO2018049261A1 (en) 2016-09-09 2018-03-15 Icellhealth Consulting Llc Oncolytic virus expressing immune checkpoint modulators
US9924705B2 (en) 2012-03-28 2018-03-27 Kymab Limited Animal models and therapeutic molecules
WO2018057051A1 (en) 2016-09-24 2018-03-29 Abvitro Llc Affinity-oligonucleotide conjugates and uses thereof
US9963716B2 (en) 2011-09-26 2018-05-08 Kymab Limited Chimaeric surrogate light chains (SLC) comprising human VpreB
WO2018087720A1 (en) 2016-11-14 2018-05-17 Novartis Ag Compositions, methods, and therapeutic uses related to fusogenic protein minion
US9975966B2 (en) 2014-09-26 2018-05-22 Chugai Seiyaku Kabushiki Kaisha Cytotoxicity-inducing theraputic agent
EP3327035A1 (en) 2010-06-22 2018-05-30 Precision Biologics Inc. Colon and pancreas cancer specific antigens and antibodies
WO2018097308A1 (en) 2016-11-28 2018-05-31 中外製薬株式会社 Ligand-binding molecule having adjustable ligand binding activity
US9999620B2 (en) 2010-08-16 2018-06-19 Duke University CaMKK-β as a target for treating cancer
EP3336104A1 (en) 2012-12-28 2018-06-20 Precision Biologics, Inc. Humanized monoclonal antibodies and methods of use for the diagnosis and treatment of colon and pancreas cancer
WO2018109228A1 (en) 2016-12-16 2018-06-21 Adrenomed Ag Anti-adrenomedullin (adm) antibody or anti-adm antibody fragment or anti-adm non-ig scaffold for use in intervention and therapy of congestion in a patient in need thereof
EP3339324A1 (en) 2016-12-22 2018-06-27 sphingotec GmbH Anti-adrenomedullin (adm) antibody or anti-adm antibody fragment or anti-adm non-ig scaffold for use in intervention and therapy of congestion in a patient in need thereof
EP3338794A1 (en) 2012-07-13 2018-06-27 The Trustees of the University of Pennsylvania Toxicity management for anti-tumor activity of cars
WO2018129451A2 (en) 2017-01-09 2018-07-12 Merrimack Pharmaceuticals, Inc. Anti-fgfr antibodies and methods of use
US10034921B2 (en) 2013-02-13 2018-07-31 Laboratoire Français Du Fractionnement Et Des Biotechnologies Proteins with modified glycosylation and methods of production thereof
WO2018141910A1 (en) 2017-02-02 2018-08-09 Amgen Research (Munich) Gmbh Low ph pharmaceutical composition comprising t cell engaging antibody constructs
WO2018146594A1 (en) 2017-02-08 2018-08-16 Novartis Ag Fgf21 mimetic antibodies and uses thereof
US10053510B2 (en) 2013-05-24 2018-08-21 Promis Neurosciences Inc. FasR antibodies and methods of use
WO2018151821A1 (en) 2017-02-17 2018-08-23 Bristol-Myers Squibb Company Antibodies to alpha-synuclein and uses thereof
US10059746B2 (en) 2011-04-04 2018-08-28 University Of Iowa Research Foundation Methods of improving vaccine immunogenicity
US10077304B2 (en) 2013-08-14 2018-09-18 The Governing Council Of The University Of Toronto Antibodies against frizzled receptor
WO2018174274A1 (en) 2017-03-24 2018-09-27 全薬工業株式会社 ANTI-IgM/B CELL SURFACE ANTIGEN BISPECIFIC ANTIBODY
WO2018175460A1 (en) 2017-03-24 2018-09-27 Novartis Ag Methods for preventing and treating heart disease
WO2018181870A1 (en) 2017-03-31 2018-10-04 公立大学法人奈良県立医科大学 Medicinal composition usable for preventing and/or treating blood coagulation factor ix abnormality, comprising multispecific antigen binding molecule replacing function of blood coagulation factor viii
WO2018187613A2 (en) 2017-04-07 2018-10-11 Bristol-Myers Squibb Company Anti-icos agonist antibodies and uses thereof
WO2018199214A1 (en) 2017-04-27 2018-11-01 中外製薬株式会社 Coagulation factor ix with improved pharmacokinetics
WO2018204907A1 (en) 2017-05-05 2018-11-08 Amgen Inc. Pharmaceutical composition comprising bispecific antibody constructs for improved storage and administration
WO2018203545A1 (en) 2017-05-02 2018-11-08 国立研究開発法人国立精神・神経医療研究センター Method for predicting and evaluating therapeutic effect in diseases related to il-6 and neutrophils
US10132818B2 (en) 2014-07-08 2018-11-20 New York University Tau imaging ligands and their uses in the diagnosis and treatment of tauopathy
WO2018218222A1 (en) 2017-05-26 2018-11-29 Goldfless Stephen Jacob High-throughput polynucleotide library sequencing and transcriptome analysis
US10149461B2 (en) 2008-10-27 2018-12-11 Revivicor, Inc. Immunocompromised ungulates
US10150811B2 (en) 2011-10-13 2018-12-11 Aerpio Therapeutics, Inc. Methods for treating vascular leak syndrome and cancer
US10149462B2 (en) 2013-10-01 2018-12-11 Kymab Limited Animal models and therapeutic molecules
WO2018229715A1 (en) 2017-06-16 2018-12-20 Novartis Ag Compositions comprising anti-cd32b antibodies and methods of use thereof
WO2018229251A1 (en) 2017-06-16 2018-12-20 Imba - Institut Für Molekulare Biotechnologie Gmbh Blood vessel organoid, methods of producing and using said organoids
EP3421486A1 (en) 2012-06-22 2019-01-02 The Trustees Of Dartmouth College Novel vista-ig constructs and the use of vista-ig for treatment of autoimmune, allergic and inflammatory disorders
WO2019003104A1 (en) 2017-06-28 2019-01-03 Novartis Ag Methods for preventing and treating urinary incontinence
US10174110B2 (en) 2013-02-13 2019-01-08 Laboratoire Français Du Fractionnement Et Des Biotechnologies Highly galactosylated anti-TNF-α antibodies and uses thereof
WO2019016247A2 (en) 2017-07-20 2019-01-24 H. Lundbeck A/S Agents, uses and methods for treatment
EP3435087A1 (en) 2010-07-16 2019-01-30 Bioatla LLC Novel methods of protein evolution
US10196439B2 (en) 2015-07-13 2019-02-05 H. Lundbeck A/S Antibodies specific for hyperphosphorylated tau and methods of use thereof
WO2019051335A1 (en) 2017-09-07 2019-03-14 Juno Therapeutics, Inc. Methods of identifying cellular attributes related to outcomes associated with cell therapy
US10232039B2 (en) 2011-04-12 2019-03-19 Duke University Compositions and methods for the treatment of tissue fibrosis
WO2019057992A2 (en) 2017-09-25 2019-03-28 Adrenomed Ag Anti-adrenomedullin (adm) binder for use in therapy or prevention of symptoms of illness
US10251377B2 (en) 2012-03-28 2019-04-09 Kymab Limited Transgenic non-human vertebrate for the expression of class-switched, fully human, antibodies
WO2019077082A1 (en) 2017-10-18 2019-04-25 Adrenomed Ag Therapy monitoring under treatment with an anti-adrenomedullin (adm) binder
WO2019078344A1 (en) 2017-10-20 2019-04-25 学校法人兵庫医科大学 Anti-il-6 receptor antibody-containing medicinal composition for preventing post-surgical adhesion
WO2019081983A1 (en) 2017-10-25 2019-05-02 Novartis Ag Antibodies targeting cd32b and methods of use thereof
WO2019081595A2 (en) 2017-10-25 2019-05-02 Sphingotec Therapeutics Gmbh Dpp3 binder directed to and binding to specific dpp3-epitopes and its use in the prevention or treatment of diseases / acute conditions that are associated with oxidative stress
US10301391B2 (en) 2016-02-03 2019-05-28 Amgen Research (Munich) Gmbh BCMA and CD3 bispecific T cell engaging antibody constructs
WO2019107384A1 (en) 2017-11-28 2019-06-06 中外製薬株式会社 Ligand-binding molecule having adjustable ligand-binding activity
WO2019109016A1 (en) 2017-12-01 2019-06-06 Millennium Pharmaceuticals, Inc. Biomarkers and methods for treatment with nae inhibitors
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
EP3498296A1 (en) 2012-10-08 2019-06-19 Prothena Biosciences Limited Antibodies recognizing alpha-synuclein
WO2019118426A1 (en) 2017-12-11 2019-06-20 Amgen Inc. Continuous manufacturing process for bispecific antibody products
US10329265B2 (en) 2014-08-22 2019-06-25 Duke University TRPA1 and TRPV4 inhibitors and methods of using the same for organ-specific inflammation and itch
WO2019133961A1 (en) 2017-12-29 2019-07-04 Amgen Inc. Bispecific antibody construct directed to muc17 and cd3
WO2019131988A1 (en) 2017-12-28 2019-07-04 Chugai Seiyaku Kabushiki Kaisha Cytotoxicity-inducing therapeutic agent
WO2019140216A1 (en) 2018-01-12 2019-07-18 Amgen Inc. Pac1 antibodies and uses thereof
WO2019140229A1 (en) 2018-01-12 2019-07-18 Bristol-Myers Squibb Company Antibodies against tim3 and uses thereof
US10363290B2 (en) 2014-10-17 2019-07-30 Kodiak Sciences Inc. Butyrylcholinesterase zwitterionic polymer conjugates
US10370455B2 (en) 2014-12-05 2019-08-06 Immunext, Inc. Identification of VSIG8 as the putative VISTA receptor (V-R) and use thereof to produce VISTA/VSIG8 agonists and antagonists
EP3520816A2 (en) 2009-10-23 2019-08-07 Millennium Pharmaceuticals, Inc. Anti-gcc antibody molecules and related compositions and methods
WO2019151418A1 (en) 2018-01-31 2019-08-08 元一 加藤 Therapeutic agent for asthma containing il-6 inhibitor
WO2019150309A1 (en) 2018-02-02 2019-08-08 Hammack Scott Modulators of gpr68 and uses thereof for treating and preventing diseases
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
WO2019154900A1 (en) 2018-02-08 2019-08-15 Sphingotec Gmbh Adrenomedullin (adm) for diagnosis and/or prediction of dementia and anti-adrenomedullin binder for use in therapy or prevention of dementia
US10421824B2 (en) 2013-03-13 2019-09-24 Amgen Inc. Proteins specific for BAFF and B7RP1
US10421823B2 (en) 2013-03-13 2019-09-24 Amgen Inc. Proteins specific for BAFF and B7RP1 and uses thereof
WO2019191416A1 (en) 2018-03-29 2019-10-03 Bristol-Myers Squibb Company Methods of purifying monomeric monoclonal antibodies
EP3553089A1 (en) 2012-05-10 2019-10-16 Bioatla, LLC Multi-specific monoclonal antibodies
US10472415B2 (en) 2016-07-12 2019-11-12 H. Lundbeck A/S Antibodies specific for hyperphosphorylated tau and methods of use
EP3569614A1 (en) 2018-05-18 2019-11-20 Julius-Maximilians-Universität Würzburg Compounds and methods for the immobilization of myostatin-inhibitors on the extracellular matrix by transglutaminase
WO2019225568A1 (en) 2018-05-21 2019-11-28 中外製薬株式会社 Lyophilized formulation sealed in glass vial
WO2019229658A1 (en) 2018-05-30 2019-12-05 Novartis Ag Entpd2 antibodies, combination therapies, and methods of using the antibodies and combination therapies
USRE47770E1 (en) 2002-07-18 2019-12-17 Merus N.V. Recombinant production of mixtures of antibodies
WO2019243555A1 (en) 2018-06-21 2019-12-26 Charité - Universitätsmedizin Berlin Complement anaphylatoxin binders and their use in treatment of a subject having an ocular wound and/or fibrosis
WO2020016433A1 (en) 2018-07-20 2020-01-23 Aicuris Gmbh & Co. Kg Methods for screening and identifying agents that inhibit or modulate the nuclear egress complex of herpesviruses
WO2020025792A1 (en) 2018-08-03 2020-02-06 Amgen Research (Munich) Gmbh Antibody constructs for cldn18.2 and cd3
WO2020025532A1 (en) 2018-07-30 2020-02-06 Amgen Research (Munich) Gmbh Prolonged administration of a bispecific antibody construct binding to cd33 and cd3
EP3613774A1 (en) 2010-06-09 2020-02-26 Genmab A/S Antibodies against human cd38
WO2020041360A1 (en) 2018-08-21 2020-02-27 Quidel Corporation Dbpa antibodies and uses thereof
WO2020043670A1 (en) 2018-08-27 2020-03-05 Affimed Gmbh Cryopreserved nk cells preloaded with an antibody construct
WO2020061210A1 (en) 2018-09-18 2020-03-26 Merrimack Pharmaceuticals, Inc. Anti-tnfr2 antibodies and uses thereof
EP3632931A1 (en) 2014-11-07 2020-04-08 Sesen Bio, Inc. Improved il-6 antibodies
WO2020077212A1 (en) 2018-10-11 2020-04-16 Amgen Inc. Downstream processing of bispecific antibody constructs
WO2020079580A1 (en) 2018-10-15 2020-04-23 Novartis Ag Trem2 stabilizing antibodies
WO2020102501A1 (en) 2018-11-16 2020-05-22 Bristol-Myers Squibb Company Anti-nkg2a antibodies and uses thereof
US10667501B2 (en) 2012-05-17 2020-06-02 Kymab Limited Transgenic non-human vertebrate for the in vivo production of dual specificity immunoglobulins or hypermutated heavy chain only immunoglobulins
WO2020112870A1 (en) 2018-11-28 2020-06-04 Forty Seven, Inc. Genetically modified hspcs resistant to ablation regime
WO2020128039A2 (en) 2018-12-21 2020-06-25 4TEEN4 Pharmaceuticals GmbH Therapy guidance and/or therapy monitoring for a treatment with angiotensin-receptor-agonist and/or a precursor thereof
US10702608B2 (en) 2013-09-08 2020-07-07 Kodiak Sciences Inc. Factor VIII zwitterionic polymer conjugates
WO2020153467A1 (en) 2019-01-24 2020-07-30 中外製薬株式会社 Novel cancer antigens and antibodies of said antigens
WO2020154293A1 (en) 2019-01-22 2020-07-30 Bristol-Myers Squibb Company Antibodies against il-7r alpha subunit and uses thereof
US10745467B2 (en) 2010-03-26 2020-08-18 The Trustees Of Dartmouth College VISTA-Ig for treatment of autoimmune, allergic and inflammatory disorders
US10745487B2 (en) 2016-03-22 2020-08-18 Bionomics Limited Method of treating cancer by administering an anti-LGR5 monoclonal antibody
WO2020175689A1 (en) 2019-02-28 2020-09-03 学校法人順天堂 Antibody capable of binding to truncated mutant calreticulin, and diagnostic, prophylactic or therapeutic drug for myeloproliferative neoplasms
WO2020180712A1 (en) 2019-03-01 2020-09-10 Merrimack Pharmaceuticals, Inc. Anti-tnfr2 antibodies and uses thereof
US10781254B2 (en) 2010-03-26 2020-09-22 The Trustees Of Dartmouth College VISTA regulatory T cell mediator protein, VISTA binding agents and use thereof
US10781264B2 (en) 2016-02-03 2020-09-22 Amgen Research (Munich) Gmbh PSMA and CD3 bispecific T cell engaging antibody constructs
WO2020189748A1 (en) 2019-03-19 2020-09-24 中外製薬株式会社 Antigen-binding molecule containing antigen-binding domain of which binding activity to antigen is changed depending on mta, and library for obtaining said antigen-binding domain
WO2020205469A1 (en) 2019-03-29 2020-10-08 Bristol-Myers Squibb Company Methods of measuring hydrophobicity of chromatographic resins
WO2020209318A1 (en) 2019-04-10 2020-10-15 中外製薬株式会社 Method for purifying fc region-modified antibody
WO2020213665A1 (en) 2019-04-17 2020-10-22 国立大学法人広島大学 Therapeutic agent for urological cancer which is characterized by being administered with il-6 inhibitor and ccr2 inhibitor in combination
WO2020246563A1 (en) 2019-06-05 2020-12-10 中外製薬株式会社 Antibody cleavage site-binding molecule
WO2020252442A1 (en) 2019-06-13 2020-12-17 Amgen Inc. Automated biomass-based perfusion control in the manufacturing of biologics
WO2020250159A1 (en) 2019-06-12 2020-12-17 Novartis Ag Natriuretic peptide receptor 1 antibodies and methods of use
US10870701B2 (en) 2016-03-15 2020-12-22 Generon (Shanghai) Corporation Ltd. Multispecific fab fusion proteins and use thereof
US10899836B2 (en) 2016-02-12 2021-01-26 Janssen Pharmaceutica Nv Method of identifying anti-VISTA antibodies
WO2021021676A1 (en) 2019-07-26 2021-02-04 Amgen Inc. Anti-il13 antigen binding proteins
US10934571B2 (en) 2002-07-18 2021-03-02 Merus N.V. Recombinant production of mixtures of antibodies
US10933115B2 (en) 2012-06-22 2021-03-02 The Trustees Of Dartmouth College VISTA antagonist and methods of use
WO2021038078A1 (en) 2019-08-30 2021-03-04 4TEEN4 Pharmaceuticals GmbH Therapy guidance and/or therapy monitoring for treatment of shock
WO2021050640A1 (en) 2019-09-10 2021-03-18 Amgen Inc. Purification method for bispecific antigen-binding polypeptides with enhanced protein l capture dynamic binding capacity
WO2021053560A1 (en) 2019-09-18 2021-03-25 Novartis Ag Combination therapy with entpd2 and cd73 antibodies
WO2021053559A1 (en) 2019-09-18 2021-03-25 Novartis Ag Entpd2 antibodies, combination therapies, and methods of using the antibodies and combination therapies
US11009509B2 (en) 2015-06-24 2021-05-18 Janssen Pharmaceutica Nv Anti-VISTA antibodies and fragments
WO2021097344A1 (en) 2019-11-13 2021-05-20 Amgen Inc. Method for reduced aggregate formation in downstream processing of bispecific antigen-binding molecules
US11014987B2 (en) 2013-12-24 2021-05-25 Janssen Pharmaceutics Nv Anti-vista antibodies and fragments, uses thereof, and methods of identifying same
US11013800B2 (en) 2011-05-16 2021-05-25 Evive Biotech Ltd. Multi-specific Fab fusion proteins comprising a CD3-binding Fab fragment with N-terminal fusion to binding domains and methods of use
WO2021127528A1 (en) 2019-12-20 2021-06-24 Amgen Inc. Mesothelin-targeted cd40 agonistic multispecific antibody constructs for the treatment of solid tumors
WO2021127547A2 (en) 2019-12-19 2021-06-24 Quidel Corporation Monoclonal antibody fusions
EP3842457A1 (en) 2015-09-09 2021-06-30 Novartis AG Thymic stromal lymphopoietin (tslp)-binding molecules and methods of using the molecules
WO2021130383A1 (en) 2019-12-27 2021-07-01 Affimed Gmbh Method for the production of bispecific fcyriii x cd30 antibody construct
US11051497B2 (en) 2011-09-19 2021-07-06 Kymab Limited Manipulation of immunoglobulin gene diversity and multi-antibody therapeutics
WO2021142191A1 (en) 2020-01-08 2021-07-15 Regeneron Pharmaceuticals, Inc. Treatment of fibrodysplasia ossificans progressiva
US11066465B2 (en) 2015-12-30 2021-07-20 Kodiak Sciences Inc. Antibodies and conjugates thereof
WO2021150824A1 (en) 2020-01-22 2021-07-29 Amgen Research (Munich) Gmbh Combinations of antibody constructs and inhibitors of cytokine release syndrome and uses thereof
EP3871689A1 (en) 2020-02-26 2021-09-01 sphingotec GmbH Anti-adm-antibodies binding to the free n-terminus for accelerated transition of adm-gly to bio-adm in patients with adm-gly/ bio-adm ratio above a threshold and combination with vitamin c
WO2021170880A2 (en) 2020-02-27 2021-09-02 Adrenomed Ag Anti-adrenomedullin (adm) binder for use in therapy of patients in shock
WO2021170876A1 (en) 2020-02-27 2021-09-02 Adrenomed Ag Anti-adrenomedullin (adm) antibody or anti-adm antibody fragment or anti-adm non-ig scaffold for use in therapy or prevention of shock
WO2021170838A1 (en) 2020-02-27 2021-09-02 4TEEN4 Pharmaceuticals GmbH Dpp3 for therapy guidance, monitoring and stratification of nt-adm antibodies in patients with shock
WO2021183861A1 (en) 2020-03-12 2021-09-16 Amgen Inc. Method for treatment and prophylaxis of crs in patients comprising a combination of bispecifc antibodies binding to cds x cancer cell and tnfalpha or il-6 inhibitor
US11123426B2 (en) 2014-06-11 2021-09-21 The Trustees Of Dartmouth College Use of vista agonists and antagonists to suppress or enhance humoral immunity
WO2021188851A1 (en) 2020-03-19 2021-09-23 Amgen Inc. Antibodies against mucin 17 and uses thereof
WO2021185785A1 (en) 2020-03-16 2021-09-23 Sphingotec Gmbh Pro-adrenomedullin or fragment thereof in patients infected with corona virus and treatments with binder against adrenomedullin
WO2021185786A1 (en) 2020-03-16 2021-09-23 4TEEN4 Pharmaceuticals GmbH Dpp3 in patients infected with coronavirus
WO2021193928A1 (en) 2020-03-27 2021-09-30 株式会社PhotoQ3 Pharmaceutical drug for destroying tumor cells
WO2021206078A1 (en) 2020-04-06 2021-10-14 株式会社PhotoQ3 Medicine for killing tumor cells
WO2021210667A1 (en) 2020-04-17 2021-10-21 中外製薬株式会社 Bispecific antigen-binding molecule, composition associated therewith, and use, kit, and method for producing composition
WO2021220215A1 (en) 2020-05-01 2021-11-04 Novartis Ag Engineered immunoglobulins
WO2021220218A1 (en) 2020-05-01 2021-11-04 Novartis Ag Immunoglobulin variants
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
US11180557B2 (en) 2012-06-22 2021-11-23 King's College London Vista modulators for diagnosis and treatment of cancer
US11180555B2 (en) 2014-09-16 2021-11-23 Ubi Us Holdings, Llc. Antibodies directed against CD4 for the treatment and functional cure of HIV
WO2021236638A1 (en) 2020-05-19 2021-11-25 Amgen Inc. Mageb2 binding constructs
WO2021235537A1 (en) 2020-05-22 2021-11-25 中外製薬株式会社 Antibody for neutralizing substance having coagulation factor viii (f.viii) function-substituting activity
WO2021243320A2 (en) 2020-05-29 2021-12-02 Amgen Inc. Adverse effects-mitigating administration of a bispecific antibody construct binding to cd33 and cd3
WO2021247591A1 (en) 2020-06-02 2021-12-09 Arcus Biosciences, Inc. Antibodies to tigit
EP3922993A1 (en) 2020-06-12 2021-12-15 4TEEN4 Pharmaceuticals GmbH Dpp3 in patients infected with coronavirus
WO2021261546A1 (en) 2020-06-24 2021-12-30 国立大学法人 東京大学 Photosensitizing dye
US11229628B2 (en) 2015-01-09 2022-01-25 Duke University TRPA1 and TRPV4 inhibitors and methods of using the same for organ-specific inflammation and itch
US11237165B2 (en) 2008-06-27 2022-02-01 Merus N.V. Antibody producing non-human animals
WO2022025030A1 (en) 2020-07-28 2022-02-03 中外製薬株式会社 Prefilled syringe preparation with needle, provided with needle shield and including novel modified antibody
WO2022025220A1 (en) 2020-07-31 2022-02-03 中外製薬株式会社 Pharmaceutical composition including cell expressing chimeric receptor
US11242393B2 (en) 2018-03-23 2022-02-08 Bristol-Myers Squibb Company Antibodies against MICA and/or MICB and uses thereof
US11242392B2 (en) 2013-12-24 2022-02-08 Janssen Pharmaceutica Nv Anti-vista antibodies and fragments
EP3961214A1 (en) 2010-12-31 2022-03-02 BioAtla, Inc. Comprehensive monoclonal antibody generation
WO2022045247A1 (en) 2020-08-27 2022-03-03 学校法人順天堂 Anti-truncated mutant calr-cd3 bispecific antibody and pharmaceutical composition
US11292839B2 (en) 2016-08-13 2022-04-05 Ubi Us Holdings, Llc Treatment and sustained virologic remission of HIV infection by antibodies to CD4 in HAART stabilized patients
WO2022074206A1 (en) 2020-10-08 2022-04-14 Affimed Gmbh Trispecific binders
WO2022096698A1 (en) 2020-11-06 2022-05-12 Amgen Inc. Polypeptide constructs binding to cd3
WO2022096704A1 (en) 2020-11-06 2022-05-12 Amgen Inc. Antigen binding domain with reduced clipping rate
WO2022097065A2 (en) 2020-11-06 2022-05-12 Novartis Ag ANTIBODY Fc VARIANTS
WO2022096700A1 (en) 2020-11-06 2022-05-12 Amgen Research (Munich) Gmbh Polypeptide constructs selectively binding to cldn6 and cd3
WO2022096716A2 (en) 2020-11-06 2022-05-12 Amgen Inc. Multitargeting bispecific antigen-binding molecules of increased selectivity
WO2022117893A2 (en) 2020-12-02 2022-06-09 S-Form Pharma Combination therapy for patients having acute and/or persistent dyspnea
US11359005B2 (en) 2017-03-30 2022-06-14 The Johns Hopkins University Supramolecular high affinity protein-binding system for purification of biomacromolecules
WO2022130182A1 (en) 2020-12-14 2022-06-23 Novartis Ag Reversal binding agents for anti-natriuretic peptide receptor 1 (npr1) antibodies and uses thereof
US11408095B2 (en) 2011-09-26 2022-08-09 Merus N.V. Generation of binding molecules
US11434302B2 (en) 2016-02-03 2022-09-06 Amgen Research (Munich) Gmbh Bispecific T cell engaging antibody constructs
EP4056993A1 (en) 2014-08-20 2022-09-14 Chugai Seiyaku Kabushiki Kaisha Method for measuring viscosity of protein solution
WO2022191306A1 (en) 2021-03-12 2022-09-15 中外製薬株式会社 Pharmaceutical composition for treatment or prevention of myasthenia gravis
WO2022212831A1 (en) 2021-04-02 2022-10-06 Amgen Inc. Mageb2 binding constructs
US11484604B2 (en) 2020-08-07 2022-11-01 Fortis Therapeutics, Inc. Immunoconjugates targeting CD46 and methods of use thereof
WO2022234102A1 (en) 2021-05-06 2022-11-10 Amgen Research (Munich) Gmbh Cd20 and cd22 targeting antigen-binding molecules for use in proliferative diseases
US11525000B2 (en) 2016-04-15 2022-12-13 Immunext, Inc. Anti-human VISTA antibodies and use thereof
US11564380B2 (en) 2009-07-08 2023-01-31 Kymab Limited Animal models and therapeutic molecules
WO2023007023A1 (en) 2021-07-30 2023-02-02 Affimed Gmbh Duplexbodies
US11584790B2 (en) 2017-04-14 2023-02-21 Kodiak Sciences Inc. Complement factor D antagonist antibodies and conjugates thereof
EP4144898A1 (en) 2021-09-07 2023-03-08 New/era/mabs GmbH Method for selecting or screening antibodies from an antibody library
WO2023058723A1 (en) 2021-10-08 2023-04-13 中外製薬株式会社 Method for preparing prefilled syringe formulation
WO2023057871A1 (en) 2021-10-04 2023-04-13 Novartis Ag Surfactant stabilizers
WO2023079493A1 (en) 2021-11-03 2023-05-11 Affimed Gmbh Bispecific cd16a binders
WO2023078968A1 (en) 2021-11-03 2023-05-11 Affimed Gmbh Bispecific cd16a binders
US11707056B2 (en) 2013-05-02 2023-07-25 Kymab Limited Animals, repertoires and methods
EP4218816A2 (en) 2014-06-20 2023-08-02 Chugai Seiyaku Kabushiki Kaisha Pharmaceutical composition for use in prevention and/or treatment of disease that develops or progresses as a result of decrease or loss of activity of blood coagulation factor viii and/or activated blood coagulation factor viii
EP4223784A2 (en) 2015-09-02 2023-08-09 The Regents of the University of Colorado, a body corporate Compositions and methods for modulating t-cell mediated immune response
US11745165B2 (en) 2017-08-18 2023-09-05 The Johns Hopkins University Supramolecular filamentous assemblies for protein purification
US11753479B2 (en) 2014-03-04 2023-09-12 Kymab Limited Nucleic acids encoding anti-OX40L antibodies
WO2023175035A1 (en) 2022-03-15 2023-09-21 Adrenomed Ag Stable aqueous formulation of an anti-adrenomedullin (adm) antibody or anti-adm antibody fragment
EP4249066A2 (en) 2014-12-23 2023-09-27 Bristol-Myers Squibb Company Antibodies to tigit
EP4248976A2 (en) 2007-08-23 2023-09-27 Amgen Inc. Antigen binding proteins to proprotein convertase subtilisin kexin type 9 (pcsk9)
US11779604B2 (en) 2016-11-03 2023-10-10 Kymab Limited Antibodies, combinations comprising antibodies, biomarkers, uses and methods
EP4269440A2 (en) 2015-02-27 2023-11-01 Chugai Seiyaku Kabushiki Kaisha Composition for treating il-6-related diseases
WO2023209568A1 (en) 2022-04-26 2023-11-02 Novartis Ag Multispecific antibodies targeting il-13 and il-18
WO2023218027A1 (en) 2022-05-12 2023-11-16 Amgen Research (Munich) Gmbh Multichain multitargeting bispecific antigen-binding molecules of increased selectivity
US11819531B2 (en) 2009-12-18 2023-11-21 Kodiak Sciences Inc. Multifunctional zwitterionic polymer conjugates
US11873334B2 (en) 2018-09-24 2024-01-16 EyePoint Pharmaceuticals, Inc. Method of treating ocular conditions by administering an antibody that activates Tie2 and binds VEGF
WO2024023369A1 (en) 2022-07-29 2024-02-01 Adrenomed Ag Anti-adrenomedullin (adm) antibody or anti-adm antibody fragment or anti-adm non-ig scaffold for use in therapy or prevention of shock
WO2024023368A1 (en) 2022-07-29 2024-02-01 4TEEN4 Pharmaceuticals GmbH Prediction of an increase of dpp3 in a patient with septic shock
US11912784B2 (en) 2019-10-10 2024-02-27 Kodiak Sciences Inc. Methods of treating an eye disorder
US11926659B2 (en) 2019-03-03 2024-03-12 Prothena Biosciences Limited Antibodies recognizing tau
WO2024059675A2 (en) 2022-09-14 2024-03-21 Amgen Inc. Bispecific molecule stabilizing composition
EP4345109A1 (en) 2022-09-30 2024-04-03 AdrenoMed AG Anti-adrenomedullin (adm) binder for use in therapy of pediatric patients with congenital heart disease

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3456192A1 (en) * 2008-06-27 2019-03-20 Merus N.V. Antibody producing non-human mammals
US8940272B2 (en) * 2010-10-11 2015-01-27 University Of Zurich Human anti-tau antibodies
SG11201403326VA (en) * 2011-12-20 2014-07-30 Regeneron Pharma Humanized light chain mice
JP6884860B2 (en) * 2016-11-04 2021-06-09 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 animal with engineered immunoglobulin lambda light chain locus

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1990004036A1 (en) * 1988-10-12 1990-04-19 Medical Research Council Production of antibodies from transgenic animals

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1991000906A1 (en) * 1989-07-12 1991-01-24 Genetics Institute, Inc. Chimeric and transgenic animals capable of producing human antibodies
EP0463151B1 (en) * 1990-01-12 1996-06-12 Cell Genesys, Inc. Generation of xenogeneic antibodies
ES2108048T3 (en) * 1990-08-29 1997-12-16 Genpharm Int PRODUCTION AND USE OF LOWER TRANSGENIC ANIMALS CAPABLE OF PRODUCING HETEROLOGICAL ANTIBODIES.

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1990004036A1 (en) * 1988-10-12 1990-04-19 Medical Research Council Production of antibodies from transgenic animals

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
CHEMICAL REVIEWS, Volume 90, No. 4, issued June 1990, UHLMANN et al., "Antisense Oligonucleotides: A New Therapeutic Principle", pages 544-584. *
NATURE, Volume 342, issued 23 November 1989, ZIJLSTRA et al., "Germline Transmission of a Disrupted B2-Microglobulin Gene Produced by Homologous Recombination in Embryonic Stem Cells", pages 435-438. *
NATURE, Volume 350, issued 04 April 1991, KITAMURA et al., "A B Cell-Deficient Mouse by Targeted Disruption of the Membrane Exon of the Immunoglobulin U Chain Gene", pages 423-426. *
See also references of EP0746609A4 *

Cited By (1291)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6713610B1 (en) 1990-01-12 2004-03-30 Raju Kucherlapati Human antibodies derived from immunized xenomice
US7501552B2 (en) 1991-08-28 2009-03-10 Medarex, Inc. Transgenic non-human animals for producing chimeric antibodies
EP0754225A4 (en) * 1993-04-26 2001-01-31 Genpharm Int Transgenic non-human animals capable of producing heterologous antibodies
EP0754225A1 (en) * 1993-04-26 1997-01-22 GenPharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
US5827690A (en) * 1993-12-20 1998-10-27 Genzyme Transgenics Corporatiion Transgenic production of antibodies in milk
US5849992A (en) * 1993-12-20 1998-12-15 Genzyme Transgenics Corporation Transgenic production of antibodies in milk
US8293480B2 (en) 1994-03-09 2012-10-23 Genpharm International Transgenic non-human animals for producing chimeric antibodies
US8158419B2 (en) 1994-03-09 2012-04-17 Medarex, Inc. Transgenic non-human animals for producing chimeric antibodies
US6319499B1 (en) 1994-07-26 2001-11-20 Amgen Inc. Methods for activating an erythropoietin receptor using antibodies
US7081523B2 (en) 1994-07-26 2006-07-25 Amgen Inc. Antibodies which activate an erythropoietin receptor
US5885574A (en) * 1994-07-26 1999-03-23 Amgen Inc. Antibodies which activate an erythropoietin receptor
US7468268B2 (en) 1995-02-20 2008-12-23 Daiichi Sankyo Co., Ltd. Nucleic acid molecules encoding osteoclastogenesis inhibitory factor proteins
US6855808B2 (en) 1995-02-20 2005-02-15 Sankyo Co., Ltd. Proteins and methods for producing the proteins
US7276344B2 (en) 1995-02-20 2007-10-02 Sankyo Co., Ltd. Methods for using the osteoclastogenesis inhibitory factor (OCIF) protein
EP0817835A4 (en) * 1995-03-29 2005-01-05 Abgenix Inc Production of antibodies using cre-mediated site-specific recombination
US7145056B2 (en) 1995-03-29 2006-12-05 Abgenix, Inc. Production of antibodies using cre-mediated site-specific recombination
EP0817835A1 (en) * 1995-03-29 1998-01-14 Xenotech Incorporated Production of antibodies using cre-mediated site-specific recombination
EP1709970A1 (en) * 1995-04-27 2006-10-11 Abgenix, Inc. Human antibodies against EGFR, derived from immunized xenomice
EP0822830A1 (en) * 1995-04-27 1998-02-11 Abgenix, Inc. Human antibodies derived from immunized xenomice
AU2008202860B9 (en) * 1995-04-27 2012-03-29 Amgen Fremont Inc. Human Antibodies Derived From Immunized Xenomice
AU2003227322B2 (en) * 1995-04-27 2005-12-01 Amgen Fremont Inc. Human Antibodies Derived From Immunized Xenomice
AU2008202860B2 (en) * 1995-04-27 2012-03-01 Amgen Fremont Inc. Human Antibodies Derived From Immunized Xenomice
EP0822830A4 (en) * 1995-04-27 2003-03-19 Abgenix Inc Human antibodies derived from immunized xenomice
EP0854917A4 (en) * 1995-10-10 2002-07-24 Genpharm Int Transgenic non-human animals capable of producing heterologous antibodies
CN102827283B (en) * 1995-10-10 2014-11-12 真药物国际公司 Transgenic non-human animals capable of producing heterologous antibodies
EP1813672A3 (en) * 1995-10-10 2008-05-28 GenPharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
EP0854917A1 (en) * 1995-10-10 1998-07-29 GenPharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
EP2186888A1 (en) * 1995-10-10 2010-05-19 GenPharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
CN102827283A (en) * 1995-10-10 2012-12-19 真药物国际公司 Transgenic non-human animals capable of producing heterologous antibodies
EP1813672A2 (en) * 1995-10-10 2007-08-01 GenPharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
EP2298816A2 (en) 1995-12-05 2011-03-23 Amgen Inc. Apoptosis induced by an antibody against Her2
US6046310A (en) * 1996-03-13 2000-04-04 Protein Design Labs., Inc. FAS ligand fusion proteins and their uses
EP1854481A2 (en) 1996-04-23 2007-11-14 Chugai Seiyaku Kabushiki Kaisha Cerebral stroke/cerebral edema preventive or remedy containing IL-8 binding inhibitor as active ingredient
US5912176A (en) * 1996-06-03 1999-06-15 United Biomedical, Inc. Antibodies against a host cell antigen complex for pre and post exposure protection from infection by HIV
US5961976A (en) * 1996-06-03 1999-10-05 United Biomedical, Inc. Antibodies against a host cell antigen complex for pre- and post-exposure protection from infection by HIV
US7722873B2 (en) 1996-10-10 2010-05-25 Genpharm International, Inc. Heterologous antibodies which bind human CD4
US8231877B2 (en) 1996-10-10 2012-07-31 Genpharm International, Inc. Heterologous antibodies which bind human CD4
EP1972194A1 (en) 1996-12-03 2008-09-24 Amgen Fremont Inc. Transgenic mammals having human ig loci including plural vh and vk regions and antibodies produced therefrom
US8809051B2 (en) 1996-12-03 2014-08-19 Amgen Fremont Inc. Transgenic mammals having human Ig loci including plural VH and Vκ regions and antibodies produced therefrom
US7820877B2 (en) 1996-12-03 2010-10-26 Amgen Fremont Inc. Transgenic mammals having human IG loci including plural Vh and Vk regions and antibodies produced therefrom
EP2305027A2 (en) 1996-12-03 2011-04-06 Amgen Fremont Inc. Transgenic mammals having human Ig loci including plural VH and Vkappa regions and antibodies produced therefrom
EP2314625A1 (en) 1996-12-03 2011-04-27 Amgen Fremont Inc. Transgenic mammals having human Ig loci including plural VH and Vkappa regions and antibodies produced therefrom
US7064244B2 (en) 1996-12-03 2006-06-20 Abgenix, Inc. Transgenic mammals having human Ig loci including plural VH and VK regions and antibodies produced therefrom
US7932375B2 (en) 1996-12-23 2011-04-26 Immunex Corporation Kits for detecting rank nucleic acids
US7411050B2 (en) 1996-12-23 2008-08-12 Immunex Corporation Monoclonal blocking antibody to human RANKL
US7744886B2 (en) 1996-12-23 2010-06-29 Immunex Corporation Methods for interfering with rank signaling
US8377690B2 (en) 1996-12-23 2013-02-19 Immunex Corporation Cells and methods for producing blocking antibodies to human RANKL
US8715683B2 (en) 1996-12-23 2014-05-06 Immunex Corporation RANK ligand polypeptides
EP2011514A1 (en) 1997-03-21 2009-01-07 Chugai Seiyaku Kabushiki Kaisha A preventive or therapeutic agent for sensitized T cell-mediated diseases comprising IL-6 antagonist as an active ingredient
EP2322216A1 (en) 1997-03-21 2011-05-18 Chugai Seiyaku Kabushiki Kaisha A preventive or therapeutic agent for sensitized T cell-mediated diseases comprising IL-6 antagonist as an active ingredient
US7449185B2 (en) 1997-04-15 2008-11-11 Daiichi Sankyo Company, Limited Antibodies to OCIF-binding molecules
US7192718B2 (en) 1997-04-15 2007-03-20 Sankyo Co. Ltd Methods for identifying a substance which specifically binds to an osteoclastogenesis inhibitory factor-binding molecule and modulates the biological activity thereof
US7527790B2 (en) 1997-04-15 2009-05-05 Daiichi Sankyo Company, Limited Methods of reducing bone absorption comprising administering an antibody that binds OBM
US7097834B1 (en) 1997-04-16 2006-08-29 Amgen Inc. Osteoprotegerin binding proteins
EP2311955A2 (en) 1997-04-16 2011-04-20 Amgen Inc. Osteoprotegerin binding proteins and their receptors
US5843678A (en) * 1997-04-16 1998-12-01 Amgen Inc. Osteoprotegerin binding proteins
EP2332993A1 (en) 1997-05-05 2011-06-15 Amgen Fremont Inc. Human monoclonal antibody to the human epidermal growth factor receptor
EP1878746A2 (en) 1997-05-05 2008-01-16 Amgen Fremont Inc. Human monoclonal antibodies to epidermal growth factor receptor
WO1998054348A1 (en) * 1997-05-31 1998-12-03 Babraham Institute Telomere-associated chromosome fragmentation
EP1916020A2 (en) 1997-08-15 2008-04-30 Chugai Seiyaku Kabushiki Kaisha Preventives and/or remedies for systemic lupus erythematosus containing anti-IL-6 receptor antibody as the active ingredient
WO1999015696A1 (en) * 1997-09-19 1999-04-01 Medelys Laboratories Inc. Method and kit for early diagnosis of autoimmunity and lymphoma in central nervous system
EP0921189A1 (en) * 1997-11-14 1999-06-09 Sankyo Company Limited Transgenic animal allergy models and methods for their use
US6118044A (en) * 1997-11-14 2000-09-12 Sankyo Company, Limited Transgenic animal allergy models and methods for their use
JP4677334B2 (en) * 1997-12-02 2011-04-27 ヤンセン アルツハイマー イミュノセラピー A preventive and therapeutic agent for amyloidogenic diseases
US8535673B2 (en) 1997-12-02 2013-09-17 Janssen Alzheimer Immunotherapy Prevention and treatment of amyloidogenic disease
US8642044B2 (en) 1997-12-02 2014-02-04 Janssen Alzheimer Immunotherapy Prevention and treatment of amyloidogenic disease
JP2006077030A (en) * 1997-12-02 2006-03-23 Neuralab Ltd Preventing and treating agent of amyloidogenic disease
US7964192B1 (en) 1997-12-02 2011-06-21 Janssen Alzheimer Immunotherapy Prevention and treatment of amyloidgenic disease
EP2305282A2 (en) 1997-12-02 2011-04-06 Janssen Alzheimer Immunotherapy Prevention and treatment of amyloidogenic disease
EP1994937A2 (en) 1997-12-02 2008-11-26 Elan Pharma International Limited Prevention and treatment of amyloidogenic disease
US8034339B2 (en) 1997-12-02 2011-10-11 Janssen Alzheimer Immunotherapy Prevention and treatment of amyloidogenic disease
US8034348B2 (en) 1997-12-02 2011-10-11 Janssen Alzheimer Immunotherapy Prevention and treatment of amyloidogenic disease
US9051363B2 (en) 1997-12-02 2015-06-09 Janssen Sciences Ireland Uc Humanized antibodies that recognize beta amyloid peptide
EP3553085A1 (en) 1998-12-23 2019-10-16 Pfizer Inc Human momoclonal antibodies to ctla-4
EP2112166A2 (en) 1998-12-23 2009-10-28 Pfizer Inc. Human monoclonal antibodies to CTLA-4
US8883984B2 (en) 1998-12-23 2014-11-11 Amgen Fremont Inc. Human monoclonal antibodies to CTLA-4
US7132281B2 (en) 1998-12-23 2006-11-07 Amgen Fremont Inc. Methods and host cells for producing 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
US7824679B2 (en) 1998-12-23 2010-11-02 Amgen Fremont Inc. Human monoclonal antibodies to CTLA-4
US8143379B2 (en) 1998-12-23 2012-03-27 Amgen Fremont Inc. Human monoclonal antibodies to CTLA-4
US8491895B2 (en) 1998-12-23 2013-07-23 Amgen Fremont Inc. Methods of treating cancer with human monoclonal antibodies to CTLA-4
US6682736B1 (en) 1998-12-23 2004-01-27 Abgenix, Inc. Human monoclonal antibodies to CTLA-4
US7807797B2 (en) 1998-12-23 2010-10-05 Amgen Fremont Inc. Human monoclonal antibodies to CTLA-4
US7411057B2 (en) 1998-12-23 2008-08-12 Amgen Fremont Inc. Nucleic acids encoding human monoclonal antibodies to CTLA-4
US9963508B2 (en) 1998-12-23 2018-05-08 Amgen Fremont Inc. Human monoclonal antibodies to CTLA-4
EP1604997A1 (en) * 1999-02-05 2005-12-14 Therapeutic Human Polyclonals, Inc. Human polyclonal antibodies from transgenic nonhuman animals
EP1151010B1 (en) * 1999-02-05 2005-10-26 Therapeutic Human Polyclonals, Inc. Human polyclonal antibodies from transgenic nonhuman animals
EP1167537A1 (en) * 1999-03-30 2002-01-02 Japan Tobacco Inc. Process for producing monoclonal antibody
US8236530B2 (en) 1999-03-30 2012-08-07 Japan Tobacco Inc. Method for preparing monoclonal antibody
EP1167537A4 (en) * 1999-03-30 2005-05-04 Japan Tobacco Inc Process for producing monoclonal antibody
JP4488579B2 (en) * 1999-03-30 2010-06-23 日本たばこ産業株式会社 Method for producing monoclonal antibody
JP2000342279A (en) * 1999-03-30 2000-12-12 Japan Tobacco Inc Production of monoclonal antibody
US6864235B1 (en) 1999-04-01 2005-03-08 Eva A. Turley Compositions and methods for treating cellular response to injury and other proliferating cell disorders regulated by hyaladherin and hyaluronans
US6911429B2 (en) 1999-04-01 2005-06-28 Transition Therapeutics Inc. Compositions and methods for treating cellular response to injury and other proliferating cell disorders regulated by hyaladherin and hyaluronans
EP2305302A2 (en) 1999-05-28 2011-04-06 Janssen Alzheimer Immunotherapy Prevention and treatment of amyloidogenic disease
EP2364719A1 (en) 1999-06-01 2011-09-14 Janssen Alzheimer Immunotherapy Prevention and treatment of amyloidogenic diseases
EP2457439A1 (en) 1999-06-10 2012-05-30 Amgen Fremont Inc. Transgenic non-human animals for producing specific isotypes of human antibodies via non-cognate switch regions
JP2014003984A (en) * 1999-06-10 2014-01-16 Amgen Fremont Inc Transgenic animal for producing specific isotypes of human antibodies via non-cognate switch regions
JP2003501103A (en) * 1999-06-10 2003-01-14 アブジェニックス インク. Transgenic animals for producing specific isotypes of human antibodies via non-homologous switch regions
JP2011087594A (en) * 1999-06-10 2011-05-06 Amgen Fremont Inc Transgenic animal for producing specific isotype of human antibody via non-cognate switch region
EP2865754A1 (en) 1999-06-14 2015-04-29 BP Corporation North America Inc. Synthetic ligation reassembly in directed evolution
US8287863B2 (en) 1999-08-23 2012-10-16 Chugai Seiyaku Kabushiki Kaisha Method for treating myeloma utilizing an expression enhancer for HM1.24 antigen
US8017114B2 (en) 1999-08-24 2011-09-13 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
US6984720B1 (en) 1999-08-24 2006-01-10 Medarex, Inc. Human CTLA-4 antibodies
WO2001018200A1 (en) 1999-09-06 2001-03-15 Chugai Seiyaku Kabushiki Kaisha Tsg-like gene
EP2316848A1 (en) 1999-09-21 2011-05-04 Chugai Seiyaku Kabushiki Kaisha Transporter genes OATP-B, C, D and E
EP2351483A1 (en) 1999-10-01 2011-08-03 Chugai Seiyaku Kabushiki Kaisha Prevention and treatment of blood coagulation-related diseases
EP1987842A1 (en) 2000-04-28 2008-11-05 Chugai Seiyaku Kabushiki Kaisha Cell proliferation inhibitor
EP2026073A1 (en) 2000-04-29 2009-02-18 University Of Iowa Research Foundation Diagnostics and therapeutics for macular degeneration-related disorders
US9095626B2 (en) 2000-05-08 2015-08-04 Celldex Therapeutics, Inc. Monoclonal antibodies to dendritic cells
US7560534B2 (en) 2000-05-08 2009-07-14 Celldex Research Corporation Molecular conjugates comprising human monoclonal antibodies to dendritic cells
US8142790B2 (en) 2000-05-08 2012-03-27 Celldex Research Corporation Methods of using molecular conjugates comprising monoclonal antibodies to dendritic cells
EP1916303A1 (en) 2000-11-30 2008-04-30 Medarex, Inc. Nucleic acids encoding rearranged human immunoglobulin sequences from transgenic transchromosomal mice
EP2045267A2 (en) 2000-12-06 2009-04-08 Elan Pharma International Limited Humanized antibodies that recognize beta amyloid peptide
US7931897B2 (en) 2001-02-07 2011-04-26 Chugai Seiyaku Kabushiki Kaisha Therapeutic agent for hematopoietic tumors
US8834876B2 (en) 2001-02-07 2014-09-16 Chugai Seiyaku Kabushiki Kaisha Therapeutic agent for hematopoietic tumors
EP2786657B1 (en) 2001-02-16 2018-02-07 Regeneron Pharmaceuticals, Inc. A method of producing an antibody comprising a human variable region and a rodent constant region.
EP3640261A1 (en) 2001-04-02 2020-04-22 Chugai Seiyaku Kabushiki Kaisha Therapeutic agent for chronic arthritides diseases of childhood-related diseases
EP2298812A2 (en) 2001-04-02 2011-03-23 Chugai Seiyaku Kabushiki Kaisha Remedies for juvenile chronic arthritis and related diseases
EP1972638A1 (en) 2001-04-02 2008-09-24 Chugai Seiyaku Kabushiki Kaisha Remedies for juvenile chronic arthritis and related diseases
EP3492100A1 (en) 2001-06-26 2019-06-05 Amgen Inc. Antibodies to opgl
EP2087908A1 (en) 2001-06-26 2009-08-12 Amgen, Inc. Antibodies to opgl
US6894149B2 (en) 2001-10-11 2005-05-17 Protein Design Labs, Inc. Anti-HLA-DA antibodies and the methods of using thereof
EP3698626A1 (en) 2001-11-30 2020-08-26 Amgen Fremont Inc. Transgenic animals bearing human ig lambda light chain genes
US7435871B2 (en) 2001-11-30 2008-10-14 Amgen Fremont Inc. Transgenic animals bearing human Igλ light chain genes
EP2319301A2 (en) 2001-11-30 2011-05-11 Amgen Fremont Inc. Transgenic animals bearing human Ig lambda light chain genes
EP3269235A1 (en) 2001-11-30 2018-01-17 Amgen Fremont Inc. Transgenic animals bearing human ig lambda light chain genes
EP2311489A2 (en) 2002-02-14 2011-04-20 Chugai Seiyaku Kabushiki Kaisha Formulation of antibody-containing solutions comprising a sugar as a stabilizer
EP3578168A1 (en) 2002-02-14 2019-12-11 Chugai Seiyaku Kabushiki Kaisha Formulation of antibody-containing solutions comprising a sugar as a stabilizer
EP3192528A1 (en) 2002-02-14 2017-07-19 Chugai Seiyaku Kabushiki Kaisha Formulation of anti-il6r antibody-containing solutions comprising a sugar as a stabilizer
US8921527B2 (en) 2002-02-14 2014-12-30 Chugai Seiyaku Kabushiki Kaisha Antibody-containing solution formulations
WO2003068259A1 (en) 2002-02-14 2003-08-21 Chugai Seiyaku Kabushiki Kaisha Antibody-containing solution pharmaceuticals
EP2110434A1 (en) 2002-02-25 2009-10-21 Genentech, Inc. Type-1 cytokine receptor GLM-R
US8128928B2 (en) 2002-03-12 2012-03-06 Wyeth Llc Humanized antibodies that recognize beta amyloid peptide
EP2314316A1 (en) 2002-04-05 2011-04-27 Amgen, Inc Human anti-OPGL neutralizing antibodies as selective OPGL pathway inhibitors
US8367063B2 (en) 2002-04-05 2013-02-05 Amgen, Inc. Human anti-OPGL neutralizing antibodies as selective OPGL pathway inhibitors
US8455629B2 (en) 2002-04-05 2013-06-04 Amgen Inc. Human anti-OPGL neutralizing antibodies as selective OPGL pathway inhibitors
US7718776B2 (en) 2002-04-05 2010-05-18 Amgen Inc. Human anti-OPGL neutralizing antibodies as selective OPGL pathway inhibitors
US7452535B2 (en) 2002-04-12 2008-11-18 Medarex, Inc. Methods of treatment using CTLA-4 antibodies
WO2003097082A2 (en) 2002-05-17 2003-11-27 Protein Design Labs Treatment of crohn's disease or psoriasis using anti-inteferon gamma antibodies
EP2316922A1 (en) 2002-05-24 2011-05-04 Schering Corporation Neutralizing human anti-IGFR antibody
EP2316921A1 (en) 2002-05-24 2011-05-04 Schering Corporation Neutralizing human anti-IGFR antibody
EP3269377A1 (en) 2002-06-28 2018-01-17 The Government of the U.S.A. as represented by The Secretary of the dept. of Health & Human Services Method for the treatment of multiple sclerosis
EP2314627A2 (en) 2002-06-28 2011-04-27 The Government of the U.S.A. as represented by The Secretary of the dept. of Health & Human Services Method for the treatment of multiple sclerosis
US10934571B2 (en) 2002-07-18 2021-03-02 Merus N.V. Recombinant production of mixtures of antibodies
USRE47770E1 (en) 2002-07-18 2019-12-17 Merus N.V. Recombinant production of mixtures of antibodies
WO2004019966A1 (en) 2002-08-27 2004-03-11 Chugai Seiyaku Kabushiki Kaisha Method of stabilizing protein solution preparation
WO2004022739A1 (en) 2002-09-04 2004-03-18 Chugai Seiyaku Kabushiki Kaisha Antibody against n-terminal peptide or c-terminal peptide of gpc3 solubilized in blood
EP3020414A1 (en) 2002-09-06 2016-05-18 Amgen, Inc Therapeutic anti-il-1r1 monoclonal antibody
EP2213685A1 (en) 2002-09-06 2010-08-04 Amgen Inc. Therapeutic anti-IL-1R1 monoclonal antibody
EP2277543A1 (en) 2002-09-06 2011-01-26 Amgen, Inc Therapeutic anti-IL-1R1 monoclonal antibody
EP3225625A1 (en) 2002-09-11 2017-10-04 Chugai Seiyaku Kabushiki Kaisha Protein purification method
EP2261230A1 (en) 2002-09-11 2010-12-15 Chugai Seiyaku Kabushiki Kaisha Protein purification method
US9535076B2 (en) 2002-09-12 2017-01-03 The Regents Of The University Of California Methods and compositions for eliciting an amyloid-selective immune response
EP2270210A2 (en) 2002-09-30 2011-01-05 Oncotherapy Science, Inc. Method for diagnosing non-small cell lung cancers
EP2270503A2 (en) 2002-09-30 2011-01-05 Oncotherapy Science, Inc. Method for diagnosing non-small cell lung cancers
EP2264186A1 (en) 2002-09-30 2010-12-22 Oncotherapy Science, Inc. Method for diagnosing non-small cell lung cancers
EP2264457A1 (en) 2002-09-30 2010-12-22 Oncotherapy Science, Inc. Method for diagnosing non-small cell lung cancers
EP2270501A2 (en) 2002-09-30 2011-01-05 Oncotherapy Science, Inc. Method for diagnosing non-small cell lung cancers
EP2264187A1 (en) 2002-09-30 2010-12-22 Oncotherapy Science, Inc. Method for diagnosing non-small cell lung cancers
EP2270498A2 (en) 2002-09-30 2011-01-05 Oncotherapy Science, Inc. Method for diagnosing non-small cell lung cancers
EP2264185A1 (en) 2002-09-30 2010-12-22 Oncotherapy Science, Inc. Method for diagnosing non-small cell lung cancers
EP2270216A2 (en) 2002-09-30 2011-01-05 Oncotherapy Science, Inc. Method for diagnosing non-small cell lung cancers
EP2270502A2 (en) 2002-09-30 2011-01-05 Oncotherapy Science, Inc. Method for diagnosing non-small cell lung cancers
EP2270215A2 (en) 2002-09-30 2011-01-05 Oncotherapy Science, Inc. Method for diagnosing non-small cell lung cancers
EP2270214A2 (en) 2002-09-30 2011-01-05 Oncotherapy Science, Inc. Method for diagnosing non-small cell lung cancers
EP2270220A2 (en) 2002-09-30 2011-01-05 Oncotherapy Science, Inc. Method for diagnosing non-small cell lung cancers
EP2264190A1 (en) 2002-09-30 2010-12-22 Oncotherapy Science, Inc. Method for diagnosing non-small cell lung cancers
EP2270217A2 (en) 2002-09-30 2011-01-05 Oncotherapy Science, Inc. Method for diagnosing non-small cell lung cancers
EP2270499A1 (en) 2002-09-30 2011-01-05 Oncotherapy Science, Inc. Method for diagnosing non-small cell lung cancers
EP2270500A1 (en) 2002-09-30 2011-01-05 Oncotherapy Science, Inc. Method for diagnosing non-small cell lung cancers
EP2270213A2 (en) 2002-09-30 2011-01-05 Oncotherapy Science, Inc. Method for diagnosing non-small cell lung cancers
EP2270222A2 (en) 2002-09-30 2011-01-05 Oncotherapy Science, Inc. Method for diagnosing non-small cell lung cancers
EP2270211A2 (en) 2002-09-30 2011-01-05 Oncotherapy Science, Inc. Method for diagnosing non-small cell lung cancers
EP2270223A2 (en) 2002-09-30 2011-01-05 Oncotherapy Science, Inc. Method for diagnosing non-small cell lung cancers
EP2298811A2 (en) 2002-10-16 2011-03-23 Amgen Inc. Human anti-ifn-gamma neutralizing antibodies as selective ifn-gamma pathway inhibitors
WO2004035747A2 (en) 2002-10-16 2004-04-29 Amgen Inc. Human anti-ifn-ϝ neutralizing antibodies as selective ifn-ϝ pathway inhibitors
WO2004050683A2 (en) 2002-12-02 2004-06-17 Abgenix, Inc. Antibodies directed to tumor necrosis factor and uses thereof
US9243064B2 (en) 2003-01-31 2016-01-26 Celldex Therapeutics Inc. Antibody vaccine conjugates and uses therefor
US9259459B2 (en) 2003-01-31 2016-02-16 Celldex Therapeutics Inc. Antibody vaccine conjugates and uses therefor
EP2221064A1 (en) 2003-03-12 2010-08-25 Janssen Alzheimer Immunotherapy Humanized antibodies that recognize beta amyloid peptide
WO2004084823A2 (en) 2003-03-19 2004-10-07 Abgenix, Inc. Antibodies against t cell immunoglobulin domain and mucin domain 1 (tim-1) antigen and uses thereof
EP3000886A1 (en) 2003-03-19 2016-03-30 Amgen Fremont Inc. Antibodies against t cell immunoglobulin domain and mucin domain 1 (tim-1) antigen and uses thereof
EP2368577A2 (en) 2003-04-28 2011-09-28 Chugai Seiyaku Kabushiki Kaisha Methods for treating interleukin-6 related diseases
EP3167901A1 (en) 2003-04-28 2017-05-17 Chugai Seiyaku Kabushiki Kaisha Methods for treating interleukin-6 related diseases
EP3903819A1 (en) 2003-04-28 2021-11-03 Chugai Seiyaku Kabushiki Kaisha Methods for treating interleukin-6 related diseases
EP4098279A1 (en) 2003-04-28 2022-12-07 Chugai Seiyaku Kabushiki Kaisha Methods for treating interleukin-6 related diseases
EP2853272A1 (en) 2003-05-08 2015-04-01 AbbVie Biotherapeutics Inc. Therapeutic use of anti-CS1 antibodies
EP3275463A1 (en) 2003-05-08 2018-01-31 AbbVie Biotherapeutics Inc. Therapeutic use of anti-cs1 antibodies
EP2371391A1 (en) 2003-05-08 2011-10-05 Abbott Biotherapeutics Corp. Therapeutic use of anti-CS1 antibodies
US10670599B2 (en) 2003-05-30 2020-06-02 Merus N.V. Method for selecting a single cell expressing a heterogeneous combination of antibodies
US9738701B2 (en) 2003-05-30 2017-08-22 Merus N.V. Method for selecting a single cell expressing a heterogeneous combination of antibodies
US10605808B2 (en) 2003-05-30 2020-03-31 Merus N.V. Antibody producing non-human animals
EP2338914A1 (en) 2003-06-04 2011-06-29 Fibrogen, Inc. Connective tissue growth factor antibodies
EP2322549A1 (en) 2003-06-04 2011-05-18 Fibrogen, Inc. Connective tissue growth factor antibodies
EP2508608A1 (en) 2003-06-09 2012-10-10 Alnylam Pharmaceuticals Inc. Method of treating neurodegenerative disease
EP2228445A1 (en) 2003-06-18 2010-09-15 Chugai Seiyaku Kabushiki Kaisha Fucose Transporter
WO2005017155A1 (en) 2003-06-18 2005-02-24 Chugai Seiyaku Kabushiki Kaisha Fucose transporter
EP3679951A1 (en) 2003-06-27 2020-07-15 Amgen Fremont Inc. Antibodies directed to the deletion mutants of epidermal growth factor receptor and uses thereof
EP3011971A1 (en) 2003-06-27 2016-04-27 Amgen Fremont Inc. Antibodies directed to the deletion mutants of epidermal growth factor receptor and uses thereof
EP2457587A1 (en) 2003-06-27 2012-05-30 Amgen Fremont Inc. Anitbodies directed to the deletion mutants of epidermal growth factor receptor and uses thereof
EP2457586A1 (en) 2003-06-27 2012-05-30 Amgen Fremont Inc. Antibodies directed to the deletion mutants of epidermal growth factor receptor and uses thereof
EP3037105A1 (en) 2003-06-27 2016-06-29 Amgen Fremont Inc. Antibodies directed to the deletion mutants of epidermal growth factor receptor and uses thereof
US8153410B2 (en) 2003-07-07 2012-04-10 Fox Chase Cancer Center Alternate morpheein forms of allosteric proteins as a target for the development of bioactive molecules
EP2341067A1 (en) 2003-07-18 2011-07-06 Amgen, Inc Specific binding agents to hepatocyte growth factor
EP2311468A1 (en) 2003-08-08 2011-04-20 Perseus Proteomics Inc. Gene overexpressed in cancer
WO2005014818A1 (en) 2003-08-08 2005-02-17 Perseus Proteomics Inc. Gene overexpressed in cancer
WO2005016111A2 (en) 2003-08-08 2005-02-24 Abgenix, Inc. Antibodies directed to parathyroid hormone (pth) and uses thereof
EP2289944A2 (en) 2003-10-10 2011-03-02 Chugai Seiyaku Kabushiki Kaisha Bispecific antibody substituting for functional proteins
EP3085783A1 (en) 2003-10-10 2016-10-26 Chugai Seiyaku Kabushiki Kaisha Bispecific antibody substituting for functional proteins
EP2311945A1 (en) 2003-10-14 2011-04-20 Chugai Seiyaku Kabushiki Kaisha Bispecific antibodies substituting for functional proteins
WO2005035754A1 (en) 2003-10-14 2005-04-21 Chugai Seiyaku Kabushiki Kaisha Double specific antibodies substituting for functional protein
WO2005054467A1 (en) 2003-12-03 2005-06-16 Chugai Seiyaku Kabushiki Kaisha EXPRESSION SYSTEM WITH THE USE OF MAMMALIAN β-ACTIN PROMOTER
EP2383295A1 (en) 2003-12-10 2011-11-02 Medarex, Inc. IP-10 antibodies and their uses
EP2418220A2 (en) 2003-12-10 2012-02-15 Medarex, Inc. Interferon alpha antibodies and their uses
EP2865687A1 (en) 2003-12-10 2015-04-29 E. R. Squibb & Sons, L.L.C. IP-10 antibodies and their uses
WO2005056605A1 (en) 2003-12-12 2005-06-23 Chugai Seiyaku Kabushiki Kaisha Modified antibodies recognizing trimer receptor or higher
US8597615B2 (en) 2004-03-19 2013-12-03 Amgen Fremont Inc. Methods of monitoring a human anti-human antibody response and inhibitors thereof
WO2005092926A2 (en) 2004-03-19 2005-10-06 Amgen Inc. Reducing the risk of human and anti-human antibodies through v gene manipulation
US8198508B2 (en) 2004-03-19 2012-06-12 Amgen Fremont, Inc. Reducing the risk of human anti-human antibodies through V gene manipulation
EP2418224A2 (en) 2004-03-19 2012-02-15 Amgen Inc. Reducing the risk of human and anti-human antibodies through V gene manipulation
US7625549B2 (en) 2004-03-19 2009-12-01 Amgen Fremont Inc. Determining the risk of human anti-human antibodies in transgenic mice
EP2343384A2 (en) 2004-03-23 2011-07-13 Oncotherapy Science, Inc. Method for diagnosing non-small cell lung cancer
EP2301576A1 (en) 2004-03-29 2011-03-30 Abbott Biotherapeutics Corp. Therapeutic use of Anti-CS1 Antibodies
EP2062917A2 (en) 2004-04-09 2009-05-27 Abbott Laboratories Antibodies to erythropoietin receptor and uses thereof
EP2204191A1 (en) 2004-04-15 2010-07-07 Galaxy Biotech, LLC Monoclonal antibodies to hepatocyte growth factor
US7863029B2 (en) 2004-06-04 2011-01-04 Fox Chase Cancer Center Alternate morpheeins of allosteric proteins as a target for the development of bioactive molecules
EP2662390A1 (en) 2004-06-21 2013-11-13 Medarex, L.L.C. Interferon alpha receptor 1 antibodies and their uses
WO2006002177A2 (en) 2004-06-21 2006-01-05 Medarex, Inc. Interferon alpha receptor 1 antibodies and their uses
EP2287195A2 (en) 2004-07-01 2011-02-23 Novo Nordisk A/S Pan-KIR2DL NK-receptor antibodies and their use in diagnostik and therapy
WO2006003179A2 (en) 2004-07-01 2006-01-12 Novo Nordisk A/S Antibodies binding to receptors kir2dl1, -2, 3 but not kir2ds4 and their therapeutic use
US7973134B2 (en) 2004-07-07 2011-07-05 Cell Signaling Technology, Inc. Reagents for the detection of protein phosphorylation in anaplastic large cell lymphoma signaling pathways
EP2314317A2 (en) 2004-07-09 2011-04-27 Chugai Seiyaku Kabushiki Kaisha Anti-glypican 3 antibody
EP2216046A2 (en) 2004-07-09 2010-08-11 Chugai Seiyaku Kabushiki Kaisha Anti-glypican 3 antibody
EP2898897A2 (en) 2004-07-09 2015-07-29 Chugai Seiyaku Kabushiki Kaisha Anti-glypican 3 antibody
US7935790B2 (en) 2004-10-04 2011-05-03 Cell Singaling Technology, Inc. Reagents for the detection of protein phosphorylation in T-cell receptor signaling pathways
EP2272539A2 (en) 2004-10-19 2011-01-12 Elan Pharmaceuticals, Inc. Truncated fragments of alpha-synuclein in lewy body disease
EP2527456A1 (en) 2004-10-22 2012-11-28 Revivicor Inc. Transgenic porcines lacking endogenous immunoglobulin light chain
US9585374B2 (en) 2004-10-22 2017-03-07 Revivicor, Inc. Ungulates with genetically modified immune systems
US11085054B2 (en) 2004-10-22 2021-08-10 Revivicor, Inc. Ungulates with genetically modified immune systems
EP2465872A2 (en) 2004-10-25 2012-06-20 Merck Sharp & Dohme Corporation Anti-ADDL Antibodies and uses thereof
WO2006055638A2 (en) 2004-11-17 2006-05-26 Abgenix, Inc. Fully human monoclonal antibodies to il-13
US8916165B2 (en) 2004-12-15 2014-12-23 Janssen Alzheimer Immunotherapy Humanized Aβ antibodies for use in improving cognition
WO2006068975A2 (en) 2004-12-20 2006-06-29 Abgenix, Inc. Binding proteins specific for human matriptase
US7807789B2 (en) 2004-12-21 2010-10-05 Cell Signaling Technology, Inc. Reagents for the detection of protein phosphorylation in EGFR-signaling pathways
EP2284194A1 (en) 2004-12-21 2011-02-16 AstraZeneca AB Antibodies directed to angiopoietin-2 and uses thereof
EP3699191A1 (en) 2004-12-21 2020-08-26 MedImmune Limited Antibodies directed to angiopoietin-2 and uses thereof
EP2208783A1 (en) 2004-12-22 2010-07-21 Chugai Seiyaku Kabushiki Kaisha Method of producing an antibody using a cell in which the function of fucose transporter is inhibited
EP2216404A1 (en) 2004-12-22 2010-08-11 Chugai Seiyaku Kabushiki Kaisha Method of producing an antibody using a cell in which the function of fucose transporter is inhibited
EP2361933A2 (en) 2005-01-26 2011-08-31 Amgen Fremont Inc. Antibodies against interleukin-1 beta
EP2384767A1 (en) 2005-03-24 2011-11-09 Millennium Pharmaceuticals, Inc. Antibodies that bind OV064 and methods of use therefor
EP3050963A1 (en) 2005-03-31 2016-08-03 Chugai Seiyaku Kabushiki Kaisha Process for production of polypeptide by regulation of assembly
EP3623473A1 (en) 2005-03-31 2020-03-18 Chugai Seiyaku Kabushiki Kaisha Process for production of polypeptide by regulation of assembly
EP2824183A1 (en) 2005-04-08 2015-01-14 Chugai Seiyaku Kabushiki Kaisha Antibody substituting for function of blood coagulation factor VIII
EP2418278A2 (en) 2005-05-09 2012-02-15 Ono Pharmaceutical Co., Ltd. Human monoclonal antibodies to programmed death 1(PD-1) and methods for treating cancer using anti-PD-1 antibodies alone or in combination with other immunotherapeutics
EP3530736A2 (en) 2005-05-09 2019-08-28 ONO Pharmaceutical Co., Ltd. Human monoclonal antibodies to programmed death 1 (pd-1) and methods for treating cancer using anti-pd-1 antibodies alone or in combination with other immunotherapeutics
EP2439273A2 (en) 2005-05-09 2012-04-11 Ono Pharmaceutical Co., Ltd. Human monoclonal antibodies to programmed death 1(PD-1) and methods for treating cancer using anti-PD-1 antibodies alone or in combination with other immunotherapeutics
EP2439272A2 (en) 2005-05-09 2012-04-11 Ono Pharmaceutical Co., Ltd. Human monoclonal antibodies to programmed death 1(PD-1) and methods for treating cancer using anti-PD-1 antibodies alone or in combination with other immunotherapeutics
EP2161336A1 (en) 2005-05-09 2010-03-10 ONO Pharmaceutical Co., Ltd. Human monoclonal antibodies to programmed death 1(PD-1) and methods for treating cancer using anti-PD-1 antibodies alone or in combination with other immunotherapeutics
WO2006132363A1 (en) 2005-06-10 2006-12-14 Chugai Seiyaku Kabushiki Kaisha Stabilizer for protein preparation comprising meglumine and use thereof
EP2982379A1 (en) 2005-07-01 2016-02-10 E. R. Squibb & Sons, L.L.C. Human monoclonal antibodies to programmed death ligand 1 (pd-l1)
EP2397497A2 (en) 2005-07-18 2011-12-21 Amgen, Inc Human anti-B7RP1 neutralizing antibodies
EP2388276A2 (en) 2005-07-18 2011-11-23 Amgen, Inc Human anti-B7RP1 neutralizing antibodies
EP2397499A2 (en) 2005-07-18 2011-12-21 Amgen, Inc Human anti-B7RP1 neutralizing antibodies
US10072090B2 (en) 2005-07-18 2018-09-11 Amgen Inc. Human anti-B7RP1 neutralizing antibodies
EP2397498A2 (en) 2005-07-18 2011-12-21 Amgen, Inc Human anti-B7RP1 neutralizing antibodies
EP2397496A2 (en) 2005-07-18 2011-12-21 Amgen, Inc Human anti-B7RP1 neutralizing antibodies
US9266945B2 (en) 2005-07-18 2016-02-23 Amgen Inc. Human anti-B7RP1 neutralizing antibodies
US8981071B2 (en) 2005-07-18 2015-03-17 Amgen, Inc. Human anti-B7RP1 neutralizing antibodies
EP2388277A2 (en) 2005-07-18 2011-11-23 Amgen, Inc Human anti-B7RP1 neutralizing antibodies
US7868140B2 (en) 2005-07-18 2011-01-11 Amgen Inc. Human anti-B7RP1 neutralizing antibodies
US9309315B2 (en) 2005-08-18 2016-04-12 Genmab A/S Therapy with CD4 binding peptides and radiation
US7888480B2 (en) 2005-08-31 2011-02-15 Cell Signaling Technology, Inc. Reagents for the detection of protein phosphorylation in leukemia signaling pathways
US8193328B2 (en) 2005-09-08 2012-06-05 Philadelphia Health & Education Corporation Identification of modulators of serine protease inhibitor Kazal and their use as anti-cancer and anti-viral agents
WO2007038637A2 (en) 2005-09-26 2007-04-05 Medarex, Inc. Human monoclonal antibodies to cd70
WO2007043641A1 (en) 2005-10-14 2007-04-19 Fukuoka University Inhibitor of transplanted islet dysfunction in islet transplantation
EP2402372A2 (en) 2005-10-18 2012-01-04 Medella Therapeutics Ltd Therapeutic agent
WO2007046489A1 (en) 2005-10-21 2007-04-26 Chugai Seiyaku Kabushiki Kaisha Therapeutic agent for heart disease
EP2532677A1 (en) 2005-10-21 2012-12-12 Novartis AG Human antibodies against il13 and therapeutic uses
EP2532679A1 (en) 2005-10-21 2012-12-12 Novartis AG Human antibodies against il13 and therapeutic uses
EP2548583A2 (en) 2005-11-10 2013-01-23 Curagen Corporation Method of treating ovarian and renal cancer using antibodies against t cell immunoglobulin domain and mucin domain 1 (tim-1) antigen
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
EP2816060A1 (en) 2005-11-14 2014-12-24 Amgen Inc. Rankl antibody-PTH/PTHRP chimeric molecules
EP2366715A2 (en) 2005-11-14 2011-09-21 Amgen Inc. Rankl Antibody-PTH/PTHRP Chimeric Molecules
WO2007055378A1 (en) 2005-11-14 2007-05-18 Cell Signals Inc. Method for treatment or prevention of disease associated with functional disorder of regulatory t cell
WO2007058194A1 (en) 2005-11-15 2007-05-24 National Hospital Organization Inhibitor of cytotoxic t cell induction
WO2007061029A1 (en) 2005-11-25 2007-05-31 Keio University Therapeutic agent for prostate cancer
US9829494B2 (en) 2005-12-01 2017-11-28 Adrenomed Ag Methods of treatment using ADM antibodies
US9062111B2 (en) 2005-12-07 2015-06-23 Medarex, L.L.C. CTLA-4 antibody dosage escalation regimens
US9573999B2 (en) 2005-12-07 2017-02-21 E. R. Squibb & Sons, L.L.C. CTLA-4 antibody dosage escalation regimens
WO2007067992A2 (en) 2005-12-08 2007-06-14 Medarex, Inc. Human monoclonal antibodies to fucosyl-gm1 and methods for using anti-fucosyl-gm1
EP2476706A2 (en) 2005-12-12 2012-07-18 Bayer HealthCare LLC Anti-MN antibodies and methods of using same
EP2404616A2 (en) 2005-12-13 2012-01-11 AstraZeneca AB Binding proteins specific for insulin-like growth factors and uses thereof
US8372412B2 (en) 2005-12-23 2013-02-12 Rapid Biosensor Systems Limited Bioassay and peptides for use therein
WO2007074880A1 (en) 2005-12-28 2007-07-05 Chugai Seiyaku Kabushiki Kaisha Antibody-containing stabilizing preparation
WO2007077028A2 (en) 2005-12-30 2007-07-12 U3 Pharma Ag Antibodies directed to her-3 and uses thereof
EP3950715A1 (en) 2005-12-30 2022-02-09 Amgen Inc. Antibodies directed to her-3 and uses thereof
EP3196213A2 (en) 2005-12-30 2017-07-26 Daiichi Sankyo Europe GmbH Antibodies directed to her-3 and uses thereof
EP2993187A2 (en) 2005-12-30 2016-03-09 U3 Pharma GmbH Antibodies directed to her-3 and uses thereof
EP2463305A1 (en) 2006-01-12 2012-06-13 Alexion Pharmaceuticals, Inc. Antibodies to OX-2/CD200 and uses thereof
EP3101033A1 (en) 2006-01-12 2016-12-07 Alexion Pharmaceuticals, Inc. Antibodies to ox-2/cd200 and uses thereof
WO2007084672A2 (en) 2006-01-17 2007-07-26 Medarex, Inc. Monoclonal antibodies against cd30 lacking in fucosyl and xylosyl residues
WO2007086490A1 (en) 2006-01-27 2007-08-02 Keio University Remedy for disease associated with choroidal angiogenesis
EP3135298A1 (en) 2006-01-27 2017-03-01 Keio University Remedy for disease associated with choroidal angiogenesis
EP2505058A1 (en) 2006-03-31 2012-10-03 Medarex, Inc. Transgenic animals expressing chimeric antibodies for use in preparing human antibodies
US8232449B2 (en) 2006-03-31 2012-07-31 Medarex, Inc. Transgenic animals expressing chimeric antibodies for use in preparing human antibodies
EP4342995A2 (en) 2006-03-31 2024-03-27 Chugai Seiyaku Kabushiki Kaisha Methods for controlling blood pharmacokinetics of antibodies
US7910798B2 (en) 2006-03-31 2011-03-22 Medarex, Inc. Transgenic animals expressing chimeric antibodies for use in preparing human antibodies
WO2007117410A2 (en) 2006-03-31 2007-10-18 Medarex, Inc. Transgenic animals expressing chimeric antibodies for use in preparing human antibodies
EP3345616A1 (en) 2006-03-31 2018-07-11 Chugai Seiyaku Kabushiki Kaisha Antibody modification method for purifying bispecific antibody
EP4001409A1 (en) 2006-03-31 2022-05-25 Chugai Seiyaku Kabushiki Kaisha Methods for controlling blood pharmacokinetics of antibodies
WO2007114319A1 (en) 2006-03-31 2007-10-11 Chugai Seiyaku Kabushiki Kaisha Method for control of blood kinetics of antibody
EP4218801A2 (en) 2006-03-31 2023-08-02 Chugai Seiyaku Kabushiki Kaisha Antibody modification method for purifying bispecific antibody
US9220244B2 (en) 2006-03-31 2015-12-29 E. R. Squibb & Sons, L.L.C. Transgenic animals expressing chimeric antibodies for use in preparing human antibodies
WO2007114325A1 (en) 2006-03-31 2007-10-11 Chugai Seiyaku Kabushiki Kaisha Antibody modification method for purifying bispecific antibody
EP2476704A2 (en) 2006-04-01 2012-07-18 Galaxy Biotech, LLC Humanized monoclonal antibodies to heptocyte growth factor
US9926367B2 (en) 2006-04-07 2018-03-27 Aerpio Therapeutics, Inc. Antibodies that bind human protein tyrosine phosphatase beta (HPTPbeta) and uses thereof
WO2007116962A1 (en) 2006-04-07 2007-10-18 Osaka University Muscle regeneration promoter
EP3252079A1 (en) 2006-04-07 2017-12-06 Aerpio Therapeutics, Inc. Antibodies that bind human protein tyrosine phosphatase beta (hptp-ss) and uses thereof
US8071323B2 (en) 2006-04-07 2011-12-06 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Human monoclonal antibodies that bind human insulin like growth factors and their use
US11814425B2 (en) 2006-04-07 2023-11-14 Eye Point Pharmaceuticals, Inc. Antibodies that bind human protein tyrosine phosphatase beta (HPTPbeta) and uses thereof
EP2371865A2 (en) 2006-04-07 2011-10-05 Warner Chilcott Company, LLC Antibodies that bind human protein tyrosine phosphatase beta (HPTP-ß) and uses thereof
EP2357202A1 (en) 2006-04-10 2011-08-17 AstraZeneca AB Targeted binding agents directed to Upar and uses thereof
WO2007119796A1 (en) 2006-04-14 2007-10-25 Medical And Biological Laboratories Co., Ltd. Mutant polypeptide having effector function
US8784810B2 (en) 2006-04-18 2014-07-22 Janssen Alzheimer Immunotherapy Treatment of amyloidogenic diseases
WO2007129457A1 (en) 2006-04-25 2007-11-15 The University Of Tokyo Therapeutic agents for alzheimer's disease and cancer
WO2008042024A2 (en) 2006-06-01 2008-04-10 Elan Pharmaceuticals, Inc. Neuroactive fragments of app
EP2371393A1 (en) 2006-06-08 2011-10-05 Chugai Seiyaku Kabushiki Kaisha Preventive or remedy for inflammatory disease
WO2007142325A1 (en) 2006-06-08 2007-12-13 Chugai Seiyaku Kabushiki Kaisha Preventive or remedy for inflammatory disease
WO2007145227A1 (en) 2006-06-14 2007-12-21 Chugai Seiyaku Kabushiki Kaisha Hematopoietic stem cell proliferation promoter
WO2008007755A1 (en) 2006-07-13 2008-01-17 Chugai Seiyaku Kabushiki Kaisha Cell death inducer
EP2574625A1 (en) 2006-07-21 2013-04-03 Chugai Seiyaku Kabushiki Kaisha Remedy for renal disease
WO2008010556A1 (en) 2006-07-21 2008-01-24 Chugai Seiyaku Kabushiki Kaisha Remedy for renal disease
EP2420513A1 (en) 2006-08-03 2012-02-22 MedImmune Limited Targeted binding agents directed to PDGFR-alpha and uses thereof
US8894998B2 (en) 2006-08-03 2014-11-25 Medimmune Limited Antibodies directed to αVβ6 and uses thereof
EP2420514A1 (en) 2006-08-03 2012-02-22 MedImmune Limited Targeted binding agents directed to PDGFR-alpha and uses thereof
US8398975B2 (en) 2006-08-03 2013-03-19 Medimmune Limited Antibodies directed to αVβ6 and uses thereof
US7939636B2 (en) 2006-08-11 2011-05-10 Cell Signaling Technology, Inc. Reagents for the detection of protein phosphorylation in c-Src signaling pathways
EP3111955A1 (en) 2006-08-14 2017-01-04 Chugai Seiyaku Kabushiki Kaisha Diagnosis and treatment of cancer using anti-desmoglein-3 antibody
EP2548576A1 (en) 2006-08-14 2013-01-23 Forerunner Pharma Research Co., Ltd. Diagnosis of cancer using anti-desmoglein-3 antibodies
WO2008030611A2 (en) 2006-09-05 2008-03-13 Medarex, Inc. Antibodies to bone morphogenic proteins and receptors therefor and methods for their use
EP2628750A2 (en) 2006-09-08 2013-08-21 The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc. Targeted identification of immunogenic peptides
WO2008032833A1 (en) 2006-09-14 2008-03-20 Medical & Biological Laboratories Co., Ltd. Antibody having enhanced adcc activity and method for production thereof
EP2486941A1 (en) 2006-10-02 2012-08-15 Medarex, Inc. Human antibodies that bind CXCR4 and uses thereof
WO2008047723A1 (en) 2006-10-12 2008-04-24 Forerunner Pharma Research Co., Ltd. Diagnosis and treatment of cancer using anti-ereg antibody
EP2530090A2 (en) 2006-10-19 2012-12-05 CSL Limited Anti-IL-13R alpha 1 antibodies and their uses thereof
EP3040347A2 (en) 2006-10-20 2016-07-06 Chugai Seiyaku Kabushiki Kaisha Pharmaceutical composition comprising anti-hb-egf antibody as active ingredient
WO2008047914A1 (en) 2006-10-20 2008-04-24 Forerunner Pharma Research Co., Ltd. Cancer therapeutic agent comprising anti-hb-egf antibody as active ingredient
WO2008047925A1 (en) 2006-10-20 2008-04-24 Forerunner Pharma Research Co., Ltd. Pharmaceutical composition comprising anti-hb-egf antibody as active ingredient
WO2008076560A2 (en) 2006-11-15 2008-06-26 Medarex, Inc. Human monoclonal antibodies to btla and methods of use
WO2008070569A2 (en) 2006-12-01 2008-06-12 Medarex, Inc. Human antibodies that bind cd22 and uses thereof
WO2009054863A2 (en) 2006-12-13 2009-04-30 Medarex, Inc. Human antibodies that bind cd19 and uses thereof
WO2008072723A1 (en) 2006-12-14 2008-06-19 Forerunner Pharma Research Co., Ltd. ANTI-Claudin-3 MONOCLONAL ANTIBODY, AND TREATMENT AND DIAGNOSIS OF CANCER USING THE SAME
WO2008074004A2 (en) 2006-12-14 2008-06-19 Medarex, Inc. Human antibodies that bind cd70 and uses thereof
WO2008081942A1 (en) 2007-01-05 2008-07-10 The University Of Tokyo Diagnosis and treatment of cancer by using anti-prg-3 antibody
WO2008090901A1 (en) 2007-01-23 2008-07-31 Shinshu University Chronic rejection inhibitor
WO2008099920A1 (en) 2007-02-15 2008-08-21 Kyushu University, National University Corporation Therapeutic agent for interstitial pulmonary disease comprising anti-hmgb-1 antibody
EP3248617A2 (en) 2007-02-16 2017-11-29 Merrimack Pharmaceuticals, Inc. Antibodies against erbb3 and uses thereof
EP2647388A1 (en) 2007-02-16 2013-10-09 Merrimack Pharmaceuticals, Inc. Antibodies Against ERBB3 and Uses Thereof
EP2716301A2 (en) 2007-02-16 2014-04-09 Merrimack Pharmaceuticals, Inc. Antibodies against ERBB3 and uses thereof
WO2008105560A1 (en) 2007-02-27 2008-09-04 Forerunner Pharma Research Co., Ltd. Pharmaceutical composition comprising anti-grp78 antibody as active ingredient
EP1972639A2 (en) 2007-03-07 2008-09-24 Cell Signaling Technology, Inc. Reagents for the detection of protein phosphorylation in carcinoma signaling pathways
WO2008111597A1 (en) 2007-03-12 2008-09-18 Chugai Seiyaku Kabushiki Kaisha Remedy for chemotherapy-resistant cancer containing hla class i-recognizing antibody as the active ingredient and use of the same
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
EP1975184A2 (en) 2007-03-26 2008-10-01 Albrecht Moritz Serine or threonine phosphorylation sites
US8003097B2 (en) 2007-04-18 2011-08-23 Janssen Alzheimer Immunotherapy Treatment of cerebral amyloid angiopathy
EP2145902A2 (en) 2007-04-19 2010-01-20 Peter Hornbeck Tyrosine phosphorylation sites and antibodies specific for them
EP1983002A2 (en) 2007-04-19 2008-10-22 Peter Hornbeck Tyrosine phosphorylation sites and antibodies specific for them
US7977462B2 (en) 2007-04-19 2011-07-12 Cell Signaling Technology, Inc. Tyrosine phosphorylation sites
EP1983003A2 (en) 2007-04-19 2008-10-22 Peter Hornbeck Tyrosine phosphorylation sites and antibodies specific for them
EP2692737A1 (en) 2007-04-20 2014-02-05 Biotie Therapies Corp. Fully human anti-vap-1 monoclonal antibodies
EP2123679A2 (en) 2007-05-01 2009-11-25 Peter Hornbeck Tyrosine phosphorylation sites
US7875705B2 (en) 2007-05-28 2011-01-25 The University Of Tokyo Tumor diagnostic agent used in PET comprising anti-ROBO1 antibody
US7709215B2 (en) 2007-06-01 2010-05-04 Cytonics Corporation Method for diagnosing and treating acute joint injury
WO2008153926A2 (en) 2007-06-05 2008-12-18 Yale University Inhibitors of receptor tyrosine kinases and methods of use thereof
US9289511B2 (en) 2007-06-21 2016-03-22 The Boeing Company Bioconjugated nanoparticles
WO2009001840A1 (en) 2007-06-25 2008-12-31 Forerunner Pharma Research Co., Ltd. Anti-prominin-1 antibody having adcc activity or cdc activity
EP3246045A1 (en) 2007-07-26 2017-11-22 Osaka University Therapeutic agents for ocular inflammatory disease comprising interleukin 6 receptor inhibitor as active ingredient
WO2009014263A1 (en) 2007-07-26 2009-01-29 Osaka University Agent for treatment of ophthalmia containing interleukin-6 receptor inhibitor as active ingredient
US8613920B2 (en) 2007-07-27 2013-12-24 Janssen Alzheimer Immunotherapy Treatment of amyloidogenic diseases
WO2009054873A2 (en) 2007-08-02 2009-04-30 Novimmune S.A. Anti-rantes antibodies and methods of use thereof
EP3255144A1 (en) 2007-08-10 2017-12-13 E. R. Squibb & Sons, L.L.C. Recombineering construct for preparing transgenic mice capable of producing human immunoglobulin
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
US8859741B2 (en) 2007-08-23 2014-10-14 Amgen Inc. Antigen binding proteins to proprotein convertase subtilisin kexin type 9 (PCSK9)
US8030457B2 (en) 2007-08-23 2011-10-04 Amgen, Inc. Antigen binding proteins to proprotein convertase subtilisin kexin type 9 (PCSK9)
US9920134B2 (en) 2007-08-23 2018-03-20 Amgen Inc. Monoclonal antibodies to proprotein convertase subtilisin kexin type 9 (PCSK9)
US8981064B2 (en) 2007-08-23 2015-03-17 Amgen Inc. Antigen binding proteins to proprotein convertase subtilisin kexin type 9 (PCSK9)
EP3202791A1 (en) 2007-08-23 2017-08-09 Amgen, Inc Antigen binding proteins to proprotein convertase subtilistin kexin type 9 (pcsk9)
US8871913B2 (en) 2007-08-23 2014-10-28 Amgen Inc. Antigen binding proteins to proprotein convertase subtilisin kexin type 9 (PCSK9)
US8871914B2 (en) 2007-08-23 2014-10-28 Amgen, Inc. Antigen binding proteins to proprotein convertase subtilisin kexin type 9 (PCSK9)
US9493576B2 (en) 2007-08-23 2016-11-15 Amgen Inc. Antigen binding proteins to proprotein convertase subtilisin kexin type 9 (PCSK9)
EP4248976A2 (en) 2007-08-23 2023-09-27 Amgen Inc. Antigen binding proteins to proprotein convertase subtilisin kexin type 9 (pcsk9)
US9045547B2 (en) 2007-08-23 2015-06-02 Amgen Inc. Methods of using antigen binding proteins to proprotein convertase subtilisin kexin type 9 (PCSK9)
US8168762B2 (en) 2007-08-23 2012-05-01 Amgen Inc. Antigen binding proteins to proprotein convertase subtilisin kexin type 9 (PCSK9)
US9056915B2 (en) 2007-08-23 2015-06-16 Amgen Inc. Antigen binding proteins to proprotein convertase subtilisin kexin type 9 (PCSK9)
US8883983B2 (en) 2007-08-23 2014-11-11 Amgen Inc. Antigen binding proteins to proprotein convertase subtilisin kexin type 9 (PCSK9)
EP3666797A1 (en) 2007-08-23 2020-06-17 Amgen, Inc Antigen binding proteins to proprotein convertase subtilisin kexin type 9 (pcsk9)
DE202008018562U1 (en) 2007-08-23 2015-11-02 Amgen Inc. Antigen-binding Proteins Against Proprotein Convertase Subtilisin Kexin Type 9 (PCSK9)
US8889834B2 (en) 2007-08-23 2014-11-18 Amgen Inc. Antigen binding proteins to proprotein convertase subtilisin kexin type 9 (PCSK9)
US9555087B2 (en) 2007-09-04 2017-01-31 Compugen Ltd Polypeptides and polynucleotides, and uses thereof as a drug target for producing drugs and biologics
EP2769728A1 (en) 2007-09-04 2014-08-27 Compugen Ltd. Polypeptides and polynucleotides, and uses thereof as a drug target for producing drugs and biologics
EP2769729A1 (en) 2007-09-04 2014-08-27 Compugen Ltd. Polypeptides and polynucleotides, and uses thereof as a drug target for producing drugs and biologics
US9375466B2 (en) 2007-09-04 2016-06-28 Compugen Ltd Polypeptides and polynucleotides, and uses thereof as a drug target for producing drugs and biologics
US9107862B2 (en) 2007-09-04 2015-08-18 Compugen Ltd. Polypeptides and polynucleotides, and uses thereof as a drug target for producing drugs and biologics
WO2009032845A2 (en) 2007-09-04 2009-03-12 Compugen, Ltd. Polypeptides and polynucleotides, and uses thereof as a drug target for producing drugs and biologics
US10098934B2 (en) 2007-09-04 2018-10-16 Compugen Ltd Polypeptides and polynucleotides, and uses thereof as a drug target for producing drugs and biologics
EP2236519A1 (en) 2007-09-18 2010-10-06 Amgen, Inc Human GM-CSF antigen binding proteins
EP3127921A1 (en) 2007-09-26 2017-02-08 Chugai Seiyaku Kabushiki Kaisha Method of modifying isoelectric point of antibody via amino acid substition in cdr
EP4339294A2 (en) 2007-09-26 2024-03-20 Chugai Seiyaku Kabushiki Kaisha Method of modifying isoelectric point of antibody via amino acid substitution in cdr
EP3689912A1 (en) 2007-09-26 2020-08-05 Chugai Seiyaku Kabushiki Kaisha Method of modifying isoelectric point of antibody via amino acid substitution in cdr
EP2497783A2 (en) 2007-09-26 2012-09-12 U3 Pharma GmbH Heparin-binding epidermal growth factor-like growth factor antigen binding proteins
WO2009040134A1 (en) 2007-09-26 2009-04-02 U3 Pharma Gmbh Heparin-binding epidermal growth factor-like growth factor antigen binding proteins
EP2617736A1 (en) 2007-09-28 2013-07-24 Chugai Seiyaku Kabushiki Kaisha Anti-glypican 3 antibody having improved kinetics in plasma
US8497355B2 (en) 2007-09-28 2013-07-30 Chugai Seiyaku Kabushiki Kaisha Anti-glypican-3 antibody having improved kinetics in plasma
WO2009041062A1 (en) 2007-09-28 2009-04-02 Chugai Seiyaku Kabushiki Kaisha Anti-glypican-3 antibody having improved kinetics in plasma
EP2584043A2 (en) 2007-09-28 2013-04-24 Chugai Seiyaku Kabushiki Kaisha Anti-glypican 3 antibody having improved kinetics in plasma
WO2009044774A1 (en) 2007-10-02 2009-04-09 Chugai Seiyaku Kabushiki Kaisha Remedy for graft-versus-host disease comprising interleukin-6 receptor inhibitor as the active ingredient
EP2586796A1 (en) 2007-10-12 2013-05-01 Novartis AG Compositions and methods for use for antibodies against sclerostin
WO2009051108A1 (en) 2007-10-15 2009-04-23 Chugai Seiyaku Kabushiki Kaisha Method for production of antibody
US9644025B2 (en) 2007-10-17 2017-05-09 Wyeth Llc Immunotherapy regimes dependent on ApoE status
EP2952524A1 (en) 2007-10-17 2015-12-09 Janssen Sciences Ireland UC Immunotherapy regimes dependent on apoe status
WO2009052439A2 (en) 2007-10-17 2009-04-23 Elan Pharma International Limited Immunotherapy regimes dependent on apoe status
EP3072963A1 (en) 2007-10-18 2016-09-28 Cell Signaling Technology, Inc. Translocation and mutant ros kinase in human non-small cell lung carcinoma
EP3741851A1 (en) 2007-10-18 2020-11-25 Cell Signaling Technology, Inc. Translocation and mutant ros kinase in human non-small cell lung carcinoma
WO2009054435A1 (en) 2007-10-24 2009-04-30 Otsuka Chemical Co., Ltd. Polypeptide having enhanced effector function
EP3305324A1 (en) 2007-11-02 2018-04-11 Novartis AG Molecules and methods for modulating low-density-lipoprotein receptor-related protein 6 (lrp6)
EP2567709A2 (en) 2007-11-02 2013-03-13 Novartis AG Molecules and methods for modulating low-density-lipoprotein receptor-related protein 6 (LRP6)
US8586720B2 (en) 2007-11-07 2013-11-19 Celldex Therapeutics Inc. Antibodies that bind human dendritic and epithelial cell 205 (DEC-205)
US8236318B2 (en) 2007-11-07 2012-08-07 Celldex Therapeutics Inc. Antibodies that bind human dendritic and epithelial cell 205 (DEC-205)
US8362214B2 (en) 2007-11-07 2013-01-29 Celldex Therapeutics Inc. Antibodies that bind human dendritic and epithelial cell 205 (DEC-205)
US9624300B2 (en) 2007-11-07 2017-04-18 Celldex Therapeutics Inc. Antibodies that bind human dendritic and epithelial cell 205 (DEC-205)
EP2918605A1 (en) 2007-11-12 2015-09-16 U3 Pharma GmbH Axl antibodies
WO2009063970A1 (en) 2007-11-14 2009-05-22 Forerunner Pharma Research Co., Ltd. Diagnosis and treatment of cancer using anti-gpr49 antibody
EP2853544A1 (en) 2007-11-15 2015-04-01 Chugai Seiyaku Kabushiki Kaisha Monoclonal antibody capable of binding to anexelekto, and use thereof
EP2062920A2 (en) 2007-11-21 2009-05-27 Peter Hornbeck Protein phosphorylation by basophilic serine/threonine kinases in insulin signalling pathways
EP3095862A1 (en) 2007-12-05 2016-11-23 Chugai Seiyaku Kabushiki Kaisha Anti-nr10 antibody and use thereof
WO2009072604A1 (en) 2007-12-05 2009-06-11 Chugai Seiyaku Kabushiki Kaisha Anti-nr10 antibody and use thereof
WO2009072598A1 (en) 2007-12-05 2009-06-11 Chugai Seiyaku Kabushiki Kaisha Therapeutic agent for pruritus
WO2009075344A1 (en) 2007-12-12 2009-06-18 Japan As Represented By Director General Of Agency Of National Cancer Center Therapeutic agent for mll leukemia and moz leukemia of which molecular target is m-csf receptor, and use thereof
EP2769993A1 (en) 2007-12-14 2014-08-27 Novo Nordisk A/S Antibodies against human NKG2D and uses thereof
US8779088B2 (en) 2007-12-17 2014-07-15 Marfl Ab Vaccine for the treatment of Mycobacterium related disorders
WO2009078799A1 (en) 2007-12-17 2009-06-25 Marfl Ab New vaccine for the treatment of mycobacterium related disorders
WO2009084659A1 (en) 2007-12-27 2009-07-09 Chugai Seiyaku Kabushiki Kaisha Solution preparation containing antibody at high concentration
EP2730659A2 (en) 2007-12-28 2014-05-14 Elan Pharmaceuticals Inc. Treatment and Prophylaxis of Amyloidosis
WO2009086539A2 (en) 2007-12-28 2009-07-09 Elan Pharmaceuticals, Inc. Treatment and prophylaxis of amyloidosis
EP3693470A1 (en) 2007-12-28 2020-08-12 Prothena Therapeutics Limited Treatment and prophylaxis of amyloidosis
EP3064512A2 (en) 2008-01-11 2016-09-07 The University of Tokyo Anti-cldn6 antibody
EP4282428A2 (en) 2008-01-11 2023-11-29 The University of Tokyo Anti-cldn6 antibody
WO2009087978A1 (en) 2008-01-11 2009-07-16 The University Of Tokyo Anti-cldn6 antibody
FR2926438A1 (en) * 2008-01-22 2009-07-24 Univ Limoges NON-HUMAN TRANSGENIC MAMMAL FOR THE CONSTANT REGION OF THE HEAVY CHAIN OF HUMAN CLASS G IMMUNOGLOBULINS AND ITS APPLICATIONS
WO2009106773A3 (en) * 2008-01-22 2009-10-22 Universite De Limoges Transgenic mice for the c gamma gene human class-g immunoglobulins
EP2641612A1 (en) 2008-02-05 2013-09-25 Bristol-Myers Squibb Company Alpha 5 - beta 1 antibodies and their uses
EP2650017A2 (en) 2008-02-05 2013-10-16 Bristol-Myers Squibb Company Alpha 5 - beta 1 antibodies and their uses
WO2009122667A1 (en) 2008-04-04 2009-10-08 中外製薬株式会社 Therapeutic for hepatic cancer
EP3023502A1 (en) 2008-04-10 2016-05-25 Cell Signaling Technology, Inc. Compositions and methods for detecting egfr mutations in cancer
EP3521311A1 (en) 2008-04-11 2019-08-07 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule capable of binding to two or more antigen molecules repeatedly
EP3056513A1 (en) 2008-04-11 2016-08-17 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule capable of binding to two or more antigen molecules repeatedly
EP3514180A1 (en) 2008-04-11 2019-07-24 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule capable of binding to two or more antigen molecules repeatedly
EP2708559A2 (en) 2008-04-11 2014-03-19 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule capable of binding to two or more antigen molecules repeatedly
WO2009125825A1 (en) 2008-04-11 2009-10-15 中外製薬株式会社 Antigen-binding molecule capable of binding to two or more antigen molecules repeatedly
EP4238993A2 (en) 2008-04-11 2023-09-06 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule capable of binding to two or more antigen molecules repeatedly
EP2708558A2 (en) 2008-04-11 2014-03-19 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule capable of binding to two or more antigen molecules repeatedly
WO2009148928A1 (en) 2008-05-29 2009-12-10 Galaxy Biotech, Llc Monoclonal antibodies to basic fibroblast growth factor
WO2009147781A1 (en) 2008-06-02 2009-12-10 国立大学法人東京大学 Antitumor agent
WO2009148148A1 (en) 2008-06-05 2009-12-10 国立がんセンター総長が代表する日本国 Neuroinvasion inhibitor
WO2009154025A1 (en) 2008-06-20 2009-12-23 国立大学法人岡山大学 ANTIBODY AGAINST OXIDIZED LDL/β2GPI COMPLEX AND USE OF THE SAME
US8575314B2 (en) 2008-06-20 2013-11-05 National University Corporation Okayama University Antibody against oxidized LDL/β2GPI complex and use of the same
US11237165B2 (en) 2008-06-27 2022-02-01 Merus N.V. Antibody producing non-human animals
US11785924B2 (en) 2008-06-27 2023-10-17 Merus N.V. Antibody producing non-human animals
EP2815766A1 (en) 2008-08-05 2014-12-24 Novartis AG Compositions and methods for antibodies targeting complement protein C5
WO2010015608A1 (en) 2008-08-05 2010-02-11 Novartis Ag Compositions and methods for antibodies targeting complement protein c5
EP2837388A1 (en) 2008-08-05 2015-02-18 Novartis AG Compositions and methods for antibodies targeting complement protein C5
EP3597216A1 (en) 2008-08-11 2020-01-22 E. R. Squibb & Sons, L.L.C. Human antibodies that bind lymphocyte activation gene-3 (lag-3) and uses thereof
EP2905030A1 (en) 2008-08-11 2015-08-12 E. R. Squibb & Sons, L.L.C. Human antibodies that bind lymphocyte activation gene-3 (LAG-3) and uses thereof
EP4147714A1 (en) 2008-08-11 2023-03-15 E. R. Squibb & Sons, L.L.C. Human antibodies that bind lymphocyte activation gene-3 (lag-3) and uses thereof
EP2927244A1 (en) 2008-09-19 2015-10-07 MedImmune, LLC Antibodies directed to DLL4 and uses thereof
US10555506B2 (en) 2008-09-30 2020-02-11 Ablexis, Llc Non-human mammals for the production of chimeric antibodies
US10575504B2 (en) 2008-09-30 2020-03-03 Ablexis, Llc Non-human mammals for the production of chimeric antibodies
US9346873B2 (en) 2008-09-30 2016-05-24 Ablexis, Llc Non-human mammals for the production of chimeric antibodies
US10561123B2 (en) 2008-09-30 2020-02-18 Ablexis, Llc Non-human mammals for the production of chimeric antibodies
US10492476B2 (en) 2008-09-30 2019-12-03 Ablexis, Llc Non-human mammals for the production of chimeric antibodies
US10638736B2 (en) 2008-09-30 2020-05-05 Ablexis, Llc Non-human mammals for the production of chimeric antibodies
US10149461B2 (en) 2008-10-27 2018-12-11 Revivicor, Inc. Immunocompromised ungulates
US9067981B1 (en) 2008-10-30 2015-06-30 Janssen Sciences Ireland Uc Hybrid amyloid-beta antibodies
WO2010054265A2 (en) 2008-11-07 2010-05-14 Galaxy Biotech, Llc. Monoclonal antibodies to fibroblast growth factor receptor 2
EP3290052A1 (en) 2008-11-07 2018-03-07 Galaxy Biotech, LLC Monoclonal antibodies to fibroblast growth factor receptor 2
EP2842573A1 (en) 2008-11-07 2015-03-04 Galaxy Biotech, LLC Monoclonal antibodies to Fibroblast Growth Factor receptor 2
EP3783024A1 (en) 2008-11-07 2021-02-24 Galaxy Biotech, LLC Monoclonal antibodies to fibroblast growth factor receptor 2
EP2894165A1 (en) 2008-11-10 2015-07-15 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
EP3974448A1 (en) 2008-11-10 2022-03-30 Alexion Pharmaceuticals, Inc. Methods and compositions for treating complementassociated disorders
EP3121197A1 (en) 2008-11-10 2017-01-25 Alexion Pharmaceuticals, Inc. Methods and compositions for treating complement-associated disorders
EP2894166A1 (en) 2008-11-10 2015-07-15 Alexion Pharmaceuticals, Inc. Methods and compositions for treating complement-associated disorders
WO2010054403A1 (en) 2008-11-10 2010-05-14 Alexion Pharmaceuticals, Inc. Methods and compositions for treating complement-associated disorders
WO2010064697A1 (en) 2008-12-05 2010-06-10 中外製薬株式会社 Anti-nr10 antibody, and use thereof
EP2949672A1 (en) 2008-12-05 2015-12-02 Chugai Seiyaku Kabushiki Kaisha Anti-nr10 antibody, and use thereof
WO2010067308A2 (en) 2008-12-08 2010-06-17 Compugen Ltd. Polypeptides and polynucleotides, and uses thereof as a drug target for producing drugs and biologics
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
US9714297B2 (en) 2008-12-09 2017-07-25 Genmab A/S Human antibodies against tissue factor and methods of use thereof
US9150658B2 (en) 2008-12-09 2015-10-06 Genmab A/S Human antibodies against tissue factor and methods of use thereof
WO2010070263A1 (en) * 2008-12-18 2010-06-24 Erasmus University Medical Center Rotterdam Non-human transgenic animals expressing humanised antibodies and use therof
CN102292445A (en) * 2008-12-18 2011-12-21 伊拉兹马斯大学鹿特丹医学中心 Non-human transgenic animals expressing humanised antibodies and use therof
US20110314563A1 (en) * 2008-12-18 2011-12-22 Kingdon Craig R Antibody production
US9131669B2 (en) 2008-12-18 2015-09-15 Erasmus University Medical Center Antibody production
JP2012512647A (en) * 2008-12-18 2012-06-07 エラスムス・ユニヴァーシティ・メディカル・センター・ロッテルダム Non-human transgenic animals expressing humanized antibodies and uses thereof
US11877565B2 (en) 2008-12-18 2024-01-23 Erasmus University Medical Center Antibody production
WO2010074049A1 (en) 2008-12-22 2010-07-01 株式会社 未来創薬研究所 Anti-hs6st2 antibody and use thereof
WO2010072740A2 (en) 2008-12-23 2010-07-01 Astrazeneca Ab TARGETED BINDING AGENTS DIRECTED TO α5β1 AND USES THEREOF
WO2010073694A1 (en) 2008-12-25 2010-07-01 国立大学法人東京大学 Diagnosis of treatment of cancer using anti-tm4sf20 antibody
WO2010074192A1 (en) 2008-12-26 2010-07-01 国立大学法人東京大学 Diagnosis and treatment of cancer using anti-lgr7 antibody
WO2010085590A1 (en) 2009-01-23 2010-07-29 Biosynexus Incorporated Opsonic and protective antibodies specific for lipoteichoic acid gram positive bacteria
WO2010093928A2 (en) 2009-02-12 2010-08-19 Cell Signaling Technology, Inc. Mutant ros expression in human cancer
EP2881402A1 (en) 2009-02-12 2015-06-10 Cell Signaling Technology, Inc. Mutant ROS expression in human liver cancer
EP3266795A1 (en) 2009-02-12 2018-01-10 Cell Signaling Technology, Inc. Method for detecting a fig-ros fusion polynucleotide
WO2010102175A1 (en) 2009-03-05 2010-09-10 Medarex, Inc. Fully human antibodies specific to cadm1
WO2010112458A1 (en) 2009-03-31 2010-10-07 Novartis Ag Composition and methods of use for therapeutic antibodies specific for the il-12 receptore betal subunit
EP3831407A1 (en) 2009-04-08 2021-06-09 LipUm AB New methods for treatment of inflammatory diseases
EP3061463A1 (en) 2009-04-08 2016-08-31 LipUm AB New methods for treatment of inflammatory diseases
WO2010117325A1 (en) 2009-04-08 2010-10-14 Olle Hernell New methods for treatment of inflammatory diseases
WO2010119691A1 (en) 2009-04-16 2010-10-21 国立大学法人東京大学 Diagnosis and treatment of cancer using anti-tmprss11e antibody
WO2010119991A2 (en) 2009-04-17 2010-10-21 Takeda Pharmaceutical Company Limited Novel method of treating cancer
US9062116B2 (en) 2009-04-23 2015-06-23 Infinity Pharmaceuticals, Inc. Anti-fatty acid amide hydrolase-2 antibodies and uses thereof
EP3275900A1 (en) 2009-04-27 2018-01-31 Novartis AG Compositions and methods for increasing muscle growth
WO2010125003A1 (en) 2009-04-27 2010-11-04 Novartis Ag Compositions and methods for increasing muscle growth
WO2010126137A1 (en) 2009-05-01 2010-11-04 国立大学法人 東京大学 Anti-cadherin antibody
US9017683B2 (en) 2009-05-01 2015-04-28 The University Of Tokyo Highly effective anti-cadherin antibody for induction of antibody-dependent cellular cytotoxicity in vivo
US8455249B2 (en) 2009-05-01 2013-06-04 The University Of Tokyo Highly effective anti-cadherin antibody for induction of antibody-dependent cellular cytotoxicity in vivo
WO2010128398A1 (en) 2009-05-04 2010-11-11 Pangenetics 110 B.V. Antibodies against nerve growth factor (ngf) with enhanced in vivo stability
WO2010128407A2 (en) 2009-05-05 2010-11-11 Novimmune S.A. Anti-il-17f antibodies and methods of use thereof
WO2010137654A1 (en) 2009-05-29 2010-12-02 株式会社未来創薬研究所 Pharmaceutical composition containing antagonist of egf family ligand as component
WO2010151632A1 (en) 2009-06-25 2010-12-29 Bristol-Myers Squibb Company Protein purifacation by caprylic acid (octanoic acid ) precipitation
US11812731B2 (en) 2009-07-08 2023-11-14 Kymab Ltd. Animal models and therapeutic molecules
US10064398B2 (en) 2009-07-08 2018-09-04 Kymab Limited Animal models and therapeutic molecules
US9434782B2 (en) 2009-07-08 2016-09-06 Kymab Limited Animal models and therapeutic molecules
US10165763B2 (en) 2009-07-08 2019-01-01 Kymab Limited Animal models and therapeutic molecules
US11564380B2 (en) 2009-07-08 2023-01-31 Kymab Limited Animal models and therapeutic molecules
US9447177B2 (en) 2009-07-08 2016-09-20 Kymab Limited Transgenic mouse homozygous for chimeric IgH locus
US9505827B2 (en) 2009-07-08 2016-11-29 Kymab Limited Animal models and therapeutic molecules
US11606941B2 (en) 2009-07-08 2023-03-21 Kymab Limited Animal models and therapeutic molecules
US9504236B2 (en) 2009-07-08 2016-11-29 Kymab Limited Animal models and therapeutic molecules
EP2907873A1 (en) 2009-07-17 2015-08-19 Bioatla LLC Simulataneous, integrated selection and evolution of antibody/protein performance and expression in production hosts
EP3636759A1 (en) 2009-07-17 2020-04-15 BioAtla LLC Simultaneous, integrated selection and evolution of antibody/protein performance and expression in production hosts
EP3406717A1 (en) 2009-07-17 2018-11-28 Bioatla LLC Simultaneous, integrated selection and evolution of antibody/protein performance and expression in production hosts
EP3156485A1 (en) 2009-07-17 2017-04-19 Bioatla LLC Simultaneous, integrated selection and evolution of antibody/protein performance and expression in production hosts
EP3042957A1 (en) 2009-07-17 2016-07-13 Bioatla LLC Simultaneous, integrated selection and evolution of antibody/protein performance and expression in production hosts
WO2011014438A1 (en) 2009-07-31 2011-02-03 N.V. Organon Fully human antibodies to btla
WO2011013786A1 (en) 2009-07-31 2011-02-03 Maeda Shin Cancer metastasis inhibitor
EP3199551A2 (en) 2009-07-31 2017-08-02 E. R. Squibb & Sons, L.L.C. Fully human antibodies to btla
WO2011017294A1 (en) 2009-08-07 2011-02-10 Schering Corporation Human anti-rankl antibodies
WO2011021381A1 (en) 2009-08-17 2011-02-24 株式会社未来創薬研究所 Pharmaceutical composition containing anti-hb-egf antibody as active ingredient
WO2011021146A1 (en) 2009-08-20 2011-02-24 Pfizer Inc. Osteopontin antibodies
WO2011037983A1 (en) 2009-09-23 2011-03-31 Medarex, Inc. Cation exchange chromatography
US11292814B2 (en) 2009-09-23 2022-04-05 E.R. Squibb & Sons, L.L.C. Cation exchange chromatography methods
WO2011037160A1 (en) 2009-09-24 2011-03-31 中外製薬株式会社 Antibody capable of recognizing hla class i
WO2011049758A1 (en) 2009-10-09 2011-04-28 Amgen Inc. Human anti-ngf neutralizing antibodies as selective ngf pathway inhibitors
EP3028717A1 (en) 2009-10-09 2016-06-08 Amgen, Inc Human anti-ngf neutralizing antibodies as selective ngf pathway inhibitors
EP3421493A1 (en) 2009-10-16 2019-01-02 Progastrine et Cancers S.à r.l. Monoclonal antibodies to progastrin and their uses
WO2011045080A2 (en) 2009-10-16 2011-04-21 Biorealites S.A.S. Monoclonal antibodies to progastrin and their uses
EP3520816A2 (en) 2009-10-23 2019-08-07 Millennium Pharmaceuticals, Inc. Anti-gcc antibody molecules and related compositions and methods
WO2011057188A1 (en) 2009-11-06 2011-05-12 Idexx Laboratories, Inc. Canine anti-cd20 antibodies
WO2011057250A1 (en) 2009-11-09 2011-05-12 Alexion Pharmaceuticals, Inc. Reagents and methods for detecting pnh type ii white blood cells and their identification as risk factors for thrombotic disorders
WO2011060206A2 (en) 2009-11-13 2011-05-19 U3 Pharma Gmbh Material and methods for treating or preventing her-3 associated diseases
EP3670539A1 (en) 2009-11-13 2020-06-24 Daiichi Sankyo Europe GmbH Material and methods for treating or preventing her-3 associated diseases
EP2719708A2 (en) 2009-11-13 2014-04-16 U3 Pharma GmbH Material and methods for treating or preventing HER-3 associated diseases
EP2896632A2 (en) 2009-11-13 2015-07-22 U3 Pharma GmbH Material and methods for treating or preventing HER-3 associated diseases
EP3351558A2 (en) 2009-11-13 2018-07-25 Daiichi Sankyo Europe GmbH Material and methods for treating or preventing her-3 associated diseases
WO2011062926A2 (en) 2009-11-17 2011-05-26 Medarex, Inc. Methods for enhanced protein production
EP3431608A2 (en) 2009-11-17 2019-01-23 E. R. Squibb & Sons, L.L.C. Method for enhanced protein production
EP3255153A1 (en) 2009-11-17 2017-12-13 E. R. Squibb & Sons, L.L.C. Methods for enhanced protein production
US8765415B2 (en) 2009-11-17 2014-07-01 Medarex, L.L.C. Methods for enhanced protein production
EP3279215A1 (en) 2009-11-24 2018-02-07 MedImmune Limited Targeted binding agents against b7-h1
WO2011067711A2 (en) 2009-12-01 2011-06-09 Compugen Ltd Novel heparanase splice variant
US11819531B2 (en) 2009-12-18 2023-11-21 Kodiak Sciences Inc. Multifunctional zwitterionic polymer conjugates
WO2011085343A1 (en) 2010-01-11 2011-07-14 Alexion Pharmaceuticals, Inc Biomarkers of immunomodulatory effects in humans treated with anti-cd200 antibodies
EP3378486A2 (en) 2010-01-20 2018-09-26 Chugai Seiyaku Kabushiki Kaisha Stabilized antibody-containing liquid formulations
WO2011090088A1 (en) 2010-01-20 2011-07-28 中外製薬株式会社 Solution preparation containing stabilized antibody
EP3892292A2 (en) 2010-01-20 2021-10-13 Chugai Seiyaku Kabushiki Kaisha Stabilized antibody-containing liquid formulations
EP3342786A1 (en) 2010-01-29 2018-07-04 Chugai Seiyaku Kabushiki Kaisha Anti-dll3 antibody
EP3907242A1 (en) 2010-01-29 2021-11-10 Chugai Seiyaku Kabushiki Kaisha Anti-dll3 antibody
WO2011093097A1 (en) 2010-01-29 2011-08-04 株式会社未来創薬研究所 Anti-dll3 antibody
WO2011092989A1 (en) 2010-01-29 2011-08-04 東レ株式会社 Polylactic acid-based resin sheet
WO2011097511A1 (en) 2010-02-05 2011-08-11 The United States Of America, As Represented By The Secretary Department Of Health & Human Services REGULATORY B CELLS (tBREGS) AND THEIR USE
WO2011099524A1 (en) 2010-02-10 2011-08-18 富士フイルムRiファーマ株式会社 Radioactive metal-labeled anti-cadherin antibody
EP4219560A2 (en) 2010-02-18 2023-08-02 The Regents of The University of California Integrin alpha v beta 8 neutralizing antibody
WO2011103490A2 (en) 2010-02-18 2011-08-25 The Regents Of The University Of California INTEGRIN αVβ8 NEUTRALIZING ANTIBODY
WO2011105573A1 (en) 2010-02-26 2011-09-01 株式会社未来創薬研究所 Anti-icam3 antibody and use thereof
WO2011109398A2 (en) 2010-03-02 2011-09-09 President And Fellows Of Harvard College Methods and compositions for treatment of angelman syndrome and autism spectrum disorders
WO2011108714A1 (en) 2010-03-04 2011-09-09 中外製薬株式会社 Antibody constant region variant
WO2011116090A1 (en) 2010-03-17 2011-09-22 Abbott Research B.V. Anti-nerve growth factor (ngf) antibody compositions
US10745467B2 (en) 2010-03-26 2020-08-18 The Trustees Of Dartmouth College VISTA-Ig for treatment of autoimmune, allergic and inflammatory disorders
US10781254B2 (en) 2010-03-26 2020-09-22 The Trustees Of Dartmouth College VISTA regulatory T cell mediator protein, VISTA binding agents and use thereof
WO2011122011A2 (en) 2010-03-30 2011-10-06 Chugai Seiyaku Kabushiki Kaisha Antibodies with modified affinity to fcrn that promote antigen clearance
EP3181581A1 (en) 2010-03-30 2017-06-21 Chugai Seiyaku Kabushiki Kaisha Antibodies with modified affinity to fcrn that promote antigen clearance
EP3702368A1 (en) 2010-03-30 2020-09-02 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecules that promote antigen clearance
US11104744B2 (en) 2010-03-31 2021-08-31 Ablexis, Llc Genetic engineering of non-human animals for the production of chimeric antibodies
US10526420B2 (en) 2010-03-31 2020-01-07 Ablexis, Llc Genetic engineering of non-human animals for the production of chimeric antibodies
US10494445B2 (en) 2010-03-31 2019-12-03 Ablexis, Llc Genetic engineering of non-human animals for the production of chimeric antibodies
US9580491B2 (en) 2010-03-31 2017-02-28 Ablexis, Llc Genetic engineering of non-human animals for the production of chimeric antibodies
US10662255B2 (en) 2010-03-31 2020-05-26 Ablexis, Llc Genetic engineering of non-human animals for the production of chimeric antibodies
US11104743B2 (en) 2010-03-31 2021-08-31 Ablexis, Llc Genetic engineering of non-human animals for the production of chimeric antibodies
US10618977B2 (en) 2010-03-31 2020-04-14 Ablexis, Llc Genetic engineering of non-human animals for the production of chimeric antibodies
US10836832B2 (en) 2010-03-31 2020-11-17 Ablexis, Llc Genetic engineering of non-human animals for the production of chimeric antibodies
US10604587B2 (en) 2010-03-31 2020-03-31 Ablexis, Llc Genetic engineering of non-human animals for the production of chimeric antibodies
US10829564B2 (en) 2010-03-31 2020-11-10 Ablexis, Llc Genetic engineering of non-human animals for the production of chimeric antibodies
US11352444B2 (en) 2010-03-31 2022-06-07 Ablexis, Llc Genetic engineering of non-human animals for the production of chimeric antibodies
US10626188B2 (en) 2010-03-31 2020-04-21 Ablexis, Llc Genetic engineering of non-human animals for the production of chimeric antibodies
US11220555B2 (en) 2010-03-31 2022-01-11 Ablexis, Llc Genetic engineering of non-human animals for the production of chimeric antibodies
US11242409B2 (en) 2010-03-31 2022-02-08 Ablexis, Llc Genetic engineering of non-human animals for the production of chimeric antibodies
EP3165540A1 (en) 2010-04-13 2017-05-10 Celldex Therapeutics, Inc. Antibodies that bind human cd27 and uses thereof
US11407821B2 (en) 2010-04-30 2022-08-09 Alexion Pharmaceuticals, Inc. Anti-C5A antibodies
US9963503B2 (en) 2010-04-30 2018-05-08 Alexion Pharmaceuticals, Inc. Methods of producing anti-C5a antibodies
US9309310B2 (en) 2010-04-30 2016-04-12 Alexion Pharmaceuticals, Inc. Nucleic acids encoding anti-C5a antibodies
US10450370B2 (en) 2010-04-30 2019-10-22 Alexion Pharmaceuticals, Inc. Anti-C5a antibodies
US9469690B2 (en) 2010-04-30 2016-10-18 Alexion Pharmaceuticals, Inc. Methods of treating complement-associated disorders with anti-C5a antibodies
WO2011137395A1 (en) 2010-04-30 2011-11-03 Rother Russell P Anti-c5a antibodies and methods for using the antibodies
US9011852B2 (en) 2010-04-30 2015-04-21 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
US9434784B1 (en) 2010-04-30 2016-09-06 Alexion Pharmaceuticals, Inc. Nucleic acids encodng anti-C5A antibodies
EP2824111A2 (en) 2010-04-30 2015-01-14 Alexion Pharmaceuticals, Inc. Anti-C5A Antibodies and Methods for Using the Antibodies
US9371378B1 (en) 2010-04-30 2016-06-21 Alexion Pharmaceuticals, Inc. Anti-C5a antibodies
WO2011140151A1 (en) 2010-05-04 2011-11-10 Dyax Corp. Antibodies against epidermal growth factor receptor (egfr)
WO2011140254A1 (en) 2010-05-04 2011-11-10 Adimab, Llc Antibodies against epidermal growth factor receptor (egfr) and uses thereof
WO2011138392A1 (en) 2010-05-06 2011-11-10 Novartis Ag Compositions and methods of use for therapeutic low density lipoprotein -related protein 6 (lrp6) antibodies
EP3345926A1 (en) 2010-05-06 2018-07-11 Novartis AG Compositions and methods of use for therapeutic low density lipoprotein-related protein 6 (lrp6) antibodies
EP4234698A2 (en) 2010-05-06 2023-08-30 Novartis AG Compositions and methods of use for therapeutic low density lipoprotein-related protein 6 (lrp6) antibodies
WO2011138391A1 (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
WO2011149046A1 (en) 2010-05-28 2011-12-01 独立行政法人国立がん研究センター Therapeutic agent for pancreatic cancer
EP4115906A1 (en) 2010-05-28 2023-01-11 Chugai Seiyaku Kabushiki Kaisha Antitumor t cell response enhancer
WO2011149051A1 (en) 2010-05-28 2011-12-01 中外製薬株式会社 Antitumor t cell response enhancer
EP3613774A1 (en) 2010-06-09 2020-02-26 Genmab A/S Antibodies against human cd38
US9168314B2 (en) 2010-06-15 2015-10-27 Genmab A/S Human antibody drug conjugates against tissue factor
US9492565B2 (en) 2010-06-15 2016-11-15 Genmab A/S Human antibody drug conjugates against tissue factor
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
EP3327035A1 (en) 2010-06-22 2018-05-30 Precision Biologics Inc. Colon and pancreas cancer specific antigens and antibodies
WO2012003338A1 (en) 2010-07-01 2012-01-05 Takeda Pharmaceutical Company Limited COMBINATION OF A cMET INHIBITOR AND AN ANTIBODY TO HGF AND/OR cMET
WO2012009442A2 (en) 2010-07-14 2012-01-19 Merck Sharp & Dohme Corp. Anti-addl monoclonal antibody and uses thereof
EP3435087A1 (en) 2010-07-16 2019-01-30 Bioatla LLC Novel methods of protein evolution
WO2012018404A2 (en) 2010-08-06 2012-02-09 U3 Pharma Gmbh Use of her3 binding agents in prostate treatment
WO2012019132A2 (en) 2010-08-06 2012-02-09 Cell Signaling Technology, Inc. Anaplastic lymphoma kinase in kidney cancer
US9999620B2 (en) 2010-08-16 2018-06-19 Duke University CaMKK-β as a target for treating cancer
WO2012022814A1 (en) 2010-08-20 2012-02-23 Novartis Ag Antibodies for epidermal growth factor receptor 3 (her3)
WO2012035518A1 (en) 2010-09-17 2012-03-22 Compugen Ltd. Compositions and methods for treatment of drug resistant multiple myeloma
EP4289445A2 (en) 2010-09-17 2023-12-13 Takeda Pharmaceutical Company Limited Stabilization of immunoglobulins through aqueous formulation with histidine at weak acidic to neutral ph
WO2012037534A1 (en) 2010-09-17 2012-03-22 Baxter International Inc. Stabilization of immunoglobulins through aqueous formulation with histidine at weak acidic to neutral ph
WO2012037530A1 (en) 2010-09-17 2012-03-22 Baxter International Inc. Stabilization of immunoglobulins and other proteins through aqueous formulations with sodium chloride at weak acidic to neutral ph
WO2012038505A1 (en) 2010-09-22 2012-03-29 Imba - Institut Für Molekulare Biotechnologie Gmbh Breast cancer diagnostics
WO2012038504A2 (en) 2010-09-22 2012-03-29 Imba - Institut Für Molekulare Biotechnologie Gmbh Breast cancer therapeutics
EP2433644A1 (en) 2010-09-22 2012-03-28 IMBA-Institut für Molekulare Biotechnologie GmbH Breast cancer therapeutics
EP2434285A1 (en) 2010-09-22 2012-03-28 IMBA-Institut für Molekulare Biotechnologie GmbH Breast cancer diagnostics
WO2012040617A2 (en) 2010-09-23 2012-03-29 Neogenix Oncology, Inc. Colon and pancreas cancer peptidomimetics
WO2012045703A1 (en) 2010-10-05 2012-04-12 Novartis Ag Anti-il12rbeta1 antibodies and their use in treating autoimmune and inflammatory disorders
US9662377B2 (en) 2010-10-25 2017-05-30 Regents Of The University Of Minneosta Therapeutic composition for treatment of glioblastoma
US9364505B2 (en) 2010-10-25 2016-06-14 Regents Of The University Of Minnesota Therapeutic composition for treatment of glioblastoma
WO2012061120A1 (en) 2010-10-25 2012-05-10 Regents Of The University Of Minnesota Therapeutic composition for treatment of glioblastoma
EP3708586A1 (en) 2010-10-29 2020-09-16 Perseus Proteomics Inc. Anti-cdh3 antibody having high internalization capacity
EP3404043A1 (en) 2010-10-29 2018-11-21 Perseus Proteomics Inc. Anti-cdh3 antibody having high internalization capacity
WO2012057328A1 (en) 2010-10-29 2012-05-03 株式会社ペルセウスプロテオミクス Anti-cdh3 antibody having high internalizing capability
EP3318633A1 (en) 2010-11-17 2018-05-09 Chugai Seiyaku Kabushiki Kaisha Multi-specific antigen-binding molecule having alternative function to function of blood coagulation factor viii
WO2012067176A1 (en) 2010-11-17 2012-05-24 中外製薬株式会社 Multi-specific antigen-binding molecule having alternative function to function of blood coagulation factor viii
WO2012066293A1 (en) 2010-11-17 2012-05-24 Biotecnol Inc Therapeutic agent
WO2012069466A1 (en) 2010-11-24 2012-05-31 Novartis Ag Multispecific molecules
EP4279512A2 (en) 2010-11-30 2023-11-22 Chugai Seiyaku Kabushiki Kaisha Cytotoxicity-inducing therapeutic agent
EP4279511A2 (en) 2010-11-30 2023-11-22 Chugai Seiyaku Kabushiki Kaisha Cytotoxicity-inducing therapeutic agent
EP4279513A2 (en) 2010-11-30 2023-11-22 Chugai Seiyaku Kabushiki Kaisha Cytotoxicity-inducing therapeutic agent
EP3434767A1 (en) 2010-11-30 2019-01-30 Chugai Seiyaku Kabushiki Kaisha Cytotoxicity-inducing therapeutic agent
EP4231014A2 (en) 2010-11-30 2023-08-23 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule capable of binding to plurality of antigen molecules repeatedly
WO2012073985A1 (en) 2010-11-30 2012-06-07 中外製薬株式会社 Cytotoxicity-inducing therapeutic agent
WO2012073992A1 (en) 2010-11-30 2012-06-07 中外製薬株式会社 Antigen-binding molecule capable of binding to plurality of antigen molecules repeatedly
EP4303236A2 (en) 2010-11-30 2024-01-10 Chugai Seiyaku Kabushiki Kaisha Cytotoxicity-inducing therapeutic agent
EP4303237A2 (en) 2010-11-30 2024-01-10 Chugai Seiyaku Kabushiki Kaisha Cytotoxicity-inducing therapeutic agent
EP3461847A1 (en) 2010-12-06 2019-04-03 Seattle Genetics, Inc. Humanized antibodies to liv-1 and use of same to treat cancer
EP3786185A1 (en) 2010-12-06 2021-03-03 Seagen Inc. Humanized antibodies to liv-1 and use of same to treat cancer
EP3156420A1 (en) 2010-12-06 2017-04-19 Seattle Genetics, Inc. Humanized antibodies to liv-1 and use of same to treat cancer
EP3961214A1 (en) 2010-12-31 2022-03-02 BioAtla, Inc. Comprehensive monoclonal antibody generation
WO2012102679A1 (en) 2011-01-24 2012-08-02 National University Of Singapore Pathogenic mycobacteria-derived mannose-capped lipoarabinomannan antigen binding proteins
WO2012106634A1 (en) 2011-02-03 2012-08-09 Alexion Pharmaceuticals, Inc. Use of an anti-cd200 antibody for prolonging the survival of allografts
EP3604330A1 (en) 2011-02-25 2020-02-05 Chugai Seiyaku Kabushiki Kaisha Fcgammariib-specific fc antibody
WO2012115241A1 (en) 2011-02-25 2012-08-30 中外製薬株式会社 Fcγriib-specific fc antibody
WO2012122484A1 (en) 2011-03-09 2012-09-13 Roberto Polakiewicz Methods and reagents for creating monoclonal antibodies
EP3345922A1 (en) 2011-03-09 2018-07-11 Cell Signaling Technology, Inc. Methods and reagents for creating monoclonal antibodies
EP2500073A1 (en) 2011-03-17 2012-09-19 ChromaCon AG Method for identification and purification of multi-specific polypeptides
WO2012123520A1 (en) 2011-03-17 2012-09-20 Chromacon Ag Method for identification and purification of multi-specific polypeptides
EP3825325A2 (en) 2011-03-30 2021-05-26 Chugai Seiyaku Kabushiki Kaisha Retention of antigen-binding molecules in blood plasma and method for modifying immunogenicity
WO2012133782A1 (en) 2011-03-30 2012-10-04 中外製薬株式会社 Retention of antigen-binding molecules in blood plasma and method for modifying immunogenicity
US10059746B2 (en) 2011-04-04 2018-08-28 University Of Iowa Research Foundation Methods of improving vaccine immunogenicity
WO2012138997A1 (en) 2011-04-07 2012-10-11 Amgen Inc. Novel egfr binding proteins
US10232039B2 (en) 2011-04-12 2019-03-19 Duke University Compositions and methods for the treatment of tissue fibrosis
US9676846B2 (en) 2011-04-12 2017-06-13 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
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
WO2012140627A1 (en) 2011-04-15 2012-10-18 Compugen Ltd. Polypeptides and polynucleotides, and uses thereof for treatment of immune related disorders and cancer
WO2012144208A1 (en) 2011-04-18 2012-10-26 国立大学法人東京大学 Diagnosis and treatment of cancer using anti-itm2a antibody
EP3597218A1 (en) 2011-05-10 2020-01-22 Amgen, Inc Methods of treating or preventing cholesterol related disorders
WO2012154999A1 (en) 2011-05-10 2012-11-15 Amgen Inc. Methods of treating or preventing cholesterol related disorders
US11013800B2 (en) 2011-05-16 2021-05-25 Evive Biotech Ltd. Multi-specific Fab fusion proteins comprising a CD3-binding Fab fragment with N-terminal fusion to binding domains and methods of use
EP3492918A1 (en) 2011-05-23 2019-06-05 Cell Signaling Technology, Inc. Ros kinase in lung cancer
EP3182128A1 (en) 2011-05-23 2017-06-21 Cell Signaling Technology, Inc. Ros kinase in lung cancer
WO2012162373A1 (en) 2011-05-23 2012-11-29 Cell Signaling Technology, Inc. Ros kinase in lung cancer
WO2012160448A2 (en) 2011-05-25 2012-11-29 Innate Pharma, S.A. Anti-kir antibodies for the treatment of inflammatory disorders
WO2012172495A1 (en) 2011-06-14 2012-12-20 Novartis Ag Compositions and methods for antibodies targeting tem8
EP3281640A1 (en) 2011-06-17 2018-02-14 President and Fellows of Harvard College Frizzled 2 as a target for therapeutic antibodies in the treatment of cancer
WO2012174446A1 (en) 2011-06-17 2012-12-20 President And Fellows Of Harvard College Frizzled 2 as a target for therapeutic antibodies in the treatment of cancer
EP4011913A1 (en) 2011-06-30 2022-06-15 Chugai Seiyaku Kabushiki Kaisha Heterodimerized polypeptide
US9428574B2 (en) 2011-06-30 2016-08-30 Compugen Ltd. Polypeptides and uses thereof for treatment of autoimmune disorders and infection
WO2013002362A1 (en) 2011-06-30 2013-01-03 中外製薬株式会社 Heterodimerized polypeptide
WO2013006437A1 (en) 2011-07-01 2013-01-10 Novartis Ag Method for treating metabolic disorders
EP3090759A1 (en) 2011-07-05 2016-11-09 Merrimack Pharmaceuticals, Inc. Antibodies against epidermal growth factor receptor (egfr) and uses thereof
WO2013006547A2 (en) 2011-07-05 2013-01-10 Merrimack Pharmaceuticals, Inc. Antibodies against epidermal growth factor receptor (egfr) and uses thereof
WO2013010955A1 (en) 2011-07-15 2013-01-24 Morphosys Ag Antibodies that are cross-reactive for macrophage migration inhibitory factor (mif) and d-dopachrome tautomerase (d-dt)
WO2013012855A1 (en) 2011-07-18 2013-01-24 Amgen Inc. Apelin antigen-binding proteins and uses thereof
WO2013012022A1 (en) 2011-07-19 2013-01-24 中外製薬株式会社 Stable protein-containing preparation containing argininamide or analogous compound thereof
WO2013017656A1 (en) 2011-08-02 2013-02-07 Medizinische Universität Wien Antagonists of ribonucleases for treating obesity
WO2013017691A1 (en) 2011-08-04 2013-02-07 Medizinische Universität Innsbruck Cahgt1p inhibitors for use in the treatment of candidiasis
WO2013027802A1 (en) 2011-08-23 2013-02-28 中外製薬株式会社 Novel anti-ddr1 antibody having anti-tumor activity
WO2013035824A1 (en) 2011-09-07 2013-03-14 ファーマロジカルズ・リサーチ プライベート リミテッド Cancer stem cell isolation
US11051497B2 (en) 2011-09-19 2021-07-06 Kymab Limited Manipulation of immunoglobulin gene diversity and multi-antibody therapeutics
US11408095B2 (en) 2011-09-26 2022-08-09 Merus N.V. Generation of binding molecules
US9963716B2 (en) 2011-09-26 2018-05-08 Kymab Limited Chimaeric surrogate light chains (SLC) comprising human VpreB
EP3680251A1 (en) 2011-09-30 2020-07-15 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecules for promoting elimination of antigens
EP3549956A2 (en) 2011-09-30 2019-10-09 Chugai Seiyaku Kabushiki Kaisha Therapeutic antigen-binding molecule with a fcrn-binding domain that promotes antigen clearance
WO2013047748A1 (en) 2011-09-30 2013-04-04 中外製薬株式会社 Antigen-binding molecule promoting disappearance of antigens having plurality of biological activities
WO2013046704A2 (en) 2011-09-30 2013-04-04 Chugai Seiyaku Kabushiki Kaisha THERAPEUTIC ANTIGEN-BINDING MOLECULE WITH A FcRn-BINDING DOMAIN THAT PROMOTES ANTIGEN CLEARANCE
WO2013047729A1 (en) 2011-09-30 2013-04-04 中外製薬株式会社 Antigen-binding molecule inducing immune response to target antigen
EP3939996A1 (en) 2011-09-30 2022-01-19 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule promoting disappearance of antigens having plurality of biological activities
WO2013046722A1 (en) 2011-09-30 2013-04-04 中外製薬株式会社 Ion concentration-dependent binding molecule library
WO2013047752A1 (en) 2011-09-30 2013-04-04 中外製薬株式会社 Antigen-binding molecule for promoting loss of antigens
EP4324850A2 (en) 2011-09-30 2024-02-21 Chugai Seiyaku Kabushiki Kaisha Therapeutic antigen-binding molecule with a fcrn-binding domain that promotes antigen clearance
EP3617313A1 (en) 2011-10-05 2020-03-04 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule for promoting clearance from plasma of antigen comprising saccharide chain receptor-binding domain
WO2013051294A1 (en) 2011-10-05 2013-04-11 中外製薬株式会社 Antigen-binding molecule for promoting clearance from plasma of antigen comprising saccharide chain receptor-binding domain
WO2013056233A1 (en) 2011-10-13 2013-04-18 Aerpio Therapeutics, Inc. Treatment of ocular disease
EP3505182A1 (en) 2011-10-13 2019-07-03 Aerpio Therapeutics, Inc. Methods for treating vascular leak syndrome and cancer
US10150811B2 (en) 2011-10-13 2018-12-11 Aerpio Therapeutics, Inc. Methods for treating vascular leak syndrome and cancer
EP3501536A1 (en) 2011-10-13 2019-06-26 Aerpio Therapeutics, Inc. Treatment of ocular disease
US10329357B2 (en) 2011-10-13 2019-06-25 Aerpio Therapeutics, Inc. Treatment of ocular disease
US10815300B2 (en) 2011-10-13 2020-10-27 Aerpio Pharmaceuticals, Inc. Methods for treating vascular leak syndrome and cancer
EP3653222A1 (en) 2011-10-14 2020-05-20 Novartis AG Antibodies and methods for wnt pathway-related diseases
WO2013054307A2 (en) 2011-10-14 2013-04-18 Novartis Ag Antibodies and methods for wnt pathway-related diseases
WO2013062083A1 (en) 2011-10-28 2013-05-02 ファーマロジカルズ・リサーチ プライベート リミテッド Cancer stem cell-specific molecule
EP3603671A2 (en) 2011-10-28 2020-02-05 Chugai Seiyaku Kabushiki Kaisha Cancer stem cell-specific molecule
WO2013063496A1 (en) 2011-10-28 2013-05-02 Millennium Pharmaceuticals, Inc. Biomarkers of response to nae inhibitors
WO2013065708A1 (en) 2011-10-31 2013-05-10 中外製薬株式会社 Antigen-binding molecule having regulated conjugation between heavy-chain and light-chain
US9220774B2 (en) 2011-11-01 2015-12-29 Bionomics Inc. Methods of treating cancer by administering anti-GPR49 antibodies
US10598653B2 (en) 2011-11-01 2020-03-24 Bionomics Inc. Methods of blocking cancer stem cell growth
US10196442B2 (en) 2011-11-01 2019-02-05 Bionomics Inc. Methods of inhibiting growth of a colon cancer tumor in a subject by administering monoclonal antibodies to G protein-coupled receptor 49 (GPR49)
WO2013067060A1 (en) 2011-11-01 2013-05-10 Bionomics, Inc. Anti-gpr49 antibodies
US9221906B2 (en) 2011-11-01 2015-12-29 Bionomics Inc. Methods of inhibiting solid tumor growth by administering GPR49 antibodies
WO2013067055A1 (en) 2011-11-01 2013-05-10 Bionomics, Inc. Methods of blocking cancer stem cell growth
WO2013067054A1 (en) 2011-11-01 2013-05-10 Bionomics, Inc. Antibodies and methods of treating cancer
WO2013067057A1 (en) 2011-11-01 2013-05-10 Bionomics, Inc. Anti-gpr49 antibodies
US9221907B2 (en) 2011-11-01 2015-12-29 Bionomics Inc. Anti-GPR49 monoclonal antibodies
WO2013071163A2 (en) 2011-11-11 2013-05-16 Millennium Pharamaceuticals, Inc. Biomarkers of response to proteasome inhibitors
WO2013071142A1 (en) 2011-11-11 2013-05-16 Millennium Pharmaceuticals, Inc. Biomarkers of response to proteasome inhibitors
WO2013072512A1 (en) 2011-11-16 2013-05-23 Adrenomed Ag Anti-adrenomedullin (adm) antibody or anti-adm antibody fragment or an anti-adm non-ig scaffold for use in therapy
US11673949B2 (en) 2011-11-16 2023-06-13 Adrenomed Ag Method of modulating the adrenomedullin (ADM) activity of a patient by administering to the patient an anti-ADM antibody or fragment thereof that specifically binds to mature human ADM
WO2013072509A1 (en) 2011-11-16 2013-05-23 Adrenomed Ag Adrenomedullin assays and methods for determining mature adrenomedullin
US9304127B2 (en) 2011-11-16 2016-04-05 Adrenomed Ag Anti-adrenomedullin (ADM) antibody or anti-ADM antibody fragment for use in therapy
WO2013072513A1 (en) 2011-11-16 2013-05-23 Adrenomed Ag Anti-adrenomedullin (adm) antibody or anti-adm antibody fragment or anti-adm non-ig scaffold for use in therapy of an acute disease or acute condition of a patient for stabilizing the circulation
WO2013075048A1 (en) 2011-11-16 2013-05-23 Amgen Inc. Methods of treating epidermal growth factor deletion mutant viii related disorders
EP3553084A1 (en) 2011-11-16 2019-10-16 AdrenoMed AG Anti-adrenomedullin (adm) antibody or anti-adm antibody fragment or anti-adm non-ig scaffold for prevention or reduction of organ dysfunction or organ failure in a patient having a chronic or acute disease or acute condition
US9140696B2 (en) 2011-11-16 2015-09-22 Adrenomed Ag Anti-adrenomedullin (ADM) antibody or anti-ADM antibody fragment or anti-ADM non-IG scaffold for reducing the risk of mortality in a patient having a chronic or acute disease or acute condition
US9402900B2 (en) 2011-11-16 2016-08-02 Adrenomed Ag Methods of modulating adrenomedullin by administering an anti-adrenomedullin (ADM) antibody
US10221238B2 (en) 2011-11-16 2019-03-05 Adrenomed Ag Method of modulating the activity of adrenomedullin in a subject in need of therapeutic intervention for organ dysfunction or organ failure associated with adranomedullin (ADM) activity by administering an anti-adrenomedullin (ADM) antibody or an anti-ADM antibody fragment to the subject
EP2594587A1 (en) 2011-11-16 2013-05-22 AdrenoMed AG Anti-Adrenomedullin (ADM) antibody or anti-ADM antibody fragment or anti-ADM non-Ig scaffold for reducing the risk of mortality in a patient having a chronic or acute disease or acute condition
US10227405B2 (en) 2011-11-16 2019-03-12 Adrenomed Ag Methods of modulating the activity of adrenomedullin in a subject in need of regulation of fluid balance by administering an anti-adrenomedullin (ADM) antibody or an anti-ADM antibody fragment
WO2013072510A1 (en) 2011-11-16 2013-05-23 Adrenomed Ag Anti-adrenomedullin (adm) antibody or anti-adm antibody fragment or anti-adm non-ig scaffold for reducing the risk of mortality in a patient having a chronic or acute disease or acute condition
WO2013072514A1 (en) 2011-11-16 2013-05-23 Adrenomed Ag Anti-adrenomedullin (adm) antibody or anti-adm antibody fragment or anti-adm non-ig scaffold for regulating the fluid balance in a patient having a chronic or acute disease
EP4086283A1 (en) 2011-11-16 2022-11-09 AdrenoMed AG Anti-adrenomedullin (adm) antibody or anti-adm antibody fragment or anti-adm non-ig scaffold for prevention or reduction of organ dysfunction or organ failure in a patient having a chronic or acute disease or acute condition
US10800842B2 (en) 2011-11-16 2020-10-13 Adrenomed Ag Anti-adrenomedullin (ADM) monoclonal antibodies and anti-ADM monoclonal antibody fragments that bind to adrenomedullin
EP2594588A1 (en) 2011-11-16 2013-05-22 AdrenoMed AG Anti-Adrenomedullin (ADM) antibody or anti-ADM antibody fragment or an anti-ADM non-Ig scaffold for use in therapy
WO2013072511A1 (en) 2011-11-16 2013-05-23 Adrenomed Ag Anti-adrenomedullin (adm) antibody or anti-adm antibody fragment or anti-adm non-ig scaffold for prevention or reduction of organ dysfunction or organ failure in a patient having a chronic or acute disease or acute condition
WO2013081143A1 (en) 2011-11-30 2013-06-06 中外製薬株式会社 Drug containing carrier into cell for forming immune complex
EP3517550A1 (en) 2011-11-30 2019-07-31 Chugai Seiyaku Kabushiki Kaisha Drug containing carrier into cell for forming immune complex
WO2013084147A2 (en) 2011-12-05 2013-06-13 Novartis Ag Antibodies for epidermal growth factor receptor 3 (her3)
EP3590538A1 (en) 2011-12-05 2020-01-08 Novartis AG Antibodies for epidermal growth factor receptor 3 (her3)
WO2013084148A2 (en) 2011-12-05 2013-06-13 Novartis Ag Antibodies for epidermal growth factor receptor 3 (her3) directed to domain ii of her3
EP3330288A1 (en) 2011-12-21 2018-06-06 Novartis AG Compositions and methods for antibodies targeting factor p
WO2013093762A1 (en) 2011-12-21 2013-06-27 Novartis Ag Compositions and methods for antibodies targeting factor p
WO2013100120A1 (en) 2011-12-28 2013-07-04 中外製薬株式会社 Humanized anti-epiregulin antibody, and cancer therapeutic agent comprising said antibody as active ingredient
US9617336B2 (en) 2012-02-01 2017-04-11 Compugen Ltd C10RF32 antibodies, and uses thereof for treatment of cancer
EP3578569A1 (en) 2012-02-06 2019-12-11 Inhibrx, Inc. Cd47 antibodies and methods of use thereof
US9045541B2 (en) 2012-02-06 2015-06-02 Inhibrx Llc CD47 antibodies and methods of use thereof
US9663575B2 (en) 2012-02-06 2017-05-30 Inhibrx, Lp CD47 antibodies and methods of use thereof
WO2013118858A1 (en) 2012-02-09 2013-08-15 中外製薬株式会社 Modified fc region of antibody
WO2013125667A1 (en) 2012-02-24 2013-08-29 中外製薬株式会社 ANTIGEN-BINDING MOLECULE FOR PROMOTING DISAPPEARANCE OF ANTIGEN VIA FcγRIIB
EP3738980A1 (en) 2012-02-24 2020-11-18 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule for promoting disappearance of antigen via fc gamma riib
US11297811B2 (en) 2012-03-28 2022-04-12 Kymab Limited Transgenic non-human vertebrate for the expression of class-switched, fully human, antibodies
US10774155B2 (en) 2012-03-28 2020-09-15 Kymab Limited Animal models and therapeutic molecules
US9924705B2 (en) 2012-03-28 2018-03-27 Kymab Limited Animal models and therapeutic molecules
US9938358B2 (en) 2012-03-28 2018-04-10 Kymab Limited Animal models and therapeutic molecules
US10251377B2 (en) 2012-03-28 2019-04-09 Kymab Limited Transgenic non-human vertebrate for the expression of class-switched, fully human, antibodies
US9896516B2 (en) 2012-03-28 2018-02-20 Kymab Limited Animal models and therapeutic molecules
US9938357B2 (en) 2012-03-28 2018-04-10 Kymab Limited Animal models and therapeutic molecules
WO2013147153A1 (en) 2012-03-29 2013-10-03 株式会社未来創薬研究所 Anti-lamp5 antibody and utilization thereof
WO2013150623A1 (en) 2012-04-04 2013-10-10 株式会社ペルセウスプロテオミクス Conjugate of anti-cdh3 (p-cadherin) antibody and drug
EP3336181A1 (en) 2012-04-18 2018-06-20 Cell Signaling Technology, Inc. Egfr and ros1 in cancer
WO2013158859A1 (en) 2012-04-18 2013-10-24 Cell Signaling Technology, Inc. Egfr and ros1 in cancer
US11926859B2 (en) 2012-04-20 2024-03-12 Merus N.V. Methods and means for the production of Ig-like molecules
US10337045B2 (en) 2012-04-20 2019-07-02 Merus N.V. Methods and means for the production of Ig-like molecules
US10752929B2 (en) 2012-04-20 2020-08-25 Merus N.V. Methods and means for the production of ig-like molecules
US10329596B2 (en) 2012-04-20 2019-06-25 Merus N.V. Methods and means for the production of Ig-like molecules
US9758805B2 (en) 2012-04-20 2017-09-12 Merus N.V. Methods and means for the production of Ig-like molecules
EP3656399A1 (en) 2012-05-03 2020-05-27 Amgen, Inc Stable formulations containing anti-pcsk9 antibodies
WO2013166448A1 (en) 2012-05-03 2013-11-07 Amgen Inc. Stable formulations containing anti-pcsk9 antibodies
EP3553089A1 (en) 2012-05-10 2019-10-16 Bioatla, LLC Multi-specific monoclonal antibodies
US10667501B2 (en) 2012-05-17 2020-06-02 Kymab Limited Transgenic non-human vertebrate for the in vivo production of dual specificity immunoglobulins or hypermutated heavy chain only immunoglobulins
WO2013173496A2 (en) 2012-05-18 2013-11-21 Seattle Genetics, Inc. Cd33 antibodies and use of same to treat cancer
EP3421048A1 (en) 2012-05-18 2019-01-02 Seattle Genetics, Inc. Cd33 antibodies and use of same to treat cancer
WO2013180200A1 (en) 2012-05-30 2013-12-05 中外製薬株式会社 Target-tissue-specific antigen-binding molecule
EP3892638A1 (en) 2012-05-30 2021-10-13 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule for eliminating aggregated antigens
EP3795215A1 (en) 2012-05-30 2021-03-24 Chugai Seiyaku Kabushiki Kaisha Target tissue-specific antigen-binding molecule
WO2013180201A1 (en) 2012-05-30 2013-12-05 中外製薬株式会社 Antigen-binding molecule for eliminating aggregated antigens
WO2013188448A2 (en) 2012-06-11 2013-12-19 Amgen Inc. Dual receptor antagonistic antigen-binding proteins and uses thereof
EP3540070A1 (en) 2012-06-11 2019-09-18 Amgen Inc. Dual receptor antagonistic antigen-binding proteins and uses thereof
EP3498857A1 (en) 2012-06-11 2019-06-19 Amgen, Inc. Dual receptor antagonistic antigen-binding proteins and uses thereof
EP4310191A2 (en) 2012-06-14 2024-01-24 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule containing modified fc region
WO2013187495A1 (en) 2012-06-14 2013-12-19 中外製薬株式会社 ANTIGEN-BINDING MOLECULE CONTAINING MODIFIED Fc REGION
US11180557B2 (en) 2012-06-22 2021-11-23 King's College London Vista modulators for diagnosis and treatment of cancer
US11752189B2 (en) 2012-06-22 2023-09-12 The Trustees Of Dartmouth College Vista antagonist and methods of use
US10933115B2 (en) 2012-06-22 2021-03-02 The Trustees Of Dartmouth College VISTA antagonist and methods of use
EP3421486A1 (en) 2012-06-22 2019-01-02 The Trustees Of Dartmouth College Novel vista-ig constructs and the use of vista-ig for treatment of autoimmune, allergic and inflammatory disorders
EP3275899A1 (en) 2012-07-02 2018-01-31 Bristol-Myers Squibb Company Optimization of antibodies that bind lymphocyte activation gene-3 (lag-3), and uses thereof
WO2014008218A1 (en) 2012-07-02 2014-01-09 Bristol-Myers Squibb Company Optimization of antibodies that bind lymphocyte activation gene-3 (lag-3), and uses thereof
EP3795592A1 (en) 2012-07-02 2021-03-24 Bristol-Myers Squibb Company Optimization of antibodies that bind lymphocyte activation gene-3 (lag-3), and uses thereof
US9701675B2 (en) 2012-07-06 2017-07-11 Duke University Activation of TRPV4 ion channel by physical stimuli and critical role for TRPV4 in organ-specific inflammation and itch
US9290489B2 (en) 2012-07-06 2016-03-22 Duke University Activation of TRPV4 ion channel by physical stimuli and critical role for TRPV4 in organ-specific inflammation and itch
WO2014007402A1 (en) 2012-07-06 2014-01-09 京都府公立大学法人 Differentiation marker for and differentiation control for ocular cells
EP4019041A1 (en) 2012-07-13 2022-06-29 The Trustees of the University of Pennsylvania Toxicity management for anti-tumor activity of cars
EP3338794A1 (en) 2012-07-13 2018-06-27 The Trustees of the University of Pennsylvania Toxicity management for anti-tumor activity of cars
US11273219B2 (en) 2012-07-13 2022-03-15 The Trustees Of The University Of Pennsylvania Toxicity management for anti-tumor activity of CARs
US10603378B2 (en) 2012-07-13 2020-03-31 The Trustees Of The University Of Pennsylvania Toxicity management for anti-tumor activity of CARs
EP3721901A1 (en) 2012-07-13 2020-10-14 The Trustees of the University of Pennsylvania Toxicity management for anti-tumor activity of cars
WO2014030750A1 (en) 2012-08-24 2014-02-27 中外製薬株式会社 MOUSE FcγRII-SPECIFIC Fc ANTIBODY
EP3597747A1 (en) 2012-08-24 2020-01-22 Chugai Seiyaku Kabushiki Kaisha Mouse fcgammarii-specific fc antibody
WO2014030728A1 (en) 2012-08-24 2014-02-27 中外製薬株式会社 Fcγriib-specific fc region variant
EP3721900A1 (en) 2012-08-24 2020-10-14 Chugai Seiyaku Kabushiki Kaisha Fcgammariib-specific fc region variant
WO2014039983A1 (en) 2012-09-07 2014-03-13 The Trustees Of Dartmouth College Vista modulators for diagnosis and treatment of cancer
WO2014037899A2 (en) 2012-09-07 2014-03-13 Novartis Ag Il-18 binding molecules
US11529416B2 (en) 2012-09-07 2022-12-20 Kings College London Vista modulators for diagnosis and treatment of cancer
EP3725805A1 (en) 2012-09-07 2020-10-21 Novartis AG Il-18 binding molecules
WO2014051022A1 (en) 2012-09-27 2014-04-03 中外製薬株式会社 Fgfr3 fusion gene and pharmaceutical drug targeting same
WO2014050926A1 (en) 2012-09-28 2014-04-03 中外製薬株式会社 Method for evaluating blood coagulation reaction
WO2014055543A2 (en) 2012-10-01 2014-04-10 Millennium Pharmaceuticals, Inc. Biomarkers and methods to predict response to inhibitors and uses thereof
EP3498296A1 (en) 2012-10-08 2019-06-19 Prothena Biosciences Limited Antibodies recognizing alpha-synuclein
US9593165B2 (en) 2012-11-08 2017-03-14 University Of Miyazaki Antibody capable of specifically recognizing transferrin receptor
WO2014073641A1 (en) 2012-11-08 2014-05-15 国立大学法人 宮崎大学 Antibody capable of specifically recognizing transferrin receptor
US11459386B2 (en) 2012-11-08 2022-10-04 Sesen Bio, Inc. IL-6 antagonists and uses thereof
US9951130B2 (en) 2012-11-08 2018-04-24 Eleven Biotherapeutics, Inc. IL-6 antagonists and uses thereof
WO2014074905A1 (en) 2012-11-08 2014-05-15 Eleven Biotherapeutics, Inc. Il-6 antagonists and uses thereof
WO2014084859A1 (en) 2012-11-30 2014-06-05 Novartis Ag Molecules and methods for modulating tmem16a activities
WO2014089111A1 (en) 2012-12-05 2014-06-12 Novartis Ag Compositions and methods for antibodies targeting epo
EP3851454A1 (en) 2012-12-05 2021-07-21 Novartis AG Compositions and methods for antibodies targeting epo
WO2014099997A1 (en) 2012-12-18 2014-06-26 Novartis Ag Compositions and methods that utilize a peptide tag that binds to hyaluronan
WO2014104165A1 (en) 2012-12-27 2014-07-03 中外製薬株式会社 Heterodimerized polypeptide
EP3336104A1 (en) 2012-12-28 2018-06-20 Precision Biologics, Inc. Humanized monoclonal antibodies and methods of use for the diagnosis and treatment of colon and pancreas cancer
WO2014114800A1 (en) 2013-01-25 2014-07-31 Amgen Research (Munich) Gmbh Antibody constructs for cdh19 and cd3
EP3699194A1 (en) 2013-01-25 2020-08-26 Amgen Research (Munich) GmbH Antibody constructs for cdh19 and cd3
US11498964B2 (en) 2013-01-25 2022-11-15 Amgen Research (Munich) Gmbh Antibody constructs for CDH19 and CD3
WO2014114801A1 (en) 2013-01-25 2014-07-31 Amgen Inc. Antibodies targeting cdh19 for melanoma
EP4137518A1 (en) 2013-02-06 2023-02-22 Inhibrx, Inc. Non-platelet depleting and non-red blood cell depleting cd47 antibodies and methods of use thereof
WO2014123580A1 (en) 2013-02-06 2014-08-14 Inhibrx Llc Non-platelet depleting and non-red blood cell depleting cd47 antibodies and methods of use thereof
WO2015198217A2 (en) 2013-02-08 2015-12-30 Novartis Ag Compositions and methods for long-acting antibodies targeting il-17
EP3656786A1 (en) 2013-02-08 2020-05-27 Novartis AG Anti-il-17a antibodies and their use in treating autoimmune and inflammatory disorders
WO2014122613A1 (en) 2013-02-08 2014-08-14 Novartis Ag Anti-il-17a antibodies and their use in treating autoimmune and inflammatory disorders
US10174110B2 (en) 2013-02-13 2019-01-08 Laboratoire Français Du Fractionnement Et Des Biotechnologies Highly galactosylated anti-TNF-α antibodies and uses thereof
US10034921B2 (en) 2013-02-13 2018-07-31 Laboratoire Français Du Fractionnement Et Des Biotechnologies Proteins with modified glycosylation and methods of production thereof
US10421824B2 (en) 2013-03-13 2019-09-24 Amgen Inc. Proteins specific for BAFF and B7RP1
WO2014165271A2 (en) 2013-03-13 2014-10-09 Neotope Biosciences Limited Tau immunotherapy
US11492417B2 (en) 2013-03-13 2022-11-08 Amgen Inc. Proteins specific for BAFF and B7RP1 and uses thereof
US10421823B2 (en) 2013-03-13 2019-09-24 Amgen Inc. Proteins specific for BAFF and B7RP1 and uses thereof
EP3689904A1 (en) 2013-03-13 2020-08-05 Prothena Biosciences Limited Tau immunotherapy
US11643457B2 (en) 2013-03-13 2023-05-09 Prothena Biosciences Limited Tau immunotherapy
WO2014141189A1 (en) 2013-03-14 2014-09-18 Erasmus University Medical Center Transgenic non-human mammal for antibody production
EP3841876A1 (en) 2013-03-14 2021-06-30 Erasmus University Medical Center Rotterdam Transgenic mouse for antibody production
EP3611189A1 (en) 2013-03-14 2020-02-19 Novartis AG Antibodies against notch 3
WO2014159239A2 (en) 2013-03-14 2014-10-02 Novartis Ag Antibodies against notch 3
US9980470B2 (en) 2013-03-14 2018-05-29 Erasmus University Medical Center Antibody production
US10392614B2 (en) 2013-03-15 2019-08-27 Abvitro Llc Methods of single-cell barcoding and sequencing
US11118176B2 (en) 2013-03-15 2021-09-14 Abvitro Llc Single cell bar-coding for antibody discovery
WO2014140368A1 (en) 2013-03-15 2014-09-18 Amgen Research (Munich) Gmbh Antibody constructs for influenza m2 and cd3
WO2014140358A1 (en) 2013-03-15 2014-09-18 Amgen Research (Munich) Gmbh Single chain binding molecules comprising n-terminal abp
US9816088B2 (en) 2013-03-15 2017-11-14 Abvitro Llc Single cell bar-coding for antibody discovery
US10876107B2 (en) 2013-03-15 2020-12-29 Abvitro Llc Single cell bar-coding for antibody discovery
US10119134B2 (en) 2013-03-15 2018-11-06 Abvitro Llc Single cell bar-coding for antibody discovery
EP3653642A1 (en) 2013-03-15 2020-05-20 Amgen Research (Munich) GmbH Single chain binding molecules comprising n-terminal abp
US11297810B2 (en) 2013-03-18 2022-04-12 Kymab Limited Animal models and therapeutic molecules
US10226033B2 (en) 2013-03-18 2019-03-12 Kymab Limited Animal models and therapeutic molecules
US9788534B2 (en) 2013-03-18 2017-10-17 Kymab Limited Animal models and therapeutic molecules
WO2014147153A1 (en) 2013-03-20 2014-09-25 Sphingotec Gmbh Adrenomedullin to guide therapy of blood pressure decline
WO2014163101A1 (en) 2013-04-02 2014-10-09 中外製薬株式会社 Fc region variant
EP3783017A1 (en) 2013-04-02 2021-02-24 Chugai Seiyaku Kabushiki Kaisha Fc region variant
US9783618B2 (en) 2013-05-01 2017-10-10 Kymab Limited Manipulation of immunoglobulin gene diversity and multi-antibody therapeutics
US11820810B2 (en) 2013-05-02 2023-11-21 Kymab Limited Antibodies, variable domains and chains tailored for human use
US10730930B2 (en) 2013-05-02 2020-08-04 Kymab Limited Antibodies, variable domains and chains tailored for human use
US9783593B2 (en) 2013-05-02 2017-10-10 Kymab Limited Antibodies, variable domains and chains tailored for human use
US11707056B2 (en) 2013-05-02 2023-07-25 Kymab Limited Animals, repertoires and methods
WO2014185550A1 (en) 2013-05-16 2014-11-20 Kyoto University Method for determining prognosis of cancer
US10053510B2 (en) 2013-05-24 2018-08-21 Promis Neurosciences Inc. FasR antibodies and methods of use
WO2014200018A1 (en) 2013-06-11 2014-12-18 独立行政法人 国立精神・神経医療研究センター Method for predicting post-therapy prognosis of relapsing-remitting multiple sclerosis (rrms) patient, and method for determining applicability of novel therapy
WO2014205300A2 (en) 2013-06-21 2014-12-24 Novartis Ag Lectin-like oxidized ldl receptor1 antibodies and methods of use
WO2014208482A1 (en) 2013-06-24 2014-12-31 中外製薬株式会社 Therapeutic agent comprising humanized anti-epiregulin antibody as active ingredient for non-small-cell lung carcinoma excluding adenocarcinoma
WO2014209384A1 (en) 2013-06-28 2014-12-31 Amgen Inc. Methods for treating homozygous familial hypercholesterolema
US9803017B2 (en) 2013-07-05 2017-10-31 University Of Washington Through Its Center For Commercialization Soluble MIC neutralizing monoclonal antibody for treating cancer
EP3699200A1 (en) 2013-07-15 2020-08-26 Cell Signaling Technology, Inc. Anti-mucin 1 binding agents and uses thereof
WO2015009740A2 (en) 2013-07-15 2015-01-22 Cell Signaling Technology, Inc. Anti-mucin 1 binding agents and uses thereof
US10689442B2 (en) 2013-08-14 2020-06-23 Sachdev Sidhu Antibodies against Frizzled receptor
US10077304B2 (en) 2013-08-14 2018-09-18 The Governing Council Of The University Of Toronto Antibodies against frizzled receptor
EP3705494A2 (en) 2013-08-14 2020-09-09 Sachdev Sidhu Antibodies against frizzled proteins and methods of use thereof
WO2015022658A2 (en) 2013-08-14 2015-02-19 Novartis Ag Methods of treating sporadic inclusion body myositis
US10702608B2 (en) 2013-09-08 2020-07-07 Kodiak Sciences Inc. Factor VIII zwitterionic polymer conjugates
WO2015041310A1 (en) 2013-09-20 2015-03-26 中外製薬株式会社 Treatment for hemorrhagic diseases by anti-protein-c antibody
WO2015046467A1 (en) 2013-09-27 2015-04-02 中外製薬株式会社 Method for producing polypeptide heteromultimer
US11399522B2 (en) 2013-10-01 2022-08-02 Kymab Limited Animal models and therapeutic molecules
US10149462B2 (en) 2013-10-01 2018-12-11 Kymab Limited Animal models and therapeutic molecules
WO2015068847A1 (en) 2013-11-11 2015-05-14 中外製薬株式会社 Antigen-binding molecule containing modified antibody variable region
EP3763813A1 (en) 2013-12-04 2021-01-13 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecules, the antigen-binding activity of which varies according to the concentration of compounds, and libraries of said molecules
WO2015083764A1 (en) 2013-12-04 2015-06-11 中外製薬株式会社 Antigen-binding molecules, the antigen-binding activity of which varies according to the concentration of compounds, and libraries of said molecules
US10618971B2 (en) 2013-12-17 2020-04-14 Kymab Limited Targeting rare human PCSK9 variants for cholesterol treatment
US8883157B1 (en) 2013-12-17 2014-11-11 Kymab Limited Targeting rare human PCSK9 variants for cholesterol treatment
US10611849B2 (en) 2013-12-17 2020-04-07 Kymab Limited Precision medicine by targeting rare human PCSK9 variants for cholesterol treatment
US9040052B1 (en) 2013-12-17 2015-05-26 Kymab Limited Precision Medicine by targeting rare human PCSK9 variants for cholesterol treatment
US11434305B2 (en) 2013-12-17 2022-09-06 Kymab Limited Precision medicine by targeting rare human PCSK9 variants for cholesterol treatment
US11242392B2 (en) 2013-12-24 2022-02-08 Janssen Pharmaceutica Nv Anti-vista antibodies and fragments
US11014987B2 (en) 2013-12-24 2021-05-25 Janssen Pharmaceutics Nv Anti-vista antibodies and fragments, uses thereof, and methods of identifying same
WO2015099165A1 (en) 2013-12-27 2015-07-02 中外製薬株式会社 Method for purifying antibody having low isoelectric point
WO2015099127A1 (en) 2013-12-27 2015-07-02 中外製薬株式会社 Fgfr gatekeeper mutant gene and drug targeting same
EP3581179A1 (en) 2013-12-27 2019-12-18 Chugai Seiyaku Kabushiki Kaisha Fgfr gatekeeper mutant gene and drug targeting same
WO2015105888A1 (en) 2014-01-07 2015-07-16 Bioatla, Llc Proteins targeting orthologs
US11293929B2 (en) 2014-01-07 2022-04-05 Bioatla, Inc. Proteins targeting orthologs
EP3447493A1 (en) 2014-01-07 2019-02-27 Bioatla, LLC Proteins targeting orthologs
WO2015121383A1 (en) 2014-02-12 2015-08-20 Michael Uhlin Bispecific antibodies for use in stem cell transplantation
US11753479B2 (en) 2014-03-04 2023-09-12 Kymab Limited Nucleic acids encoding anti-OX40L antibodies
US11773175B2 (en) 2014-03-04 2023-10-03 Kymab Limited Antibodies, uses and methods
WO2015136469A1 (en) 2014-03-12 2015-09-17 Prothena Biosciences Limited Anti-mcam antibodies and associated methods of use
WO2015136472A1 (en) 2014-03-12 2015-09-17 Prothena Biosciences Limited Anti-laminin4 antibodies specific for lg4-5
WO2015136470A1 (en) 2014-03-12 2015-09-17 Prothena Biosciences Limited Anti-mcam antibodies and associated methods of use
WO2015136471A1 (en) 2014-03-12 2015-09-17 Prothena Biosciences Limited Anti-laminin4 antibodies specific for lg1-3
US9546214B2 (en) 2014-04-04 2017-01-17 Bionomics, Inc. Humanized antibodies that bind LGR5
US10358500B2 (en) 2014-04-04 2019-07-23 Bionomics Inc. Humanized antibodies that bind LGR5
EP3461495A1 (en) 2014-04-24 2019-04-03 Novartis AG Methods of improving or accelerating physical recovery after surgery for hip fracture
WO2015162590A1 (en) 2014-04-24 2015-10-29 Novartis Ag Methods of improving or accelerating physical recovery after surgery for hip fracture
EP3610924A1 (en) 2014-06-06 2020-02-19 Bristol-Myers Squibb Company Antibodies against glucocorticoid-induced tumor necrosis factor receptor (gitr) and uses thereof
WO2015187835A2 (en) 2014-06-06 2015-12-10 Bristol-Myers Squibb Company Antibodies against glucocorticoid-induced tumor necrosis factor receptor (gitr) and uses thereof
EP3998079A1 (en) 2014-06-06 2022-05-18 Bristol-Myers Squibb Company Antibodies against glucocorticoid-induced tumor necrosis factor receptor (gitr) and uses thereof
US11123426B2 (en) 2014-06-11 2021-09-21 The Trustees Of Dartmouth College Use of vista agonists and antagonists to suppress or enhance humoral immunity
WO2015195917A1 (en) 2014-06-18 2015-12-23 Mersana Therapeutics, Inc. Monoclonal antibodies against her2 epitope and methods of use thereof
EP4285917A2 (en) 2014-06-18 2023-12-06 Mersana Therapeutics, Inc. Monoclonal antibodies against her2 epitope and methods of use thereof
EP4218816A2 (en) 2014-06-20 2023-08-02 Chugai Seiyaku Kabushiki Kaisha Pharmaceutical composition for use in prevention and/or treatment of disease that develops or progresses as a result of decrease or loss of activity of blood coagulation factor viii and/or activated blood coagulation factor viii
WO2015198240A2 (en) 2014-06-25 2015-12-30 Novartis Ag Compositions and methods for long acting proteins
WO2015198243A2 (en) 2014-06-25 2015-12-30 Novartis Ag Compositions and methods for long acting proteins
US11155610B2 (en) 2014-06-28 2021-10-26 Kodiak Sciences Inc. Dual PDGF/VEGF antagonists
US9840553B2 (en) 2014-06-28 2017-12-12 Kodiak Sciences Inc. Dual PDGF/VEGF antagonists
US10132818B2 (en) 2014-07-08 2018-11-20 New York University Tau imaging ligands and their uses in the diagnosis and treatment of tauopathy
US10859582B2 (en) 2014-07-08 2020-12-08 New York University Tau imaging ligands and their uses in the diagnosis and treatment of tauopathy
US11519920B2 (en) 2014-07-08 2022-12-06 New York University Tau imaging ligands and their uses in the diagnosis and treatment of tauopathy
US10618955B2 (en) 2014-07-15 2020-04-14 Kymab Limited Methods for treating neurodegenerative disease using anti-PD-1 antibodies
US9187562B1 (en) 2014-07-15 2015-11-17 Kymab Limited Methods for treating anaemia
US9914769B2 (en) 2014-07-15 2018-03-13 Kymab Limited Precision medicine for cholesterol treatment
US9023359B1 (en) 2014-07-15 2015-05-05 Kymab Limited Targeting rare human PCSK9 variants for cholesterol treatment
US9017678B1 (en) 2014-07-15 2015-04-28 Kymab Limited Method of treating rheumatoid arthritis using antibody to IL6R
US11555066B2 (en) 2014-07-15 2023-01-17 Kymab Limited Precision medicine for cholesterol treatment
US9109034B1 (en) 2014-07-15 2015-08-18 Kymab Limited Precision medicine by targeting PD-L1 variants for treatment of cancer
US8999341B1 (en) 2014-07-15 2015-04-07 Kymab Limited Targeting rare human PCSK9 variants for cholesterol treatment
US9394568B2 (en) 2014-07-15 2016-07-19 Kymab Limited Methods of treating anaemia
US8992927B1 (en) 2014-07-15 2015-03-31 Kymab Limited Targeting human NAV1.7 variants for treatment of pain
US9034332B1 (en) 2014-07-15 2015-05-19 Kymab Limited Precision medicine by targeting rare human PCSK9 variants for cholesterol treatment
US9068012B1 (en) 2014-07-15 2015-06-30 Kymab Limited Targeting rare human PCSK9 variants for cholesterol treatment
US8986694B1 (en) 2014-07-15 2015-03-24 Kymab Limited Targeting human nav1.7 variants for treatment of pain
US10711059B2 (en) 2014-07-15 2020-07-14 Kymab Limited Methods for treating neurodegenerative diseases using anti-PD-L1 antibodies
US8986691B1 (en) 2014-07-15 2015-03-24 Kymab Limited Method of treating atopic dermatitis or asthma using antibody to IL4RA
US8980273B1 (en) 2014-07-15 2015-03-17 Kymab Limited Method of treating atopic dermatitis or asthma using antibody to IL4RA
US9067998B1 (en) 2014-07-15 2015-06-30 Kymab Limited Targeting PD-1 variants for treatment of cancer
US9428578B2 (en) 2014-07-15 2016-08-30 Kymab Limited Methods of treating anaemia
US9062105B1 (en) 2014-07-15 2015-06-23 Kymab Limited Precision Medicine by targeting VEGF-A variants for treatment of retinopathy
US9139648B1 (en) 2014-07-15 2015-09-22 Kymab Limited Precision medicine by targeting human NAV1.9 variants for treatment of pain
US9034331B1 (en) 2014-07-15 2015-05-19 Kymab Limited Targeting rare human PCSK9 variants for cholesterol treatment
US8945560B1 (en) 2014-07-15 2015-02-03 Kymab Limited Method of treating rheumatoid arthritis using antibody to IL6R
US9045548B1 (en) 2014-07-15 2015-06-02 Kymab Limited Precision Medicine by targeting rare human PCSK9 variants for cholesterol treatment
US9150660B1 (en) 2014-07-15 2015-10-06 Kymab Limited Precision Medicine by targeting human NAV1.8 variants for treatment of pain
US9439963B2 (en) 2014-07-15 2016-09-13 Kymab Limited Methods of treating anaemia
US9045545B1 (en) 2014-07-15 2015-06-02 Kymab Limited Precision medicine by targeting PD-L1 variants for treatment of cancer
US9303089B2 (en) 2014-07-15 2016-04-05 Kymab Limited Methods of treating anaemia
US9051378B1 (en) 2014-07-15 2015-06-09 Kymab Limited Targeting rare human PCSK9 variants for cholesterol treatment
US11661462B2 (en) 2014-07-31 2023-05-30 Amgen Research (Munich) Gmbh Optimized cross-species specific bispecific single chain antibody contructs
US9765157B2 (en) 2014-07-31 2017-09-19 Amgen Research (Munich) Gmbh Antibody constructs for CDH19 and CD3
WO2016016415A1 (en) 2014-07-31 2016-02-04 Amgen Research (Munich) Gmbh Bispecific single chain antibody construct with enhanced tissue distribution
WO2016016412A1 (en) 2014-07-31 2016-02-04 Amgen Research (Munich) Gmbh Antibody constructs for cdh19 and cd3
WO2016016859A1 (en) 2014-07-31 2016-02-04 Amgen Research (Munich) Gmbh Optimized cross-species specific bispecific single chain antibody constructs
WO2016016442A1 (en) 2014-08-01 2016-02-04 INSERM (Institut National de la Santé et de la Recherche Médicale) An anti-cd45rc antibody for use as drug
WO2016020880A2 (en) 2014-08-07 2016-02-11 Novartis Ag Angiopoietin-like 4 antibodies and methods of use
WO2016020882A2 (en) 2014-08-07 2016-02-11 Novartis Ag Angiopoetin-like 4 (angptl4) antibodies and methods of use
EP4122957A1 (en) 2014-08-07 2023-01-25 Novartis AG Angiopoietin-like 4 antibodies and methods of use
EP4056993A1 (en) 2014-08-20 2022-09-14 Chugai Seiyaku Kabushiki Kaisha Method for measuring viscosity of protein solution
US11014896B2 (en) 2014-08-22 2021-05-25 Duke University TRPA1 and TRPV4 inhibitors and methods of using the same for organ-specific inflammation and itch
US10329265B2 (en) 2014-08-22 2019-06-25 Duke University TRPA1 and TRPV4 inhibitors and methods of using the same for organ-specific inflammation and itch
WO2016039796A2 (en) 2014-09-12 2016-03-17 Regeneron Pharmaceuticals, Inc. Treatment of fibrodysplasia ossificans progressiva
EP3604335A1 (en) 2014-09-12 2020-02-05 Regeneron Pharmaceuticals, Inc. Treatment of fibrodysplasia ossificans progressiva
EP3950944A1 (en) 2014-09-15 2022-02-09 AbVitro LLC High-throughput nucleotide library sequencing
WO2016044227A1 (en) 2014-09-15 2016-03-24 Abvitro, Inc. High-throughput nucleotide library sequencing
US10590483B2 (en) 2014-09-15 2020-03-17 Abvitro Llc High-throughput nucleotide library sequencing
EP3536786A1 (en) 2014-09-15 2019-09-11 AbVitro LLC High-throughput nucleotide library sequencing
US11180555B2 (en) 2014-09-16 2021-11-23 Ubi Us Holdings, Llc. Antibodies directed against CD4 for the treatment and functional cure of HIV
US9975966B2 (en) 2014-09-26 2018-05-22 Chugai Seiyaku Kabushiki Kaisha Cytotoxicity-inducing theraputic agent
US11001643B2 (en) 2014-09-26 2021-05-11 Chugai Seiyaku Kabushiki Kaisha Cytotoxicity-inducing therapeutic agent
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
US10363290B2 (en) 2014-10-17 2019-07-30 Kodiak Sciences Inc. Butyrylcholinesterase zwitterionic polymer conjugates
US11071771B2 (en) 2014-10-17 2021-07-27 Kodiak Sciences Inc. Butyrylcholinesterase zwitterionic polymer conjugates
US10738078B2 (en) 2014-11-03 2020-08-11 Bristol-Myers Squibb Company Use of caprylic acid precipitation for protein purification
WO2016073401A1 (en) 2014-11-03 2016-05-12 Bristol-Myers Squibb Company Use of caprylic acid precipitation for protein purification
EP4268843A2 (en) 2014-11-07 2023-11-01 F. Hoffmann-La Roche Ltd Improved il-6 antibodies
WO2016073894A1 (en) 2014-11-07 2016-05-12 Eleven Biotherapeutics, Inc. Therapeutic agents with increased ocular retention
US11142571B2 (en) 2014-11-07 2021-10-12 Sesen Bio, Inc. IL-6 antibodies
EP3632931A1 (en) 2014-11-07 2020-04-08 Sesen Bio, Inc. Improved il-6 antibodies
WO2016081748A2 (en) 2014-11-21 2016-05-26 Bristol-Myers Squibb Company Antibodies against cd73 and uses thereof
EP3725808A1 (en) 2014-11-21 2020-10-21 Bristol-Myers Squibb Company Antibodies against cd73 and uses thereof
US10370455B2 (en) 2014-12-05 2019-08-06 Immunext, Inc. Identification of VSIG8 as the putative VISTA receptor (V-R) and use thereof to produce VISTA/VSIG8 agonists and antagonists
WO2016098079A2 (en) 2014-12-19 2016-06-23 Novartis Ag Compositions and methods for antibodies targeting bmp6
EP4249066A2 (en) 2014-12-23 2023-09-27 Bristol-Myers Squibb Company Antibodies to tigit
US11229628B2 (en) 2015-01-09 2022-01-25 Duke University TRPA1 and TRPV4 inhibitors and methods of using the same for organ-specific inflammation and itch
EP4134379A1 (en) 2015-01-28 2023-02-15 Prothena Biosciences Limited Anti-transthyretin antibodies
WO2016120809A1 (en) 2015-01-28 2016-08-04 Prothena Biosciences Limited Anti-transthyretin antibodies
WO2016120810A1 (en) 2015-01-28 2016-08-04 Prothena Biosciences Limited Anti-transthyretin antibodies
WO2016120811A1 (en) 2015-01-28 2016-08-04 Prothena Biosciences Limited Anti-transthyretin antibodies
EP3816179A2 (en) 2015-02-05 2021-05-05 Chugai Seiyaku Kabushiki Kaisha Fc region variant comprising a modified fcrn-binding domain
WO2016125495A1 (en) 2015-02-05 2016-08-11 Chugai Seiyaku Kabushiki Kaisha Antibodies comprising an ion concentration dependent antigen-binding domain, fc region variants, il-8-binding antibodies, and uses therof
EP4269440A2 (en) 2015-02-27 2023-11-01 Chugai Seiyaku Kabushiki Kaisha Composition for treating il-6-related diseases
EP4194071A1 (en) 2015-03-13 2023-06-14 Bristol-Myers Squibb Company Use of alkaline washes during chromatography to remove impurities
WO2016149088A1 (en) 2015-03-13 2016-09-22 Bristol-Myers Squibb Company Use of alkaline washes during chromatography to remove impurities
WO2016153983A1 (en) 2015-03-20 2016-09-29 Bristol-Myers Squibb Company Use of dextran for protein purification
WO2016153978A1 (en) 2015-03-20 2016-09-29 Bristol-Myers Squibb Company Use of dextran to enhance protein purification by affinity chromatography
WO2016166360A1 (en) 2015-04-17 2016-10-20 Bayer Pharma Aktiengesellschaft Bispecific antibody constructs for cdh3 and cd3
WO2016166014A1 (en) 2015-04-17 2016-10-20 F. Hoffmann-La Roche Ag Combination therapy with coagulation factors and multispecific antibodies
US11926666B2 (en) 2015-04-17 2024-03-12 Amgen Research (Munich) Gmbh Bispecific antibody constructs for CDH3 and CD3
US11028171B2 (en) 2015-04-17 2021-06-08 Amgen Research (Munich) Gmbh Bispecific antibody constructs for CDH3 and CD3
EP4276116A2 (en) 2015-04-17 2023-11-15 Amgen Research (Munich) GmbH Bispecific antibody constructs for cdh3 and cd3
WO2016172726A1 (en) 2015-04-24 2016-10-27 The Regents Of The University Of California Modulators of ror1-ror2 binding
WO2016176341A1 (en) 2015-04-29 2016-11-03 Regeneron Pharmaceuticals, Inc. Treatment of fibrodysplasia ossificans progressiva
WO2016196228A1 (en) 2015-05-29 2016-12-08 Bristol-Myers Squibb Company Antibodies against ox40 and uses thereof
WO2016193872A2 (en) 2015-06-05 2016-12-08 Novartis Ag Antibodies targeting bone morphogenetic protein 9 (bmp9) and methods therefor
US11009509B2 (en) 2015-06-24 2021-05-18 Janssen Pharmaceutica Nv Anti-VISTA antibodies and fragments
WO2017004016A1 (en) 2015-06-29 2017-01-05 The Rockefeller University Antibodies to cd40 with enhanced agonist activity
US10562962B2 (en) 2015-07-13 2020-02-18 H. Lundbeck A/S Antibodies specific for hyperphosphorylated tau and methods of use thereof
US10934348B2 (en) 2015-07-13 2021-03-02 H. Lundbeck A/S Antibodies specific for hyperphosphorylated tau and methods of use thereof
US11739140B2 (en) 2015-07-13 2023-08-29 H. Lundbeck A/S Antibodies specific for hyperphosphorylated tau and methods of use thereof
US10196439B2 (en) 2015-07-13 2019-02-05 H. Lundbeck A/S Antibodies specific for hyperphosphorylated tau and methods of use thereof
EP4327885A2 (en) 2015-07-31 2024-02-28 Amgen Research (Munich) GmbH Antibody constructs for msln and cd3
US11447567B2 (en) 2015-07-31 2022-09-20 Amgen Research (Munich) Gmbh Antibody constructs for FLT3 and CD3
WO2017021370A1 (en) 2015-07-31 2017-02-09 Amgen Research (Munich) Gmbh Bispecific antibody constructs binding egfrviii and cd3
WO2017021356A1 (en) 2015-07-31 2017-02-09 Amgen Research (Munich) Gmbh Bispecific antibody constructs binding mesothelin and cd3
US10683351B2 (en) 2015-07-31 2020-06-16 Amgen Research (Munich) Gmbh Antibody constructs for DLL3 and CD3
WO2017021362A1 (en) 2015-07-31 2017-02-09 Amgen Research (Munich) Gmbh Antibody constructs for flt3 and cd3
WO2017021354A1 (en) 2015-07-31 2017-02-09 Amgen Research (Munich) Gmbh Antibody constructs for cd70 and cd3
US10851170B2 (en) 2015-07-31 2020-12-01 Amgen Research (Munich) Gmbh Antibody constructs for CD70 and CD3
WO2017021349A1 (en) 2015-07-31 2017-02-09 Amgen Research (Munich) Gmbh Bispecific antibody constructs binding dll3 and cd3
EP3912999A1 (en) 2015-07-31 2021-11-24 Amgen Research (Munich) GmbH Bispecific antibody constructs binding egfrviii and cd3
US10519241B2 (en) 2015-07-31 2019-12-31 Amgen Research (Munich) Gmbh Antibody constructs for EGFRVIII and CD3
US10294300B2 (en) 2015-07-31 2019-05-21 Amgen Research (Munich) Gmbh Antibody constructs for DLL3 and CD3
US11591396B2 (en) 2015-07-31 2023-02-28 Amgen Research (Munich) Gmbh Antibody constructs for DLL3 and CD3
US11884720B2 (en) 2015-07-31 2024-01-30 Amgen Research (Munich) Gmbh Antibody constructs for MSLN and CD3
US11155629B2 (en) 2015-07-31 2021-10-26 Amgen Research (Munich) Gmbh Method for treating glioblastoma or glioma with antibody constructs for EGFRVIII and CD3
EP3865514A1 (en) 2015-07-31 2021-08-18 Amgen Research (Munich) GmbH Bispecific antibody constructs binding dll3 and cd3
EP4219562A2 (en) 2015-07-31 2023-08-02 Amgen Research (Munich) GmbH Antibody constructs for flt3 and cd3
WO2017021893A1 (en) 2015-08-03 2017-02-09 Novartis Ag Methods of treating fgf21-associated disorders
EP4223784A2 (en) 2015-09-02 2023-08-09 The Regents of the University of Colorado, a body corporate Compositions and methods for modulating t-cell mediated immune response
EP3842457A1 (en) 2015-09-09 2021-06-30 Novartis AG Thymic stromal lymphopoietin (tslp)-binding molecules and methods of using the molecules
WO2017042701A1 (en) 2015-09-09 2017-03-16 Novartis Ag Thymic stromal lymphopoietin (tslp)-binding antibodies and methods of using the antibodies
WO2017046774A2 (en) 2015-09-16 2017-03-23 Prothena Biosciences Limited Use of anti-mcam antibodies for treatment or prophylaxis of giant cell arteritis, polymyalgia rheumatica or takayasu's arteritis
WO2017046776A2 (en) 2015-09-16 2017-03-23 Prothena Biosciences Limited Use of anti-mcam antibodies for treatment or prophylaxis of giant cell arteritis, polymyalgia rheumatica or takayasu's arteritis
WO2017046994A1 (en) 2015-09-18 2017-03-23 Chugai Seiyaku Kabushiki Kaisha Il-8-binding antibodies and uses thereof
EP3933047A1 (en) 2015-09-24 2022-01-05 AbVitro LLC Affinity-oligonucleotide conjugates and uses thereof
WO2017053905A1 (en) 2015-09-24 2017-03-30 Abvitro Llc Affinity-oligonucleotide conjugates and uses thereof
WO2017053902A1 (en) 2015-09-25 2017-03-30 Abvitro Llc High throughput process for t cell receptor target identification of natively-paired t cell receptor sequences
WO2017075484A2 (en) 2015-10-30 2017-05-04 Galaxy Biotech, Llc Highly potent antibodies binding to death receptor 4 and death receptor 5
WO2017086367A1 (en) 2015-11-18 2017-05-26 中外製薬株式会社 Combination therapy using t cell redirection antigen binding molecule against cell having immunosuppressing function
WO2017086419A1 (en) 2015-11-18 2017-05-26 中外製薬株式会社 Method for enhancing humoral immune response
WO2017087678A2 (en) 2015-11-19 2017-05-26 Bristol-Myers Squibb Company Antibodies against glucocorticoid-induced tumor necrosis factor receptor (gitr) and uses thereof
WO2017095875A1 (en) 2015-11-30 2017-06-08 Bristol-Myers Squibb Company Anti human ip-10 antibodies and their uses
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
WO2017103895A1 (en) 2015-12-18 2017-06-22 Novartis Ag Antibodies targeting cd32b and methods of use thereof
WO2017110980A1 (en) 2015-12-25 2017-06-29 中外製薬株式会社 Antibody having enhanced activity, and method for modifying same
US11066465B2 (en) 2015-12-30 2021-07-20 Kodiak Sciences Inc. Antibodies and conjugates thereof
WO2017125897A1 (en) 2016-01-21 2017-07-27 Novartis Ag Multispecific molecules targeting cll-1
EP3851457A1 (en) 2016-01-21 2021-07-21 Novartis AG Multispecific molecules targeting cll-1
US11352433B2 (en) 2016-02-03 2022-06-07 Amgen Research (Munich) Gmbh BCMA and CD3 bispecific T cell engaging antibody constructs
EP4206228A1 (en) 2016-02-03 2023-07-05 Amgen Research (Munich) GmbH Psma and cd3 bispecific t cell engaging constructs
US11434302B2 (en) 2016-02-03 2022-09-06 Amgen Research (Munich) Gmbh Bispecific T cell engaging antibody constructs
US10301391B2 (en) 2016-02-03 2019-05-28 Amgen Research (Munich) Gmbh BCMA and CD3 bispecific T cell engaging antibody constructs
EP4039709A1 (en) 2016-02-03 2022-08-10 Amgen Research (Munich) GmbH Bcma and cd3 bispecific t cell engaging antibody constructs
US10781264B2 (en) 2016-02-03 2020-09-22 Amgen Research (Munich) Gmbh PSMA and CD3 bispecific T cell engaging antibody constructs
US10899836B2 (en) 2016-02-12 2021-01-26 Janssen Pharmaceutica Nv Method of identifying anti-VISTA antibodies
WO2017149513A1 (en) 2016-03-03 2017-09-08 Prothena Biosciences Limited Anti-mcam antibodies and associated methods of use
WO2017152085A1 (en) 2016-03-04 2017-09-08 Bristol-Myers Squibb Company Combination therapy with anti-cd73 antibodies
WO2017151176A1 (en) 2016-03-04 2017-09-08 The Rockefeller University Antibodies to cd40 with enhanced agonist activity
WO2017153953A1 (en) 2016-03-09 2017-09-14 Prothena Biosciences Limited Use of anti-mcam antibodies for treatment or prophylaxis of granulomatous lung diseases
WO2017153955A1 (en) 2016-03-09 2017-09-14 Prothena Biosciences Limited Use of anti-mcam antibodies for treatment or prophylaxis of granulomatous lung diseases
WO2017159287A1 (en) 2016-03-14 2017-09-21 中外製薬株式会社 Cell injury inducing therapeutic drug for use in cancer therapy
EP4302782A2 (en) 2016-03-15 2024-01-10 Mersana Therapeutics, Inc. Napi2b-targeted antibody-drug conjugates and methods of use thereof
WO2017160754A1 (en) 2016-03-15 2017-09-21 Mersana Therapeutics,Inc. Napi2b-targeted antibody-drug conjugates and methods of use thereof
US10870701B2 (en) 2016-03-15 2020-12-22 Generon (Shanghai) Corporation Ltd. Multispecific fab fusion proteins and use thereof
US10745487B2 (en) 2016-03-22 2020-08-18 Bionomics Limited Method of treating cancer by administering an anti-LGR5 monoclonal antibody
WO2017177200A1 (en) 2016-04-07 2017-10-12 Duke University Small molecule dual-inhibitors of trpv4 and trpa1 for sanitizing and anesthetizing
US11564911B2 (en) 2016-04-07 2023-01-31 Duke University Small molecule dual-inhibitors of TRPV4 and TRPA1 for sanitizing and anesthetizing
US11649283B2 (en) 2016-04-15 2023-05-16 Immunext, Inc. Anti-human vista antibodies and use thereof
US11603402B2 (en) 2016-04-15 2023-03-14 Immunext, Inc. Anti-human vista antibodies and use thereof
US11603403B2 (en) 2016-04-15 2023-03-14 Immunext, Inc. Anti-human vista antibodies and use thereof
US11525000B2 (en) 2016-04-15 2022-12-13 Immunext, Inc. Anti-human VISTA antibodies and use thereof
US11053318B2 (en) 2016-04-19 2021-07-06 Amgen Research (Munich) Gmbh Administration of a bispecific construct binding to CD33 and CD3 for use in a method for the treatment of myeloid leukemia
WO2017182427A1 (en) 2016-04-19 2017-10-26 Amgen Research (Munich) Gmbh Administration of a bispecific construct binding to cd33 and cd3 for use in a method for the treatment of myeloid leukemia
EP3896450A1 (en) 2016-04-21 2021-10-20 4TEEN4 Pharmaceuticals GmbH Methods for determining dpp3 and therapeutic methods
EP3854817A1 (en) 2016-04-21 2021-07-28 4TEEN4 Pharmaceuticals GmbH Methods for determining dpp3 and therapeutic methods
DE112017002105T5 (en) 2016-04-21 2019-04-25 Sphingotec Therapeutics Gmbh Method for the determination of DPP3 and therapeutic methods
WO2017182561A1 (en) 2016-04-21 2017-10-26 Sphingotec Therapeutics Gmbh Methods for determining dpp3 and therapeutic methods
US11584791B2 (en) 2016-05-02 2023-02-21 Prothena Biosciences Limited Antibodies recognizing tau
WO2017191561A1 (en) 2016-05-02 2017-11-09 Prothena Biosciences Limited Antibodies recognizing tau
WO2017191560A1 (en) 2016-05-02 2017-11-09 Prothena Biosciences Limited Antibodies recognizing tau
WO2017196663A1 (en) 2016-05-09 2017-11-16 Bristol-Myers Squibb Company Tl1a antibodies and uses thereof
US11767364B2 (en) 2016-05-09 2023-09-26 Bristol-Myers Squibb Company TL1A antibodies and methods of treatment
US10968279B2 (en) 2016-05-09 2021-04-06 Bristol-Myers Squibb Company TL1A antibodies and uses thereof
WO2017208210A1 (en) 2016-06-03 2017-12-07 Prothena Biosciences Limited Anti-mcam antibodies and associated methods of use
WO2017214167A1 (en) 2016-06-06 2017-12-14 City Of Hope Baff-r targeted chimeric antigen receptor-modified t-cells and uses thereof
WO2017214170A2 (en) 2016-06-06 2017-12-14 City Of Hope Baff-r antibodies and uses thereof
WO2017216724A1 (en) 2016-06-15 2017-12-21 Novartis Ag Methods for treating disease using inhibitors of bone morphogenetic protein 6 (bmp6)
WO2018002081A1 (en) 2016-06-27 2018-01-04 Aicuris Anti-Infective Cures Gmbh Hcmv entry inhibitors
WO2018007924A2 (en) 2016-07-02 2018-01-11 Prothena Biosciences Limited Anti-transthyretin antibodies
WO2018007922A2 (en) 2016-07-02 2018-01-11 Prothena Biosciences Limited Anti-transthyretin antibodies
WO2018007923A2 (en) 2016-07-02 2018-01-11 Prothena Biosciences Limited Anti-transthyretin antibodies
WO2018007588A1 (en) 2016-07-08 2018-01-11 Sphingotec Gmbh Adrenomedullin for assessing congestion in a subject with acute heart failure
EP4231018A2 (en) 2016-07-08 2023-08-23 SphingoTec GmbH Adrenomedullin for assessing congestion in a subject with acute heart failure
US10647762B2 (en) 2016-07-12 2020-05-12 H. Lundbeck A/S Antibodies specific for hyperphosphorylated tau and methods of use thereof
US10487142B2 (en) 2016-07-12 2019-11-26 H. Lundbeck A/S Antibodies specific for hyperphosphorylated tau and methods of use thereof
US10472415B2 (en) 2016-07-12 2019-11-12 H. Lundbeck A/S Antibodies specific for hyperphosphorylated tau and methods of use
US11111290B2 (en) 2016-07-12 2021-09-07 H. Lundbeck A/S Antibodies specific for hyperphosphorylated tau and methods of use thereof
US10533052B2 (en) 2016-07-14 2020-01-14 Bristol-Myers Squibb Company Antibodies against TIM3 and uses thereof
WO2018013818A2 (en) 2016-07-14 2018-01-18 Bristol-Myers Squibb Company Antibodies against tim3 and uses thereof
US11591392B2 (en) 2016-07-14 2023-02-28 Bristol-Myers Squibb Company Antibodies against TIM3 and uses thereof
US10077306B2 (en) 2016-07-14 2018-09-18 Bristol-Myers Squibb Company Antibodies against TIM3 and uses thereof
WO2018029586A1 (en) 2016-08-07 2018-02-15 Novartis Ag Mrna-mediated immunization methods
WO2018031726A1 (en) 2016-08-12 2018-02-15 Bristol-Myers Squibb Company Methods of purifying proteins
US11292839B2 (en) 2016-08-13 2022-04-05 Ubi Us Holdings, Llc Treatment and sustained virologic remission of HIV infection by antibodies to CD4 in HAART stabilized patients
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
WO2018049248A1 (en) 2016-09-09 2018-03-15 Icellhealth Consulting Llc Oncolytic virus equipped with bispecific engager molecules
WO2018049261A1 (en) 2016-09-09 2018-03-15 Icellhealth Consulting Llc Oncolytic virus expressing immune checkpoint modulators
WO2018057051A1 (en) 2016-09-24 2018-03-29 Abvitro Llc Affinity-oligonucleotide conjugates and uses thereof
US11779604B2 (en) 2016-11-03 2023-10-10 Kymab Limited Antibodies, combinations comprising antibodies, biomarkers, uses and methods
WO2018087720A1 (en) 2016-11-14 2018-05-17 Novartis Ag Compositions, methods, and therapeutic uses related to fusogenic protein minion
WO2018097308A1 (en) 2016-11-28 2018-05-31 中外製薬株式会社 Ligand-binding molecule having adjustable ligand binding activity
WO2018109228A1 (en) 2016-12-16 2018-06-21 Adrenomed Ag Anti-adrenomedullin (adm) antibody or anti-adm antibody fragment or anti-adm non-ig scaffold for use in intervention and therapy of congestion in a patient in need thereof
EP3339324A1 (en) 2016-12-22 2018-06-27 sphingotec GmbH Anti-adrenomedullin (adm) antibody or anti-adm antibody fragment or anti-adm non-ig scaffold for use in intervention and therapy of congestion in a patient in need thereof
WO2018129451A2 (en) 2017-01-09 2018-07-12 Merrimack Pharmaceuticals, Inc. Anti-fgfr antibodies and methods of use
WO2018141910A1 (en) 2017-02-02 2018-08-09 Amgen Research (Munich) Gmbh Low ph pharmaceutical composition comprising t cell engaging antibody constructs
WO2018146594A1 (en) 2017-02-08 2018-08-16 Novartis Ag Fgf21 mimetic antibodies and uses thereof
US11827695B2 (en) 2017-02-17 2023-11-28 Bristol-Myers Squibb Company Antibodies to alpha-synuclein and uses thereof
WO2018151821A1 (en) 2017-02-17 2018-08-23 Bristol-Myers Squibb Company Antibodies to alpha-synuclein and uses thereof
US11142570B2 (en) 2017-02-17 2021-10-12 Bristol-Myers Squibb Company Antibodies to alpha-synuclein and uses thereof
WO2018175460A1 (en) 2017-03-24 2018-09-27 Novartis Ag Methods for preventing and treating heart disease
WO2018174274A1 (en) 2017-03-24 2018-09-27 全薬工業株式会社 ANTI-IgM/B CELL SURFACE ANTIGEN BISPECIFIC ANTIBODY
US11359005B2 (en) 2017-03-30 2022-06-14 The Johns Hopkins University Supramolecular high affinity protein-binding system for purification of biomacromolecules
WO2018181870A1 (en) 2017-03-31 2018-10-04 公立大学法人奈良県立医科大学 Medicinal composition usable for preventing and/or treating blood coagulation factor ix abnormality, comprising multispecific antigen binding molecule replacing function of blood coagulation factor viii
WO2018187613A2 (en) 2017-04-07 2018-10-11 Bristol-Myers Squibb Company Anti-icos agonist antibodies and uses thereof
US11584790B2 (en) 2017-04-14 2023-02-21 Kodiak Sciences Inc. Complement factor D antagonist antibodies and conjugates thereof
WO2018199214A1 (en) 2017-04-27 2018-11-01 中外製薬株式会社 Coagulation factor ix with improved pharmacokinetics
WO2018203545A1 (en) 2017-05-02 2018-11-08 国立研究開発法人国立精神・神経医療研究センター Method for predicting and evaluating therapeutic effect in diseases related to il-6 and neutrophils
WO2018204907A1 (en) 2017-05-05 2018-11-08 Amgen Inc. Pharmaceutical composition comprising bispecific antibody constructs for improved storage and administration
US11918650B2 (en) 2017-05-05 2024-03-05 Amgen Inc. Pharmaceutical composition comprising bispecific antibody constructs for improved storage and administration
WO2018218222A1 (en) 2017-05-26 2018-11-29 Goldfless Stephen Jacob High-throughput polynucleotide library sequencing and transcriptome analysis
WO2018229251A1 (en) 2017-06-16 2018-12-20 Imba - Institut Für Molekulare Biotechnologie Gmbh Blood vessel organoid, methods of producing and using said organoids
WO2018229715A1 (en) 2017-06-16 2018-12-20 Novartis Ag Compositions comprising anti-cd32b antibodies and methods of use thereof
WO2019003104A1 (en) 2017-06-28 2019-01-03 Novartis Ag Methods for preventing and treating urinary incontinence
WO2019016247A2 (en) 2017-07-20 2019-01-24 H. Lundbeck A/S Agents, uses and methods for treatment
US11745165B2 (en) 2017-08-18 2023-09-05 The Johns Hopkins University Supramolecular filamentous assemblies for protein purification
WO2019051335A1 (en) 2017-09-07 2019-03-14 Juno Therapeutics, Inc. Methods of identifying cellular attributes related to outcomes associated with cell therapy
EP4159230A1 (en) 2017-09-25 2023-04-05 AdrenoMed AG Anti-adrenomedullin (adm) binder for use in therapy or prevention of symptoms of illness
WO2019057992A2 (en) 2017-09-25 2019-03-28 Adrenomed Ag Anti-adrenomedullin (adm) binder for use in therapy or prevention of symptoms of illness
EP3854414A1 (en) 2017-09-25 2021-07-28 AdrenoMed AG Anti-adrenomedullin (adm) binder for use in therapy or prevention of symptoms of illness
EP4159229A1 (en) 2017-09-25 2023-04-05 AdrenoMed AG Anti-adrenomedullin (adm) binder for use in therapy or prevention of symptoms of illness
WO2019077082A1 (en) 2017-10-18 2019-04-25 Adrenomed Ag Therapy monitoring under treatment with an anti-adrenomedullin (adm) binder
WO2019078344A1 (en) 2017-10-20 2019-04-25 学校法人兵庫医科大学 Anti-il-6 receptor antibody-containing medicinal composition for preventing post-surgical adhesion
WO2019081983A1 (en) 2017-10-25 2019-05-02 Novartis Ag Antibodies targeting cd32b and methods of use thereof
WO2019081595A2 (en) 2017-10-25 2019-05-02 Sphingotec Therapeutics Gmbh Dpp3 binder directed to and binding to specific dpp3-epitopes and its use in the prevention or treatment of diseases / acute conditions that are associated with oxidative stress
WO2019107384A1 (en) 2017-11-28 2019-06-06 中外製薬株式会社 Ligand-binding molecule having adjustable ligand-binding activity
WO2019109016A1 (en) 2017-12-01 2019-06-06 Millennium Pharmaceuticals, Inc. Biomarkers and methods for treatment with nae inhibitors
WO2019118426A1 (en) 2017-12-11 2019-06-20 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
WO2019131988A1 (en) 2017-12-28 2019-07-04 Chugai Seiyaku Kabushiki Kaisha Cytotoxicity-inducing therapeutic agent
WO2019133961A1 (en) 2017-12-29 2019-07-04 Amgen Inc. Bispecific antibody construct directed to muc17 and cd3
WO2019140229A1 (en) 2018-01-12 2019-07-18 Bristol-Myers Squibb Company Antibodies against tim3 and uses thereof
WO2019140216A1 (en) 2018-01-12 2019-07-18 Amgen Inc. Pac1 antibodies and uses thereof
WO2019151418A1 (en) 2018-01-31 2019-08-08 元一 加藤 Therapeutic agent for asthma containing il-6 inhibitor
WO2019150309A1 (en) 2018-02-02 2019-08-08 Hammack Scott Modulators of gpr68 and uses thereof for treating and preventing diseases
WO2019154900A1 (en) 2018-02-08 2019-08-15 Sphingotec Gmbh Adrenomedullin (adm) for diagnosis and/or prediction of dementia and anti-adrenomedullin binder for use in therapy or prevention of dementia
US11242393B2 (en) 2018-03-23 2022-02-08 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
WO2019219923A1 (en) 2018-05-18 2019-11-21 Julius-Maximilians-Universität Würzburg Compounds and methods for the immobilization of myostatin-inhibitors on the extracellular matrix by transglutaminase
EP3569614A1 (en) 2018-05-18 2019-11-20 Julius-Maximilians-Universität Würzburg Compounds and methods for the immobilization of myostatin-inhibitors on the extracellular matrix by transglutaminase
WO2019225568A1 (en) 2018-05-21 2019-11-28 中外製薬株式会社 Lyophilized formulation sealed in glass vial
WO2019229658A1 (en) 2018-05-30 2019-12-05 Novartis Ag Entpd2 antibodies, combination therapies, and methods of using the antibodies and combination therapies
WO2019243555A1 (en) 2018-06-21 2019-12-26 Charité - Universitätsmedizin Berlin Complement anaphylatoxin binders and their use in treatment of a subject having an ocular wound and/or fibrosis
EP3586865A1 (en) 2018-06-21 2020-01-01 Charité - Universitätsmedizin Berlin Complement anaphylatoxin binders and their use in treatment of a subject having an ocular wound and/or fibrosis
WO2020016433A1 (en) 2018-07-20 2020-01-23 Aicuris Gmbh & Co. Kg Methods for screening and identifying agents that inhibit or modulate the nuclear egress complex of herpesviruses
WO2020025532A1 (en) 2018-07-30 2020-02-06 Amgen Research (Munich) Gmbh Prolonged administration of a bispecific antibody construct binding to cd33 and cd3
WO2020025792A1 (en) 2018-08-03 2020-02-06 Amgen Research (Munich) Gmbh Antibody constructs for cldn18.2 and cd3
US11692031B2 (en) 2018-08-03 2023-07-04 Amgen Research (Munich) Gmbh Antibody constructs for CLDN18.2 and CD3
WO2020041360A1 (en) 2018-08-21 2020-02-27 Quidel Corporation Dbpa antibodies and uses thereof
WO2020043670A1 (en) 2018-08-27 2020-03-05 Affimed Gmbh Cryopreserved nk cells preloaded with an antibody construct
WO2020061210A1 (en) 2018-09-18 2020-03-26 Merrimack Pharmaceuticals, Inc. Anti-tnfr2 antibodies and uses thereof
US11873334B2 (en) 2018-09-24 2024-01-16 EyePoint Pharmaceuticals, Inc. Method of treating ocular conditions by administering an antibody that activates Tie2 and binds VEGF
WO2020077212A1 (en) 2018-10-11 2020-04-16 Amgen Inc. Downstream processing of bispecific antibody constructs
WO2020079580A1 (en) 2018-10-15 2020-04-23 Novartis Ag Trem2 stabilizing antibodies
WO2020102501A1 (en) 2018-11-16 2020-05-22 Bristol-Myers Squibb Company Anti-nkg2a antibodies and uses thereof
WO2020112870A1 (en) 2018-11-28 2020-06-04 Forty Seven, Inc. Genetically modified hspcs resistant to ablation regime
WO2020128039A2 (en) 2018-12-21 2020-06-25 4TEEN4 Pharmaceuticals GmbH Therapy guidance and/or therapy monitoring for a treatment with angiotensin-receptor-agonist and/or a precursor thereof
WO2020154293A1 (en) 2019-01-22 2020-07-30 Bristol-Myers Squibb Company Antibodies against il-7r alpha subunit and uses thereof
US11008395B2 (en) 2019-01-22 2021-05-18 Bristol Myers-Squibb Company Antibodies against IL-7R alpha subunit and uses thereof
US11919962B2 (en) 2019-01-22 2024-03-05 Bristol Myers-Squibb Company Antibodies against IL-7R alpha subunit and uses thereof
WO2020153467A1 (en) 2019-01-24 2020-07-30 中外製薬株式会社 Novel cancer antigens and antibodies of said antigens
WO2020175689A1 (en) 2019-02-28 2020-09-03 学校法人順天堂 Antibody capable of binding to truncated mutant calreticulin, and diagnostic, prophylactic or therapeutic drug for myeloproliferative neoplasms
WO2020180712A1 (en) 2019-03-01 2020-09-10 Merrimack Pharmaceuticals, Inc. Anti-tnfr2 antibodies and uses thereof
US11926659B2 (en) 2019-03-03 2024-03-12 Prothena Biosciences Limited Antibodies recognizing tau
WO2020189748A1 (en) 2019-03-19 2020-09-24 中外製薬株式会社 Antigen-binding molecule containing antigen-binding domain of which binding activity to antigen is changed depending on mta, and library for obtaining said antigen-binding domain
WO2020205469A1 (en) 2019-03-29 2020-10-08 Bristol-Myers Squibb Company Methods of measuring hydrophobicity of chromatographic resins
WO2020209318A1 (en) 2019-04-10 2020-10-15 中外製薬株式会社 Method for purifying fc region-modified antibody
WO2020213665A1 (en) 2019-04-17 2020-10-22 国立大学法人広島大学 Therapeutic agent for urological cancer which is characterized by being administered with il-6 inhibitor and ccr2 inhibitor in combination
WO2020246563A1 (en) 2019-06-05 2020-12-10 中外製薬株式会社 Antibody cleavage site-binding molecule
WO2020250159A1 (en) 2019-06-12 2020-12-17 Novartis Ag Natriuretic peptide receptor 1 antibodies and methods of use
WO2020252442A1 (en) 2019-06-13 2020-12-17 Amgen Inc. Automated biomass-based perfusion control in the manufacturing of biologics
WO2021021676A1 (en) 2019-07-26 2021-02-04 Amgen Inc. Anti-il13 antigen binding proteins
WO2021038078A1 (en) 2019-08-30 2021-03-04 4TEEN4 Pharmaceuticals GmbH Therapy guidance and/or therapy monitoring for treatment of shock
WO2021050640A1 (en) 2019-09-10 2021-03-18 Amgen Inc. Purification method for bispecific antigen-binding polypeptides with enhanced protein l capture dynamic binding capacity
WO2021053559A1 (en) 2019-09-18 2021-03-25 Novartis Ag Entpd2 antibodies, combination therapies, and methods of using the antibodies and combination therapies
WO2021053560A1 (en) 2019-09-18 2021-03-25 Novartis Ag Combination therapy with entpd2 and cd73 antibodies
US11912784B2 (en) 2019-10-10 2024-02-27 Kodiak Sciences Inc. Methods of treating an eye disorder
WO2021097344A1 (en) 2019-11-13 2021-05-20 Amgen Inc. Method for reduced aggregate formation in downstream processing of bispecific antigen-binding molecules
WO2021127547A2 (en) 2019-12-19 2021-06-24 Quidel Corporation Monoclonal antibody fusions
WO2021127528A1 (en) 2019-12-20 2021-06-24 Amgen Inc. Mesothelin-targeted cd40 agonistic multispecific antibody constructs for the treatment of solid tumors
WO2021130383A1 (en) 2019-12-27 2021-07-01 Affimed Gmbh Method for the production of bispecific fcyriii x cd30 antibody construct
WO2021142191A1 (en) 2020-01-08 2021-07-15 Regeneron Pharmaceuticals, Inc. Treatment of fibrodysplasia ossificans progressiva
WO2021150824A1 (en) 2020-01-22 2021-07-29 Amgen Research (Munich) Gmbh Combinations of antibody constructs and inhibitors of cytokine release syndrome and uses thereof
EP3871689A1 (en) 2020-02-26 2021-09-01 sphingotec GmbH Anti-adm-antibodies binding to the free n-terminus for accelerated transition of adm-gly to bio-adm in patients with adm-gly/ bio-adm ratio above a threshold and combination with vitamin c
WO2021170763A1 (en) 2020-02-26 2021-09-02 Sphingotec Gmbh Anti-adm-antibodies binding to the free n-terminus for accelerated transition of adm-gly to bio-adm in patients with adm-gly/ bio-adm ratio above a threshold and combination with vitamin c
WO2021170838A1 (en) 2020-02-27 2021-09-02 4TEEN4 Pharmaceuticals GmbH Dpp3 for therapy guidance, monitoring and stratification of nt-adm antibodies in patients with shock
WO2021170876A1 (en) 2020-02-27 2021-09-02 Adrenomed Ag Anti-adrenomedullin (adm) antibody or anti-adm antibody fragment or anti-adm non-ig scaffold for use in therapy or prevention of shock
WO2021170880A2 (en) 2020-02-27 2021-09-02 Adrenomed Ag Anti-adrenomedullin (adm) binder for use in therapy of patients in shock
WO2021183861A1 (en) 2020-03-12 2021-09-16 Amgen Inc. Method for treatment and prophylaxis of crs in patients comprising a combination of bispecifc antibodies binding to cds x cancer cell and tnfalpha or il-6 inhibitor
WO2021185786A1 (en) 2020-03-16 2021-09-23 4TEEN4 Pharmaceuticals GmbH Dpp3 in patients infected with coronavirus
WO2021185784A1 (en) 2020-03-16 2021-09-23 Sphingotec Gmbh Pro-adrenomedullin or fragment thereof in patients infected with corona virus and treatments with binder against adrenomedullin
WO2021185785A1 (en) 2020-03-16 2021-09-23 Sphingotec Gmbh Pro-adrenomedullin or fragment thereof in patients infected with corona virus and treatments with binder against adrenomedullin
WO2021188851A1 (en) 2020-03-19 2021-09-23 Amgen Inc. Antibodies against mucin 17 and uses thereof
WO2021193928A1 (en) 2020-03-27 2021-09-30 株式会社PhotoQ3 Pharmaceutical drug for destroying tumor cells
WO2021206078A1 (en) 2020-04-06 2021-10-14 株式会社PhotoQ3 Medicine for killing tumor cells
WO2021210667A1 (en) 2020-04-17 2021-10-21 中外製薬株式会社 Bispecific antigen-binding molecule, composition associated therewith, and use, kit, and method for producing composition
WO2021220215A1 (en) 2020-05-01 2021-11-04 Novartis Ag Engineered immunoglobulins
WO2021220218A1 (en) 2020-05-01 2021-11-04 Novartis Ag Immunoglobulin variants
EP3909601A1 (en) 2020-05-11 2021-11-17 LeukoCom GmbH A novel antibody binding specifically to human ceacam1/3/5 and use thereof
WO2021228746A1 (en) 2020-05-11 2021-11-18 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
WO2021236638A1 (en) 2020-05-19 2021-11-25 Amgen Inc. Mageb2 binding constructs
WO2021235537A1 (en) 2020-05-22 2021-11-25 中外製薬株式会社 Antibody for neutralizing substance having coagulation factor viii (f.viii) function-substituting activity
WO2021243320A2 (en) 2020-05-29 2021-12-02 Amgen Inc. Adverse effects-mitigating administration of a bispecific antibody construct binding to cd33 and cd3
WO2021247591A1 (en) 2020-06-02 2021-12-09 Arcus Biosciences, Inc. Antibodies to tigit
EP3922993A1 (en) 2020-06-12 2021-12-15 4TEEN4 Pharmaceuticals GmbH Dpp3 in patients infected with coronavirus
WO2021261546A1 (en) 2020-06-24 2021-12-30 国立大学法人 東京大学 Photosensitizing dye
WO2022025030A1 (en) 2020-07-28 2022-02-03 中外製薬株式会社 Prefilled syringe preparation with needle, provided with needle shield and including novel modified antibody
WO2022025220A1 (en) 2020-07-31 2022-02-03 中外製薬株式会社 Pharmaceutical composition including cell expressing chimeric receptor
US11484604B2 (en) 2020-08-07 2022-11-01 Fortis Therapeutics, Inc. Immunoconjugates targeting CD46 and methods of use thereof
WO2022045247A1 (en) 2020-08-27 2022-03-03 学校法人順天堂 Anti-truncated mutant calr-cd3 bispecific antibody and pharmaceutical composition
WO2022074206A1 (en) 2020-10-08 2022-04-14 Affimed Gmbh Trispecific binders
WO2022096698A1 (en) 2020-11-06 2022-05-12 Amgen Inc. Polypeptide constructs binding to cd3
WO2022096700A1 (en) 2020-11-06 2022-05-12 Amgen Research (Munich) Gmbh Polypeptide constructs selectively binding to cldn6 and cd3
WO2022096704A1 (en) 2020-11-06 2022-05-12 Amgen Inc. Antigen binding domain with reduced clipping rate
WO2022097065A2 (en) 2020-11-06 2022-05-12 Novartis Ag ANTIBODY Fc VARIANTS
WO2022096716A2 (en) 2020-11-06 2022-05-12 Amgen Inc. Multitargeting bispecific antigen-binding molecules of increased selectivity
EP4023218A1 (en) 2020-12-02 2022-07-06 S-Form Pharma Combination therapy for patients having acute and/or persistent dyspnea
WO2022117893A2 (en) 2020-12-02 2022-06-09 S-Form Pharma Combination therapy for patients having acute and/or persistent dyspnea
WO2022130182A1 (en) 2020-12-14 2022-06-23 Novartis Ag Reversal binding agents for anti-natriuretic peptide receptor 1 (npr1) antibodies and uses thereof
WO2022191306A1 (en) 2021-03-12 2022-09-15 中外製薬株式会社 Pharmaceutical composition for treatment or prevention of myasthenia gravis
WO2022212831A1 (en) 2021-04-02 2022-10-06 Amgen Inc. Mageb2 binding constructs
WO2022234102A1 (en) 2021-05-06 2022-11-10 Amgen Research (Munich) Gmbh Cd20 and cd22 targeting antigen-binding molecules for use in proliferative diseases
WO2023007023A1 (en) 2021-07-30 2023-02-02 Affimed Gmbh Duplexbodies
EP4144898A1 (en) 2021-09-07 2023-03-08 New/era/mabs GmbH Method for selecting or screening antibodies from an antibody library
WO2023036852A1 (en) 2021-09-07 2023-03-16 new/era/mabs GmbH Method for selecting or screening antibodies and antigens from an antibody library
WO2023057871A1 (en) 2021-10-04 2023-04-13 Novartis Ag Surfactant stabilizers
WO2023058723A1 (en) 2021-10-08 2023-04-13 中外製薬株式会社 Method for preparing prefilled syringe formulation
WO2023078968A1 (en) 2021-11-03 2023-05-11 Affimed Gmbh Bispecific cd16a binders
WO2023079493A1 (en) 2021-11-03 2023-05-11 Affimed Gmbh Bispecific cd16a binders
WO2023175035A1 (en) 2022-03-15 2023-09-21 Adrenomed Ag Stable aqueous formulation of an anti-adrenomedullin (adm) antibody or anti-adm antibody fragment
WO2023209568A1 (en) 2022-04-26 2023-11-02 Novartis Ag Multispecific antibodies targeting il-13 and il-18
WO2023218027A1 (en) 2022-05-12 2023-11-16 Amgen Research (Munich) Gmbh Multichain multitargeting bispecific antigen-binding molecules of increased selectivity
WO2024023368A1 (en) 2022-07-29 2024-02-01 4TEEN4 Pharmaceuticals GmbH Prediction of an increase of dpp3 in a patient with septic shock
WO2024023369A1 (en) 2022-07-29 2024-02-01 Adrenomed Ag Anti-adrenomedullin (adm) antibody or anti-adm antibody fragment or anti-adm non-ig scaffold for use in therapy or prevention of shock
WO2024059675A2 (en) 2022-09-14 2024-03-21 Amgen Inc. Bispecific molecule stabilizing composition
EP4345109A1 (en) 2022-09-30 2024-04-03 AdrenoMed AG Anti-adrenomedullin (adm) binder for use in therapy of pediatric patients with congenital heart disease

Also Published As

Publication number Publication date
CA2124967A1 (en) 1993-06-24
CA2124967C (en) 2008-04-08
JP2004008218A (en) 2004-01-15
EP0746609A4 (en) 1997-12-17
EP0746609A1 (en) 1996-12-11
AU3328493A (en) 1993-07-19
JPH07503132A (en) 1995-04-06

Similar Documents

Publication Publication Date Title
US5545806A (en) Ransgenic non-human animals for producing heterologous antibodies
US5661016A (en) Transgenic non-human animals capable of producing heterologous antibodies of various isotypes
US5789650A (en) Transgenic non-human animals for producing heterologous antibodies
US5877397A (en) Transgenic non-human animals capable of producing heterologous antibodies of various isotypes
CA2124967C (en) Transgenic non-human animals capable of producing heterologous antibodies
US5569825A (en) Transgenic non-human animals capable of producing heterologous antibodies of various isotypes
US5814318A (en) Transgenic non-human animals for producing heterologous antibodies
US5625126A (en) Transgenic non-human animals for producing heterologous antibodies
US5770429A (en) Transgenic non-human animals capable of producing heterologous antibodies
CA2161351C (en) Transgenic non-human animals capable of producing heterologous antibodies
US5633425A (en) Transgenic non-human animals capable of producing heterologous antibodies
US7084260B1 (en) High affinity human antibodies and human antibodies against human antigens
US8158419B2 (en) Transgenic non-human animals for producing chimeric antibodies
US5874299A (en) Transgenic non-human animals capable of producing heterologous antibodies
AU747370B2 (en) Transgenic non-human animals capable of producing heterologous antibo dies
WO1997013852A9 (en) Transgenic non-human animals capable of producing heterologous antibodies
EP1288229A2 (en) Transgenic non human animals capable of producing heterologous antibodies
AU720612B2 (en) Transgenic non-human animals capable of producing heterologous antibodies
AU2003204055B2 (en) Transgenic non-human animals capable of producing heterologous antibodies

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AT AU BB BG BR CA CH CS DE DK ES FI GB HU JP KP KR LK LU MG MN MW NL NO NZ PL RO RU SD SE UA US

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): AT BE CH DE DK ES FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN ML MR SN TD TG

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

Ref document number: 2124967

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 1993901143

Country of ref document: EP

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

WWP Wipo information: published in national office

Ref document number: 1993901143

Country of ref document: EP

WWW Wipo information: withdrawn in national office

Ref document number: 1993901143

Country of ref document: EP