WO1994025585A1 - 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
WO1994025585A1
WO1994025585A1 PCT/US1994/004580 US9404580W WO9425585A1 WO 1994025585 A1 WO1994025585 A1 WO 1994025585A1 US 9404580 W US9404580 W US 9404580W WO 9425585 A1 WO9425585 A1 WO 9425585A1
Authority
WO
WIPO (PCT)
Prior art keywords
human
heavy chain
transgene
gene
sequence
Prior art date
Application number
PCT/US1994/004580
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 US08/053,131 external-priority patent/US5661016A/en
Priority claimed from US08/096,762 external-priority patent/US5814318A/en
Application filed by Genpharm International, Inc. filed Critical Genpharm International, Inc.
Priority to AU68194/94A priority Critical patent/AU6819494A/en
Priority to CA2161351A priority patent/CA2161351C/en
Priority to EP94916581A priority patent/EP0754225A4/en
Priority to JP6524481A priority patent/JPH08509612A/en
Publication of WO1994025585A1 publication Critical patent/WO1994025585A1/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

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/8509Vectors or expression systems specially adapted for eukaryotic hosts for animal cells for producing genetically modified animals, e.g. transgenic
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K67/00Rearing or breeding animals, not otherwise provided for; New breeds of animals
    • A01K67/027New breeds of vertebrates
    • A01K67/0275Genetically 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
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/46Hybrid immunoglobulins
    • C07K16/461Igs containing Ig-regions, -domains or -residues form different species
    • C07K16/462Igs containing a variable region (Fv) from one specie and a constant region (Fc) from another
    • 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
    • A01K2217/30Animal model comprising expression system for selective cell killing, e.g. toxins, enzyme dependent prodrug therapy using ganciclovir
    • 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
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered

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 producing heterologous antibodies wherein a variable region sequence comprises somatic mutation as compared to germline rearranged variable region sequences, transgenic nonhuman animals which produce antibodies having a human primary sequence and which bind to human antigens, hybridomas made from B cells of such transgenic animals, and monclonal antibodies expressed by such hybridomas.
  • HAMA human anti-mouse antibodies
  • 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 produced.
  • antibodies reactive with self antigens may be produced.
  • 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 animals harboring such transgenes that are able to undergo functional V-D-J rearrangement to generate recombinational diversity and junctional diversity.
  • transgenes and transgenic animals preferably include cis-acting sequences that are able to undergo functional V-D-J rearrangement to generate recombinational diversity and junctional diversity.
  • 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 ⁇ 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
  • transgene 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 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
  • heterologous transgenes that undergo successful isotype switching.
  • Transgenic animals that cannot switch isotypes are limited to producing heterologous 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 immunoglobulin
  • 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 (l) functional B cell development may occur, and (2)
  • therapeutically useful heterologous antibodies may be any therapeutically useful heterologous antibodies.
  • immunoglobulin transgene capable of functional V-D-J recombination and/or capable of isotype switching could fulfill these needs.
  • transgenic nonhuman animals which are capable of producing a heterologous antibody, such as a human antibody.
  • 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.
  • 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, lgG3, IgG4, IgM, IgA1, IgA2, IgA sec , IgD, of IgE.
  • IgG1, IgG2, lgG3, IgG4, IgM, IgA1, IgA2, IgA sec , 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
  • 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 classswitching 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.
  • 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
  • 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.
  • 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 XhoI and SalI 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.
  • Such endogenous gene segments include diversity, joining and constant region gene segments.
  • 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.
  • 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.
  • the invention provides transgenic non-human animals comprising: a homozygous pair of functionally disrupted endogenous heavy chain alleles, a homozygous pair of
  • immunoglobulin heavy chain transgene wherein said animal makes an antibody response following immunization with an antigen, such as a human antigen (e.g., CD4).
  • an antigen such as a human antigen (e.g., CD4).
  • the invention also provides such a transgenic non-human animal wherein said functionally disrupted endogenous heavy chain allele is a J H region homologous recombination knockout, said functionally disrupted endogenous light chain allele is a J ⁇ region
  • immunoglobulin heavy chain transgene is the HC1 or HC2 human minigene transgene
  • said heterologous light chain transgene is the KC2 or KC1e human ⁇ transgene
  • said antigen is a human antigen
  • the invention also provides various embodiments for suppressing, ablating, and/or functionally disrupting the endogenous nonhuman immunoglobulin loci.
  • the invention also provides transgenic mice expressing both human sequence heavy chains and chimeric heavy chains comprising a human sequence heavy chain variable region and a murine sequence heavy chain constant region.
  • Such chimeric heavy chains are generally produced by transswitching between a functionally rearranged human transgene and an endogenous murine heavy chain constant region (e.g., ⁇ 1, ⁇ 2a, ⁇ 2b, ⁇ 3).
  • Antibodies comprising such chimeric heavy chains typically in combination with a transgene-encoded human sequence light chain or endogenous murine light chain, are formed in response to immunization with a predetermined antigen.
  • the transgenic mice of these embodiments can comprise B cells which produce (express) a human sequence heavy chain at a first timepoint and trans-switch to produce (express) a chimeric heavy chain composed of a human variable region and a murine constant region (e.g., ⁇ 1, ⁇ 2a, ⁇ 2b, ⁇ 3) at a second (subsequent) timepoint; such human sequence and chimeric heavy chains are incorporated into functional
  • the transgenic mice of these embodiments can comprise B cells which express a human sequence heavy chain and subsequently switch (via trans-switching or cis-switching) to express a chimeric or isotype-switched heavy chain composed of a human variable region and a alternative constant region (e.g., murine ⁇ 1, ⁇ 2a, ⁇ 2b, ⁇ 3; human ⁇ , ⁇ , ⁇ ); such human sequence and chimeric or isotype-switched heavy chains are incorporated into functional antibodies with light chains (human or mouse); such antibodies are present in the serum of such transgenic mice.
  • a human sequence heavy chain and subsequently switch (via trans-switching or cis-switching) to express a chimeric or isotype-switched heavy chain composed of a human variable region and a alternative constant region (e.g., murine ⁇ 1, ⁇ 2a, ⁇ 2b, ⁇ 3; human ⁇ , ⁇ , ⁇ ); such human sequence and chimeric or isotype-
  • 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 ⁇ 1 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. 20A through 20E depict the structure of a kappa light chain targeting vector.
  • Figs. 21A through 21F 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.
  • Figs. 37A-37C depict 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 HC1 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 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. 42 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. 43 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. 44 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. 45 shows formation of a light chain minilocus by homologous recombination between two overlapping fragments which were co-injected.
  • Fig. 46 shows ELISA results for monoclonal antibodies reactive with CEA and non-CEA antigens showing the specificity of antigen binding.
  • Fig. 47 shows the DNA sequences of 10 cDNAs amplified by PCR to amplify transcripts having a human VDJ and a murine constant region sequence.
  • Fig. 48 shows ELISA results for various dilutions of serum obtained from mice bearing both a human heavy chain minilocus transgene and a human ⁇ minilocus transgene; the mouse was immunized with human CD4 and the data shown
  • Fig. 49 shows relative distribution of lymphocytes staining for human ⁇ or mouse ⁇ as determined by FACS for three mouse genotypes.
  • Fig. 50 shows relative distribution of lymphocytes staining for human ⁇ or mouse ⁇ as determined by FACS for three mouse genotypes.
  • Fig. 51 shows relative distribution of lymphocytes staining for mouse ⁇ as determined by FACS for three mouse genotypes.
  • Fig. 52 shows relative distribution of lymphocytes staining for mouse ⁇ or human ⁇ as determined by FACS for four mouse genotypes.
  • Fig. 53 shows the amounts of human ⁇ , human ⁇ , human ⁇ , mouse ⁇ , mouse ⁇ , mouse ⁇ , and mouse ⁇ chains in the serum of unimmunized 0011 mice.
  • Fig. 54 shows a scatter plot showing the amounts of human ⁇ , human ⁇ , human ⁇ , mouse ⁇ , mouse y, mouse ⁇ , and mouse ⁇ chains in the serum of unimmunized 0011 mice of various genotypes.
  • Fig. 55 shows the titres of antibodies comprising human ⁇ , human ⁇ , or human ⁇ chains in anti-CD4 antibodies in the serum taken at three weeks or seven weeks post-immunization following immunization of a 0011 mouse with human CD4.
  • Fig. 56 shows a schematic representation of the human heavy chain minilocus transgenes pHC1 and pHC2, and the light chain minilocus transgenes pKC1, pKC1e, and the light chain minilocus transgene created by homologous recombination between pKC2 and Co4 at the site indicated.
  • Fig. 57 shows a linkage map of the murine lambda light chain locus as taken from Storb et al. (1989) op.cit.; the stippled boxes represent a pseudogene.
  • Fig. 58 shows a schematic representation of
  • Fig. 59 schematically shows the structure of a homologous recombination targeting transgene for deleting genes, such as heavy chain constant region genes.
  • Fig. 60 shows a map of the BALB/c murine heavy chain locus as taken from Immunoglobulin Genes, Honjo, T, Alt, FW, and Rabbits TH (eds.) Academic Press, NY (1989) p. 129.
  • Structural genes are shown by closed boxes in the top line; second and third lines show restriction sites with symbols indicated.
  • Fig. 61 shows a nucleotide sequence of mouse heavy chain locus ⁇ constant region gene.
  • Fig. 62 shows the construction of a frameshift vector (plasmid B) for introducing a two bp frameshift into the murine heavy chain locus J 4 gene.
  • Fig. 63 shows isotype specific response of transgenic animals during hyperimmunization. The relative levels of reactive human ⁇ and ⁇ 1 are indicated by a
  • Fig. 64A and 64B show expression of transgene encoded ⁇ 1 isotype mediated by class switch recombination.
  • FIG. 64A shows a Southern blot of PacI/SfiI digested DNA isolated from three transgene expressing hybridomas. From left to right: clone 92-09A-5H1-5, human ⁇ 1 + / ⁇ -; clone 92-90A-4G2-2, human ⁇ 1 + / ⁇ -; clone 92-09A-4F7-A5-2, human ⁇ 1-, ⁇ + . All three hybridomas are derived from a ⁇ month old male mouse
  • Fig. 64B is a diagram of two possible deletional mechanisms by which a class switch from ⁇ to ⁇ 1 can occur.
  • the human ⁇ gene is flanked by 400 bp direct repeats ( ⁇ and ⁇ ) which can recombine to delete ⁇ . Class switching by this mechanism will always generate a 6.4 kb PacI/SfiI fragment, while class switching by recombination between the ⁇ and the ⁇ 1 switch regions will generate a PacI/Sfil fragment between 4 and ⁇ kb, with size variation between individual switch events.
  • the two ⁇ 1 expressing hybridomas examined in Fig. 64A appear to have undergone recombination between the ⁇ and ⁇ 1 switch regions.
  • Fig. 65 shows chimeric human/mouse immunoglobulin heavy chains generated by trans-switching.
  • cDNA clones of trans-switch products were generated by reverse transcription and PCR amplification of a mixture of spleen and lymph node RNA isolated from a hyperimmunized HC1 transgenic-JHD mouse (#2357; see legend to Fig. 63 for description of animal and immunization schedule).
  • the partial nucleotide sequence of 10 randomly picked clones is shown. Lower case letters indicate germline encoded, capital letters indicate nucleotides that cannot be assigned to known germline sequences; these may be somatic mutations, N nucleotides, or truncated D segments. Both face type indicates mouse ⁇ sequences.
  • Figs. 66A and 66B show that the rearranged VH251 transgene undergoes somatic mutation in a hyperimmunized.
  • sequence is shown at the top; nucleotide changes from germline are given for each clone. A period indicates identity with germline sequence, capital letters indicate no identified germline origin. The sequences are grouped according to J segment usage. The germline sequence of each of the J segments if shown. Lower case letters within CDR3 sequences indicate identity to known D segment included in the HC1 transgene. The assigned D segments are indicated at the end of each sequence. Unassigned sequences could be derived from N region addition or somatic mutation; or in some cases they are simply too short to distinguish random N nucleotides from known D segments.
  • Fig. 66A primary response 13 randomly picked VH251- ⁇ 1 cDNA clones.
  • a 4 week old female HC1 line 26-JHD mouse (#2599) was given a single injection of KLH and complete Freunds adjuvant; spleen cell RNA was isolated 5 days later.
  • the overall frequency of somatic mutations within the V segment is 0.06% (2/3,198 bp).
  • Fig. 66B secondary response 13 randomly picked VH251- ⁇ 1 cDNA clones.
  • a 2 month old female HC1 line 26-JHD mouse (#3204) was given 3 injections of HEL and Freunds adjuvant over one month (a primary injection with complete adjuvant and boosts with incomplete at one week and 3 weeks); spleen and lymph node RNA was isolated 4 months later.
  • the overall frequency of somatic mutations within the V segment is 1.6% (52/3,198 bp).
  • Figs. 67A and 67B show that extensive somatic mutation is confined to ⁇ 1 sequences: somatic mutation and class switching occur within the same population of B cells.
  • Fig. 67A IgM: 23 randomly picked VH251- ⁇ cDNA clones. Nucleotide sequence of 156 bp segment including CDRs 1 and 2 surrounding residues. The overall level of somatic mutation is 0.1% (5/3,744 bp).
  • Fig 67B IgG: 23 randomly picked VH251- ⁇ I cDNA clones. Nucleotide sequence of segment including CDRs 1 through 3 and surrounding
  • Fig. 68 indicates that VH51P1 and VH56P1 show extensive somatic mutation of in an unimmunized mouse.
  • the overall frequency of somatic mutation with the 19 VH56p1 segments is 2.2%
  • Double transgenic mice with disrupted endogenous Ig loci contain human IgM/c positive B cells. FACS of cells isolated from spleens of 4 mice with different genotypes.
  • Left column control mouse (#9944, 6 wk old female JH+/-, JC ⁇ +/- ; heterozygous wild-type mouse heavy and ⁇ -light chain loci, non-transgenic).
  • Second column human heavy chain transgenic (#9877, 6 wk old female JH-/-, JC ⁇ -/-, HC2 line 2550 +; homozygous for disrupted mouse heavy and ⁇ -light chain loci, hemizygous for HC2 transgene).
  • Third column human ⁇ -light chain transgenic (#9878, 6 wk old female JH-/-, JC ⁇ -/-, KCo4 line 4437 +; homozygous for
  • Second row spleen cells stained for expression of human ⁇ heavy chain (x-axis) and human ⁇ light chain (y-axis).
  • Third row spleen cells stained for expression of mouse ⁇ heavy chain (x-axis) and mouse ⁇ light chain (y-axis).
  • Bottom row histogram of spleen cells stained for expression of mouse B220 antigen (log fluorescence: x-axis; cell number: y-axis).
  • Fig. 70 Secreted immunoglobulin levels in the serum of double transgenic mice. Human ⁇ , ⁇ , and ⁇ , and mouse ⁇ and ⁇ from 18 individual HC2/KCo4 double transgenic mice homozygous for endogenous heavy and ⁇ -light chain locus disruption. Mice: (+) HC2 line 2550 (-5 copies of HC2 per integration), KCo4 line 4436 (1-2 copies of KCo4 per
  • HC2 line 2550, KCo4 line 4437 (-10 copies of KCo4 per integration);
  • HC2 line 2550, KCo4 line 4583 (-5 copies of KCo4 per integration);
  • D HC2 line 2572 (30-50 copies of HC2 per integration, KCo4 line 4437;
  • HC2 line 5467 (20-30 copies of HC2 per integration, KCo4 line 4437.
  • Figs. 71A and 71B show human antibody responses to human antigens.
  • Fig. 71A Primary response to recombinant human soluble CD4. Levels of human IgM and human ⁇ light chain are reported for prebleed (0) and post-immunization (•) serum from four double transgenic mice.
  • Fig. 71B Switching to human IgG occurs in vivo . Human IgG (circles) was detected with peroxidase conjugated polyclonal anti-human IgG used in the presence of 1.5 ⁇ /ml excess IgE, ⁇ and 1% normal mouse serum to inhibit non-specific cross-reactivity.
  • Fig. 72 shows FACS analysis of human PBL with a hybridoma supernatant that discriminates human CD4+
  • lymphocytes from human CD8+ lymphocytes from human CD8+ lymphocytes.
  • Fig. 73 shows human ⁇ -CD4 IgM anf IgG in transgenic mouse serum.
  • Fig. 74 shows competition binding experiments comparing a transgenic mouse ⁇ -human CD4 hybridoma monoclonal, 2C11-8, to the RPA-TA and Leu-3A monoclonals.
  • Fig. 75 shows production data for Ig expression of cultured 2C11-8 hybridoma.
  • Table 1 depicts the sequence of vector pGPe.
  • Table 2 depicts the sequence of gene V H 49.8.
  • 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.
  • Table 10 shows the genotypes of several 0011 mice.
  • Table 11 shows transgene V and J segment usage.
  • Table 12 shows the occurrence of somatic mutation in the HC2 heavy chain transgene in transgenic mice.
  • 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
  • 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 copending applications U.S.S.N. 07/990,860, filed 16 December 1992, U.S.S.N. 07/810,279 filed 17 December 1991, U.S.S.N. 07/904,068 filed 23 June 1992; U.S.S.N. 0 ⁇ /853,408, filed 18 March 1992, U.S.S.N. 0 ⁇ /5 ⁇ 4, ⁇ 48 filed August 29, 1990,
  • 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.
  • 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.
  • the invention also provides a method for identifying candidate hybridomas which secrete a monoclonal antibody comprising a human immunoglobulin chain consisting essentially of a human VDJ sequence in polypeptide linkage to a human constant region sequence.
  • candidate hybridomas are identified from a pool of hybridoma clones comprising: (1) hybridoma clones that express immunoglobulin chains consisting essentially of a human VDJ region and a human constant region, and (2) trans-switched hybridomas that express heterohybrid immunoglobulin chains consisting essentially of a human VDJ region and a murine constant region.
  • the supernatant(s) of individual or pooled hybridoma clones is contacted with a predetermined antigen, typically an antigen which is
  • a solid substrate e.g., a microtitre well
  • An antibody that specifically binds to human constant regions is also contacted with the hybridoma supernatant and predetermined antigen under binding conditions so that the antibody selectively binds to at least one human constant region epitope but substantially does not bind to murine constant region epitopes; thus forming complexes consisting essentially of hybridoma supernatant (transgenic monoclonal antibody) bound to a predetermined antigen and to an antibody that specifically binds human constant regions (and which may be labeled with a detectable label or
  • 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 various cells of the immune system, some phagocytic cells and the first component (C1q) of the classical complement system.
  • 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, and generally from a species other than that 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
  • heterohybrid antibody 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., ⁇ , e, 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 ⁇ 10 7 M -1 , and (2) to preferentially bind to the predetermined antigen with an affinity that is at least two-fold greater than its affinity for binding to a non-specific antigen (e.g., BSA, casein) other than the predetermined antigen or a closely-related 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.
  • nucleic acids For nucleic acids, the term "substantial homology" indicates that two nucleic acids, or designated 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 cells, 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 (198 ⁇ ).
  • 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.
  • DNA sequences substantially
  • 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.
  • 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,
  • 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 transgenic 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 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: ⁇ 305- ⁇ 316 (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 or a portion of a D region gene segment in the resultant
  • 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.
  • 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
  • a minilocus comprises a portion of the genomic immunoglobulin locus having at least one internal (i.e., not at a terminus of the portion) deletion of a non-essential DNA portion (e.g., intervening sequence; intron or portion thereof) as compared to the naturally-occurring germline Ig locus.
  • 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 gene (C H ) segments.
  • C H heavy or light constant region gene
  • 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 lgA2.
  • the ⁇ and ⁇ constant region gene segments define the IgD and IgE antibody classes, respectively.
  • 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.
  • 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.
  • a transgene includes the D region gene DHQ52
  • a transgene sequence is recognizable as containing a D region sequence, specifically a DHQ52 sequence.
  • a transgene includes the D region gene DHQ52
  • a transgene-encoded 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 may be recognizable as containing a D region
  • D region segments may be incorporated in VDJ joining to various extents and in various reading frames, a comparison of the D region area of a heavy chain variable region to the D region segments present in the transgene is necessary to determine the incorporation of particular D segments.
  • potential exonuclease digestion during recombination may lead to imprecise V-D and D-J joints during V-D-J recombination.
  • D region sequences may be recognizable but may not correspond identically to a consecutive D region sequence in the transgene.
  • 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
  • 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.
  • 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 ⁇ 0 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
  • CDR1, 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
  • 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 antigen binding domain.
  • 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 is heterologous to that normally encountered in the nonhuman animal used to practice the invention.
  • 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 transgene 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
  • IFN interferon 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
  • 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.
  • 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 ⁇ ).
  • 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 (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))
  • 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 structure: two direct repeat sequences flank each of two clusters of 49-bp tandem repeats. (See M. R.
  • 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 ⁇ 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: in the mouse, IL-4 decreases IgM, IgG2a, IgG2b, and IgG3 expression and increases IgE and IgG1 expression; while IFN ⁇ selectively stimulates IgG2a expression and antagonizes the IL-4-induced increase in IgE and IgG1 expression (Coffraan et al., J. Immunol. 136: 949 (1986) and Snapper et al.,
  • lymphokines actually promote switch recombination.
  • the observed induction of the ⁇ 1 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.
  • a 5' flanking sequence from one switch region can be operably linked to a different switch region for transgene construction (e.g., a 5' flanking sequence from the human S ⁇ 1 switch can be grafted immediately upstream of the S ⁇ 1 switch; a murine S ⁇ 1 flanking region can be grafted adjacent to a human ⁇ 1 switch sequence; or the murine S ⁇ 1 switch can be grafted onto the human ⁇ 1 coding region), 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.
  • Monoclonal antibodies can be obtained by various techniques familiar to those skilled in the art. Briefly, spleen cells from an animal immunized with a desired antigen are immortalized, commonly by fusion with a myeloma cell (see. Kohler and Milstein, Eur. J. Immunol., 6:511-519 (1976)).
  • 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.
  • 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 is estimated to consist of approximately 200 V gene segments (current data supports the existence of about 50-100 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.
  • 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 ⁇ pa 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. 2:727-738 (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 and in copending application, entitled “Transgenic Non-Human Animals Capable of Producing Heterologous Antibodies,” filed August 29, 1990, under U.S.S.N. 07/574,748.
  • 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
  • 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. Homologous recombination of overlapping DNA fragments to form transgenes in vivo is further described in commonly assigned U.S. Patent Application entitled "Intracellular Generation of DNA by
  • 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 typically capable 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 homologous 9 kb subregions, linked in tandem (Siebenlist, et al . ( 1981) ,
  • 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 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 in copending applications, U.S.S.N. 07/574,748 filed August 29, 1990, PCT/US91/06185 filed August 28, 1991, and U.S.S.N.
  • V segments, D segments, J segments, and C genes can be isolated as described in copending applications U.S.S.N. 07/574,748 filed August 29, 1990 and PCT/US91/06185 filed August 28, 1991.
  • 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 ⁇ 5 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 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.
  • 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 , ⁇ ).
  • 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.
  • 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.
  • 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 y2 . S y3 , S ⁇ 4 , S ⁇ , S a2 . 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 , a 1 , ⁇ 2 , or e 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
  • Trans-switching to endogenous nonhuman heavy chain constant region genes can occur and produce chimeric heavy chains and antibodies comprising such chimeric human/mouse heavy chains.
  • Such chimeric antibodies may be desired for certain uses described herein or may be undesirable.
  • 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.
  • targeting vectors are used based on the design employed by Jaenisch and co-workers (Zijlstra, et al. (1989), Nature, 342, 435-438) for the successful disruption of the mouse 02-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 seguence 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 KpnI 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 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 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
  • 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 and thereby 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. Natl. 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 (e.g., murine) 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 are selected so as to react specifically with one or more endogenous (e.g., murine) Ig chains but to have minimal or no cross-reactivity with heterologous Ig chains encoded by an Ig transgene of the invention.
  • Suitable antibody sources for antibody comprise:
  • monoclonal antibodies such as a monoclonal antibody that specifically binds to a murine ⁇ , ⁇ , ⁇ , or ⁇ chains but does not react with the human immunoglobulin chain (s) encoded by a human Ig transgene of the invention;
  • polyclonal antiserum or mixtures thereof typically such antiserum/antisera is monospecific for binding to a single species of endogenous Ig chain (e.g., murine ⁇ , murine ⁇ , murine ⁇ , murine ⁇ ) or to multiple species of endogenous Ig chain, and most preferably such antisera
  • polyclonal antibodies are preferred, and such substantially monospecific polyclonal antibodies can be advantageously produced from an antiserum raised against human immunoglobulin (s) by pre-adsorption with antibodies derived from the nonhuman animal species (e.g., murine) and/or, for example, by affinity chromatography of the antiserum or purified fraction thereof on an affinity resin containing immobilized human Ig (wherein the bound fraction is enriched for the desired anti-human Ig in the antiserum; the bound fraction is typically eluted with conditions of low pH or a chaotropic salt solution).
  • Cell separation and/or complement fixation can be employed to provide the enhancement of antibody-directed cell depletion of lymphocytes expressing endogenous (e.g., murine) immunoglobulin chains.
  • endogenous (e.g., murine) immunoglobulin chains e.g., murine
  • antibodies are employed for ex vivo depletion of murine Ig-expressing explanted hematopoietic cells and/or B-lineage lymphocytes obtained from a transgenic mouse harboring a human Ig transgene.
  • hematopoietic cells and/or B-lineage lymphocytes are explanted from a transgenic nonhuman animal harboring a human Ig transgene (preferably harboring both a human heavy chain transgene and a human light chain transgene) and the explanted cells are incubated with an antibody (or antibodies) which (1) binds to an endogenous immunoglobulin (e.g., murine ⁇ and/or ⁇ ) and (2) lacks substantial binding to human immunoglobulin chains encoded by the transgene(s).
  • an antibody or antibodies
  • Such antibodies are referred to as "suppression antibodies” for clarity.
  • the explanted cell population is selectively
  • suppression antibody e.g., by ADCC,.by complement fixation, or by a toxin linked to the suppression antibody
  • clonal anergy induced by the suppression antibody and the like.
  • antibodies used for antibody suppression of endogenous Ig chain production will be capable of fixing complement. It is frequently preferable that such antibodies may be selected so as to react well with a convenient
  • complement source for ex vivo/in vitro depletion such as rabbit or guinea pig complement.
  • the suppressor antibodies possess effector functions in the nonhuman transgenic animal species; thus, a suppression antibody comprising murine effector functions (e.g., ADCC and complement fixation) generally would be preferred for use in transgenic mice.
  • a suppression antibody that specifically binds to a predetermined endogenous
  • immunoglobulin chain is used for ex vivo/in vitro depletion of lymphocytes expressing an endogenous immunoglobulin.
  • a cellular explant e.g., lymphocyte sample
  • a suppression antibody e.g., by immobilization, complement fixation, and the like
  • cells specifically binding to the suppression antibody are depleted (e.g., by immobilization, complement fixation, and the like), thus generating a cell subpopulation depleted in cells expressing endogenous (nonhuman) immunoglobulins (e.g., lymphocytes expressing murine Ig).
  • the resultant depleted lymphocyte population (T cells, human Ig-positive B-cells, etc.) can be transferred into a immunocompatible (i.e., MHC-compatible) nonhuman animal of the same species and which is substantially incapable of producing endogenous antibody (e.g., SCID mice, RAG-1 or RAG-2 knockout mice).
  • a immunocompatible nonhuman animal of the same species e.g., SCID mice, RAG-1 or RAG-2 knockout mice.
  • endogenous antibody e.g., SCID mice, RAG-1 or RAG-2 knockout mice.
  • B-cells producing such antibodies.
  • B-cells may be used to generate hybridomas by
  • Antibody suppression can be used in combination with other endogenous Ig
  • inactivation/suppression methods e.g., J H knockout, C H knockout, D-region ablation, antisense suppression,
  • VDJ human variable region
  • endogenous heavy chain locus by any of various methods, including but not limited to the following: (1) functionally disrupting and/or deleting by homologous recombination at least one and preferably all of the endogenous heavy chain constant region genes, (2) mutating at least one and
  • endogenous heavy chain constant region genes to encode a termination codon (or frameshift) to produce a truncated or frameshifted product (if trans-switched), and other methods and strategies apparent to those of skill in the art.
  • Deletion of a substantial portion or all of the heavy chain constant region genes and/or D-region genes may be accomplished by various methods, including sequential deletion by homologous recombination targeting vectors, especially of the "hit-and-run” type and the like.
  • functional disruption and/or deletion of at least one endogenous light chain locus e.g., ⁇
  • constant region genes is often preferable.
  • the heterologous transgene comprises a frameshift in the J segment(s) and a compensating frameshift (i.e., to regenerate the original reading frame) in the initial region (i.e., amino-terminal coding portion) of one or more (preferably all) of the transgene constant region genes.
  • Antisense suppression and antibody suppression may also be used to effect a substantially complete functional inactivation of endogenous Ig gene product expression (e.g., murine heavy and light chain sequences) and/or trans-switched antibodies (e.g., human variable/murine constant chimeric antibodies).
  • endogenous Ig gene product expression e.g., murine heavy and light chain sequences
  • trans-switched antibodies e.g., human variable/murine constant chimeric antibodies
  • suppression strategies may be used to effect essentially total suppression of endogenous (e.g., murine) Ig chain expression.
  • trans-switched immunoglobulin it may be desirable to produce a trans-switched immunoglobulin.
  • trans-switched heavy chains can be chimeric (i.e., a non-murine (human) variable region and a murine constant region).
  • Antibodies comprising such chimeric trans-switched
  • immunoglobulins can be used for a variety of applications where it is desirable to have a non-human (e.g., murine) constant region (e.g., for retention of effector functions in the host, for the presence of murine immunological
  • a non-human (e.g., murine) constant region e.g., for retention of effector functions in the host, for the presence of murine immunological
  • a human variable region repertoire may possess advantages as compared to the murine variable region repertoire with respect to certain antigens.
  • the human V H , D, J H , V L , and J L genes have been selected for during evolution for their ability to encode immunoglobulins that bind certain
  • antigens which provided evolutionary selective pressure for the murine repertoire can be distinct from those antigens which provided evolutionary pressure to shape the human repertoire.
  • Other repertoire advantages may exist, making the human variable region repertoire advantageous when combined with a murine constant region (e.g., a trans-switched murine) isotype.
  • a murine constant region e.g., a trans-switched murine
  • the presence of a murine constant region can afford advantages over a human constant region.
  • a murine y constant region linked to a human variable region by trans-switching may provide an antibody which possesses murine effector functions (e.g., ADCC, murine complement fixation) so that such a chimeric antibody (preferably monoclonal) which is reactive with a predetermined antigen (e.g., human IL-2 receptor) may be tested in a mouse disease model, such as a mouse model of graft-versus-host disease wherein the T lymphocytes in the mouse express a functional human IL-2 receptor.
  • murine effector functions e.g., ADCC, murine complement fixation
  • a chimeric antibody preferably monoclonal
  • a predetermined antigen e.g., human IL-2 receptor
  • the human variable region encoding sequence may be isolated (e.g., by PCR amplification or cDNA cloning from the source (hybridoma clone)) and spliced to a sequence encoding a desired human constant region to encode a human sequence antibody more suitable for human therapeutic uses where immunogenicity is preferably minimized.
  • the polynucleotide(s) having the resultant fully human encoding sequence(s) can be expressed in a host cell (e.g., from an expression vector in a mammalian cell) and purified for pharmaceutical formulation.
  • the chimeric antibodies may be used directly without replacing the murine constant region with a human constant region.
  • Other variations and uses of trans-switched chimeric antibodies will be evident to those of skill in the art.
  • the present invention provides transgenic nonhuman animals containing B lymphocytes which express chimeric antibodies, generally resulting from trans-switching between a human heavy chain transgene and an endogenous murine heavy chain constant region gene.
  • Such chimeric antibodies comprise a human sequence variable region and a murine constant region, generally a murine switched (i.e., non- ⁇ , non- ⁇ ) isotype.
  • the transgenic nonhuman animals capable of making chimeric
  • antibodies to a predetermined antigen are usually also.
  • the animal is homozygous for a functionally disrupted heavy chain locus and/or light chain locus but retains one or more endogenous heavy chain constant region gene(s) capable of trans-switching (e.g., ⁇ , ⁇ , e) and
  • Such a mouse is immunized with a predetermined antigen, usually in combination with an adjuvant, and an immune response comprising a
  • the serum of such an immunized animal can comprise such chimeric antibodies at concentrations of about at least 1 ⁇ g/ml, often about at least 10 ⁇ g/ml, frequently at least 30 ⁇ g/ml, and up to 50 to 100 ⁇ g/ml or more.
  • antibodies comprising chimeric human variable/mouse constant region heavy chains typically also comprises antibodies which comprise human variable/human constant region (complete human sequence) heavy chains.
  • Chimeric trans-switched antibodies usually comprise (1) a chimeric heavy chain composed of a human variable region and a murine constant region (typically a murine gamma) and (2) a human transgene-encoded light chain (typically kappa) or a murine light chain (typically lambda in a kappa knockout background).
  • Such chimeric trans-switched antibodies generally bind to a predetermined antigen (e.g., the immunogen) with an affinity of about at least 1 ⁇ 10 7 M -1 , preferably with an affinity of about at least 5 ⁇ 10 7 M -1 , more preferably with an affinity of at least 1 ⁇ 10 8 M -1 to 1 ⁇ 10 9 M -1 or more.
  • a predetermined antigen e.g., the immunogen
  • the predetermined antigen is a human protein, such as for example a human cell surface antigen (e.g., CD4, CD8, IL-2 receptor, EGF receptor, PDGF receptor), other human biological macromolecule (e.g., thrombomodulin, protein C, carbohydrate antigen, sialyl Lewis antigen, L-selectin), or nonhuman disease associated macromolecule (e.g., bacterial LPS, virion capsid protein or envelope glycoprotein) and the like.
  • a human cell surface antigen e.g., CD4, CD8, IL-2 receptor, EGF receptor, PDGF receptor
  • other human biological macromolecule e.g., thrombomodulin, protein C, carbohydrate antigen, sialyl Lewis antigen, L-selectin
  • nonhuman disease associated macromolecule e.g., bacterial LPS, virion capsid protein or envelope glycoprotein
  • the invention provides transgenic nonhuman animals comprising a genome comprising: (1) a homozygous functionally disrupted endogenous heavy chain locus comprising at least one murine constant region gene capable of trans-switching (e.g., in cis linkage to a functional switch recombination sequence and typically to a functional enhancer), (2) a human heavy chain transgene capable of rearranging to encode end express a functional human heavy chain variable region and capable of trans-switching (e.g., having a cis-linked RSS); optionally further comprising (3) a human light chain (e.g., kappa) transgene capable of rearranging to encode a functional human light chain variable region and expressing a human sequence light chain; optionally further comprising (4) a homozygous functionally disrupted endogenous light chain locus ( ⁇ , preferably ⁇ and ⁇ ); and optionally further comprising (5) a serum comprising an antibody comprising a chimeric heavy chain composed of a human sequence variable region
  • Such transgenic mice may further comprise a serum comprising chimeric antibodies which bind a predetermined human antigen (e.g., CD4, CD8, CEA) with an affinity of about at least 1 ⁇ 10 4 M -1 , preferably with an affinity of about at least 5 ⁇ 10 4 M -1 , more preferably with an affinity of at least 1 ⁇ 10 7 M -1 to 1 ⁇ 10 9 M -1 or more.
  • a predetermined human antigen e.g., CD4, CD8, CEA
  • hybridomas can be made wherein the monoclonal antibodies produced thereby have an affinity of at least 8 ⁇ 10 7 M -1 .
  • Chimeric antibodies comprising a heavy chain composed of a murine constant region and a .human variable region, often capable of binding to a nonhuman antigen, may also be present in the serum or as an antibody secreted from a hybridoma.
  • such chimeric antibodies can be generated by trans-switching, wherein a human transgene encoding a human variable region (encoded by productive V-D-J rearrangement in vivo) and a human constant region, typically human ⁇ ,
  • immunoglobulin constant gene switch sequence thereby operably linking the transgene-encoded human variable region with a heavy chain constant region which is not encoded by said transgene, typically an endogenous murine immunoglobulin heavy chain constant region or a heterologous (e.g., human) heavy chain constant region encoded on a second transgene.
  • cis-switching refers to isotype-switching by
  • trans-switching involves recombination between a transgene RSS and an RSS element outside the transgene, often on a different chromosome than the chromosome which harbors the transgene.
  • Trans-switching generally occurs between an RSS of an expressed transgene heavy chain constant region gene and either an RSS of an endogenous murine constant region gene (of a non- ⁇ isotype, typically ⁇ ) or an RSS of a human constant region gene contained on a second transgene, often integrated on a separate chromosome.
  • a non-chimeric antibody having a substantially fully human sequence is produced.
  • a polynucleotide encoding a human heavy chain constant region (e.g., ⁇ 1) and an operably linked RSS (e.g., a ⁇ 1 RSS) can be introduced (e.g., transfected) into a population of hybridoma cells generated from a
  • transgenic mouse B-cell (or B cell population) expressing an antibody comprising a transgene-encoded human ⁇ chain.
  • the resultant hybridoma cells can be selected for the presence of the introduced polynucleotide and/or for the expression of trans-switched antibody comprising a heavy chain having the variable region (idiotype/antigen reactivity) of the human ⁇ chain and having the constant region encoded by the introduced polynucleotide sequence (human ⁇ 1).
  • Trans-switch comprising a heavy chain having the variable region (idiotype/antigen reactivity) of the human ⁇ chain and having the constant region encoded by the introduced polynucleotide sequence (human ⁇ 1).
  • the invention also provides a method for producing such chimeric trans-switched antibodies comprising the step of immunizing with a predetermined antigen a transgenic mouse comprising a genome comprising: (1) a homozygous functionally disrupted endogenous heavy chain locus comprising at least one murine constant region gene capable of trans-switching (e.g., ⁇ 2a, ⁇ 2b, ⁇ 1, ⁇ 3), (2) a human heavy chain transgene capable of rearranging to encode a functional human heavy chain variable region and expressing a human sequence heavy chain and capable of undergoing isotype switching (and/or trans-switching), and optionally further comprising (3) a human light chain (e.g., kappa) transgene capable of rearranging to encode a functional human light (e.g., kappa) chain variable region and expressing a human sequence light chain, and optionally further comprising (4) a homozygous functionally disrupted endogenous light chain locus (typically ⁇ ,
  • a serum comprising an antibody comprising a chimeric heavy chain composed of a human sequence variable region encoded by a human transgene and a murine constant region sequence encoded by an endogenous murine heavy chain constant region gene (e.g., ⁇ 1, ⁇ 2a, ⁇ 2b, ⁇ 3).
  • trans-switching and cis-switching is associated with the process of somatic mutation.
  • Somatic mutation expands the range of antibody affinities encoded by clonal progeny of a B-cell.
  • hybridoma cell population which expresses a first antibody comprising a heavy chain comprising a first human heavy chain variable region in polypeptide linkage to a first human heavy chain constant region (e.g., ⁇ )
  • a hybridoma cell clonal variant which express an antibody comprising a heavy chain containing said first human heavy chain variable region in polypeptide linkage to a second heavy chain constant region (e.g., a human ⁇ , ⁇ , or e constant region).
  • a hybridoma cell population which expresses a first antibody comprising a heavy chain comprising a first human heavy chain variable region in polypeptide linkage to a first human heavy chain constant region (e.g., ⁇ )
  • hybridoma cell clonal variants which express an antibody comprising a heavy chain containing said first human heavy chain variable region in polypeptide linkage to a second heavy chain constant region (e.g., a human ⁇ , ⁇ , or e constant region).
  • trans- or cis- as through the administration of agents that promote isotype switching, such as T-cell-derived lymphokines (e.g., IL-4 and IFN ⁇ ), by introduction of a polynucleotide comprising a functional RSS and a heterologous (e.g. human) heavy chain constant region gene to serve as a substrate for trans-switching, or by a combination of the above, and the like.
  • agents that promote isotype switching such as T-cell-derived lymphokines (e.g., IL-4 and IFN ⁇ )
  • a polynucleotide comprising a functional RSS and a heterologous (e.g. human) heavy chain constant region gene to serve as a substrate for trans-switching, or by a combination of the above, and the like.
  • Class switching and affinity maturation take place within the same population of B cells derived from transgenic animals of the present invention. Therefore, identification of class-switched B cells (or hybridomas derived therefrom) can be used as a screening step for obtaining high affinity monoclonal antibodies.
  • a variety of approaches can be
  • a single continuous human genomic fragment comprising both ⁇ and ⁇ constant region genes with the associated RSS elements and switch regulatory elements (e.g., sterile transcript promoter) can be used as a transgene.
  • some portions of the desired single contiguous human genomic fragment can be difficult to clone efficiently, such as due to instability problems when replicated in a cloning host or the like; in particular, the region between ⁇ and ⁇ 3 can prove difficult to clone efficiently, especially as a contiguous fragment comprising the ⁇ gene, ⁇ 3 gene, a V gene, D gene segments, and J gene segments.
  • a discontinuous human transgene composed of a human ⁇ gene, human ⁇ 3 gene, a human V gene(s), human D gene segments, and human J gene segments, with one or more deletions of an intervening (intronic) or otherwise nonessential sequence (e.g., one or more V, D, and/or J segment and/or one or more non- ⁇ constant region gene(s)).
  • minigenes have several advantages as compared to isolating a single contiguous segment of genomic DNA spanning all of the essential elements for efficient
  • Such a minigene avoids the necessity of isolating large pieces of DNA which may contain sequences which are difficult to clone
  • miniloci comprising elements necessary for isotype switching (e.g., human ⁇ sterile transcript promoter) for producing cis- or trans-switching, can advantageously undergo somatic mutation and class switching in vivo.
  • isotype switching e.g., human ⁇ sterile transcript promoter
  • hybridoma clones producing antibodies having high binding affinity are obtained by selecting, from a pool of hybridoma cells derived from B cells of transgenic mice harboring a human heavy chain transgene capable of isotype switching (see, supra) and substantially lacking endogenous murine heavy chain loci capable of undergoing productive (in-frame) V-D-J rearrangement, hybridomas which express an antibody comprising a heavy chain comprising a human sequence heavy chain variable region in polypeptide linkage to a human (or mouse) non- ⁇ heavy chain constant region; said antibodys are termed "switched antibodies” as they comprise a "switched heavy chain” which is produced as a consequence of cis-switching and/or trans-switching in vivo or in cell culture.
  • Hybridomas producing switched antibodies generally have undergone
  • hybridomas secreting a human sequence antibody having substantial binding affinity greater than 1 ⁇ 10 7 M -1 to 1 ⁇ 10 8 M -1 ) for a predetermined antigen and wherein said human sequence antibody comprises human immunoglobulin
  • variable region (s) can be selected by a method comprising a two-step process. One step is to identify and isolate
  • hybridoma cells which secrete immunoglobulins which comprise a switched heavy chain e.g., by binding hybridoma cells to an immobilized immunoglobulin which specifically binds a switched heavy chain and does not substantially bind to an unswitched isotype, e.g., ⁇ ).
  • the other step is to identify hybridoma cells which bind to the predetermined antigen with substantial binding affinity (e.g., by ELISA of hybridoma clone
  • hybridoma cells which bind predetermined antigen which bind predetermined antigen.
  • Hybridoma cells which express switched antibodies that have substantial binding affinity for the predetermined antigen are isolated and cultured under suitable growth conditions known in the art, typically as individual selected clones.
  • the method comprises the step of culturing said selected clones under conditions suitable for expression of monocloanl antibodies; said monoclonal antibodies are collected and can be administered for therapeutic, prophylactic, and/or
  • the selected hybridoma clones can serve as a source of DNA or RNA for isolating immunoglobulin sequences which encode immunoglobulins (e.g. a variable region) that bind to (or confer binding to) the predetermined antigen.
  • immunoglobulins e.g. a variable region
  • the human variable region encoding sequence may be isolated (e.g., by PCR amplification or cDNA cloning from the source (hybridoma clone)) and spliced to a sequence encoding a desired human constant region to encode a human sequence antibody more suitable for human therapeutic uses where immunogenicity is preferably minimized.
  • polynucleotide (s) having the resultant fully human encoding sequence(s) can be expressed in a host cell (e.g., from an expression vector in a mammalian cell) and purified for pharmaceutical formulation.
  • a heterologous transgene capable of encoding a human immunoglobulin advantageously comprises a cis-linked enhancer which is not derived from the mouse genome, and/or which is not naturally associated in cis with the exons of the heterologous transgene.
  • a human ⁇ transgene e.g., a ⁇ minilocus
  • a human V ⁇ gene can advantageously comprise a human V ⁇ gene, a human J ⁇ gene, a human C ⁇ gene, and a xenoenhancer
  • said xenoenhancer comprises a human heavy chain intronic enhancer and/or a murine heavy chain intronic enhancer, typically located between a J ⁇ gene and the C ⁇ gene, or located downstream of the C ⁇ gene.
  • the mouse heavy chain J- ⁇ intronic enhancer (Banerji et al. (1983) Cell 33: 729) can be isolated on a 0.9 kb Xbal fragment of the plasmid pKVe2 (see, infra).
  • the human heavy chain J- ⁇ intronic enhancer (Hayday et al. (1984) Nature 307: 334) can be isolated as a 1.4 kb Mlul/HindIII fragment (see, infra).
  • Addition of a transcriptionally active xenoenhancer to a transgene such as a combined xenoenhancer consisting essentially of a human J- ⁇ intronic enhancer linked in cis to a mouse J- ⁇ intronic enhancer, can confer high levels of expression of the transgene, especially where said transgene encodes a light chain, such as human ⁇ .
  • a rat 3' enhancer can be advantageously included in a minilocus construct capable of encoding a human heavy chain.
  • 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.
  • a heavy chain transgene having a predetermined repertoire may comprise, for example, human V H genes which are preferentially used in antibody responses to the predetermined antigen type in humans.
  • some V H genes may be excluded from a defined repertoire for various reasons (e.g., have a low likelihood of encoding high affinity V regions for the predetermined antigen; have a low propensity to undergo somatic mutation and affinity sharpening; or are immunogenic to certain humans).
  • 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 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. Transgenic animals containing rearranged heavy and rearranged light immunoglobulin transgenes.
  • transgenic animal 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 ⁇ 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.
  • mice are derived according to Hogan, et al., "Manipulating the Mouse Embryo: A Laboratory Manual",
  • 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
  • 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.
  • Nuclei are isolated from fresh human placental tissue as described by Marzluff et al., "Transcription and Translation: A Practical Approach", B.D. Hammes and
  • 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'.
  • 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.
  • the 450 kb XhoI to NotI fragment plus the approximately 400 kb Mlul to BssHII fragment have sequence overlap defined by the BssHII and XhoI 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).
  • 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.
  • 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 (SfiI) in the polylinker.
  • pGP1 is digested with Mlul and Spel to cut the recognition sequences in the polylinker portion of the plasmid.
  • a 3' pGP2 is identical to pGP1 except that it contains an additional Sfi I site located between the Mlul and SpeI sites. This allows inserts to be completely excised with SfiI as well as with NotI.
  • rat IGH 3' enhancer sequence is PCR amplified by using the following oligonucleotides:
  • pGP1 is digested with BamHI and BglII 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. D. Cloning of Cu Region
  • pGP1 is digested with BamHI and HindIII 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/HindIII cut pGP1.
  • Proper orientation of fragment (c) is checked by HindIII digestion to form pCON1 containing a 12 kb insert encoding the C ⁇ region.
  • pHIG1 contains J segments, switch and ⁇ sequences in its 18 kb insert with an SfiI 3' site and a Spel 5' site in a polylinker flanked by NotI sites, will be used for rearranged VDJ segments.
  • pCONl is identical except that it lacks the J region and contains only a 12 kb insert. The use of pCONl 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 using the following oligonucleotide which is specific for the third exon of ⁇ -I (CH3).
  • a 7.7 kb HindIII to BglII fragment (fragment (a) in Fig. 11) is cloned into HindIII/BglII cut pRE3 to form pREG1.
  • the upstream 5.3 kb HindIII fragment (fragment (b) in Fig. 11) is cloned into HindIII digested pREG1 to form pREG2. Correct orientation is confirmed by BamHI/Spel digestion.
  • the previously described plasmid pHIG1 contains human J segments and the C ⁇ constant region exons.
  • pHIG1 was digested with SfiI (Fig. 10).
  • the plasmid pREG2 was also digested with SfiI to produce a 13.5 kb insert containing human C ⁇ 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 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 pCONl with SfiI and combining that with the SfiI fragment containing the human C ⁇ region and the rat 3' enhancer by digesting pREG2 with SfiI.
  • 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
  • AAC CAC AGT GTC - 3' DXP4 5' - GCC TGA AAT GGA GCC TCA GGG CAC AGT GGG
  • DN4 5' - GCA GGG AGG ACA TGT TTA GGA TCT GAG GCC GCA CCT GAC ACC - 3'
  • a 5.2 kb XhoI 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' SfiI 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.
  • pVH1 is digested with SfiI and the resultant
  • 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.
  • 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 SpeI 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.
  • the resultant is plasmid designated pHIG5'0 (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.
  • pHIG5'0 overlap
  • 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 SfiI site to generate pHIGS' 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'.
  • pEu1 The construction of pEu1 is depicted in Fig. 16.
  • the mouse heavy chain enhancer is isolated on the Xbal to
  • This E ⁇ fragment is cloned into EcoRV/Xbal digested pGP1 by blunt end filling in EcoRI site.
  • the resultant plasmid is designated pEmul.
  • 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:
  • the 16 kb fraction is isolated from the SmaI digested gel and the 11 kb region is similarly isolated from the gel containing DNA digested with BamHI.
  • the 16 kb SmaI fraction is cloned into Lambda FIX II (Stratagene, La Jolla, California) which has been digested with XhoI, treated with klenow fragment DNA polymerase to fill in the XhoI restriction digest product. Ligation of the 16 kb SmaI fraction destroys the SmaI sites and lases XhoI sites intact.
  • 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 SmaI fragment fragment (b) in Fig. 20) is subcloned and subsequently inserted into pKapl digested with SmaI.
  • 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 digested pGP1 such that the 3' end is toward the SfiI site.
  • the resultant plasmid is designated pKAPint.
  • One or more 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 SmaI 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 ⁇ .
  • Immunoglobulin ⁇ Light Chain Genes 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. For example, 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 synthesis of oligo dT primed cDNA using the enzyme reverse transcriptase (for general methods see. Goodspeed et al.
  • the single stranded cDNA is then isolated and G residues are added to the 3' end using the enzyme
  • 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:
  • 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 GGG AAG ATG AAG ACA GAT GGT GCA
  • unique sequences that can be used as a primer for the PCR amplification of the newly synthesized DNA strand
  • AAG AAG
  • the sequence is amplified by PCR using the following two oligonucleotide primers:
  • 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 unique restriction endonuclease site is identified upstream of the rearranged V segment.
  • DNA from the Ig expressing cell line is then cut with SmaI and second enzyme (or BamHI or Kpnl if there is SmaI 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 XhoI depending on what site does not exist in fragment) and cut with the corresponding linker enzyme to give DNA fragments with BamHI, EcoRI or XhoI ends.
  • SmaI and second enzyme or BamHI or Kpnl if there is SmaI 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 XhoI depending on what site does not exist in fragment) and cut with the corresponding linker enzyme to give DNA fragments with BamHI, EcoRI or XhoI ends.
  • 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: 5' - GAG GTA CAC TGA CAT ACT GGC ATG - 3'
  • PCR amplified sequence is then purified by gel electrophoresis and used as template for dideoxy sequencing reactions using the following oligonucleotide as a primer:
  • 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.
  • 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 pGPI.
  • V segment containing clones are isolated using the unique probe o-mu, and the insert is subcloned into MluI or MluI/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 ClaI/XhoI fragment containing J ⁇ 1 together with a 13 kb XhoI 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
  • a human genomic DNA phage library was 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 VkIII 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 XhoI/SalI fragment containing V ⁇ 65.8 into the 5' XhoI 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 ⁇ 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
  • mice from KC1 injected embryos 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. All 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,
  • the kappa light chain minilocus transgene pKC2 was generated by inserting an 8 kb XhoI/SalI fragment containing V ⁇ 65.5 into the 5' XhoI 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 pKC1 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- ⁇ intronic enhancer (Banerji et al., Cell 33:729-740 (1983)) together with a 1.4 kb MluI/HindIII fragment containing the human heavy chain J- ⁇ 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);
  • 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 XhoI 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 SalI 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 XhoI site. These two oligonucleotides amplify a 0.7 kb DNA
  • the PCR amplified DNA was digested with NcoI and XhoI 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 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 ⁇ 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)).
  • This example describes the inactivation of the mouse endogenous kappa locus by homologous recombination in
  • ES embryonic stem
  • blastocysts early mouse embryos
  • 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 (XbaI/TaqI fragment; Adra et al. (1987) Gene 60: 65) in the cloning vector pGEM-7Zf(+).
  • the plasmid also includes a heterologous
  • 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 ⁇ locus: 5'- GGC TGA TGC TGC ACC AAC TGT ATC CAT CTT CCC ACC ATC CAG -3' and for the J ⁇ 5 gene segment: 5'- CTC ACG TTC GGT GCT GGG ACC AAG CTG GAG CTG AAA CGT AAG -3 ' .
  • a 1.2 kb EcoRI/SphI fragment extending 5' of the J ⁇ region was also isolated from a positive phage clone.
  • An SphI/Xbal/BglII/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 sequence was modified to a BamHI site and the TK cassette was then excised as a BamHI/HindIII fragment and subcloned into pGP1b to generate pGP1b-TK.
  • This plasmid was linearized at the XhoI site and the XhoI fragment from pNEO-K5'3', containing the neo gene flanked by genomic sequences from 5' of J ⁇ 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 K1 is shown in Fig. 20e. Generation and analysis of ES cells with targeted inactivation of a kappa allele
  • 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. Embryol. 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 K1 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 ⁇ 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
  • displayed the 1.8 kb Xbal band indicative of a homologous recombination into one of the kappa genes.
  • These ⁇ clones were further digested with the enzymes BglII, Sacl, and Pstl to verify that the vector integrated homologously into one of the kappa genes.
  • 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.
  • 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 seguences from which the J H region has been deleted and replaced by the gene for the selectable marker neo.
  • the neomycin resistance gene (neo) used for drug selection of transfected ES cells, was derived from a repaired version of the plasmid pGEM7 (KJ1).
  • KJ1 neomycin resistance gene
  • restriction fragment encompassing the mutation with the corresponding sequence from a wild-type neo clone.
  • HindIII site in the prepared pGEM7 (KJ1) was converted to a SalI site by addition of a synthetic adaptor, and the neo expression cassette excised by digestion with Xbal/SalI. The ends of the neo fragment were then blunted by treatment with the Klenow form of DNA polI, and the neo fragment was
  • 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'
  • pGMT mouse immunoglobulin heavy chain targeting
  • 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 HindIII. The TK DNA fragment was subcloned between the EcoRI and HindIII sites of pGMT, creating the plasmid pGMT-TK (Fig. 21c).
  • a 5.9 kb genomic Xbal/XhoI fragment situated 5' of the J H region, was derived from a positive genomic phage clone by limit digestion of the DNA with XhoI, and partial digestion with Xbal.
  • this XbaI 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 XhoI site of
  • J H KO1 (Fig. 21e) 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. 2If 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 ⁇ 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 Stul and probed with the 500 bp EcoRI/StuI fragment designated as probe A in Fig. 2If. This probe detects a Stul 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).
  • 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 matings: wild-type mice bearing two copies of the normal J H locus, heterozygotes carrying 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
  • the plasmid pBR322 was digested with EcoRI and StyI 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
  • 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
  • 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, NcoI, ClaI, EcoRV, Xbal, Sacl, XhoI, SphI, Pstl, BglII, EcoRI, SmaI, KpnI, HindIII, and NotI).
  • the resulting plasmid, pRE3 was digested with BamHI and HindIII, and the insert containing the rat Ig heavy chain 3' enhancer cloned into BamHI/HindIII 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 4 kb XhoI 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 ⁇ -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 XhoI cut, Klenow treated, pJM1.
  • the resulting plasmid, pJM2 (Fig.
  • 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 XhoI fragment, that includes the D elements D ⁇ 1 , D N1 , and D M2 (Ichihara et al., EMBO J. 7:4141 (1988)), was isolated from phage clone ⁇ 4.1.
  • pDH1 contains a 10.6 kb insert that includes at least ⁇ D segments and can be excised with XhoI (5') and EcoRV (3'). 4.
  • 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 ClaI and the insert isolated. This insert was ligated to the XhoI/EcoRV insert of pDH1 and XhoI/ClaI digested pGPe to generate pCOR1 (Fig. 24).
  • plasmid pCOR1 was partially digested with XhoI and the isolated XhoI/SalI insert of pVH251 cloned into the upstream XhoI site to generate the plasmid pIGM1 (Fig. 25).
  • 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 HindIII 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.
  • dideoxy sequencing reactions were carried 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 ⁇ 29.5 and ⁇ S ⁇ 1.13 were both determined to be of the ⁇ 1 subclass.
  • a 7.8 kb HindIII fragment of phage clone ⁇ 29.5, containing the ⁇ 1 coding region was cloned into pUC18.
  • the resulting plasmid, pLT1 was digested with XhoI, Klenow treated, and religated to destroy the internal XhoI site.
  • the resulting clone, pLT1xk was digested with HindIII and the insert isolated and cloned into pSP72 to generate the plasmid clone pLTlxks.
  • p ⁇ el contains all of the ⁇ 1 constant region coding exons, together with 5 kb of downstream sequences, linked to the rat heavy chain 3' enhancer.
  • a 5.3 kb HindIII fragment containing the ⁇ 1 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 ⁇ S ⁇ 1.13 and cloned into pSP72 with the polylinker XhoI site adjacent to the 5' end of the insert, to generate the plasmid clone PS ⁇ 1S.
  • the XhoI/SalI insert of PS ⁇ 1S was cloned into XhoI digested P ⁇ el to generate the plasmid clone P ⁇ e2 (Fig. 26).
  • P ⁇ e2 contains all of the ⁇ 1 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 XhoI site at the 5' end of the insert. The entire insert, together with the XhoI site and the 3' rat enhancer can be excised from vector sequences by digestion with NotI. 4 .
  • pHC1 contains 2
  • 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.
  • Phage clone ⁇ 49.8 was isolated and a 6.1 kb Xbal fragment containing the variable segment VH49.8 subcloned into pNN03 (such that the polylinker ClaI site is downstream of VH49.8 and the polylinker XhoI 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).
  • 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 SmaI and HindIII, and treated with the Klenow fragment of polymerase I. The blunt ended fragment was then cloned into ClaI 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 SalI site at the 3' end of the insert and a unique XhoI 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 HindIII/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 SmaI and
  • the ligation product was digested with SalI and ligated to SalI 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 SalI and complete digestion with XhoI, followed by purification of the 15 kb fragment on an agarose gel. The insert is then cloned into the XhoI site of P ⁇ e2 to generate the plasmid clone.
  • pVGE1 (Fig. 27).
  • pVGE1 contains two human heavy chain variable gene segments upstream of the human ⁇ 1 constant gene and associated switch region.
  • a unique SalI 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 pVGE1 is digested with SalI and the XhoI insert of pIGM1 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 XhoI 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 XhoI 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 ⁇ 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

Abstract

The invention relates to transgenic non-human animals capable of producing heterologous antibodies and methods for producing human sequence antibodies which bind to human antigens with substantial affinity.

Description

TRANSGENIC NON-HUMAN ANIMALS CAPABLE OF PRODUCING
HETEROLOGOUS ANTIBODIES CROSS-REFERENCE TO RELATED APPLICATION
This application is a continuation-in-part of 08/ 209,741 filed 9 March 1994, 08/165,699 filed 10 December 1993 and 08/161,739 filed 03 December 1993, which is a
continuation-in-part of 08/155,301 filed 18 November 1993, which is a continuation-in-part of Serial No. 08/096,762, filed 22 July 93, which is a continuation-in part of
08/053,131 filed 26 April 93, which is a CIP of Serial No. 07/990,860, filed 16 December 92, Serial No. 07/810,279 filed 17 December 91, and Serial No. 07/904,068 filed 23 June 92; which is a CIP of Serial No. 07/853,408, filed 18 March 92, all incorporated herein by reference.
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 producing heterologous antibodies wherein a variable region sequence comprises somatic mutation as compared to germline rearranged variable region sequences, transgenic nonhuman animals which produce antibodies having a human primary sequence and which bind to human antigens, hybridomas made from B cells of such transgenic animals, and monclonal antibodies expressed by such hybridomas. 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 (HAMA) 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 ĸ 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
heterologous transgenes that undergo successful isotype switching. Transgenic animals that cannot switch isotypes are limited to producing heterologous 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 (l) 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, lgG3, 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 isotypes, 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 classswitching 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 XhoI and SalI
restriction sites flanked by two unique NotI sites. This method exploits the complementary termini of XhoI and SalI 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 invention provides transgenic non-human animals comprising: a homozygous pair of functionally disrupted endogenous heavy chain alleles, a homozygous pair of
functionally disrupted endogenous light chain alleles, at least one copy of a heterologous immunoglobulin heavy chain transgene, and at least one copy of a heterologous
immunoglobulin heavy chain transgene, wherein said animal makes an antibody response following immunization with an antigen, such as a human antigen (e.g., CD4). The invention also provides such a transgenic non-human animal wherein said functionally disrupted endogenous heavy chain allele is a JH region homologous recombination knockout, said functionally disrupted endogenous light chain allele is a Jκ region
homologous recombination knockout, said heterologous
immunoglobulin heavy chain transgene is the HC1 or HC2 human minigene transgene, said heterologous light chain transgene is the KC2 or KC1e human ĸ transgene, and wherein said antigen is a human antigen.
The invention also provides various embodiments for suppressing, ablating, and/or functionally disrupting the endogenous nonhuman immunoglobulin loci.
The invention also provides transgenic mice expressing both human sequence heavy chains and chimeric heavy chains comprising a human sequence heavy chain variable region and a murine sequence heavy chain constant region. Such chimeric heavy chains are generally produced by transswitching between a functionally rearranged human transgene and an endogenous murine heavy chain constant region (e.g., γ1, γ2a, γ2b, γ3). Antibodies comprising such chimeric heavy chains, typically in combination with a transgene-encoded human sequence light chain or endogenous murine light chain, are formed in response to immunization with a predetermined antigen. The transgenic mice of these embodiments can comprise B cells which produce (express) a human sequence heavy chain at a first timepoint and trans-switch to produce (express) a chimeric heavy chain composed of a human variable region and a murine constant region (e.g., γ1, γ2a, γ2b, γ3) at a second (subsequent) timepoint; such human sequence and chimeric heavy chains are incorporated into functional
antibodies with light chains; such antibodies are present in the serum of such transgenic mice. Thus, to restate: the transgenic mice of these embodiments can comprise B cells which express a human sequence heavy chain and subsequently switch (via trans-switching or cis-switching) to express a chimeric or isotype-switched heavy chain composed of a human variable region and a alternative constant region (e.g., murine γ1, γ2a, γ2b, γ3; human γ, α, ∈); such human sequence and chimeric or isotype-switched heavy chains are incorporated into functional antibodies with light chains (human or mouse); such antibodies are present in the serum of such transgenic mice.
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 ĸ 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 ĸ 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 Cγ1 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 Jĸ and human Cĸ 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. 20A through 20E depict the structure of a kappa light chain targeting vector.
Figs. 21A through 21F 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 ĸ 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.
Figs. 37A-37C depict 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 HC1 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 the conventional single-letter notation.
Fig. 41 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. 42 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. 43 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
(heptamer/nonamer) are shown boxed.
Fig. 44 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. 45 shows formation of a light chain minilocus by homologous recombination between two overlapping fragments which were co-injected.
Fig. 46 shows ELISA results for monoclonal antibodies reactive with CEA and non-CEA antigens showing the specificity of antigen binding.
Fig. 47 shows the DNA sequences of 10 cDNAs amplified by PCR to amplify transcripts having a human VDJ and a murine constant region sequence.
Fig. 48 shows ELISA results for various dilutions of serum obtained from mice bearing both a human heavy chain minilocus transgene and a human ĸ minilocus transgene; the mouse was immunized with human CD4 and the data shown
represents antibodies reactive with human CD4 and possessing human ĸ , human μ, or human γ epitopes, respectively.
Fig. 49 shows relative distribution of lymphocytes staining for human μ or mouse μ as determined by FACS for three mouse genotypes.
Fig. 50 shows relative distribution of lymphocytes staining for human ĸ or mouse ĸ as determined by FACS for three mouse genotypes.
Fig. 51 shows relative distribution of lymphocytes staining for mouse λ as determined by FACS for three mouse genotypes.
Fig. 52 shows relative distribution of lymphocytes staining for mouse λ or human ĸ as determined by FACS for four mouse genotypes.
Fig. 53 shows the amounts of human μ, human γ, human ĸ , mouse μ, mouse γ, mouse ĸ, and mouse λ chains in the serum of unimmunized 0011 mice.
Fig. 54 shows a scatter plot showing the amounts of human μ, human γ, human ĸ , mouse μ, mouse y, mouse ĸ , and mouse λ chains in the serum of unimmunized 0011 mice of various genotypes.
Fig. 55 shows the titres of antibodies comprising human μ, human γ, or human ĸ chains in anti-CD4 antibodies in the serum taken at three weeks or seven weeks post-immunization following immunization of a 0011 mouse with human CD4.
Fig. 56 shows a schematic representation of the human heavy chain minilocus transgenes pHC1 and pHC2, and the light chain minilocus transgenes pKC1, pKC1e, and the light chain minilocus transgene created by homologous recombination between pKC2 and Co4 at the site indicated.
Fig. 57 shows a linkage map of the murine lambda light chain locus as taken from Storb et al. (1989) op.cit.; the stippled boxes represent a pseudogene. Fig. 58 shows a schematic representation of
inactivation of the murine λ locus by homologous gene
targeting.
Fig. 59 schematically shows the structure of a homologous recombination targeting transgene for deleting genes, such as heavy chain constant region genes.
Fig. 60 shows a map of the BALB/c murine heavy chain locus as taken from Immunoglobulin Genes, Honjo, T, Alt, FW, and Rabbits TH (eds.) Academic Press, NY (1989) p. 129.
Structural genes are shown by closed boxes in the top line; second and third lines show restriction sites with symbols indicated.
Fig. 61 shows a nucleotide sequence of mouse heavy chain locus α constant region gene.
Fig. 62 shows the construction of a frameshift vector (plasmid B) for introducing a two bp frameshift into the murine heavy chain locus J4 gene.
Fig. 63 shows isotype specific response of transgenic animals during hyperimmunization. The relative levels of reactive human μ and γ1 are indicated by a
colorimetric ELISA assay (y-axis). We immunized three 7-10 week old male HC1 line 57 transgenic animals (#1991, #2356, #2357), in a homozygous JHD background, by intraperitoneal injections of CEA in Freund's adjuvant. The figure depicts binding of 250 fold dilutions of pooled serum (collected prior to each injection) to CEA coated microtiter wells.
Fig. 64A and 64B show expression of transgene encoded γ1 isotype mediated by class switch recombination. The genomic structure of integrated transgenes in two
different human γ1 expressing hybridomas is consistent with recombination between the μ and γ1 switch regions. Fig. 64A shows a Southern blot of PacI/SfiI digested DNA isolated from three transgene expressing hybridomas. From left to right: clone 92-09A-5H1-5, human γ1+ / μ-; clone 92-90A-4G2-2, human γ1+/μ-; clone 92-09A-4F7-A5-2, human γ1-,μ+. All three hybridomas are derived from a γ month old male mouse
hemizygous for the HC1-57 integration, and homozygous for the JHD disruption (mouse #1991). The blot is hybridized with a probe derived from a 2.3 kb BglII/SfiI DNA fragment spanning the 3' half of the human γ1 switch region. No switch product is found in the μ expressing hybridoma, while the two γ1 expressing hybridomas, 92-09A-5H1-5 and 92-09A-4G2-2, contain switch products resulting in PacI/Sfil fragments of 5.1 and 5.3 kb respectively, Fig. 64B is a diagram of two possible deletional mechanisms by which a class switch from μ to γ1 can occur. The human μ gene is flanked by 400 bp direct repeats (σμ and ∑μ) which can recombine to delete μ. Class switching by this mechanism will always generate a 6.4 kb PacI/SfiI fragment, while class switching by recombination between the μ and the γ1 switch regions will generate a PacI/Sfil fragment between 4 and γ kb, with size variation between individual switch events. The two γ1 expressing hybridomas examined in Fig. 64A appear to have undergone recombination between the μ and γ1 switch regions.
Fig. 65 shows chimeric human/mouse immunoglobulin heavy chains generated by trans-switching. cDNA clones of trans-switch products were generated by reverse transcription and PCR amplification of a mixture of spleen and lymph node RNA isolated from a hyperimmunized HC1 transgenic-JHD mouse (#2357; see legend to Fig. 63 for description of animal and immunization schedule). The partial nucleotide sequence of 10 randomly picked clones is shown. Lower case letters indicate germline encoded, capital letters indicate nucleotides that cannot be assigned to known germline sequences; these may be somatic mutations, N nucleotides, or truncated D segments. Both face type indicates mouse γ sequences.
Figs. 66A and 66B show that the rearranged VH251 transgene undergoes somatic mutation in a hyperimmunized. The partial nucleotide sequence of IgG heavy chain variable region cDNA clones from CH1 line 26 mice exhibiting Fig. 66A primary and Fig. 66B secondary responses to antigen. Germline
sequence is shown at the top; nucleotide changes from germline are given for each clone. A period indicates identity with germline sequence, capital letters indicate no identified germline origin. The sequences are grouped according to J segment usage. The germline sequence of each of the J segments if shown. Lower case letters within CDR3 sequences indicate identity to known D segment included in the HC1 transgene. The assigned D segments are indicated at the end of each sequence. Unassigned sequences could be derived from N region addition or somatic mutation; or in some cases they are simply too short to distinguish random N nucleotides from known D segments. Fig. 66A primary response: 13 randomly picked VH251-γ1 cDNA clones. A 4 week old female HC1 line 26-JHD mouse (#2599) was given a single injection of KLH and complete Freunds adjuvant; spleen cell RNA was isolated 5 days later. The overall frequency of somatic mutations within the V segment is 0.06% (2/3,198 bp). Fig. 66B secondary response: 13 randomly picked VH251-γ1 cDNA clones. A 2 month old female HC1 line 26-JHD mouse (#3204) was given 3 injections of HEL and Freunds adjuvant over one month (a primary injection with complete adjuvant and boosts with incomplete at one week and 3 weeks); spleen and lymph node RNA was isolated 4 months later. The overall frequency of somatic mutations within the V segment is 1.6% (52/3,198 bp).
Figs. 67A and 67B show that extensive somatic mutation is confined to γ1 sequences: somatic mutation and class switching occur within the same population of B cells. Partial nucleotide sequence of VH251 cDNA clones isolated from spleen and lymph node cells of HC1 line 57 transgenic-JHD mouse (#2357) hyperimmunized against CEA (see Fig. 63 for immunization schedule). Fig. 67A: IgM: 23 randomly picked VH251-μ cDNA clones. Nucleotide sequence of 156 bp segment including CDRs 1 and 2 surrounding residues. The overall level of somatic mutation is 0.1% (5/3,744 bp). Fig 67B: IgG: 23 randomly picked VH251-γI cDNA clones. Nucleotide sequence of segment including CDRs 1 through 3 and surrounding
residues. The overall frequency of somatic mutation within the V segment is 1.1% (65/5,658 bp). For comparison with the μ sequences in Fig. 67A: the mutation frequency for first 156 nucleotides is 1.1% (41/3,588 bp). See legend to
Figs. 66A and 66B for explanation of symbols.
Fig. 68 indicates that VH51P1 and VH56P1 show extensive somatic mutation of in an unimmunized mouse. The partial nucleotide sequence of IgG heavy chain variable region cDNA clones from a 9 week old, unimmunized female HC2 line 2550 transgenic-JHD mouse (#5250). The overall frequency of somatic mutation with the 19 VH56p1 segments is 2.2%
(101/4,674 bp). The overall frequency of somatic mutation within the single VH51p1 segment is 2.0% (5/246 bp). See legend to Figs. 66A and 66B for explanation of symbols.
Fig. 69. Double transgenic mice with disrupted endogenous Ig loci contain human IgM/c positive B cells. FACS of cells isolated from spleens of 4 mice with different genotypes. Left column: control mouse (#9944, 6 wk old female JH+/-, JCκ+/- ; heterozygous wild-type mouse heavy and ĸ-light chain loci, non-transgenic). Second column: human heavy chain transgenic (#9877, 6 wk old female JH-/-, JCĸ-/-, HC2 line 2550 +; homozygous for disrupted mouse heavy and ĸ-light chain loci, hemizygous for HC2 transgene). Third column: human ĸ-light chain transgenic (#9878, 6 wk old female JH-/-, JCĸ-/-, KCo4 line 4437 +; homozygous for
disrupted mouse heavy and ĸ-light chain loci, hemizygous for KCo4 transgene). Right column: double transgenic (#9879, 6 wk old female JH-/-m JCĸ-/- , HC2 line 2550 +, KCo4 line 4437 +; homozygous for disrupted mouse heavy and ĸk-light chain loci, hemizygous for HC2 and KCo4 transgenes). Top row:
spleen cells stained for expression of mouse λ light chain (x-axis) and human ĸ light chain (y-axis). Second row: spleen cells stained for expression of human μ heavy chain (x-axis) and human ĸ light chain (y-axis). Third row: spleen cells stained for expression of mouse μ heavy chain (x-axis) and mouse ĸ light chain (y-axis). Bottom row: histogram of spleen cells stained for expression of mouse B220 antigen (log fluorescence: x-axis; cell number: y-axis). For each of the two color panels, the relative number of cells in each of the displayed quadrants is given as percent of a e-parameter gate based on propidium iodide staining and light scatter. The fraction of B220+ cells in each of the samples displayed in the bottom row is given as a percent of the lymphocyte light scatter gate. Fig. 70. Secreted immunoglobulin levels in the serum of double transgenic mice. Human μ, γ, and ĸ , and mouse γ and λ from 18 individual HC2/KCo4 double transgenic mice homozygous for endogenous heavy and ĸ-light chain locus disruption. Mice: (+) HC2 line 2550 (-5 copies of HC2 per integration), KCo4 line 4436 (1-2 copies of KCo4 per
integration); (O) HC2 line 2550, KCo4 line 4437 (-10 copies of KCo4 per integration); (x) HC2 line 2550, KCo4 line 4583 (-5 copies of KCo4 per integration); (D) HC2 line 2572 (30-50 copies of HC2 per integration, KCo4 line 4437; (Δ) HC2 line 5467 (20-30 copies of HC2 per integration, KCo4 line 4437.
Figs. 71A and 71B show human antibody responses to human antigens. Fig. 71A: Primary response to recombinant human soluble CD4. Levels of human IgM and human ĸ light chain are reported for prebleed (0) and post-immunization (•) serum from four double transgenic mice. Fig. 71B: Switching to human IgG occurs in vivo . Human IgG (circles) was detected with peroxidase conjugated polyclonal anti-human IgG used in the presence of 1.5 μ/ml excess IgE, ĸ and 1% normal mouse serum to inhibit non-specific cross-reactivity. Human ĸ light chain (squares) was detected using a peroxidase conjugated polyclonal anti-human ĸ reagent in the presence of 1% normal mouse serum. A representative result from one mouse (#9344; HC2 line 2550, KCo4 line 4436) is shown. Each point
represents an average of duplicate wells minus background absorbance.
Fig. 72 shows FACS analysis of human PBL with a hybridoma supernatant that discriminates human CD4+
lymphocytes from human CD8+ lymphocytes.
Fig. 73 shows human α-CD4 IgM anf IgG in transgenic mouse serum.
Fig. 74 shows competition binding experiments comparing a transgenic mouse α-human CD4 hybridoma monoclonal, 2C11-8, to the RPA-TA and Leu-3A monoclonals.
Fig. 75 shows production data for Ig expression of cultured 2C11-8 hybridoma.
Table 1 depicts the sequence of vector pGPe.
Table 2 depicts the sequence of gene VH49.8. 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.
Table 10 shows the genotypes of several 0011 mice.
Table 11 shows transgene V and J segment usage.
Table 12 shows the occurrence of somatic mutation in the HC2 heavy chain transgene in transgenic mice.
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. Thus, developing hybridomas secreting human immunoglobulin
chainsspecifically reactive with predetermined human antigens is problematic, since a reliable source of human antibody-secreting B cells that can evoke an antibody response against predetermined human antigens is needed.
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 copending applications U.S.S.N. 07/990,860, filed 16 December 1992, U.S.S.N. 07/810,279 filed 17 December 1991, U.S.S.N. 07/904,068 filed 23 June 1992; U.S.S.N. 0γ/853,408, filed 18 March 1992, U.S.S.N. 0γ/5γ4,γ48 filed August 29, 1990,
U.S.S.N. 0γ/5γ5,962 filed August 31, 1990, and PCT/US91/06185 filed August 28, 1991, each incorporated herein by reference) 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.
The invention also provides a method for identifying candidate hybridomas which secrete a monoclonal antibody comprising a human immunoglobulin chain consisting essentially of a human VDJ sequence in polypeptide linkage to a human constant region sequence. Such candidate hybridomas are identified from a pool of hybridoma clones comprising: (1) hybridoma clones that express immunoglobulin chains consisting essentially of a human VDJ region and a human constant region, and (2) trans-switched hybridomas that express heterohybrid immunoglobulin chains consisting essentially of a human VDJ region and a murine constant region. The supernatant(s) of individual or pooled hybridoma clones is contacted with a predetermined antigen, typically an antigen which is
immobilized by adsoption onto a solid substrate (e.g., a microtitre well), under binding conditions to select
antibodies having the predetermined antigen binding
specificity. An antibody that specifically binds to human constant regions is also contacted with the hybridoma supernatant and predetermined antigen under binding conditions so that the antibody selectively binds to at least one human constant region epitope but substantially does not bind to murine constant region epitopes; thus forming complexes consisting essentially of hybridoma supernatant (transgenic monoclonal antibody) bound to a predetermined antigen and to an antibody that specifically binds human constant regions (and which may be labeled with a detectable label or
reporter). Detection of the formation of such complexes indicates hybridoma clones or pools which express a human immunoglobulin chain.
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, and generally from a species other than that 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 , e, 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 × 107 M-1, and (2) to preferentially bind to the predetermined antigen with an affinity that is at least two-fold greater than its affinity for binding to a non-specific antigen (e.g., BSA, casein) other than the predetermined antigen or a closely-related 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.
For nucleic acids, the term "substantial homology" indicates that two nucleic acids, or designated 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 cells, 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 (198γ).
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.
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 (γ) 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 transgenic 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:γ305-γ316 (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 or a 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 spliceacceptor site or a promoter/enhancer motif. For example, a minilocus comprises a portion of the genomic immunoglobulin locus having at least one internal (i.e., not at a terminus of the portion) deletion of a non-essential DNA portion (e.g., intervening sequence; intron or portion thereof) as compared to the naturally-occurring germline Ig locus.
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 gene (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 lgA2. The δ and ∈ constant region gene segments define the IgD and IgE antibody 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 mRNAs) 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 transgene-encoded 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 may be recognizable as containing a D region
sequence, specifically a DHQ52 sequence. However, since D region segments may be incorporated in VDJ joining to various extents and in various reading frames, a comparison of the D region area of a heavy chain variable region to the D region segments present in the transgene is necessary to determine the incorporation of particular D segments. Moreover, potential exonuclease digestion during recombination may lead to imprecise V-D and D-J joints during V-D-J recombination.
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 γ0 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, 151γ-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 (CDR1, CDR2 and CDR3) also referred to as hypervariable regions 1, 2 and 3 (Kabat et al. Seguences 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 antigen 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 is heterologous to that normally encountered in the nonhuman animal used to practice the invention.
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 transgene 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. 2:621-62γ (1990); Reid et al., Proc. Natl. Acad. Sci. USA 86:840-844 (1989); Stavnezer et al., Proc. Natl. Acad. Sci. USA 85:γγ04-γγ08 (1988); and Mills et al., Nucl. Acids Res. 18:γ305-γ316 (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 S switch
regions; the sequences immediately upstream of the human Sγ1, and Sγ3 switch regions can be used to advantage, with Sγ1 generally preferred. Alternatively, or in combination, murine Ig switch sequences may be used; it may frequently be
advantageous to employ Ig switch sequences of the same species as the transgenic non-human animal. Furthermore, 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 e) 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 δ). 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 murine 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 Sμ 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 α genes produces a composite Sμ-Sα sequence. Typically, there is no specific site, either in Sμ 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: in the mouse, 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 (Coffraan et al., J. Immunol. 136: 949 (1986) and Snapper et al.,
Science 236: 944 (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 (198γ), 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:γγ04 (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 γ1 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., a 5' flanking sequence from the human Sγ1 switch can be grafted immediately upstream of the Sα1 switch; a murine Sγ1 flanking region can be grafted adjacent to a human γ1 switch sequence; or the murine Sγ1 switch can be grafted onto the human γ1 coding region), 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.
Monoclonal Antibodies
Monoclonal antibodies can be obtained by various techniques familiar to those skilled in the art. Briefly, spleen cells from an animal immunized with a desired antigen are immortalized, commonly by fusion with a myeloma cell (see. Kohler and Milstein, Eur. J. Immunol., 6:511-519 (1976)).
Alternative methods of immortalization include transformation with Epstein Barr Virus, oncogenes, or retroviruses, or other methods well known in the art. Colonies arising from single immortalized cells are screened for production of antibodies of the desired specificity and affinity for the antigen, and yield of the monoclonal antibodies produced by such cells may be enhanced by various techniques, including injection into the peritoneal cavity of a vertebrate host. Various
techniques useful in these arts are discussed, for example, in Harlow and Lane, Antibodies: A Laboratory Manual, Cold Spring Harbor, New York (1988) including: immunization of animals to produce immunoglobulins; production of monoclonal antibodies; labeling immunoglobulins for use as probes; immunoaffinity purification; and immunoassays. 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 is estimated to consist of approximately 200 V gene segments (current data supports the existence of about 50-100 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 \pa 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. 2:727-738 (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 and in copending application, entitled "Transgenic Non-Human Animals Capable of Producing Heterologous Antibodies," filed August 29, 1990, under U.S.S.N. 07/574,748.
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 U.S. Patent Application entitled "Intracellular Generation of DNA by
Homologous Recombination of DNA Fragments" filed August 29, 1990, under U.S.S.N. 07/574,γ4γ.
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 typically capable 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 immunoglobulin.
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 «S 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 SfiI/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 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 in copending applications, U.S.S.N. 07/574,748 filed August 29, 1990, PCT/US91/06185 filed August 28, 1991, and U.S.S.N.
07/810,2γ9 filed December 1γ, 1991, each of which is
incorporated herein by reference. Rearranged or unrearranged V segments, D segments, J segments, and C genes, with or without flanking sequences, can be isolated as described in copending applications U.S.S.N. 07/574,748 filed August 29, 1990 and PCT/US91/06185 filed August 28, 1991.
A minilocus light chain transgene may be similarly constructed from the human λ or ĸ immunoglobulin locus.
Thus, for example, an immunoglobulin heavy chain minilocus transgene construct, e.g., of about γ5 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, as described in copending application, U.S.S.N. 07/574,748 filed August 29, 1990.
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 non-classical isotype switching (e.g., δ-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, ∈).
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 the 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, Sy2. Sy3 , Sγ4, , Sa2. and S. 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 (6-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, a1, α2, or e CH gene, preferably a human CH gene of these isotypes, and more preferably a human γ1 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.
Trans-switching to endogenous nonhuman heavy chain constant region genes can occur and produce chimeric heavy chains and antibodies comprising such chimeric human/mouse heavy chains. Such chimeric antibodies may be desired for certain uses described herein or may be undesirable.
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 ĸ-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 and ĸ genes, targeting vectors are used based on the design employed by Jaenisch and co-workers (Zijlstra, et al. (1989), Nature, 342, 435-438) for the successful disruption of the mouse 02-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 seguence 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 KpnI 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; U.S.S.N. 07/530,165 ("New human CRIPTO gene"); WO91/09865; WO91/04753; WO90/13641; 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 and thereby 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. Natl. 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 (e.g., murine) 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. Suitable antibody sources for antibody comprise:
(1) monoclonal antibodies, such as a monoclonal antibody that specifically binds to a murine μ, γ, ĸ , or λ chains but does not react with the human immunoglobulin chain (s) encoded by a human Ig transgene of the invention;
(2) mixtures of such monoclonal antibodies, so that the mixture binds with multiple epitopes on a single species of endogenous Ig chain, with multiple endogenous Ig chains (e.g., murine μ and murine γ, or with multiple epitopes and multiple chains or endogenous immunoglobulins;
(3) polyclonal antiserum or mixtures thereof, typically such antiserum/antisera is monospecific for binding to a single species of endogenous Ig chain (e.g., murine μ, murine γ, murine ĸ , murine λ) or to multiple species of endogenous Ig chain, and most preferably such antisera
possesses negligible binding to human immunoglobulin chains encoded by a transgene of the invention; and/or
(4) a mixture of polyclonal antiserum and monoclonal antibodies binding to a single or multiple species of
endogenous Ig chain, and most preferably possessing negligible binding to human immunoglobulin chains encoded by a transgene of the invention. Generally, polyclonal antibodies are preferred, and such substantially monospecific polyclonal antibodies can be advantageously produced from an antiserum raised against human immunoglobulin (s) by pre-adsorption with antibodies derived from the nonhuman animal species (e.g., murine) and/or, for example, by affinity chromatography of the antiserum or purified fraction thereof on an affinity resin containing immobilized human Ig (wherein the bound fraction is enriched for the desired anti-human Ig in the antiserum; the bound fraction is typically eluted with conditions of low pH or a chaotropic salt solution).
Cell separation and/or complement fixation can be employed to provide the enhancement of antibody-directed cell depletion of lymphocytes expressing endogenous (e.g., murine) immunoglobulin chains. In one embodiment, for example, antibodies are employed for ex vivo depletion of murine Ig-expressing explanted hematopoietic cells and/or B-lineage lymphocytes obtained from a transgenic mouse harboring a human Ig transgene. Thus, hematopoietic cells and/or B-lineage lymphocytes are explanted from a transgenic nonhuman animal harboring a human Ig transgene (preferably harboring both a human heavy chain transgene and a human light chain transgene) and the explanted cells are incubated with an antibody (or antibodies) which (1) binds to an endogenous immunoglobulin (e.g., murine μ and/or ĸ ) and (2) lacks substantial binding to human immunoglobulin chains encoded by the transgene(s). Such antibodies are referred to as "suppression antibodies" for clarity. The explanted cell population is selectively
depleted of cells which bind to the suppression antibody(ies); such depletion can be accomplished by various methods, such as (1) physical separation to remove suppression antibody-bound cells from unbound cells (e.g., the suppression antibodies may be bound to a solid support or magnetic bead to immobilize and remove cells binding to the suppression antibody), (2)
antibody-dependent cell killing of cells bound by the
suppression antibody (e.g., by ADCC,.by complement fixation, or by a toxin linked to the suppression antibody), and (3) clonal anergy induced by the suppression antibody, and the like.
Frequently, antibodies used for antibody suppression of endogenous Ig chain production will be capable of fixing complement. It is frequently preferable that such antibodies may be selected so as to react well with a convenient
complement source for ex vivo/in vitro depletion, such as rabbit or guinea pig complement. For in vivo depletion, it is generally preferred that the suppressor antibodies possess effector functions in the nonhuman transgenic animal species; thus, a suppression antibody comprising murine effector functions (e.g., ADCC and complement fixation) generally would be preferred for use in transgenic mice.
In one variation, a suppression antibody that specifically binds to a predetermined endogenous
immunoglobulin chain is used for ex vivo/in vitro depletion of lymphocytes expressing an endogenous immunoglobulin. A cellular explant (e.g., lymphocyte sample) from a transgenic nonhuman animal harboring a human immunoglobulin transgene is contacted with a suppression antibody and cells specifically binding to the suppression antibody are depleted (e.g., by immobilization, complement fixation, and the like), thus generating a cell subpopulation depleted in cells expressing endogenous (nonhuman) immunoglobulins (e.g., lymphocytes expressing murine Ig). The resultant depleted lymphocyte population (T cells, human Ig-positive B-cells, etc.) can be transferred into a immunocompatible (i.e., MHC-compatible) nonhuman animal of the same species and which is substantially incapable of producing endogenous antibody (e.g., SCID mice, RAG-1 or RAG-2 knockout mice). The reconstituted animal
(mouse) can then be immunized with an antigen (or reimmunized with an antigen used to immunize the donor animal from which the explant was obtained) to obtain high-affinity (affinity matured) antibodies and B-cells producing such antibodies. Such B-cells may be used to generate hybridomas by
conventional cell fusion and screened. Antibody suppression can be used in combination with other endogenous Ig
inactivation/suppression methods (e.g., JH knockout, CH knockout, D-region ablation, antisense suppression,
compensated frameshift inactivation).
Complete Endogenous Ig Locus Inactivation In certain embodiments, it is desirable to effect complete inactivation of the endogenous Ig loci so that hybrid immunoglobulin chains comprising a human variable region and a non-human (e.g., murine) constant region cannot be formed
(e.g., by trans-switching between the transgene and endogenous Ig sequences). Knockout mice bearing endogenous heavy chain alleles with are functionally disrupted in the JH region only frequently exhibit trans-switching, typically wherein a rearranged human variable region (VDJ) encoded by a transgene is expressed as a fusion protein linked to an endogenous murine constant region, although other trans-switched
junctions are possible. To overcome this potential problem, it is generally desirable to completely inactivate the
endogenous heavy chain locus by any of various methods, including but not limited to the following: (1) functionally disrupting and/or deleting by homologous recombination at least one and preferably all of the endogenous heavy chain constant region genes, (2) mutating at least one and
preferably all of the endogenous heavy chain constant region genes to encode a termination codon (or frameshift) to produce a truncated or frameshifted product (if trans-switched), and other methods and strategies apparent to those of skill in the art. Deletion of a substantial portion or all of the heavy chain constant region genes and/or D-region genes may be accomplished by various methods, including sequential deletion by homologous recombination targeting vectors, especially of the "hit-and-run" type and the like. Similarly, functional disruption and/or deletion of at least one endogenous light chain locus (e.g., ĸ ) to ablate endogenous light chain
constant region genes is often preferable.
Frequently, it is desirable to employ a frameshifted transgene wherein the heterologous transgene comprises a frameshift in the J segment(s) and a compensating frameshift (i.e., to regenerate the original reading frame) in the initial region (i.e., amino-terminal coding portion) of one or more (preferably all) of the transgene constant region genes. Trans-switching to an endogenous IgH locus constant gene
(which does not comprise a compensating frameshift) will result in a truncated or missense product that results in the trans-switched B cell being deleted or non-selected, thus suppressing the trans-switched phenotype.
Antisense suppression and antibody suppression may also be used to effect a substantially complete functional inactivation of endogenous Ig gene product expression (e.g., murine heavy and light chain sequences) and/or trans-switched antibodies (e.g., human variable/murine constant chimeric antibodies).
Various combinations of the inactivation and
suppression strategies may be used to effect essentially total suppression of endogenous (e.g., murine) Ig chain expression. Trans-Switching
In some variations, it may be desirable to produce a trans-switched immunoglobulin. For example, such trans-switched heavy chains can be chimeric (i.e., a non-murine (human) variable region and a murine constant region).
Antibodies comprising such chimeric trans-switched
immunoglobulins can be used for a variety of applications where it is desirable to have a non-human (e.g., murine) constant region (e.g., for retention of effector functions in the host, for the presence of murine immunological
determinants such as for binding of a secondary antibody which does not bind human constant regions). For one example, a human variable region repertoire may possess advantages as compared to the murine variable region repertoire with respect to certain antigens. Presumably the human VH, D, JH, VL, and JL genes have been selected for during evolution for their ability to encode immunoglobulins that bind certain
evolutionarily important antigens; antigens which provided evolutionary selective pressure for the murine repertoire can be distinct from those antigens which provided evolutionary pressure to shape the human repertoire. Other repertoire adavantages may exist, making the human variable region repertoire advantageous when combined with a murine constant region (e.g., a trans-switched murine) isotype. The presence of a murine constant region can afford advantages over a human constant region. For example, a murine y constant region linked to a human variable region by trans-switching may provide an antibody which possesses murine effector functions (e.g., ADCC, murine complement fixation) so that such a chimeric antibody (preferably monoclonal) which is reactive with a predetermined antigen (e.g., human IL-2 receptor) may be tested in a mouse disease model, such as a mouse model of graft-versus-host disease wherein the T lymphocytes in the mouse express a functional human IL-2 receptor. Subsequently, the human variable region encoding sequence may be isolated (e.g., by PCR amplification or cDNA cloning from the source (hybridoma clone)) and spliced to a sequence encoding a desired human constant region to encode a human sequence antibody more suitable for human therapeutic uses where immunogenicity is preferably minimized. The polynucleotide(s) having the resultant fully human encoding sequence(s) can be expressed in a host cell (e.g., from an expression vector in a mammalian cell) and purified for pharmaceutical formulation. For some applications, the chimeric antibodies may be used directly without replacing the murine constant region with a human constant region. Other variations and uses of trans-switched chimeric antibodies will be evident to those of skill in the art.
The present invention provides transgenic nonhuman animals containing B lymphocytes which express chimeric antibodies, generally resulting from trans-switching between a human heavy chain transgene and an endogenous murine heavy chain constant region gene. Such chimeric antibodies comprise a human sequence variable region and a murine constant region, generally a murine switched (i.e., non-μ, non-δ) isotype. The transgenic nonhuman animals capable of making chimeric
antibodies to a predetermined antigen are usually also
competent to make fully human sequence antibodies if both human heavy chain and human light chain transgenes encoding human variable and human constant region genes are integrated. Most typically, the animal is homozygous for a functionally disrupted heavy chain locus and/or light chain locus but retains one or more endogenous heavy chain constant region gene(s) capable of trans-switching (e.g., γ,α, e) and
frequently retains a cis-linked enhancer. Such a mouse is immunized with a predetermined antigen, usually in combination with an adjuvant, and an immune response comprising a
detectable amount of chimeric antibodies comprising heavy chains composed of human sequence variable regions linked to murine constant region sequences is produced. Typically, the serum of such an immunized animal can comprise such chimeric antibodies at concentrations of about at least 1 μg/ml, often about at least 10 μg/ml, frequently at least 30 μg/ml, and up to 50 to 100 μg/ml or more. The antiserum containing
antibodies comprising chimeric human variable/mouse constant region heavy chains typically also comprises antibodies which comprise human variable/human constant region (complete human sequence) heavy chains. Chimeric trans-switched antibodies usually comprise (1) a chimeric heavy chain composed of a human variable region and a murine constant region (typically a murine gamma) and (2) a human transgene-encoded light chain (typically kappa) or a murine light chain (typically lambda in a kappa knockout background). Such chimeric trans-switched antibodies generally bind to a predetermined antigen (e.g., the immunogen) with an affinity of about at least 1 × 107 M-1, preferably with an affinity of about at least 5 × 107 M-1, more preferably with an affinity of at least 1 × 108 M-1 to 1 × 109 M-1 or more. Frequently, the predetermined antigen is a human protein, such as for example a human cell surface antigen (e.g., CD4, CD8, IL-2 receptor, EGF receptor, PDGF receptor), other human biological macromolecule (e.g., thrombomodulin, protein C, carbohydrate antigen, sialyl Lewis antigen, L-selectin), or nonhuman disease associated macromolecule (e.g., bacterial LPS, virion capsid protein or envelope glycoprotein) and the like.
The invention provides transgenic nonhuman animals comprising a genome comprising: (1) a homozygous functionally disrupted endogenous heavy chain locus comprising at least one murine constant region gene capable of trans-switching (e.g., in cis linkage to a functional switch recombination sequence and typically to a functional enhancer), (2) a human heavy chain transgene capable of rearranging to encode end express a functional human heavy chain variable region and capable of trans-switching (e.g., having a cis-linked RSS); optionally further comprising (3) a human light chain (e.g., kappa) transgene capable of rearranging to encode a functional human light chain variable region and expressing a human sequence light chain; optionally further comprising (4) a homozygous functionally disrupted endogenous light chain locus ( ĸ , preferably ĸ and λ); and optionally further comprising (5) a serum comprising an antibody comprising a chimeric heavy chain composed of a human sequence variable region encoded by a human transgene and a murine constant region sequence encoded by an endogenous murine heavy chain constant region gene
(e.g., γ1, γ2a, γ2b, γ3).
Such transgenic mice may further comprise a serum comprising chimeric antibodies which bind a predetermined human antigen (e.g., CD4, CD8, CEA) with an affinity of about at least 1 × 104 M-1, preferably with an affinity of about at least 5 × 104 M-1, more preferably with an affinity of at least 1 × 107 M-1 to 1 × 109 M-1 or more. Frequently, hybridomas can be made wherein the monoclonal antibodies produced thereby have an affinity of at least 8 × 107 M-1. Chimeric antibodies comprising a heavy chain composed of a murine constant region and a .human variable region, often capable of binding to a nonhuman antigen, may also be present in the serum or as an antibody secreted from a hybridoma.
Generally , such chimeric antibodies can be generated by trans-switching, wherein a human transgene encoding a human variable region (encoded by productive V-D-J rearrangement in vivo) and a human constant region, typically human μ,
undergoes switch recombination with a non-transgene
immunoglobulin constant gene switch sequence (RSS) thereby operably linking the transgene-encoded human variable region with a heavy chain constant region which is not encoded by said transgene, typically an endogenous murine immunoglobulin heavy chain constant region or a heterologous (e.g., human) heavy chain constant region encoded on a second transgene.
Whereas cis-switching refers to isotype-switching by
recombination of RSS elements within a transgene, trans-switching involves recombination between a transgene RSS and an RSS element outside the transgene, often on a different chromosome than the chromosome which harbors the transgene.
Trans-switching generally occurs between an RSS of an expressed transgene heavy chain constant region gene and either an RSS of an endogenous murine constant region gene (of a non-μ isotype, typically γ) or an RSS of a human constant region gene contained on a second transgene, often integrated on a separate chromosome.
When trans-switching occurs between an RSS of a first, expressed transgene heavy chain constant region gene (e.g., μ) and an RSS of a human heavy chain constant region gene contained on a second transgene, a non-chimeric antibody having a substantially fully human sequence is produced. For example and not limitation, a polynucleotide encoding a human heavy chain constant region (e.g., γ1) and an operably linked RSS (e.g., a γ1 RSS) can be introduced (e.g., transfected) into a population of hybridoma cells generated from a
transgenic mouse B-cell (or B cell population) expressing an antibody comprising a transgene-encoded human μ chain. The resultant hybridoma cells can be selected for the presence of the introduced polynucleotide and/or for the expression of trans-switched antibody comprising a heavy chain having the variable region (idiotype/antigen reactivity) of the human μ chain and having the constant region encoded by the introduced polynucleotide sequence (human γ1). Trans-switch
recombination between the RSS of the transgene-encoded human μ chain and the RSS of the introduced polynucleotide encoding a downstream isotype (e.g., γ1) thereby can generate a trans-switched antibody.
The invention also provides a method for producing such chimeric trans-switched antibodies comprising the step of immunizing with a predetermined antigen a transgenic mouse comprising a genome comprising: (1) a homozygous functionally disrupted endogenous heavy chain locus comprising at least one murine constant region gene capable of trans-switching (e.g., γ2a, γ2b, γ1, γ3), (2) a human heavy chain transgene capable of rearranging to encode a functional human heavy chain variable region and expressing a human sequence heavy chain and capable of undergoing isotype switching (and/or trans-switching), and optionally further comprising (3) a human light chain (e.g., kappa) transgene capable of rearranging to encode a functional human light (e.g., kappa) chain variable region and expressing a human sequence light chain, and optionally further comprising (4) a homozygous functionally disrupted endogenous light chain locus (typically ĸ ,
preferably both ĸ and λ), and optionally further comprising (5) a serum comprising an antibody comprising a chimeric heavy chain composed of a human sequence variable region encoded by a human transgene and a murine constant region sequence encoded by an endogenous murine heavy chain constant region gene (e.g., γ1, γ2a, γ2b, γ3).
Affinity Tagging: Selecting for Switched Isotypes Advantageously, trans-switching (and cis-switching) is associated with the process of somatic mutation. Somatic mutation expands the range of antibody affinities encoded by clonal progeny of a B-cell. For example, antibodies produced by hybridoma cells which have undergone switching (trans- or cis-) represent a broader range of antigen-binding affinities than is present in hybridoma cells which have not undergone switchin Thus, a hybridoma cell population (typically clonal) which expresses a first antibody comprising a heavy chain comprising a first human heavy chain variable region in polypeptide linkage to a first human heavy chain constant region (e.g., μ) can be screened for hybridoma cell clonal variants which express an antibody comprising a heavy chain containing said first human heavy chain variable region in polypeptide linkage to a second heavy chain constant region (e.g., a human γ, α, or e constant region). Such clonal variants can be produced by natural clonal variation producing cis-switching in vitro. by induction of class switching
(trans- or cis-) as through the administration of agents that promote isotype switching, such as T-cell-derived lymphokines (e.g., IL-4 and IFNγ), by introduction of a polynucleotide comprising a functional RSS and a heterologous (e.g. human) heavy chain constant region gene to serve as a substrate for trans-switching, or by a combination of the above, and the like.
Class switching and affinity maturation take place within the same population of B cells derived from transgenic animals of the present invention. Therefore, identification of class-switched B cells (or hybridomas derived therefrom) can be used as a screening step for obtaining high affinity monoclonal antibodies. A variety of approaches can be
employed to facilitate class switching events such as cis-switching (intratransgene switching), trans-switching, or both. For example, a single continuous human genomic fragment comprising both μ and γ constant region genes with the associated RSS elements and switch regulatory elements (e.g., sterile transcript promoter) can be used as a transgene.
However, some portions of the desired single contiguous human genomic fragment can be difficult to clone efficiently, such as due to instability problems when replicated in a cloning host or the like; in particular, the region between δ and γ3 can prove difficult to clone efficiently, especially as a contiguous fragment comprising the μ gene, γ3 gene, a V gene, D gene segments, and J gene segments.
Also for example, a discontinuous human transgene (minigene) composed of a human μ gene, human γ3 gene, a human V gene(s), human D gene segments, and human J gene segments, with one or more deletions of an intervening (intronic) or otherwise nonessential sequence (e.g., one or more V, D, and/or J segment and/or one or more non-μ constant region gene(s)). Such minigenes have several advantages as compared to isolating a single contiguous segment of genomic DNA spanning all of the essential elements for efficient
immunoglobulin expression and switching. For example, such a minigene avoids the necessity of isolating large pieces of DNA which may contain sequences which are difficult to clone
(e.g., unstable sequences, poison sequences, and the like). Moreover, miniloci comprising elements necessary for isotype switching (e.g., human γ sterile transcript promoter) for producing cis- or trans-switching, can advantageously undergo somatic mutation and class switching in vivo. As many
eukaryotic DNA sequences can prove difficult to clone,
omitting non-essential sequences can prove advantageous.
In a variation, hybridoma clones producing antibodies having high binding affinity (e.g., at least 1 × 107 M-1, preferably at least 1 × 108 M-1, more preferably at least 1 × 109 M-1 or greater) are obtained by selecting, from a pool of hybridoma cells derived from B cells of transgenic mice harboring a human heavy chain transgene capable of isotype switching (see, supra) and substantially lacking endogenous murine heavy chain loci capable of undergoing productive (in-frame) V-D-J rearrangement, hybridomas which express an antibody comprising a heavy chain comprising a human sequence heavy chain variable region in polypeptide linkage to a human (or mouse) non-μ heavy chain constant region; said antibodys are termed "switched antibodies" as they comprise a "switched heavy chain" which is produced as a consequence of cis-switching and/or trans-switching in vivo or in cell culture. Hybridomas producing switched antibodies generally have undergone the process of somatic mutation, and a pool of said hybridomas will generally have a broader range of antigen binding affinities from which hybridoma clones secreting high affinity antibodies can be selected.
Typically, hybridomas secreting a human sequence antibody having substantial binding affinity (greater than 1 × 107 M-1 to 1 × 108 M-1) for a predetermined antigen and wherein said human sequence antibody comprises human immunoglobulin
variable region (s) can be selected by a method comprising a two-step process. One step is to identify and isolate
hybridoma cells which secrete immunoglobulins which comprise a switched heavy chain (e.g., by binding hybridoma cells to an immobilized immunoglobulin which specifically binds a switched heavy chain and does not substantially bind to an unswitched isotype, e.g., μ). The other step is to identify hybridoma cells which bind to the predetermined antigen with substantial binding affinity (e.g., by ELISA of hybridoma clone
supernatants, FACS analysis using labeled antigen, and the like). Typically, selection of hybridomas which secrete switched antibodies is performed prior to identifying
hybridoma cells which bind predetermined antigen. Hybridoma cells which express switched antibodies that have substantial binding affinity for the predetermined antigen are isolated and cultured under suitable growth conditions known in the art, typically as individual selected clones. Optionally, the method comprises the step of culturing said selected clones under conditions suitable for expression of monocloanl antibodies; said monoclonal antibodies are collected and can be administered for therapeutic, prophylactic, and/or
diagnostic purposes.
Often, the selected hybridoma clones can serve as a source of DNA or RNA for isolating immunoglobulin sequences which encode immunoglobulins (e.g. a variable region) that bind to (or confer binding to) the predetermined antigen.
Subsequently, the human variable region encoding sequence may be isolated (e.g., by PCR amplification or cDNA cloning from the source (hybridoma clone)) and spliced to a sequence encoding a desired human constant region to encode a human sequence antibody more suitable for human therapeutic uses where immunogenicity is preferably minimized. The
polynucleotide (s) having the resultant fully human encoding sequence(s) can be expressed in a host cell (e.g., from an expression vector in a mammalian cell) and purified for pharmaceutical formulation.
Xenoenhancers
A heterologous transgene capable of encoding a human immunoglobulin (e.g., a heavy chain) advantageously comprises a cis-linked enhancer which is not derived from the mouse genome, and/or which is not naturally associated in cis with the exons of the heterologous transgene. For example, a human ĸ transgene (e.g., a ĸ minilocus) can advantageously comprise a human Vĸ gene, a human Jĸ gene, a human Cĸ gene, and a xenoenhancer, typically said xenoenhancer comprises a human heavy chain intronic enhancer and/or a murine heavy chain intronic enhancer, typically located between a Jĸ gene and the Cĸ gene, or located downstream of the Cĸ gene. For example, the mouse heavy chain J-μ intronic enhancer (Banerji et al. (1983) Cell 33: 729) can be isolated on a 0.9 kb Xbal fragment of the plasmid pKVe2 (see, infra). The human heavy chain J-μ intronic enhancer (Hayday et al. (1984) Nature 307: 334) can be isolated as a 1.4 kb Mlul/HindIII fragment (see, infra). Addition of a transcriptionally active xenoenhancer to a transgene, such as a combined xenoenhancer consisting essentially of a human J-μ intronic enhancer linked in cis to a mouse J-μ intronic enhancer, can confer high levels of expression of the transgene, especially where said transgene encodes a light chain, such as human ĸ . Similarly, a rat 3' enhancer can be advantageously included in a minilocus construct capable of encoding a human heavy chain.
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 human ĸ 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 human ĸ light chain gene will allelically and isotypically exclude the rearrangement of the endogenous mouse ĸ 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.
In some embodiments, it may be preferable to
generate mice with predetermined repertoires to limit the selection of V genes represented in the antibody response to a predetermined antigen type. A heavy chain transgene having a predetermined repertoire may comprise, for example, human VH genes which are preferentially used in antibody responses to the predetermined antigen type in humans. Alternatively, some VH genes may be excluded from a defined repertoire for various reasons (e.g., have a low likelihood of encoding high affinity V regions for the predetermined antigen; have a low propensity to undergo somatic mutation and affinity sharpening; or are immunogenic to certain humans). 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 ĸ 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 proteinase ĸ 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
Genomic ĸ Light Chain Human Ig Transgene Formed by In Vivo Homologous Recombination
A map of the human ĸ 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 XhoI 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
Genomic ĸ Light Chain Human Ig Transgene Formed bv In Vivo Homologous Recombination A 750 kb MluI 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 XhoI to NotI fragment plus the approximately 400 kb Mlul to BssHII fragment have sequence overlap defined by the BssHII and XhoI 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 (SfiI) 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 SpeI sites. This allows inserts to be completely excised with SfiI 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.
The 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/HindIII fragment that includes all human J segments (Matsuda et al., EMBO J.. 7:1047-1051 (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 10 kb HindIII/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/HindIII 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 BglII 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 HindIII digest. The resultant plasmid pHIG1 (Fig. 10) contains an 18 kb insert encoding J and Cμ segments. D. Cloning of Cu Region
pGP1 is digested with BamHI and HindIII 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/HindIII cut pGP1. Proper orientation of fragment (c) is checked by HindIII digestion to form pCON1 containing a 12 kb insert encoding the Cμ region.
Whereas pHIG1 contains J segments, switch and μ sequences in its 18 kb insert with an SfiI 3' site and a Spel 5' site in a polylinker flanked by NotI sites, will be used for rearranged VDJ segments. pCONl is identical except that it lacks the J region and contains only a 12 kb insert. The use of pCONl in the construction of fragment containing rearranged VDJ segments will be described hereinafter.
E. Cloning of 7-1 Constant Region (pREG2)
The cloning of the human γ-1 region is depicted in Fig. 16.
Yamamura et al., Proc. Natl. Acad. Sci. USA
22:2152-2156 (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 HindIII 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 HindIII 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 using the following oligonucleotide which is specific for the third exon of γ-I (CH3).
5' TGA GCC ACG AAG ACC CTG AGG
TCA AGT TCA ACT GGT ACG TGG 3'
A 7.7 kb HindIII to BglII fragment (fragment (a) in Fig. 11) is cloned into HindIII/BglII cut pRE3 to form pREG1. The upstream 5.3 kb HindIII fragment (fragment (b) in Fig. 11) is cloned into HindIII digested pREG1 to form pREG2. Correct orientation is confirmed by BamHI/Spel digestion. F. Combining Cγ and Cμ
The previously described plasmid pHIG1 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 SfiI (Fig. 10). The plasmid pREG2 was also digested with SfiI 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 pCONl with SfiI and combining that with the SfiI fragment containing the human Cγ region and the rat 3' enhancer by digesting pREG2 with SfiI. 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 XhoI 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/XhoI 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 XhoI 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' SfiI 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.
pVH1 is digested with SfiI and the resultant
fragment is cloned into the SfiI 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 SpeI 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/HindIII 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 pHIG5'0 (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 SfiI site to generate pHIGS' 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 pEu1
The construction of pEu1 is depicted in Fig. 16. The mouse heavy chain enhancer is isolated on the Xbal to
EcoRI 678 bp fragment (Banerji et al., Cell 21:729-740 (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 ĸ Light chain Minilocus
The ĸ construct contains at least one human Vκ segment, all five human Jκ segments, the human J-Cκ enhancer, human ĸ constant region exon, and, ideally, the human 3' ĸ enhancer (Meyer et al., EMBO J. 8:1959-1964 (1989)). The ĸ 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 SmaI fragment that contains the human Cκ exon and the 3' human enhancer by analogy with the mouse locus;
(b) A 5' adjacent 5 kb SmaI fragment, which
contains all five J segments;
(c) The mouse heavy chain intronic enhancer
isolated from pEu1 (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 ĸ 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 SmaI 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 SmaI digested gel and the 11 kb region is similarly isolated from the gel containing DNA digested with BamHI.
The 16 kb SmaI fraction is cloned into Lambda FIX II (Stratagene, La Jolla, California) which has been digested with XhoI, treated with klenow fragment DNA polymerase to fill in the XhoI restriction digest product. Ligation of the 16 kb SmaI fraction destroys the SmaI sites and lases XhoI 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 Cĸ specific oligo: 5' GAA CTG TGG CTG CAC CAT CTG TCT
TCA TCT TCC CGC CAT CTG 3'
A 16 kb XhoI insert that was subcloned into the XhoI cut pEu1 so that Cĸ is adjacent to the SmaI site. The
resultant plasmid was designated pKapl.
The above Cĸ specific oligonucleotide is used to probe the λ EMBL3/BamHI library to identify an 11 kb clone. A 5 kb SmaI fragment (fragment (b) in Fig. 20) is subcloned and subsequently inserted into pKapl digested with SmaI. Those plasmids containing the correct orientation of J segments, Cĸ and the Eμ enhancer are designated pKap2.
One or more Vĸ segments are thereafter subcloned into the Mlul site of pKap2 to yield the plasmid pKapH which encodes the human Vĸ segments, the human Jĸ segments, the human Cĸ 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 ĸ 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 SfiI site. The resultant plasmid is designated pKAPint. One or more Vĸ 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 SmaI 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 ĸ Light Chain Genes This example describes the cloning of immunoglobulin ĸ 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 the ĸ 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 ĸ 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 a ĸ 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 (for general methods see. Goodspeed et al.
(1989) Gene 76: 1; Dunn et al. (1989) J. Biol. Chem. 264:
13057). The single stranded cDNA is then isolated and G residues are added to the 3' end using the enzyme
polynucleotide terminal transferase. The G-tailed
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 the ĸ 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 SmaI, 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 SmaI and second enzyme (or BamHI or Kpnl if there is SmaI 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 XhoI depending on what site does not exist in fragment) and cut with the corresponding linker enzyme to give DNA fragments with BamHI, EcoRI or XhoI 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 (MluI 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 pGPI. V segment containing clones are isolated using the unique probe o-mu, and the insert is subcloned into MluI or MluI/Spel cut plasmid pCON2. The resulting plasmid is called pRMGH. EXAMPLE 8
Construction of Human ĸ 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 ClaI/XhoI fragment containing Jκ1 together with a 13 kb XhoI 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' XhoI site for cloning Vκ segments and a unique 3' SalI 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 VĸI pseudogene as it contains a non-canonical recombination heptamer.
One of the functional clones, Vk 65-8, which encodes a VkIII family gene, was used to build a light chain minilocus construct.
PKC1
The kappa light chain minilocus transgene pKC1 (Fig. 32) was generated by inserting a 7.5 kb XhoI/SalI fragment containing Vĸ 65.8 into the 5' XhoI 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 × 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 transgene (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. All 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 XhoI/SalI fragment containing Vκ 65.5 into the 5' XhoI 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 pKC1 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-μ intronic enhancer (Banerji et al., Cell 33:729-740 (1983)) together with a 1.4 kb MluI/HindIII fragment containing the human heavy chain J-μ 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 XhoI site of the vector pK31s that includes Jĸ2-4, 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 SalI sites at the 3' end for the insertion of additional cis-acting regulatory sequences.
Two synthetic oligonucleotides (o-130, o-131) were used to 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 VĸIII 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 Jĸ1 and includes an XhoI site. These two oligonucleotides amplify a 0.7 kb DNA
fragment from human spleen DNA corresponding to rearranged VĸIII genes joined to Jĸ1 segments. The PCR amplified DNA was digested with NcoI and XhoI 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 × 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 (XbaI/TaqI fragment; Adra et al. (1987) Gene 60: 65) 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 (PvuII/HindIII 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 Cĸ locus: 5'- GGC TGA TGC TGC ACC AAC TGT ATC CAT CTT CCC ACC ATC CAG -3' and for the Jĸ5 gene segment: 5'- CTC ACG TTC GGT GCT GGG ACC AAG CTG GAG CTG AAA CGT AAG -3 ' .
An 8 kb BglII/Sad fragment extending 3' of the mouse Cκ segment was isolated from a positive phage clone in two pieces, as a 1.2 kb BglII/Sad fragment and a 6.8 kb Sad fragment, and subcloned into BglII/Sad 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 SphI/Xbal/BglII/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 recombinantε, 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/HindIII fragment and subcloned into pGP1b to generate pGP1b-TK. This plasmid was linearized at the XhoI site and the XhoI fragment from pNEO-K5'3', containing the neo gene flanked by genomic sequences from 5' of Jĸ and 3' of Cĸ, 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 K1 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. Embryol. 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 K1 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 × 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, γ displayed the 1.8 kb Xbal band indicative of a homologous recombination into one of the kappa genes. These γ clones were further digested with the enzymes BglII, Sacl, and Pstl 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), BglII, Sacl, and Pstl 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, 7.5, and 5.7 kb, respectively (see Fig. 20a and e). All γ positive clones detected by the Xbal digest showed the expected BglII, Sacl, and Pstl restriction fragments diagnostic of a homologous recombination at the kappa light chain. In addition. Southern blot analysis of an NsiI 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 seguences 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 Sacl/Stul fragment, spanning the JH region, was isolated from a positive phage clone and subcloned into Sacl/SmaI 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
HindIII site in the prepared pGEM7 (KJ1) was converted to a SalI site by addition of a synthetic adaptor, and the neo expression cassette excised by digestion with Xbal/SalI. The ends of the neo fragment were then blunted by treatment with the Klenow form of DNA polI, and the neo fragment was
subcloned into the Nael site of pUC18 JH, generating the plasmid pUClδ 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 HindIII. The TK DNA fragment was subcloned between the EcoRI and HindIII 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/XhoI fragment, situated 5' of the JH region, was derived from a positive genomic phage clone by limit digestion of the DNA with XhoI, and partial digestion with Xbal. As noted in Fig. 21a, this XbaI 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/XhoI 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 XhoI 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. 2If 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 × 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 Stul and probed with the 500 bp EcoRI/StuI fragment designated as probe A in Fig. 2If. This probe detects a Stul 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 HindIII, SpeI and Hpal. Hybridization of probe A (see Fig. 21f) to Southern blots of HindIII, 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 Stul digest showed the predicted HindIII, Spel, and HpaI bands diagnostic of a targeted JH gene. In addition, Southern blot analysis of a Stul digest of all 12 clones using a neo-specific probe (probe B, Fig. 2If) 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 Stul band of approximately 3 kb in addition to the wild-type band of 4.7 kb, demonstrating germline transmission of the targeted JH 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 matings: wild-type mice bearing two copies of the normal JH locus, heterozygotes carrying 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. pGP1a
The plasmid pBR322 was digested with EcoRI and StyI 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 XhoI ClaI BamHI HindIII NotI
GCggccgcctcgagatcactatcgattaattaaggatccagcagtaagcttgcGGCCGC pGI1c
NotI SmaI XhoI SalI HindIII BamHI SacII NotI
GCggccgcatcccgggtctcgaggtcgacaagctttcgaggatccgcGGCCGC pGP1d
NotI SalI HindIII ClaI BamHI XhoI NotI
GCggccgctgtcgacaagcttatcgatggatcctcgagtgcGGCCGC pGP1f
NotI SalI HindIII EcoRI ClaI KpnI BamHI XhoI NotI GCggccgctgtcgacaagcttcgaattcagatcgatgtggtacctggatcctcgagtgcGGCCGC 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, NcoI, ClaI, EcoRV, Xbal, Sacl, XhoI, SphI, Pstl, BglII, EcoRI, SmaI, KpnI, HindIII, and NotI). The resulting plasmid, pRE3, was digested with BamHI and HindIII, and the insert containing the rat Ig heavy chain 3' enhancer cloned into BamHI/HindIII 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 imgf000116_0001
B. Construction of IgM expressing minilocus transgene. pIGM1
1. Isolation of J-μ constant region clones and construction of pJMl
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 HindIII/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 HindIII/XhoI 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/XhoI digested pNN03 to obtain the plasmid pJM1. 2. pJM2
A 4 kb XhoI 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 δ-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 XhoI cut, Klenow treated, pJM1. The resulting plasmid, pJM2 (Fig. 23), had lost the internal XhoI site but retained the 3' XhoI 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 XhoI fragment, that includes the D elements Dκ1, DN1, and DM2 (Ichihara et al., EMBO J. 7:4141 (1988)), was isolated from phage clone λ4.1. An adjacent upstream 5.2 kb XhoI fragment, that includes the D elements DLR1, Dχp1, Dχp'1, and DA1, was isolated from phage clone λ4.3. Each of these D region XhoI fragments were cloned into the SalI site of the plasmid vector pSP72 (Promega, Madison, WI) so as to destroy the XhoI site linking the two sequences. The upstream
fragment was then excised with XhoI and SmaI, and the
downstream fragment with EcoRV and XhoI. The resulting isolated fragments were ligated together with SalI digested pSP72 to give the plasmid pDH1. pDH1 contains a 10.6 kb insert that includes at least γ D segments and can be excised with XhoI (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 ClaI and the insert isolated. This insert was ligated to the XhoI/EcoRV insert of pDH1 and XhoI/ClaI digested pGPe to generate pCOR1 (Fig. 24).
5. PVH251
A 10.3 kb genomic HindIII 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. pIGM1
The plasmid pCOR1 was partially digested with XhoI and the isolated XhoI/SalI insert of pVH251 cloned into the upstream XhoI site to generate the plasmid pIGM1 (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 HindIII 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 carried 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 HindIII fragment of phage clone λ29.5, containing the γ1 coding region was cloned into pUC18. The resulting plasmid, pLT1, was digested with XhoI, Klenow treated, and religated to destroy the internal XhoI site. The resulting clone, pLT1xk, was digested with HindIII and the insert isolated and cloned into pSP72 to generate the plasmid clone pLTlxks. Digestion of pLTlxks at a polylinker XhoI 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 X29.5 into XhoI/BamHI digested pGPe to generate the plasmid clone pγβl. pγel contains all of the γ1 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 HindIII fragment containing the γ1 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 λSγ1.13 and cloned into pSP72 with the polylinker XhoI site adjacent to the 5' end of the insert, to generate the plasmid clone PSγ1S. The XhoI/SalI insert of PSγ1S was cloned into XhoI digested Pγel to generate the plasmid clone Pγe2 (Fig. 26). Pγe2 contains all of the γ1 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 XhoI site at the 5' end of the insert. The entire insert, together with the XhoI site and the 3' rat enhancer can be excised from vector sequences by digestion with NotI. 4 . PHC1
The plasmid pIGM1 was digested with XhoI and the 43 kb insert isolated and cloned into XhoI 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 γ1
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 λ49.8 was isolated and a 6.1 kb Xbal fragment containing the variable segment VH49.8 subcloned into pNN03 (such that the polylinker ClaI site is downstream of VH49.8 and the polylinker XhoI 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 imgf000122_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 SmaI and HindIII, and treated with the Klenow fragment of polymerase I. The blunt ended fragment was then cloned into ClaI 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 SalI site at the 3' end of the insert and a unique XhoI site at the 5' end.
3. pSγ1-5'
A 0.7 kb Xbal/HindIII 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 HindIII/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 SmaI and
HindIII, treated with Klenow enzyme, and ligated with the following oligonucleotide linker: 5'- ccg gtc gac egg -3'
The ligation product was digested with SalI and ligated to SalI 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 SalI and complete digestion with XhoI, followed by purification of the 15 kb fragment on an agarose gel. The insert is then cloned into the XhoI site of Pγe2 to generate the plasmid clone. pVGE1 (Fig. 27). pVGE1 contains two human heavy chain variable gene segments upstream of the human γ1 constant gene and associated switch region. A unique SalI 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 pVGE1 is digested with SalI and the XhoI insert of pIGM1 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 XhoI 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 × 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 pIGM1 transgenics, and human IgM and IgG1 in pHC1 transgenics, provides evidence that the transgene sequences function correctly in directing VDJ joining, transcription, and isotype 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 levels 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
approximate
injected transgene
animal # transgene copies per cell human IgM human IgG1
6 pIGM1 1 ++ - 7 pIGM1 0 - -
9 pIGM1 0 - -
10 pIGM1 0 - -
12 pIGM1 0 - -
15 pIGM1 10 ++ -
18 pHC1 0 - -
19 pHC1 1 - -
21 pHC1 <1 - -
26 pHC1 2 ++ +
38 pHC1 5 ++ + 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 pIGM1 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 gammal 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 XhoI 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 γ 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 imgf000130_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.
TABLE 5 J. , Segment Che >ιce
Percent Usage (± 3%)
J. Segment HC1 transgenic Human PBL
Jl 7 1
J2 3 <1
J3 17 9
J4 44 53
J5 3 15
J6 26 22
100% 100%
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 sequences 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'l 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.
TABLE 6 D Segment Choice
Percent Usage (± 3%)
D. Segment HC1 transgenic Human PBL
DLR1 <1 <1
DXP1 3 6
DXP'1 25 19
DAI <1 12
DK1 7 12
DN1 12 22
DIR2 7 4
DM2 <1 2
DLR2 3 4
DHQ52 26 2
? 17 17
100% 100% I. Functionality of VDJ joints
Table γ 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 imgf000133_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)).
TABLE 8 CDR3 Length Distribution
Percent Occurrence (± 3%)
#amino acids in CDR3 HC1 transgenic Human PBL
3-8 26 14
9-12 48 41
13-18 26 37
19-23 <1 7
>23 <1 1
100% 100%
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 λEMBL3/SP6/T7 (Clonetech Laboratories, Inc., Palo Alto, CA) are screened with a 1 kb PacI/HindIII 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,-tcc ctg aga etc tec tgt gca gcc tct gga ttc ace ttc agt-3' Clones that hybridized with both V and J-μ probes are isolated and the DNA sequence 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 XhoI site is adjacent to the 5' end of the insert sequence. A subclone containing a functional VDJ segment is digested with XhoI and Pad (Pad, a rare-cutting enzyme, recognizes a site near the J-m intronic enhancer), and the insert cloned into XhoI/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, XhoI, SalI, HindIII, 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 SalI, HindIII, ClaI, BamHI, and XhoI restriction sites flanked by NotI sites. B. Isolation of Jĸ and Cĸ 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 acg act gga gat taa acg taa gca -3' and the phage clones 136.2 and 136.5 isolated. A 7.4 kb XhoI fragment that includes the Jĸ1 segment was isolated from
136.2 and subcloned into the plasmid pNN03 to generate the plasmid clone p36.2. A neighboring 13 kb XhoI fragment that includes Jk segments 2 through 5 together with the Cĸ gene segment was isolated from phage clone 136.5 and subcloned into the plasmid pNNO3 to generate the plasmid clone p36.5.
Together these two clones span the region beginning 7.2 kb upstream of Jĸ1 and ending 9 kb downstream of Cĸ.
C. Construction of rearranged light chain transgenes
1. pCK1, a Cĸ vector for expressing rearranged variable segments
The 13 kb XhoI insert of plasmid clone p36.5
containing the Cĸ gene, together with 9 kb of downstream sequences, is cloned into the SalI site of plasmid vector pGP1c with the 5' end of the insert adjacent to the plasmid XhoI site. The resulting clone, pCK1 can accept cloned fragments containing rearranged VJĸ segments into the unique 5' XhoI 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-Cĸ intronic enhancer and may contain the human 3' ĸ enhancer. 2. pCK2, a Cĸ vector with heavy chain enhancers for
expressing rearranged variable segments
A 0.9 kb XbaI 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 HindIII and a 1.4 kb Mlul/HindIII fragment of phage clone λl.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, pMHE1, 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/HindIII fragment. This 2.3 kb fragment is isolated and cloned into pGP1c to generate pMHE2. pMHE2 is digested with SalI and the 13 kb XhoI 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/Tγ (Clonetech Laboratories, Inc., Palo Alto, CA) were screened with the human kappa light chain J region containing 3.5 kb XhoI/SmaI fragment of p36.5. Positive clones were tested for hybridization with the
following Vĸ specific oligonucleotide: oligo-65 5' -agg ttc agt ggc agt ggg tct ggg aca gac ttc act etc ace ate agc-3'
Clones that hybridized with both V and J probes are isolated and the DNA sequence of the rearranged VJĸ 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 XhoI 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 VJĸ 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. pJCK1, a Jĸ, Cĸ containing vector for constructing
minilocus transgenes
The 13 kb Cĸ containing XhoI insert of p36.5 is treated with Klenow enzyme and cloned into HindIII digested, Klenow-treated, plasmid pGP1d. A plasmid clone is selected such that the 5' end of the insert is adjacent to the vector derived ClaI site. The resulting plasmid, p36.5-1d, is digested with ClaI and Klenow-treated. The Jĸ1 containing γ.4 kb XhoI insert of p36.2 is then Klenow-treated and cloned into the ClaI, Klenow-treated p36.5-1d. A clone is selected in which the p36.2 insert is in the same orientation as the p36.5 insert. This clone, pJCK1 (Fig. 34), contains the entire human Jĸ region and Cĸ, together with 7.2 kb of upstream sequences and 9 kb of downstream sequences. The insert also contains the human J-Cĸ intronic enhancer and may contain a human 3' ĸ enhancer. The insert is flanked by a unique 3' SalI site for the purpose of cloning additional 3' flanking sequences such as heavy chain or light chain enhancers. A unique XhoI site is located at the 5' end of the insert for the purpose of cloning in unrearranged Vĸ gene segments. The unique SalI and XhoI 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 Vĸ gene segments and generation of transgenic animals expressing human Ig light chain protein
The Vĸ specific oligonucleotide, oligo-65 (discussed above), is used to probe a human placental genomic DNA library cloned into the phage vector 1EMBL3/SP6/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 donor sequences, and intact recombination
sequence. DNA fragments containing selected variable gene segments are cloned into the unique XhoI 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 lysed 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 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 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., Technigue 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 670-830 kb NotI fragment
described above containing multiple copies of V1 through V5 is isolated as described. (Berman et al. (1988), 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 pUClδ. 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 XhoI site. The plasmid clone pCK1 is used as a light chain expression cassette and rearranged VJ
sequences are cloned into the XhoI 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 (HC1); these mice are designated as line 112. Blood was washed and
resuspended in 50 mis Hanks' and diluted to 1×108 cells/ml 0.2 mis (2×107 cells) were then injected interperitoneally using a 28 gauge needle and 1 cc 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 × 107 cells was given intravenously through the tail to enhance the production of hybridomas.
Figure imgf000144_0001
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 cvtometry
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 (1x 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 XhoI 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, BglII, Asp718, XhoI, BamHI, HindIII, NotI.
Construction of pBCE1.
A 0.8 kb Xbal/BglII 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/BglII 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 BglII/EcoRI digested pGHlh. The resulting plasmid is digested with BamHI and the BamHI/BglII of pVH251N is inserted in the same
orientation as the hGH 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 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 HindIII and a 1.4 kb Mlul (klenow) /HindIII 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/HindIII fragment.
The BamHI/HindIII fragment of pMHE1 is cloned into BamHI/HindIII cut pVhgh to generate the B-cell expression vector pBCE1. This vector, depicted in Fig. 36, contains unique XhoI 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 promoterenhancer 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 pBCE1 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 Asp718 and XhoI and cloned into Asp718/XhoI digested pBCE1 to generate the antisense transgene construct pMAS1. The purified NotI insert of pMASl 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 pMASl 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 XhoI 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
Figures 37A-37D illustrate the response of three mouse littermates 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, (HC1, 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 HC1-26 line and three mice of the HC1-57 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 HC1-57 at Day 14, and later for line HC1-26 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 transgene-encoded 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 HC1-57 transgenic mice, containing multiple copies of the HC1 transgene, were bred with immunoglobulin heavy chain deletion mice to obtain mice that contain the HC1 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 XhoI 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-iniection 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'-aagg ttc agt ggc agt ggg tct ggg aca gac ttc act etc ace ate agcs'), was used to probe a human placental genomic DNA library cloned into the phage vector λEMBL3/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 . 3 f
A 3 kb Xba fragment of phage clone λ65.3 was subcloned into pUC19 so that the vector derived SalI 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 XhoI site is proximal to the 5' end of the insert and the vector derived SalI site 3'. The resulting plasmid is designated p65.5gl.
(1 c) p65.8
A 6.5 kb HindIII 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/HindIII subfragment, which was cloned into pGP1f. The resulting clone, p65.15f, has unique XhoI and SalI sites located at the respective 5' and 3' ends of the insert.
2. PKV4
The XhoI/SalI insert of p65.8 was cloned into the XhoI site of p65.15f to generate the plasmid pKV2. The
XhoI/Sall insert of p65.5gl was cloned into the XhoI site of pKV2 to generate pKV3. The XhoI/SalI insert of pKV3 was cloned into the XhoI site of p65.3f to generate the plasmid pKV4. This plasmid contains a single 21 kb XhoI/Sall insert that includes 4 functional Vκ gene segments. The entire insert can also be excised with NotI.
3. PKC1B
(3 a) pKcor Two XhoI fragments derived from human genomic DNA phage λ clones were subcloned into plasmid vectors. The first, a 13 kb Jĸ2-Jĸ5/Cĸ containing fragment, was treated with Klenow enzyme and cloned into HindIII 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 ClaI site. The second XhoI fragment, a 7.4 kb piece of DNA containing Jĸ1 was cloned into XhoI/Sall-digested pSP72, such that the 3' insert XhoI site was destroyed by ligation to the vector SalI site. The resulting clone, p36.2s, includes an insert derived ClaI site 4.5 kb upstream of Jĸ1 and a polylinker derived ClaI site downstream in place of the naturally occurring XhoI site between Jĸ1 and Jĸ2. This clone was digested with ClaI to release a 4.7 kb fragment which was cloned into ClaI digested pK-31 in the correct 5' to 3' orientation to generate a plasmid containing all 5 human Jk 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 XhoI and SalI 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. and 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 XhoI site. The resulting plasmid, p24Bf, was cut with XhoI and the 17.7 kb XhoI/Sall fragment of pKcor cloned into it in the same orientation as the enhancer fragment. The resulting plasmid, pKcorB, includes unique XhoI and SalI sites at the 5' and 3' ends of the insert respectively.
(3 c) pKC1B
The XhoI/SalI insert of pKcorB was cloned into the SalI site of p65.3f to generate the light-chain minilocus-transgene plasmid pKClB. This plasmid includes a single functional human Vĸ 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
The two NotI inserts from plasmids pKV4 and pKC1B 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 pKC1B 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.
EXAMPLE 22
This example demonstrates the successful production of a murine hybridoma clone secreting a monoclonal antibody reactive with a specific immunogen, wherein the monoclonal antibody comprises a human immunoglobulin chain encoded by a human Ig transgene.
Generation of Monoclonal Antibodies Incorporating Human Heavy Chain Transgene Product
1. Immunization of Mouse Harboring Human Heavy Chain
Transgene
A mouse containing a human heavy chain encoding transgene and homozygous for knockout (i.e., functional disruption) of the endogenous heavy chain locus (see. EXAMPLE 20, supra) was immunized with purified human CEA, and spleen cells were subsequently harvested after a suitable immune response period. The murine spleen cells were fused with mouse myeloma cells to generate hybridomas using conventional techniques (see, Kohler and Milstein, Eur. J. Immunol., 6:511-519 (1976); Harlow and Lane, Antibodies: A Laboratory Manual, Cold Spring Harbor, New York (1988)). The mouse used for immunization contained a human unrearranged heavy chain minilocus transgene which comprised a single functional VH gene (VH251), human D and J segments, human μ constant region, and human γ1 constant region genes. The transgenic line from which it originated was designated HC1-57 (supra).
One hundred μg of purified human carcinoembryonic antigen (CEA) (Cyrstal Chem, Chicago, IL or Scripps Labs, San Diego, CA) 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. An additional 20 μg of soluble CEA was administered intravenously on Day 83, followed by 50 μg alum-precipitated CEA in incomplete Freund's adjuvant on Day 92. Human heavy chain responses to CEA were confirmed in serum samples prior to fusion of spleen cells with myleoma cells. The animal was sacrificed on Day 95, the spleen removed and fused with P3X63-Ag8.653 mouse myeloma cells (ATCC CRL 1580, American Type Culture Collection, Rockville, MD) using polyethylene glycol. Two weeks later, supernates from fusion wells were screened for the presence of antibodies specifically reactive with CEA, and which contained human heavy chain μ or γ constant region epitopes by ELISA.
Briefly, purified human CEA was coated onto PVC microtitre plates at 2.5 μg/ml, and incubate with culture supernate diluted 1:4 or 1:5 in PBS, 0.5% Tween-20, 5% chicken serum. Plates were washed, followed by addition of horseradish peroxidase-conjugated goat antiserum specific for human IgG Fc or rabbit antiserum specific for human IgM Fc5Mu (Jackson ImmunoResearch, West Grove, PA). Presence of conjugate bound to captured antibody was determined, after further washing, by the addition of ABTS substrate. Two independent fusion wells were found to contain antibody with substantial binding to CEA. After cloning, both hybridomas were found to be positive for the presence of human μ chain and murine ĸ chain by ELISA. No mouse IgG or IgM were detected using similar assays.
Subcloning of the two independent parent hybridomas resulted in two clones, designated 92-09A-4F7-A5-2 and 92-09A-1D7-1-7-1. Both lines were deposited with the ATCC Patent Culture Depository under the Budapest Treaty and were assigned ATCC Designation HB 11307 and HB 11308, respectively. Culture supernatants from these cell lines were assessed for
specificity by testing for reactivity to several purified target proteins using ELISA. As shown in Fig. 46, ELISA assays for determining the reactivity of the monoclonal antibodies to various antigens demonstrate that only CEA and the CEA-related antigen NCA-2 show significant reactivity, indicating the development of a restricted reactivity for the variable regions of the heterohybrid immunoglobulin molecules.
EXAMPLE 23
This example demonstrates that a rearranged human VDJ gene encoded by a human Ig minilocus transgene may be transcribed as a transcript which includes an endogenous Ig constant region gene, for example by the mechanism of trans-switching, to encode a chimeric human/mouse Ig chain.
Identification of Trans-Switch Transcripts Encoding Chimeric Human-Mouse Heavy Chains
RNA was isolated from a hyperimmunized HC1 line 57 transgenic mouse homozygous for the endogenous heavy chain J segment deletion (supra). cDNA was synthesized according to Taylor et al. (1993) Nucleic Acids Res. 20: 6287, incorporated herein by reference, and amplified by PCR using the following two primers: o-149 (human VH251 ) :
5' -CTA GCT CGA GTC CAA GGA GTC TGT GCC GAG GTG CAG CTG (G,A,T,C)-3'
o-249 (mouse gamma):
5' -GGC GCT CGA GCT GGA CAG GG(A/C) TCC A(G/T)A GTT CCA-3'
Oligonucleotide o-149 is specific for the HC1-encoded variable gene segment VH251, while o-249 hybridizes to both mouse and human gamma sequences with the following order of specificities:
mouse γ1 = mouse γ2b = mouse γ3 > mouse γ2a >> human γ1.
DNA sequences from 10 randomly chosen clones generated from the PCR products was determined and is shown in Fig. 47. Two clones comprised human VDJ and mouse γ1; four clones comprised human VDJ and mouse γ2b; and four clones comprised human VDJ and mouse γ3. These results indicate that in a fraction of the transgenic B cells, the transgene-encoded human VDJ recombined into the endogenous murine heavy chain locus by class switching or an analogous recombination.
EXAMPLE 24
This example describes a method for screening a pool of hybridomas to discriminate clones which encode chimeric human/mouse Ig chains from clones which encode and express a human Ig chain. For example, in a pool of hybridoma clones made from a transgenic mouse comprising a human Ig heavy chain transgene and homozygous for a J region-disrupted endogenous heavy chain locus, hybridoma clones encoding trans-switched human VDJ-murine constant region heavy chains may be
identified and separated from hybridoma clones expressing human VDJ-human constant region heavy chains.
Sceening Hybridomas to Eliminate Chimeric Ig Chains
The screening process involves two stages, which may be conducted singly or optionally in combination: (1) a preliminary ELISA-based screen, and (2) a secondary molecular characterization of candidate hybridomas. Preferably, a preliminary ELISA-based screen is used for initial
identification of candidate hybridomas which express a human VDJ region and a human constant region.
Hybridomas that show positive reactivity with the antigen (e.g., the immunogen used to elicit the antibody response in the transgenic mouse) are tested using a panel of monoclonal antibodies that specifically react with mouse μ, γ, ĸ, and λ, and human μ, γ, and ĸ. Only hybridomas that are positive for human heavy and light chains, as well as negative for mouse chains, are identified as candidate hybridomas that express human immunoglobulin chains. Thus, candidate
hybridomas are shown to have reactivity with specific antigen and to possess epitopes characteristic of a human constant region.
RNA is isolated from candidate hybridomas and used to synthesize first strand cDNA. The first strand cDNA is then ligated to a unique single-stranded oligonucleotide of predetermined seguence (oligo-X) using RNA ligase (which ligates single-stranded DNA). The ligated cDNA is then amplified in two reactions by PCR using two sets of
oligonucleotide primers. Set H (heavy chain) includes an oligo that specifically anneals to either human μ or human γ1 (depending on the results of the ELISA) and an oligo that anneals to the oligo-X sequence. This prevents bias against detection of particular V segments, including mouse V segments that may have trans-rearranged into the human minilocus. A second set of primers, Set L (light chain), includes an oligo that specifically anneals to human ĸ and an oligo that anneals specifically to oligo-X. The PCR products are molecularly cloned and the DNA sequence of several are determined to ascertain whether the hybridoma is producing a unique human antibody on the basis of sequence comparison to human and murine Ig sequences.
EXAMPLE 25
This example demonstrates production of a transgenic mouse harboring a human light chain (ĸ) minilocus.
Human ĸ Minilocus transgenic mice
KC1
A 13 kb XhoI Jĸ2-Kĸ containing fragment from a phage clone (isolated from a human genomic DNA phage library by hybridization to a ĸ specific oligonucleotide, e.g., supra) was treated with Klenow enzyme and cloned into the Klenow treated HindIII site of pGP1d to produce pK-31. This
destroyed the insert XhoI sites and positioned the unique polylinker derived XhoI site at the 5' end next to Jĸ2. A unique polylinker derived ClaI site is located between this XhoI site and the inset sequences, while a unique polylinker derived SalI site is located at the 3' end of the insert. A 7.5 kb XhoI fragment, containing Jĸ1 and upstream sequences, was also isolated from a human genomic DNA phage clone
(isolated from a human genomic DNA phage library by
hybridization to a ĸ specific oligonucleotide, e.g. supra). This 7.5 kb XhoI fragment was cloned into the SalI site of pSP72 (Promega, Madison, Wisconsin), thus destroying both XhoI sites and positioning a polylinker ClaI site 3' of Jĸ1.
Digestion of the resulting clone with ClaI released a 4.7 kb fragment containing Jk1 and 4.5 kb of upstream sequences.
This 4.7 kb fragment was cloned into the ClaI site of pK-31 to create pKcor. The remaining unique 5' XhoI site is derived from polylinker sequences. A 6.5 kb XhoI/Sall DNA fragment containing the unrearranged human VĸIII gene segment 65.8 (plasmid p65.8, EXAMPLE 21) was cloned into the XhoI site of pKcor to generate the plasmid pKC1. The NotI insert of pKC1 was microinjected into 1/2 day mouse embryos to generate transgenic mice. Two independent pKC1 derived transgenic lines were established and used to breed mice containing both heavy and light chain miniloci. These lines, KC1-673 and KC1-674, were estimated by Southern blot hybridization to contain integrations of approximately 1 and 10-20 copies of the transgenes respectively.
KC1e
The plasmid pMHE1 (EXAMPLES 13 and 18) was digested with BamHI and HindIII to excise the 2.3 kb insert containing both the mouse and human heavy chain J-μ intronic enhancers. This fragment was Klenow treated, ligated to SalI linkers (New England Biolabs, Beverly, Massachusetts), and cloned into the unique 3' SalI site of pKC1 to generate the plasmid pKC1e.
The NotI insert of pKC1e was microinjected into 1/2 day mouse embryos to generate transgenic mice. Four independent pKC1e derived transgenic lines were established and used to breed mice containing both heavy and light chain miniloci. These lines, KC1e-1399, KC1e-1403, KC1e-1527, and KC1e-1536, were estimated by Southern blot hybridization to contain
integrations of approximately 20-50, 5-10, 1-5, and 3-5 copies of the transgene, respectively. PKC2
A 6.8 kb XhoI/SalI DNA fragment containing the unrearranged human VĸIII gene segment 65.5 (plasmid p65.5gl, EXAMPLE 21) was cloned into the unique 5' XhoI site of pKC1 to generate the plasmid pKC2. This minilocus transgene contains two different functional VĸIII gene segments. The NotI insert of pKC2 was microinjected into 1/2 day mouse embryos to generate transgenic mice. Five independent pKC2 derived transgenic lines were established and used to breed mice containing both heavy and light chain miniloci. These lines, KC2-1573, KC2-1579, KC2-1588, KC2-1608, and KC2-1610, were estimated by Southern blot hybridization to contain
integrations of approximately 1-5, 10-50, 1-5, 50-100, and 5-20 copies of the transgene, respectively.
EXAMPLE 26
This example shows that transgenic mice bearing the human ĸ transgene can make an antigen-induced antibody
response forming antibodies comprising a functional human ĸ chain.
Antibody Responses Associated with Human Ig ĸ Light Chain
A transgenic mouse containing the HC1-5γ human heavy chain and KC1e human ĸ transgenes was immunized with purified human soluble CD4 (a human glycoprotein antigen). Twenty μg of purified human CD4 (NEN Research products, Westwood, MA) insolublized by conjugation to polystyrene latex particles (Polysciences, Warrington, PA) was injected intraperitoneally in saline with dimethyldioctadecyl ammonium bromide
(Calbiochem, San Diego, CA) on Day 0, followed by further injections on Day 20 and Day 34.
Retro-orbital bleeds were taken on Days 25 and 40, and screened for the presence of antibodies to CD4, containing human IgM or human IgG heavy chain by ELISA. Briefly,
purified human CD4 was coated onto PVC microtitre plates at
2.5 μg/ml and incubated with culture supernate diluted 1:4/1:5 in PBS, 0.5% Tween-20, 5% chicken serum. Plates were washed, followed by addition of horseradish peroxidase-conjugated goat antiserum specific for human IgG Fc or rabbit antiserum specific for human IgM Fc5Mu (Jackson ImmunoResearch, Westr Grove, PA). Presence of conjugate bound to captured antibody was determined after further washing by addition of ABTS substrate. Human μ reactive with antigen was detected in both bleeds, while there was essentially undetectable γ reactivity. The Day 40 sample was also tested for antigen-reactive human ĸ chain using the same assay with goat anti-human ĸ peroxidase conjugate (Sigma, St. Louis, MO). CD4-binding ĸ reactivity was detected at this time point. - The assay results are shown in Fig. 48.
EXAMPLE 27
This example shows the successful generation of mice which are homozygous for functionally disrupted murine heavy and light chain loci (heavy chain and ĸ chain loci) and which concomitantly harbor a human heavy chain transgene and a human light chain transgene capable of productively rearranging to encode functional human heavy chains and functional human light chains. Such mice are termed "0011" mice, indicating by the two 0's in the first two digits that the mice lack
functional heavy and light chain loci and indicating by the l's in the second two digits that the mice are hemizygous for a human heavy chain transgene and a human light chain
transgene. This example shows that such 0011 mice are capable of making a specific antibody response to a predetermined antigen, and that such an antibody response can involve isotype switching.
0011/0012 Mice: Endogenous Ig Knockout + Human Ig Transgenes
Mice which were homozygous for a functionally disrupted endogenous heavy chain locus lacking a functional JH region (designated JHD++ or JHΔ++) and also harboring the human HC1 transgene, such as the HC1-26 transgenic mouse line described supra , were interbred with mice homozygous for a functionally disrupted endogenous kappa chain locus lacking a functional JH region (designated here as JKD++ or JKΔ++; see Example 9) to produce mice homozygous for functionally
disrupted heavy chain and kappa chain loci (heavy chain/kappa chain knockouts), designated as JHD++/JKD++ and containing a HC1 transgene. Such mice were produced by interbreeding and selected on the basis of genotype as evaluated by Southern blot of genomic DNA. These mice, designated HC1-26+/JKD++/JHD++ mice, were interbred with mice harboring a human kappa chain transgene (lines KC2-1610, KC1e-1399, and KC1e-1527; see Example 25), and Southern blot analysis of genomic DNA was used to identify offspring mice homozygous for functionally disrupted heavy and light chain loci and also hemizygous for the HC1 transgene and the KC2 or KC1e
transgene. Such mice are designated by numbers and were identified as to their genotype, with the following
abbreviations: HC1-26+ indicates hemizygosity for the HC1-26 line human heavy chain minilocus transgene integration; JHD++ indicates homozygosity for JH knockout; JKD++ indicates homozygosity for Jκ knockout; KC2-1610+ indicates hemizygosity for a KC2 human ĸ transgene integrated as in line KC2-1610; KC1e-1527+ indicates hemizygosity for a KC1e human ĸ transgene integrated as in line KC1e-1527; KC1e-1399+ indicates
hemizygosity for a KC1e human ĸ transgene integrated as in line KC1e-1399.
The resultant individual offspring were each given a numerical designation (e.g., 6295, 6907, etc.) and each was evaluated for the presence of JH knockout alleles, Jκ knockout alleles, HC1-26 transgene, and ĸ transgene (KC2 or KC1e) and determined to be either hemizygous (+) or homozygous (++) at each locus. Table 10 shows the number designation, sex, and genotypes of several of the offspring mice.
Table 10
ID No. Sex Ig Code Genotype
6295 M 0011 HC1-26+ ;JHD++;JKD++; KC2-1610+
6907 M 0011 HC1-26+ ;JHD++;JKD++;KC1e-1527+
7086 F 0011 HC1-26+ ;JHD++;JKD++;KC1e-1399+
7088 F 0011 HC1-26+ ;JHD++;JKD++;KC1e-1399+
7397 F 0011 HC1-26+ ;JHD++;JKD++;KC1e-1527+
7494 F 0012 HC1-26+ ;JHD++;JKD++; KC2-1610++
7497 M 0011 HC1-26+ ;JHD++;JKD++;KC1e-1399+
7648 F 0011 HC1-26+ ;JHD++;JKD++; KC2-1610+
7649 F 0012 HC1-26+ ;JHD++;JKD++; KC2-1610++
7654 F 0011 HC1-26+ ;JHD++;JKD++; KC2-1610+
7655 F 0011 HC1-26+ ;JHD++;JKD++; KC2-1610+
7839 F 0011 HC1-26+ ;JHD++;JKD++;KC1e-1399+
7656 F 0001 HC1-26-;JHD++;JKD++; KC2-1610+
7777 F 1100 Col-2141-;JHD+;JKD+ We removed spleens from three 6 week old female mice. Mouse # 7655 was determined by Southern blot
hybridization to be hemizygous for the HC1 (line 26) and KC2 (line 1610) transgene integrations, and homozygous for the JHΔ and JĸΔ targeted deletions of the mouse μ and ĸJ regions.
Mouse #7656 was determined by Southern blot hybridization to be hemizygous for the KC2 (line 1610) transgene integration and homozygous for the JHΔ and JĸΔ targeted deletions of the mouse μ and ĸJ regions. Mouse # 7777 was determined by
Souther blot hybridization to be hemizygous for the JHΔ and
JĸΔ targeted deletions of the mouse μ and ĸJ regions. Because of the recessive nature of these deletions, this mouse should be phenotypically wild-type. Expression of Endogenous Ig Chains in 0011 Mice
FACS analysis using a panel of antibodies reactive with either human μ, mouse μ, hman ĸ, mouse ĸ , or mouse λ was used to sort lymphocytes explanted from (1) a wildtype mouse (7777), (2) a 0001 mouse homozygous for heavy chain and kappa knockout alleles and harboring a human light chain transgene (7656), and (3) a 0011 mouse homozygous for heavy chain and kappa knockout alleles and harboring a human light chain transgene and a human heavy chain transgene (7655).
We prepared single cell suspensions from spleen and lysed the red cells with NH4Cl, as described by Mishell and
Shiigi (Mishell, B.B. & Shiigi, S.M. (eds) Selected Methods in Cellular Immunology. W.H. Freeman & Co., New York, 1980). The lymphocytes are stained with the following reagents:
propidium iodide (Molecular Probes, Eugene, OR), FITC
conjugated anti-human IgM (clone G20-127; Pharmingen, San Diego, CA), FITC conjugated anti-mouse IgM (clone R6-60.2; Pharmingen, San Diego, CA), phycoerythrin conjugated anti-human Igĸ (clone HP6062; CalTag, South San Francisco, CA), FITC conjugated anti-mouse Igλ (clone R26-46; Pharmingen, San Diego, CA) FITC conjugated anti-mouse B220 (clone RA3-6B2; Pharmingen, San Diego, CA), and Cy-Chrome conjugated anti-mouse B220 (clone RA3-6B2; Pharmingen, San Diego, CA). We analyzed the stained cells using a FACScan flow cytometer and LYSIS II software (Becton Dickinson, San Jose, CA).
Macrophages and residual red cells are excluded by gating on forward and side scatter. Dead cells are excluded by gating out propidium iodide positive cells. The flow cytometric data in Figs. 49 and 50 confirms the Southern blot hybridization data and demonstrates that mouse #7655 expresses both human μ and human ĸ and relatively little if any mouse μ or mouse ĸ. Nevertheless a significant fraction of the B cells (about 70-80%) appear to express hybrid Ig receptors consisting of human heavy and mouse λ light chains.
Fig. 49 shows the relative distribution of B cells expressing human μ or mouse μ on the cell surface; 0011 mouse (7655) lymphocytes are positive for human μ but relatively lack mouse μ; 0001 mouse (7656) lymphocytes do not express much human μ or mouse μ; wildtype mouse (7777) lymphocytes express mouse μ but lack human μ.
Fig. 50 shows the relative distribution of B cells expressing human ĸ or mouse ĸ on the cell surface; 0011 mouse (7655) lymphocytes are positive for human ĸ but relatively lack mouse ĸ; 0001 mouse (7656) lymphocytes do not express much human ĸ or mouse ĸ; wildtype mouse (7777) lymphocytes express mouse ĸ but lack human ĸ.
Fig. 51 shows the relative distribution of B cells expressing mouse λ on the cell surface; 0011 mouse (7655) lymphocytes are positive for mouse λ; 0001 mouse (7656) lymphocytes do not express significant mouse λ; wildtype mouse (7777) lymphocytes express mouse λ but at a relatively lower level than the 0011 mouse (7655).
Fig. 52 shows the relative distribution of B cells positive for endogenous mouse λ as compared to human ĸ
(transgene-encoded). The upper left panel shows the results of cells from a wildtype mouse possessing functional
endogenous heavy and light chain alleles and lacking human transgene (s); the cells are positive for mouse lambda. The upper right panel shows cells from a mouse (#5822) having a ĸ knockout background (JKD++) and harboring the human ĸ
transgene intergration of the KC1e-1399 line; the cells are positive for human ĸ or mouse λ in roughly proportional amounts. The lower left panel shows cells from a mouse
(#7132) having a ĸ knockout background (JKD++) and harboring the human ĸ transgene intergration of the KC2-1610 line; more cells are positive for mouse λ than for human ĸ, possibly indicating that the KC2-1610 transgene integration is less efficient than the KC1e-1399 transgene integration. The lower right panel shows cells from a mouse harboring a human ĸ minilocus transgene (KCo4) and lacking a functional endogenous murine ĸ allele. The data presented in Fig. 52 also
demonstrates the variability of phenotypic expression between transgenes. Such variability indicates the desirability of selecting for individual transgenes and/or transgenic lines which express one or more desired phenotypic features
resulting from the integrated transgene (e.g., isotype
switching, high level expression, low murine Ig background). Generally, single or multiple transgene species (e.g., pKC1e, pKC2, KCo4) are employed separately to form multiple
individual transgenic lines differing by: (1) transgene, (2) site(s) of transgene integration, and/or (3) genetic
background. Individual transgenic lines are examined for desired parameters, such as: (1) capability to mount an immune respone to a predetermined antigen, (2) frequency of isotype switching within transgene-encoded constant regions and/or frequency of trans-switching to endogenous (e.g., murine) Ig constant region genes, (3) expression level of transgene-encoded immmunoglobulin chains and antibodies, (4) expression level of endogenous (e.g., murine) immunoglobulin
immunoglobulin sequences, and (5) frequency of productive VDJ and VJ rearrangement. Typically, the transgenic lines which produce the largest concentrations of transgene-encoded (e.g., human) immunoglobulin chains are selected; preferably, the selected lines produce about at least 40 μg/ml of transgene-encoded heavy chain (e.g., human μ or human 7) in the serum of the transgenic animal and/or about at least 100 μg/ml of transgene-encoded light chain (e.g., human ĸ ) .
Mice were examined for their expression of human and murine immmunoglobulin chains in their unimmunized serum and in their serum following immunization with a specific antigen, human CD4. Fig. 53 shows the relative expression of human μ, human γ , murine μ, murine γ , human ĸ, murine ĸ, and murine λ chains present in the serum of four separate unimmunized 0011 mice of various genotypes (nt = not tested); human ĸ
predominates as the most abundant light chain, and human μ and murine γ (putatively a product of trans-switching) are the most abundant heavy chains, with variability between lines present, indicating the utility of a selection step to
identify advantageous genotypic combinations that minimize expression of murine chains while allowing expression of human chains. Mice #6907 and 7088 show isotype switching (cis-switching within the transgene) from human μ to human 7.
Fig. 54 shows serum immunoglobulin chain levels for human μ (huμ), human γ (turγ), human ĸ (h.uĸ), murine μ (msμ), murine γ (msγ), murine ĸ (msĸ), and murine λ (msλ) in mice of the various 0011 genotypes.
Specific Antibody Response in 0011 Mice
An 0011 mouse (#6295) was immunized with an immunogenic dose of human CD4 according to the following immunization schedule: Day 0, intraperitoneal injection of 100 μl of CD4 mouse immune serum; Day 1, inject 20 μg of human CD4 (American Bio-Tech) on latex beads with DDA in 100 μl; Day 15 inject 20 μg of human CD4 (American Bio-Tech) on latex beads with DDA in 100 μl; Day 29 inject 20 μg of human CD4 (American Bio-Tech) on latex beads with DDA in 100 μl; Day 43 inject 20 μg of human CD4 (American Bio-Tech) on latex beads with DDA in 100 μl.
Fig. 55 shows the relative antibody response to CD4 immunization at 3 weeks and 7 weeks demonstrating the presence of human μ, human ĸ, and human γ chains in the anti-CD4 response. Human γ chains are present at significantly
increased abundance in the 7 week serum, indicating that cis- switching within the heavy chain transgene (isotype switching) is occurring in a temporal relationship similar to that of isotype switching in a wildtype animal.
Fig. 56 shows a schematic compilation of various human heavy chain and light chain transgenes. EXAMPLE 28
This example provides for the targeted knockout of the murine λ light chain locus.
Targeted Inactivation of the Murine Lambda Light Chain Locus
Unlike the Ig heavy and kappa light chain loci, the murine VλJλ and Cλ gene segments are not grouped into 3 families arranged in a 5' to 3' array, but instead are
interspersed. The most 5' portion consists of two V segments (Vλ2 and VλX) which are followed, proceeding in a 3'
direction, by two constant region exons, each associated with its own J segment (Jλ2Cλ2 and the pseudogene Jλ4Cλ4). Next is the most extensively used V segment (Vλ1) which is followed by the second cluster of constant region exons (Jλ3Cλ3 and
Jλ1Cλ1,). Overall the locus spans approximate 200 kb, with intervals of -20-90 kb between the two clusters.
Expression of the lambda locus involves
rearrangement of Vλ2 or VλX predominantly to Jλ2 and only rarely further 3' to Jλ3 or Jλ1. Vλ1 can recombine with both Jλ3 and Jλ1. Thus the lambda locus can be mutated in order to fully eliminate recombination and expression of the locus.
The distance between the two lambda gene clusters makes it difficult to inactivate expression of the locus via the generation of a single compact targeted deletion, as was used in inactivating the murine Ig heavy and kappa light chain loci. Instead, a small single deletion which would eliminate expression lambda light chains spans approximately 120 kb, extending from Jλ2Cλ2 to Jλ1Cλ1 (Fig. 57). This removes all of the lambda constant region exons as well as the Vλ1 gene segment, ensuring inactivation of the locus.
Replacement type targeting vectors (Thomas and
Capecchi (1987) op.cit) are constructed in which the deleted 120 kb is replaced with the selectable marker gene, neo, in a PGK expression cassette. The marker is embedded within genomic lambda sequences flanking the deletion to provide homology to the lambda locus and can also contain the HSV-tk gene, at the end of one of the regions of homology, to allow for enrichment for cells which have homologously integrated the vectors. Lambda locus genomic clone sequences are obtained by screening of a strain 129/Sv genomic phage library isogenic to the ES line being targeted, since the use of targeting vectors isogenic to the chromosomal DNA being targeted has been reported to enhance the efficiency of homologous recombination. Targeting vectors are constructed which differ in their lengths of homology to the lambda locus. The first vector (vector 1 in Fig. 58) contains the marker gene flanked by total of approximately 8-12 kb of lambda locus sequences. For targeting events in which replacement vectors mediate addition or detection of a few kb of DNA this has been demonstrated to be a more than sufficient extent of homology (Hasty et al. (1991) op.cit; Thomas et al. (1992) op.cit.).
Vectors with an additional approximately 40-60 kb of flanking lambda sequence are also constructed (vector 2 in Fig. 58). Human Ig miniloci of at least 80 kb are routinely cloned and propagated in the plasmid vector pGP1 (Taylor et al. (1993) op.cit).
An alternative approach for inactivation of the lambda locus employs two independent mutations, for example mutations of the two constant region clusters or of the two V region loci, in the same ES cell. Since both constant regions are each contained within ~6 kb of DNA, whereas one of the V loci spans ~19 kb, targeting vectors are constructed to independently delete the Jλ2Cλ2/Jλ4Cλ4 and the Jλ3Cλ3/Jλ1Cλ1 loci. As shown in Fig. 58, each vector consists of a
selectable marker (e.g., neo or pac) in a PGK expression cassette, surrounded by a total of -8-12 kb of lambda locus genomic DNA blanking each deletion. The HSV-tk gene can be added to the targeting vectors to enrich for homologous recombination events by positive-negative selection. ES cells are targeted sequentially with the two vectors, such that clones are generated which carry a deletion of one of the constant region loci; these clones are then targeted
sequentially with the two vectors, such that clones will be generated which carry a deletion of one of the constant region loci, and these clones are then targeted to generate a
deletion of the remaining functional constant region cluster. Since both targeting events are thus being directed to the same cell, it is preferable to use a different selectable marker for the two targetings. In the schematic example shown in Fig. 58, one of the vectors contains the neo gene and the other the pac (puromycin N-acetyl transferase) gene. A third potential dominant selectable marker is the hyg (hygromycin phosphotransferase) gene. Both the pac and hyg genes can be been inserted into the PGK expression construct successfully used for targeting the neo gene into the Ig heavy and kappa light chain loci. Since the two lambda constant region clusters are tightly linked, it is important that the two mutations reside on the same chromosome. There preferably is a 50% probability of mutating the same allele by two
independent targeting events, and linkage of the mutations is established by their co-segregation during breeding of
chimeras derived from the doubly targeted ES cells.
EXAMPLE 28
This example provides for the targeted knockout of the murine heavy chain locus.
Targeted Inactivation of the Murine Heavy Chain Locus
A homologous recombination gene targeting transgene having the structure shown in Fig. 59 is used to delete at least one and preferably substantially all of the murine heavy chain locus constant region genes by gene targeting in ES cells. Fig. 59 shows a general schematic diagram of a
targeting transgene. Segment (a) is a cloned genomic DNA sequence located upstream of the constant region gene(s) to be deleted (i.e, proximal to the JH genes); segment (b) comprises a positive selection marker, such as pgk-neo; segment (c) is a cloned genomic DNA sequence located downstream of the constant region gene(s) to be deleted (i.e, distal to the constan region gene(s) and and JH genes); and segment (d), which is optional, comprises a negative selection marker gene (e.g., HSV-tk). Fig. 60 shows a map of the murine heavy chain locus as taken from Immunoglobulin Genes, Honjo, T, Alt, FW, and Rabbits TH (eds.) Academic Press, NY (1989) p. 129. A targeting transgene having a structure according to Fig. 59, wherein: (1) the (a) segment is the 11.5 kb insert of clone JH8.1 (Chen et al. (1993) Int. Immunol. 5 : 647) or an equivalent portion comprising about at least 1-4 kb of
sequence located upstream of the murine Cμ gene, (2) the (b) segment is pgk-neo as described supra, (3) the (c) segment comprises the 1674 bp sequence shown in Fig. 61 or a 4-6 kb insert isolated from a phage clone of the mouse Cα gene isolated by screening a mouse genomic clone library with the end-labeled oligonucleotide having the sequence:
5'-gtg ttg cgt gta tea get gaa ace tgg aaa cag ggt gac cag-3' and (4) the (d) segment comprises the HSV-tk expression cassette described supra.
Alternatively, a stepwise deletion of one or more heavy chain constant region genes is performed wherein a first targeting transgene comprises homology regions, i.e., segments (a) and (c), homologous to sequences flanking a constant region gene or genes, a first species of positive selection marker gene (pgk-neo), and an HSV-tk negative selection marker. Thus, the (a) segment can comprise a sequence of at least about 1-4 kb and homologous to a region located upstream of Cγ3 and the (c) segment can comprise a sequence of at least about 1-4 kb and homologous to a region located upstream of Cγ2a. This targeting transgene deletes the Cγ3, Cγ1, Cγ2b, and Cγ2a genes. This first targeting transgene is introduced into ES cells and correctly targeted recombinants are selected (e.g., with G418), producing a correctly targeted C region deletion. Negative selection for loss of the HSV-tk cassette is then performed (e.g., with ganciclovir or FIAU). The resultant correctly targeted first round C deletion
recombinants have a heavy chain locus lacking the Cγ3, Cγ1, Cγ2b, and 0γ2a genes.
A second targeting transgene comprises homology regions, i.e., segments (a) and (c), homologous to sequences flanking a constant region gene or genes, a second species of positive selection marker gene different that the first species (e.g., gpt or pac), and an HSV-tk negative selection marker. Thus, the (a) segment can comprise a sequence of at least about 1-4 kb and homologous to a region located upstream of Ce and the (c) segment can comprise a sequence of at least about 1-4 kb and homologous to a region located upstream of Cα. This targeting transgene deletes the Ce and Cα genes.
This second targeting transgene is introduced into the correctly targeted C-region recombinant ES cells obtained from the first targeting event. Cells which are correctly targeted for the second knockout event (i.e., by homologous recombination with the second targeting transgene) are
selected for with a selection drug that is specific for the second species of positive selection marker gene (e.g., mycophenolic acid to select for gpt; puromycin to select for pac). Negative selection for loss of the HSV-tk cassette is then performed (e.g., with ganciclovir or FIAU). These resultant correctly targeted second round C region
recombinants have a heavy chain locus lacking the Cγ3, Cγ1, Cγ2b, Cγ2a, C∈, and Cα genes.
Correctly targeted first-round or second-round recombinant ES cells lacking one or more C region genes are used for blastocyst injections as described (supra) and chimeric mice are produced. Germline transmission of the targeted heavy chain alleles is established, and breeding of the resultant founder mice is performed to generate mice homozygous for C-region knockouts. Such C-region knockout mice have several advantages as compared to JH knockout mice; for one example, C-region knockout mice have diminished ability (or completely lack the ability) to undergo trans-switching between a human heavy chain transgene and an
endogenous heavy chain locus constant region, thus reducing the frequency of chimeric human/mouse heavy chains in the transgenic mouse. Knockout of the murine gamma genes is preferred, although μ and delta are frequently also deleted by homologous targeting. C-region knockout can be done in conjunction with other targeted lesions int he endogenous murine heavy chain locus; a C-region deletion can be combined with a JH knockout to preclude productive VDJ rearrangement of the murine heavy chain locus and to preclude or reduce trans-switching between a human heavy chain transgene and the murine heavy chain locus, among others. For some embodiments, it may be desirable to produce mice which specifically lack one or more C-region genes of the endogenous heavy chain locus, but which retain certain other C-region genes; for example, it may be preferable to retain the murine Cα gene to allow to
production of chimeric human/mouse IgA by trans-switching, if such IgA confers an advantageous phenotype and does not substantially interfere with the desired utility of the mice. EXAMPLE 29
This example demonstrates ex vivo depletion of lymphocytes expressing an endogenous (murine) immunoglobulin from a lymphocyte sample obtained from a transgenic mouse harboring a human transgene. The lymphocytes expressing murine Ig are selectively depleted by specific binding to an anti-murine immunoglobulin antibody that lacks substantial binding to human immunoglobulins encoded by the transgene(s). Ex Vivo Depletion of Murine Ig-Expressing B-cells
A mouse homozygous for a human heavy chain minilocus transgene (HC2) and a human light chain minilocus transgene
(KCo4) is bred with a C57BL/6 (B6) inbred mouse to obtain 2211 mice (i.e., mice which: are homozygous for a functional endogenous murine heavy chain locus, are homozygous for a functional endogenous murine light chain locus, and which possess one copy of a human heavy chain transgene and one copy of a human light chain transgene). Such 2211 mice also express B6 major and minor histocompatibility antigens. These mice are primed with an immunogenic dose of an antigen, and after approximately one week spleen cells are isolated. B cells positive for murine Ig are removed by solid phase-coupled antibody-dependent cell separation according to standard methods (Wysocki et al. (1978) Proc. Natl. Acad. Sci. (U.S.A.) 75: 2844; MACS magnetic cell sorting, Miltenyi Biotec Inc., Sunnyvale, CA), followed by antibody-dependent
complement-mediated cell lysis (Selected Methods in Cellular Immunology, Mishell BB and Shiigi SM (eds.), W.H. Freeman and Company, New York, 1980, pp.211-212) to substantially remove residual cells positive for murine Ig. The remaining cells in the depleted sample (e.g., T cells, B cells positive for human Ig) are injected i.v., preferably together with additional anti-murine Ig antibody to deplete arising B cells, into a SCID/B6 or RAG/B6 mouse. The reconstitutued mouse is then further immunized for the antigen to obtain antibody and affinity matured cells for producing hybridoma clones.
EXAMPLE 30
Production of Fully Human Antibodies in Somatic Chimeras
A method is described for producing fully human antibodies in somatic chimeric mice. These mice are generated by introduction of embryonic stem (ES) cells, carrying human immunoglobulin (Ig) heavy and light chain transgenes and lacking functional murine Ig heavy and kappa light chain genes, into blastocysts from RAG-1 or RAG-2 deficient mice.
RAG-1 and RAG-2 deficient mice (Mombaerts et al. (1992) Cell 68: 869; Shinkai et al. (1992) Cell 68: 855) lack murine B and T cells due to an inability to initiate VDJ rearrangement and to assemble the gene segments encoding Igs and T cell receptors (TCR). This defect in B and T cell production can be complemented by injection of wild-type ES cells into blastocysts derived from RAG-2 deficient animals. The resulting chimeric mice produce mature B and T cells derived entirely from the injected ES cells (Chen et al.
(1993) Proc. Natl. Acad. Sci. USA 90: 4528).
Genetic manipulation of the injected ES cells is used for introducing defined mutations and/or exogenous DNA constructs into all of the B and/or T cells of the chimeras. Chen et al. (1993), Proc. Natl. Acad. Sci. USA 90:4528-4532) generated ES cells carrying a homozygous inactivation of the Ig heavy chain locus, which, when injected into RAG
blastocysts, produced chimeras which made T cells in the absence of B cells. Transfection of a rearranged murine heavy chain into the mutant ES cells results in the rescue of B cell development and the production of both B and T cells in the chimeras.
Chimeric mice which express fully human antibodies in the absence of murine Ig heavy chain or kappa light chain synthesis can be generated. Human Ig heavy and light chain constructs are introduced into ES cells homozygous for
inactivation of both the murine Ig heavy and kappa light chain genes. The ES cells are then injected into blastocysts derived from RAG2 deficient mice. The resulting chimeras contain B cells derived exclusively from the injected ES cells which are incapable of expressing murine Ig heavy and kappa light chain genes but do express human Ig genes.
Generation of ES cells Homozygous for Inactivation of the Immunoglobulin Heavy and Kappa Light Chain Genes
Mice bearing inactivated Ig heavy and kappa light chain loci were generated by targeted deletion, in ES cells, of Ig JH and Jκ/Cκ sequences, respectively according to known procedures (Chen et al. (1993) EMBO J. 12: 821; and Chen et al. (1993) Int. Immunol. op.cit). The two mutant strains of mice were bred together to generate a strain homozygous for inactivation of both Ig loci. This double mutant strain was used for derivation of ES cells. The protocol used was essentially that described by Robertson (1987, in
Teratocarcinomas and Embryonic Stem Cells: A Practical
Approach, p. 71-112, edited by E.J. Robertson, IRL Press). Briefly, blastocysts were generated by natural matings of homozygous double mutant mice. Pregnant females were
ovariectomized on day 2.5 of gestation and the "delayed" blastocysts were flushed from the uterus on day 7 of gestation and cultured on feeder cells, to help maintain their
undifferentiated state. Stem cells from the inner cell mass of the blastocysts, identifiable by their morphology, were picked, dissociated, and passaged on feeder cells. Cells with a normal karyotype were identified, and male cell lines will be tested for their ability to generate chimeras and
contribute to the germ cells of the mouse. Male ES cells are preferable to female lines since a male chimera can produce significantly more offspring.
Introduction of Human Ig Genes into Mouse Ig Heavy and Kappa Light Chain Deficient ES cells
Human immunoglobulin heavy and light chain genes are introduced into the mutant ES cells as either minilocus constructs, such as HC2 and KC-CO4, or as YAC clones, such as J1.3P. Transfection of ES cells withh human Ig DNAs is carried out by techniques such as electroporation or lipofection with a cationic lipid. In order to allow for selection of ES cells which have incorporated the human DNA, a selectable marker either is ligated to the constructs or is co-transfected with the constructs into ES cells. Since the mutant ES cells contain the neomycin phosphotransferse (neo) gene as a result of the gene targeting events which generated the Ig gene inactivations, different selectable markers, such as
hygromycin phosphotransferase (hyg) or puromycin N-acetyl transferase (pac), are used to introduce the human Ig genes into the ES cells.
The human Ig heavy and light chain genes can be introduced simultaneously or sequentially, using different selectable markers, into the mutant ES cells. Following transfection, cells are selected with the appropriate
selectable marker and drug-resistant colonies are expanded for freezing and for DNA analysis to verify and analyze the integration of the human gene sequences.
Generation of Chimeras
ES clones containing human Ig heavy and light chain genes are injected into RAG-2 blastocysts as described
(Bradley, A. (1987), m Teratocarcinomas and Embryonic Stem Cells: A Practical Approach, p. 113-151, edited by E.J.
Robertson, IRL Press) and transferred into the uteri of pseudopregnant females. Offspring are screened for the presence of human antibodies by ELISA assay of serum samples. Positive animals are used for immunization and the production of human monoclonal antibodies.
EXAMPLE 31
This example describes the introduction, via
homologous recombination in ES cells, of a targeted frameshift mutation into the murine heavy chain locus leading to a deletion of B cells which undergo switch recombination. The frameshifted mice are suitable hosts for harboring non-murine (e.g., human) transgenes encoding human sequence immunoglobulins.
The novel frameshifted mice can be used for expressing non-murine (e.g., human) sequence immunoglobulins encoded by heavy chain transgene (s) and/or light chain
transgene (s), and for the isolation of hybridomas expressing class-switched, affinity matured, human sequence antibodies from introduced transgenes, among other uses. A frameshift is introduced into one of the four mouse JH gene segments and into the first exon of the mouse μ gene. The two introduced frameshift mutations compensate for each other thus allowing for the expression of fully functional murine μ heavy chain when a B cell uses the frameshifted JH for a functional VDJ joint. None of the other three JH segments can be used for functional VDJ joining because of the frameshift in μ, which is not compensated in the remaining JH genes. Alternatively, compensating frameshifts can be engineered into multiple murine JH genes.
A mouse homozygous for a compensated, frameshifted immunoglobulin heavy chain allele has an approximately
physiological level of peripheral B cells, and an
approximately physiological level of serum IgM comprising both murine and human μ. However, B cells recruited into germinal centers frequently undergo a class switch to a non-μ isotype. Such a class switch in B cells expressing the endogenous murine μ chain leads to the expression of a non-compensated frameshift mRNA, since the remaining non-μ CH genes do not possess a compensating frameshift. The resulting B cells do not express a B cell receptor and are deleted. Hence, B cells expressing a murine heavy chain are deleted once they reach the stage of differentiation where isotype switching occurs. However, B cells expressing heavy chains encoded by a non-murine (e.g., human) transgene capable of isotype switching and which does not contain such isotype-restrictive
frameshifts are capable of further development, including isotype switching and/or affinity maturation, and the like.
Therefore, the frameshifted mouse has an impaired secondary response with regard to murine heavy chain (μ) but a significant secondary response with regard to transgene-encoded heavy chains. If a heavy chain transgene that is capable of undergoing class switching is introduced into this mutant background, the non-IgM secondary response is dominated by transgene expressing B cells. It is thus possible to isolate affinity matured human sequence immunoglobulin
expressing hybridomas from these frameshifted mice. Moreover, the frameshifted mice generally possess immunoprotective levels of murine IgM, which may be advantageous where the human heavy chain transgene can encode only a limited
repertoire of variable regions.
For making hybridomas secreting human sequence monoclonal antibodies, transgenic mutant mice are immunized; their spleens fused with a myeloma cell line; and the
resulting hybridomas screened for expression of the transgene encoded human non-μ isotype. Further, the frameshifted mouse may be advantageous over a JH deleted mouse because it will contain a functional μ switch sequence adjacent to a
transcribed VDJ which serves as an active substrate for cis-switching (Gu et al. (1993) Cell 73: 1155); thus reducing the level of trans-switched B cells that express chimeric
human/mouse antibodies.
Construction of Frameshift Vectors
Two separate frameshift vectors are built. One of the vectors is used to introduce 2 nucleotides at the 3' end of the mouse J4 gene segment, and one of the vectors is used to delete those same two nucleotides from the 5' end of exon 1 of the mouse μ gene. 1. JH vector.
A 3.4 kb XhoI/EcoRI fragment covering the mouse heavy chain J region and the μ intronic enhancer is subcloned into a plasmid vector that contains a neomycin resistance gene as well as a herpes thymidine kinase gene under the control of a phosphoglycerate kinase promoter (tk/neo cassette; Hasty et al., (1991) Nature 350: 243). This clone is then used as a substrate for generating 2 different PCR fragments using the following oligonucleotide primers: o-A1 5'- cca cac tct gca tgc tgc aga age ttt tct gta -3' o-A2 5'- ggt gac tga ggt ace ttg ace cca gta gtc cag -3' o-A3 5'-- ggt tac etc agt cac cgt etc etc aga ggt aag aat ggc etc -3'
o-A4 5' - agg etc cac cag ace tct eta gac age aac tac -3'
Oligonucleotides o-A1 and o-A2 are used to amplify a 1.2 kb fragment which is digested with SphI and Kpnl.
Oligonucleotides o-A3 and o-A4 are used to amplify a 0.6 kb fragment which is digested with Kpnl and Xbal. These two digested fragments are then cloned into SphI/Xbal digested plasmid A to produce plasmid B.
Plasmid B contains the 2 nucleotide insertion at the end of the J4 and, in addition, contains a new Kpnl site upstream of the insertion. The Kpnl site is used as a
diagnostic marker for the insertion.
Additional flanking sequences may be cloned into the 5' XhoI site and the 3' EcoRI site of plasmid B to increase its homologous recombination efficiency. The resulting plasmid is then digested with SphI, or another restriction enzyme with a single site within the insert, and
electroporated into embryonic stem cells which are then selected with G418 as described by Hasty et al. (1991) op.cit. Homologous recombinants are identified by Southern blot hybridization and then selected with FIAU as described by Hasty et al. to obtain deleted subclones which contain only the 2 base pair insertion and the new Kpnl site in JH4. These are identified by Southern blot hybridization of Kpnl digested DNA and confirmed by DNA sequence analysis of PCR amplified JH4 DNA.
The resulting mouse contains a JH4 segment that has been converted from the unmutated sequence:
...TGGGGTCAAGGAACCTCAGTCACCGTCTCCTCAG_gtaagaatggcctctcc...
TrpGlyGlnGlyThrSerValThrVAlSerSerGlu
to the mutant sequence:
...TGGGGTCAAGGTACCTCAGTCACCGTCTCCTCAGAGgtaagaatggcctctcc...
TrpGlyGlnGlyThrSerValThrVAlSerSerGlu μ Exon 1 Vector
Using similar in vitro mutagenesis methodology described above to engineer a two base pair insertion into the JH4 gene segment, PCR products and genomic subclones are assembled to create a vector containing a two base pair deletion at the 5' end of the first μ exon. In addition, to mark the mutation, a new Xmnl site is also introduced
downstream by changing an A to a G.
The sequence of the unmutated μ gene is:
...ctggtcctcagAGAGTCAGTCCTTCCCAAATGTCTTCCCCCTCGTC...
GluSerGlnSerPheProAsnValPheProLeuVal The sequence of the mutated μ gene is:
Xmnl
...ctggtcctcag_AGTCAGTCCTTCCCGAATGTCTTCCCCCTCGTC...
SerGInSerPheProAsnValPheProLeuVal
The homologous recombination vector containing the mutant sequence is linearized and electroporated into an ES cell line containing the JH4 insertion. Homologous recombinants are identified from neoraycin-resistant clones. Those homologous recombinants that contain the frameshift insertion on the same chromosome as the JH4 insertion are identified by Southern blot hybridization of Kpnl/BamHI digested DNA. The JH4 insertion is associated with a new Kpnl site that reduces the size of the J-μ intron containing Kpnl/BamHI fragment from the wild type 11.3 kb to a mutant 9 kb. The resulting clones are then selected for deletion of the inserted tk/neo cassette using FIAU. Clones containing the mutant μ exon are
identified by Southern blot hybridization of Xmnl digested DNA. The mutation is confirmed by DNA sequence analysis of PCR amplified μ exonl DNA.
Generation of Frameshifted Mice
The ES cell line containing both the two base pair insertion in JH4, and the two base pair deletion in μ exon 1, is then introduced into blastocyst stage embryos which are inserted into pseudopregnant females to generate chimeras.
Chimeric animals are bred to obtain germline transmission, and the resulting animals are bred to homozygosity to obtain mutant animals homozygous for compensated frameshifted heavy chain loci and having impaired secondary humoral immune responses in B cells expressing murine heavy chains.
A human heavy chain transgene, such as for example pHC1 or pHC2 and the like, may be bred into the murine heavy chain frameshift background by crossbreeding mice harboring such a human transgene into mice having the frameshifted murine IgH locus. Via interbreeding and backcrossing, mice homozygous at the murine IgH locus for μ-compensated
frameshifted murine IgH alleles (i.e., capable of compensated in-frame expression of only murine μ and not murine non-μ chains) and harboring at least one integrated copy of a functional human heavy chain transgene (e.g., pHC1 or pHC2) are produced. Such mice may optionally contain knockout of endogenous murine ĸ and/or λ loci as described supra, and may optionally comprise a human or other non-murine light chain transgene (e.g., pKC1e, pKC2, and the like).
Alternatively, the human transgene(s) (heavy and/or light) may comprise compensating frameshifts, so that the transgene J gene(s) contain a frameshift that is compensated by a frameshift in the transgene constant region gene(s).
Trans-switching to the endogenous constant region genes is uncompensated and produces a truncated or nonsense product; B cells expressing such uncompensated trans-switched
immunoglobulins are selected against and depleted.
EXAMPLE 32
Endogenous Heavy Chain Inactivation by D Region Ablation
This example describes a positive-negative selection homologous recombination vector for replacing the mouse germline immunoglobulin heavy chain D region with a
nonfunctional rearranged VDJ segment. The resulting allele functions within a B cell as a normal non-productive allele, with the allele undergoing intra-allele heavy chain class switching, thereby reducing the level of trans-switching to an active transgene locus.
D Region Targeting Construct
An 8-15 kb DNA fragment located upstream of the murine D region is isolated and subcloned from a mouse strain 129 phage library using an oligonucleotide probe comprising approximately 50 consecutive nucleotides of the published sequence for the DFL16.1 segment listed in GenBank. DFL16.1 is the upstream D segment (i.e., proximal to the V region gene cluster and distal to the constant region gene cluster).
Similarly, a 9.5 kb BamHI fragment containing JH3, JH4, Eμ, Sμ, and the first two coding exons of the μ constant region is isolated and subcloned from a mouse strain 129 genomic phage library.
A 5-10 kb rearranged VDJ is then isolated from a mouse hybridoma (any strain) and a synthetic linker containing a stop codon is inserted into the J segment. The stop linker within the J is preferable to an out-of-frame VDJ junction because of the possibility of V replacement rearrangements.
These three fragments are assembled together with a
PGKneo positive selection cassette and a PGKHSVtk negative selection cassette to form a positive-negative selection vector for eliminating the mouse D region in 129-derived ES cells (e.g., AB1) by homologous recombination. The targeting vector is formed by ligating the 8-15 kb DNA fragment to the positive selection cassette (e.g., PGKneo), which is itself ligated to the rearranged 5-10 kb rearranged VDJ, which is itself ligated to the 9.5 kb BamHI fragment; the negative selection cassette (e.g., PGKHSVtk) is then ligated at either end of the targeting construct. The construction of such a D region targeting vector is shown schematically in Fig. 63.
The D region targeting construct is transferred into AB1 ES cells, positive and negative selection is performed as described above, and correctly targeted ES cells are cloned. The correctly targeted ES cell clones are used for blastocyst injections and chimeric mice are produced. The chimeric mice are bred to produce founder mice harboring a D-region
inactivated heavy chain allele. Interbreeding of offspring is performed to produce homozygotes lacking a functional
endogenous heavy chain locus. Such homozygotes are used to crossbreed to mice harboring human Ig transgenes (e.g., pHC1, pHC2, pKC2, pKC1e, KCo4) to yield (by further backcrossing to the homozygotes lacking a functional D-region) mice lacking a functional endogenous heavy chain locus and harboring a human heavy transgene (and preferably also a human light chain transgene). In embodiments where some functional endogenous light chain loci remain (e.g., λ loci), it is generally preferred that transgenes contain transcriptional control sequences that direct high level expression of human light chain (e.g., ĸ) polypeptides, and thus allow the transgene locus to compete effectively with the remaining endogenous light chain (e.g., λ) loci. For example, the Co4 kappa light chain transgene is generally preferred as compared to pKC1 with regard to the ability to compete effeectively with the endogenous λ loci in the transgenic animal.
EXAMPLE 33
This example describes expansion of the human light chain transgene V gene repertoire by co-injection of a human ĸ light chain minilocus and a yeast artificial chromosome comprising a portion of the human Vĸ locus.
Introduction of Functional Human Light Chain V Segments bv Co-Injection of Vκ-Containing YAC DNA and a ĸ Minilocus
An approximately 450 kb YAC clone containing part of the human Vĸ locus was obtained as a non-amplified YAC DNA from clone 4xl7E1 of the publicly available ICRF YAC library (Larin et al. (1991) Proc. Natl. Acad. Sci. (U.S.A.) 88: 4123; Genome Analysis Laboratory, Imperial Cancer Research Fund,
London, UK). The 450 kb YAC clone was isolated without prior amplification by standard pulsed-field gel electrophoresis as per the manufacturer's specifications (CHEF DR-II
electrophoresis cell, Bio-Rad Laboratories, Richmond, CA).
Six individual pulse field gels were stained with ethidium bromide and the gel material containing the YAC clone DNA was excised from the gel and then embedded in a new (low melting point agarose in standard gel buffer) gel cast in a triangular gel tray. The resulting triangular gel (containing the six excised YAC-containing gel blocks) was extended at the apex with a narrow agarose gel with 2 M NaOAc in addition to the standard electrophoresis buffer. The gel was then placed in an electrophoresis chamber immersed in standard gel buffer. The Y-shaped gel former rises above the surface of the buffer so that current can only flow to the narrow high salt gel portion. A plexiglas block was placed over the high salt gel slice to prevent diffusion of the NaOAc into the buffer. The YAC DNA was then electrophoresed out of the original excised gel sliced (embedded) and into the narrow high salt gel portion. At the point of transition from the low salt gel to the high salt gel, there is a resistance drop that effectively halts the migration of the DNA at the apex of the triangular gel.
Following electrophoresis and staining with ethidium bromide, the concentrated YAC DNA was cut away from the rest of the gel and the agarose was digested with GELase (Epicentre Technologies, Madison, Wisconsin). Cesium chloride was then added to the resultant YAC-containing liquid to obtain a density of 1.68 g/ml. This solution was centrifuged at 37,000 rpm for 36 hours to separate the YAC DNA from any
contaminating material. 0.5 ml fractions of the resulting density gradient were isolated and the peak DNA fraction was dialyzed against 5 mM Tris (pH 7.4), 5 mM NaCl, 0.1 M EDTA.
Following dialysis, the concentration of the resulting 0.65 ml solution of YAC DNA was found to contain 2 μg/ml of DNA. This YAC DNA was mixed with purified DNA insert from plasmids pKClB and pKV4 at a ratio of 20:1:1 (micrograms YAC4×17E1:KC1B:KV4). The resulting 2 μg/ml solution was injected into the pronuclei of half-day B6CBF2 embryos, and 95 surviving microinjected embryos were transferred into the oviducts of pseudopregnant females. Twelve mice which developed from the microinjected embryos were born.
EXAMPLE 34
This example describes class-switching, somatic mutation, and B cell development in immunized transgenic mice homozygous for an inactivated endogenous immunoglobulin locus and containing the HC1 or HC2 heavy chain transgene(s).
To demonstrate that a human sequence germline configuration minilocus can functionally replace the authentic locus, we bred a mouse strain lacking endogenous IgH with strains containing human germline-configuration IgH
transgenes. The two transgene miniloci, HC1 and HC2, include one and four functional variable (V) segments respectively 10 and 16 diversity (D) segments respectively, all six joining (JH) segments, and both the μ and γ1 constant region segments. The miniloci include human cis-acting regulatory sequences╌ such as the JH-μ intronic enhancer and the μ and γ1 switch sequences╌ that are closely linked to the coding segments.
They also include an additional enhancer element derived from the 3' end of the rat IgH locus. We crossed HC1 and HC2 transgenic mice with stem-cell derived mutant mice that lack JH segments (JHD mice) as described (supra) and cannot
therefore undergo functional heavy chain rearrangements. The resulting transgenic-JHD mice contain B cells that are
dependent on the introduced heavy chain sequences.
Immunizations and hybridomas.
We immunized mice by intraperitoneal injections of 50-100μg of antigen. Antigens included human carcinoembryonic antigen (CEA; Crystal Chem, Chicago, IL), hen eggwhite
lysozyme (HEL; Pierce, Rockford, IL), and keyhole limpet hemocyanin (KLH; Pierce, Rockford, IL). For primary
injections we mixed the antigen with complete Freund's
adjuvant, for subsequent injections we used incomplete
Freund's adjuvant (Gibco BRL, Gaithersburg, MD). We fused spleen cells with the non-secreting mouse myeloma P3X63-Ag8.653 (ATCC, CRL1580). We assayed serum samples and
hybridoma supernatants for the presence of specific and non-specific antibody comprising human heavy chain sequences by ELISA. For detection of non-specific antibodies we coated microtiter wells with human heavy chain isotype specific antibody (mouse MAb α human IgG1, clone HP6069, Calbiochem, La Jolla, CA; mouse MAb α human IgM, clone CH6, The Binding Site, Birmingham, UK) and developed with peroxidase conjugated antisera (horseradish peroxidase conjugated affinity purified fab fragment from polyclonal goat α human IgG(fc), cat # 109-036-098; affinity purified horseradish peroxidase conjugated polyclonal rabbit α human IgM(fc), cat # 309-035-095. Jackson Immuno Research, West Grove, PA). For detection of antigen-specific antibodies we coated microtiter wells with antigen and developed with peroxidase-conjugated human heavy chain isotype specific antisera. We detected bound peroxidase by incubation with hydrogen peroxide and
2,2'-Azino-bis-(3-Ethylbenzthiazoline-6-Sulfonic Acid, Sigma Chem. Co., St. Louis, MO). The reaction product is measured by absorption at 415 nm, and corrected for absorption at
490 nm.
Flow cytometry.
We prepared single cell suspensions from spleen, bone marrow, and peritoneal cavity, and lysed red cells with NH4Cl, as described by Mishell and Shiigi. The lymphocytes are stained with the following reagents: Phycoerythrin conjugated anti-mouse Igĸ (clone X36; Becton Dickinson, San Jose, CA), FITC conjugated anti-mouse IgD (clone SBA 1,
Southern Biotech, AL), FITC conjugated anti-mouse CD5 (clone 53-7.3; Becton Dickinson, San Jose, CA), FITC conjugated anti-mouse Igλ (clone R26-46; Pharmingen, San Diego, CA), and Cy-Chrome conjugated anti-mouse B220 (clone RA3-6B2; Pharmingen, San Diego, CA). We analyzed the stained cells using a FACScan flow cytometer and LYSIS II software (Becton Dickinson, San Jose, CA). Most macrophages, neutrophils, and residual red cells are excluded by gating on forward and side scatter.
Rescue of B cell compartment
In the peritoneal cavity of HC1 transgenic-JHD animals we find normal levels of CD5+ B cells and
approximately one-quarter the normal level of conventional CD5- B cells. The transgenic peritoneal CD5+ B cells are similar to the so-called B-1 cells described in normal
animals: they are larger than conventional B and T
lymphocytes, they express lower levels of B220 than the conventional B cells found in the spleen, and they include a higher proportion of λ light chain expressing cells. Over 90% of the splenic B cells express ĸ, while up to 50% of the peritoneal B cells express λ. Thus, while the level of conventional B cells is uniformly reduced in all tissues, the level of B-1, which are reported to have a much greater capacity for self-renewal, appears to be normal in the HC1 transgenic-JHD animals.
Class switching.
In transgenic-JHD mice, repeated exposure to antigen results in the production of human γ1 antibodies as well as μ antibodies. We injected human CEA into transgenic-JHD mice at weekly intervals and monitored the serum levels of antigenspecific IgM and IgG1 over a period of four weeks (Fig. 63). At one week there is a detectable IgM response but no IgG1 response. However, the IgG1 response is greater than the IgM response after two weeks, and it continues to increase while the IgM response remains relatively constant. This pattern╌ an initial IgM reaction followed by an IgG reaction╌is typical of a secondary immune response; and it suggests that cis-acting sequences included in the transgene may be
responding to cytokines that direct class switching. We have considered three possible mechanisms for expression of non-μ isotypes, each of which have been discussed in the literature. These mechanisms are: alternative splicing, which does not involve deletion of the μ gene; "δ-type" switching, which involved deletion of the μ gene via homologous recombination between flanking repeat sequences; and non-homologous
recombination between switch regions. The results of our experiments, described below, are indicative of a switch region recombination model.
Two types of non-deletional alternative splicing mechanisms can be invoked to explain an isotype shift. First, it is possible that a single transcript covering both μ and γ1 is expressed from the transgene; this transcript could be alternatively spliced in response to cytokines induced by exposure to antigen. Alternative, a cytokine induced sterile transcript initiating upstream of γ1 could be trans-spliced to the μ transcript. If either of these mechanisms were
responsible for the expression of human γ1 sequences, then we would expect to be able to isolate hybridomas that express both μ and γ1. However, although we have screened several hundred hybridomas expressing either human μ or human γ1, we have not found any such double producer (μ+, γ1+) hybridomas. This indicates that expression of γ1 is accompanied by
deletion of the μ gene.
Deletion of the μ gene can be mediated by non-homologous recombination between the μ and γ1 switch regions, or by homologous recombination between the two flanking 400 bp direct repeats (σμ and ∑μ) that are included in the HC1 and HC2 transgenes. Deletional recombination between σμ and ∑μ has been reported to be responsible for the IgD+, IgM- phenotype of some human B cells. While the first mechanism, non-homologous switch recombination, should generate switch products of varying lengths, the second mechanism, σμ/∑μ recombination, should always generate the same product. We performed a Southern blot analysis of genomic DNA isolated from three hybridomas (Fig. 64A), one expressing μ and two expressing γ1. We find genomic rearrangements upstream of the transgene γ1 only in the two the γ1 switch regions (Fig. 64B) . Furthermore, neither of the observed structures is compatible with homologous recombination between σμ and ∑μ. Our results are therefore consistent with a model for γ1 isotype
expression mediated by deletional non-homologous recombination between the transgene encoded μ and γ1 switch regions.
Trans-switching.
In addition to human γ1, we find mouse γ in the serum of HC1 and HC2 transgenic-JHD mice. We have also obtained mouse γ expressing hybridomas from these animals.
Because the non-transgenic homozygous JHD animals do not express detectable levels of mouse immunoglobulins, we
attribute the expression of mouse γ in the HC1 and HC2
transgenic-JHD animals to the phenomenon of trans-switching. All of the transgenic hybridomas that we have analyzed express either mouse or human constant region sequences, but not both. It is therefore unlikely that a trans-splicing mechanism is involved. We used PCR amplification to isolate cDNA clones of trans-switch products, and determined the nucleotide sequence of 10 of the resulting clones (Fig. 65). The 5' oligonucleotide in the PCR amplification is specific for the transgene encoded VH251, and the 3' oligonucleotide is
specific for mouse γ1, γ2b, and γ3 sequences. We find
examples of trans-switch products incorporating all three of these mouse constant regions.
Somatic mutation.
Approximately 1% of the nucleotides within the variable regions of the trans-switch products shown in Fig. 7 are not germline encoded. This is presumably due to somatic mutation. Because the mutated sequence has been translocated to the endogenous locus, the cis-acting sequences directing these mutations could be located anywhere 3' of the mouse γ switch. However, as we discuss below, we also observe somatic mutation in VDJ segments that have not undergone such
translocations; and this result indicates that sequences required by heavy chain somatic mutation are included in the transgene.
To determine if the HC1 and HC2 constructs include sufficient cis-acting sequences for somatic mutation to occur in the transgenic-JHD mice, we isolated and partially
sequenced cDNA clones derived from two independent HC1
transgenic lines and one HC2 line. We find that some of the γ1 transcripts from transgenic-JHD mice contain V regions with extensive somatic mutations. The frequency of these mutated transcripts appears to increase with repeated immunizations. Figs. 66A and 66B show two sets of cDNA sequences: one set is derived form an HC1 (line 26) transgenic-JHD mouse that we immunized with a single injection of antigen 5 days before we isolated RNA; the second set is derived from an HC1 (line 26) transgenic-JHD mouse that we hyperimmunized by injecting antigen on three different days beginning 5 months before we isolated RNA; the second set is derived from an HC1 (line 26) transgenic-JHD mouse that we hyperimmunized by injecting antigen on three different days beginning 5 months before we isolated RNA. Only 2 of the 13 V regions from the 5 day post-exposure mouse contain any non-germline encoded nucleotides. Each of these V's contains only a single nucleotide change, giving an overall somatic mutation frequency of less than 0.1% for this sample. In contrast, none of the 13 V sequences from the hyperimmunized animal are completely germline, and the overall somatic mutation frequency is 1.6%.
Comparison of μ and γ1 transcripts isolated from a single tissue sample shows that the frequency of somatic mutations is higher in transgene copies that have undergone a class switch. We isolated and partially sequenced 47
independent μ and γ1 cDNA clones from a hyperimmunized CH1 line 57 transgenic-JHD mouse (Fig. 67A and 67B). Most of the μ cDNA clones are unmodified relative to the germline
sequence, while over half of the γ1 clones contain multiple non-germline encoded nucleotides. The γ1 expressing cells are distinct from the μ expressing cells and, while the two processes are not necessarily linked, class switching and somatic mutation are taking place in the same sub-population of B cells.
Although we do not find extensive somatic mutation of the VH251 gene in non-hyperimmunized CHI transgenic mice, we have found considerable somatic mutation in VH56pl and VH51pl genes in a naive HC2 transgenic mouse. We isolated spleen and lymph node RNA from an unimmunized 9 week old female HC2 transgenic animal. We individually amplified γ1 transcripts that incorporate each of the four V regions in the HC2 transgene using V and γ1 specific primers. The relative yields of each of the specific PCR products were
VH56p1>>VH51p1>VH4.21>VH251. Although this technique is not strictly quantitative, it may indicate a bias in V segment usage in the HC2 mouse. Fig. 68 shows 23 randomly picked γ1 cDNA sequences derived from PCR amplifications using an equimolar mix of all four V specific primers. Again we observe a bias toward VH56p1 (19/23 clones). In addition, the VH56pl sequences show considerable somatic mutation, with an overall frequency of 2.1% within the V gene segment.
Inspection of the CDR3 sequences reveals that although 17 of the 19 individual VH56pl clones are unique, they are derived from only 7 different VDJ recombination events. It thus appears that the VH56p1 expressing B cells are selected, perhaps by an endogenous pathogen or self antigen, in the naive animal. It may be relevant that this same gene is over-represented in the human fetal repertoire.
Summary
Upstream cis-acting sequences define the
functionality of the individual switch regions, and are necessary for class switching. Our observation╌that class switching within the HC1 transgene is largely confined to cells involved in secondary response, and does not occur randomly across the entire B cell population╌suggests that the minimal sequences contained with the transgene are
sufficient. Because the γ sequences included in this
construct begin only 116 nucleotides upstream of the start site of the γ1 sterile transcript, the switch regulatory region is compact.
Our results demonstrate that these important cis-acting regulatory elements are either closely linked to individual γ genes, or associated with the 3' heavy chain enhancer included in the HC1 and HC2 transgenes. Because the HC1 and HC2 inserts undergo transgene-autonomous class
switching╌which can serve as a marker for sequences that are likely to have been somatically mutated╌we were able to easily find hypermutated transcripts that did not originate from translocations to the endogenous locus. We found
somatically mutated γ transcripts in three independent
transgenic lines (two HC1 lines and one HC2 line). It is therefore unlikely that sequences flanking the integration sites of the transgene affect this process; instead, the transgene sequences are sufficient to direct somatic mutation.
EXAMPLE 35
This example describes the generation of hybridomas from mice homozygous for an inactivated endogenous
immunoglobulin locus and containing transgene sequences encoding a human sequence heavy chain and human sequence light chain. The hybridomas described secrete monoclonal antibodies comprising a human sequence heavy chain and a human seqeunce light chain and bind to a predetermined antigen expressed on T lymphocytes. The example also demonstrates the capacity of the mice to make a human sequence antibody in response to a human-derived immunogen, human CD4, and the suitability of such mice as a source for making hybridomas secreting human sequence monoclonal antibodies reactive with human antigens.
A. Generation of Human Ig Monoclonal Antibodies Derived from HCl Transgenic Mice Immunized with a Human CD4 Antigen
A transgenic mouse homozygous for a functionally disrupted JH locus and harboring a transgene capable of rearranging to encode a human sequence heavy chain and a transgene capable of rearranging to encode a human sequence light chain was immunized. The genotype of the mouse was HC1-26+ KC1e-1536+ JHD+/+ JKD-, indicating homozygosity for murine heavy chain inactivation and the presence of germline copies of the HC1 human sequence heavy chain transgene and the KC1e human sequence light chain transgene.
The mouse was immunized with a variant of the EL4 cell line (ATCC) expressing a mouse-human hybrid CD4 molecule encoded by a stably transfected polynucleotide. The expressed CD4 molecule comprises a substantially human-like CD4
sequence. Approximately 5 × 106 cells in 100 μl of PBS accompanied by 100 μl of Complete Freund's Adjuvant (CFA) were introduced into the mouse via intraperitoneal injection on Day 0. The inoculation was repeated on Days 7, 14, 21, 28, 60, and 77, with test bleeds on Days 18, 35, and 67. The spleen was removed on Day 81 and approximately 7.2 × 107 spleen cells were fused to approximately 1.2 × 107 fusion partner cells (P3x63Ag8.653 cell line; ATCC) by standard methods (PEG fusion) and cultured in RPMI 1640 15 % FCS, 4 mM glutamine, 1 mM sodium pyruvate plus HAT and PSN medium. Multiple fusions were performed.
Hybridomas were grown up and supernatants were tested with ELISA for binding to a commercial source of purified recombinant soluble human sequence CD4 expressed in CHO cells (American Bio-Technologies, Inc. (ABT), Cambridge, MA) and/or CD4 obtained from NEN-DuPont. The ABT sample contained a purified 55 kD human CD4 molecule comprised the V1 through V3 domains of human CD4. The recombinant human sequence CD4 (produced in CHO-K1 cells) was adsorbed to the assay plate and used to capture antibody from hybridoma supernatants, the captured antibodies were then evaluated for binding to a panel of antibodies which bind either human μ, human ĸ, human γ , murine μ, or murine ĸ.
One hybridoma was subcloned from its culture plate well, designated 1F2. The 1F2 antibody bound to the ABT CD4 preparation, was positive for human μ and human ĸ, and was negative for human γ, mouse γ, and mouse ĸ.
B. Generation of Human Ig Monclonal Antibodies Derived from HC2 Transgenic Mice Immunized with Human CD4 and Human IgE.
The heavy chain transgene, HC2, is shown in Fig. 56 and has been described supra (see, Example 34).
The human light chain transgene, KCo4, depicted in Fig. 56 is generated by the cointegration of two individually cloned DNA fragments at a single site in the mouse genome.
The fragments comprise 4 functional Vĸ segments, 5J segments, the Cĸ exon, and both the intronic and downstream enhancer elements (see Example 21) (Meyer and Neuberger (1989), EMBO J. 8:1959-1964; Judde and Max (1992), Mol. Cell Biol. 12:5206-5216). Because the two fragments share a common 3 kb sequence (see Fig. 56), they can potentially integrate into genomic DNA as a contiguous 43 kb transgene, following homologous
recombination between the overlapping sequences. It has been demonstrated that such recombination events frequently occur upon microinjection of overlapping DNA fragments (Pieper et al. (1992), Nucleic Acids Res. 20:1259-1264). Co-injected DNA's also tend to co-integrate in the zygote, and the
sequences contained within the individually cloned fragments would subsequently be jointed by DNA rearrangement during B cell development. Table 11 shows that transgene inserts from at least 2 of the transgenic lines are functional. Examples of VJ junctions incorporating each of the 4 transgene encoded V segments, and each of the 5J segments, are represented in this set of 36 clones.
Figure imgf000196_0001
Table 11. Human light chain V and J segment usage in KCo4 transgenic mice. The table shows the number of PCR clones, amplified from cDNA derived from two transgenic lines, which contain the indicated human kappa sequences. cDNA was synthesized using spleen RNA isolated from w individual KCo4 transgenic mice (mouse #8490, 3 mo., male, KCo4 line 4437; mouse #8867, 2.5 mo., female, KCo4 line 4436). The cDNA was amplified bv PCR usins a Cĸ specific oligonucleotide, 5'TAG AAG GAA TTC AGC AGG CAC ACA ACA GAG GCA GTT CCA 3' , AND A 1:3 mixture of the followms 2 Vĸ specific oliεonucleotides: 5' AGC TTC TCG AGC TCC TGC TGC TCT GTT TCC CAG GTG CC 3' and 5' CAG CTT CTC GAG CTC CTG CTA CTC TGG CTC (C,A)CA GAT ACC 3'. The PCR product was digested with Xhol and EcoRI, and cloned into a plasmid vector. Partial nucleotide sequences were determined by the dideoxy chain termination method for 18 randomly picked clones from each animal. The sequences of each clone were compared to the germline sequence of the unrearranged transgene.
Twenty-three light chain minilocus positive and 18 heavy chain positive mice developed from the injected embryos. These mice, and their progeny, were bred with mice containing targeted mutations in the endogenous mouse heavy (strain JHD) and ĸ light chain loci (strain JCKD) to obtain mice containing human heavy and ĸ light chain in the absence of functional mouse heavy and ĸ light chain loci. These mice contain only λB cells.
Table 12 shows that somatic mutation occurs in the variable regions of the transgene-encoded human heavy chain transcripts of the transgenic mice. Twenty-three cDNA clones from a HC2 transgenic mouse were partially sequenced to
determine the frequency of non-germline encoded nucleotides within the variable region. The data include only the
sequence of V segment codons 17-94 from each clone, and does not include N regions. RNA was isolated from the spleen and lymph node of mouse 5250 (HC2 line 2550 hemizygouc, JHD
homozygous). Single-stranded cDNA was synthesized and γ transcripts amplified by PCR as described [references]. The amplified cDNA was cloned into plasmid vectors, and 23
randomly picked clones were partially sequenced by the dideoxy chain-termination method. The frequency of PCR-introduced nucleotide changes is estimated from constant region sequence as <0.2%.
TABLE 12: The Variable Regions of Human γ Transcripts in HC2 Transgenic Mice Contain Non-Germline-Encoded Nucleotides
Number of non- Frequency of non-
VH Number of germline encoded germline-encoded
Segment clones nucleotides nucleotides (%)
VH251 0 ╌
VH56P1 10 100 2.1
VH51P1 1 5 2.0
VH4.21 .3 0 0.0
Flow cytometry
We analyzed the stained cells using a FACScan flow cytometer and LYSIS II software (Becton Dickinson, San Jose, CA). Spleen cells were stained with the following reagents: propidium iodide (Molecular Probes, Eugene, OR), phycoerythrin conjugated α-human Igĸ (clone HP6062; Caltag, S. San Francisco, CA), phycoerythrin conjugated α-mouse Igĸ (clone X36; Becton Dickinson, San Jose, CA), FITC conjugated α-mouse Igλ (clone R26-46; Pharmingen, San diego, CA), FITC conjugated α-mouse Igμ (clone R6-60.2; Pharmingen, San Diego, CA), FITC conjugated α-human Igμ (clone G20-127; Pharmingen, San Diego, CA), and Cy-Chrome conjugated α-mouse B220 (clone RA3-6B2;
Pharmingen, San Diego, CA).
Expression of human Ig transgenes
Figure 69 shows a flow cytometric analysis of spleen cells from
KCo4 and HC2 mice that are homozygous for both the JHD and JCKD mutations. The human sequence HC2 transgene rescued B cell development in the JHD mutant background, restoring the
relative number of B220+ cells in the spleen to approximately half that of a wild type animal. These B cells expressed cell surface immunoglobulin receptors that used transgene encoded heavy chain. The human KCo4 transgene was also functional, and competed successfully with the intact endogenous λ light chain locus. Nearly 95% of the splenic B cells in JHD/JCKD
homozygous mutant mice that contain both heavy and light chain human transgenes (double transgenic) expressed completely human cell surface IgMĸ.
Serum Ig levels were determined by ELISA done as follows: human μ: microtiter wells coated with mouse Mab α human IgM (clone CH6, The Binding Site, Birmingham, UK) and developed with peroxidase conjugated rabbit α human IgM(fc) (cat # 309-035-095, Jackson Immuno Research, West Grove, PA). Human γ: microtiter wells coated with mouse MAb α human IgG1 (clone HP6069, Calbiochem, La Jolla, CA) and developed with peroxidase conjugated goat α human IgG(fc) (cat # 109-036-098, Jackson Immuno Research, West Grove, PA). Human ĸ : microtiter wells coated with mouse Mab α human Igĸ (cat # 0173, AMAC, Inc. Igĸ (cat #A7164, Sigma Chem. Co., St. Louis, MO). Mouse 7:
microtiter wells coated with goat α mouse IgG (cat #115-006-071, Jackson Immuno Research, West Grove, PA). Mouse λ: microtiter wells coated with rat MAb α mouse Igλ (cat # 02171D, Pharmingen, San Diego, CA) and developed with peroxidase conjugated rabbit α mouse IgM(fc) (cat # 309-035-095, Jackson Immuno Research, West Grove, PA). Bound peroxidase is detected by incubation with hydrogen peroxide and 2,2'-Azino-bis-)3-Ethylbenzthiazoline-6-Sulfonic Acid, Sigma Chem. Co., St.
Louis, MO) . The reaction product is measured by absorption at 415 nm.
The double transgenic mice also express fully human antibodies in the serum. Figure 70 shows measured serum levels of immunoglobulin proteins for 18 individual double transgenic mice, homozygous for endogenous heavy and kappa light chain inactivations, derived from several different transgenic founder animals. We found detectable levels of human μ, γ1, and ĸ. We have shown supra that the expressed human γ1 results from authentic class switching by genomic recombination between the transgene μ and γ1 switch regions. Furthermore, we have found that intra-transgene class switching was accompanied by somatic mutation of the heavy chain variable regions. In addition to human immunoglobulins, we also found mouse γ and λ in the serum. The present of mouse λ protein is expected because the endogenous locus is completely intact. We have shown elsewhere that the mouse γ expression is a
consequence of trans-switch recombination of transgene VDJ segments into the endogenous heavy chain locus. This trans-switching phenomenon, which was originally demonstrated for wild-type heavy chain alleles and rearranged VDJ transgenes (Durdik et al. (1989), Proc. Natl. Acad. Sci. USA 86:2346-2350; Gerstein et al. (1990), Cell 63:537-548), occurs in the mutant JHD background because the downstream heavy chain constant regions and their respective switch elements are still intact.
The serum concentration of human IgMĸ in the double transgenic mice was approximately 0.1 mg/ml, with very little deviation between animals or between lines. However, human γ1, mouse γ, and mouse λ levels range from 0.1 to
10 micrograms/ml. The observed variation in γ levels between individual animals may be a consequence of the fact that γ is an inducible constant region. Expression presumably depends on factors such as the health of the animal, exposure to antigens, and possibly MHC type. The mouse λ serum levels are the only parameter that appears to correlate with individual transgenic lines. KCo4 line 4436 mice which have the fewest number of copies of the transgene per integration (approximately 1-2 copies) have the highest endogenous λ levels, while KCo4 line 4437 mice (-10 copies per integration) have the lowest λ levels. This is consistent with a model in which endogenous λ rearranges subsequent to the ĸ transgene, and in which the serum λ level is not selected for, but is instead a reflection of the relative size of the precursor B cell pool. Transgene loci containing multiple light chain inserts may have the opportunity to undergo more than one V to J recombination event, with an increased probability that one of them will be functional. Thus high copy lines will have a smaller pool of potential λ cells.
Immunizations with human CD4 and IgE
To test the ability of the transgenic B cells to participate in an immune response, we immunized double
transgenic mice with human protein antigens, and measured serum levels of antigen specific immunoglobulins by ELISA. Mice were immunized with 50 μg recombinant sCD4 (cat. # 013101, American Bio-Technologies Inc., Cambridge, MA) covalently linked to polystyrene beads (cat # 08226, Polysciences Inc., Warrington, PA) in complete Freund's adjuvant by intraperitoneal injection. Each of the mice are homozygous for disruptions of the
endogenous μ and ĸ loci, and hemizygous for the human heavy chain transgene HC2 line 2500 and human ĸ light chain transgene KCo4 line 4437.
Methods
Serum samples were diluted into microtiter wells coated with recombinant sCD4. Human antibodies were detected with peroxidase conjugated rabbit α human IgM(fc) (Jackson
Immuno Research, West Grove, PA) or peroxidase conjugated goat anti-human Igĸ (Sigma, St. Louis, MO). Figure 71A shows the primary response of
transgenic mice immunized with recombinant human soluble CD4. All four of the immunized animals show an antigen-specific human IgM response at one week. The CD4-specific serum
antibodies comprise both human μ heavy chain and human ĸ light chain.
To evaluate the ability of the HC2 transgene to participate in a secondary response, we hyperimmunized the transgenic mice by repeated injection with antigen, and
monitored the heavy chain isotype of the induced antibodies. Mice homozygous for the human heavy chain transgene HC2 and human ĸ light chain transgene KCo4 were immunized with 25 μg of human IgEĸ (The Binding Site, Birmingham, UK) in complete
Freund's adjuvant on day = 0. Thereafter, animals were
injected with IgEĸ in incomplete Freund's adjuvant at
approximately weekly intervals. Serum samples were diluted 1:10, and antigen-specific ELISAs were performed on human IgE, λ coated plates.
Figure 71B shows a typical time course of the immune response from these animals: we injected double
transgenic mice with human IgE in complete Freund's adjuvant, followed by weekly boosts of IgE in incomplete Freund's
adjuvant. The initial human antibody response was IgMĸ, followed by the appearance of antigen specific human IgGĸ. The induced serum antibodies in these mice showed no cross-reactivity to human IgM or BSA. The development, over time, of a human IgG
We have also tested the ability of the heavy chain transgene to undergo class switching in vitro: splenic B cells purified form animals hemizygous for the same heavy chain construct (HC2, line 2550) switch from human IgM to human IgG1 in the presence of LPS and recombinant mouse IL-4. However, in vitro switching did not take place in the presence of LPS and recombinant mouse IL-2, or LPS alone.
In a transgenic mouse immunized with human CD4, human IgG reactivity to the CD4 antigen was detectable at serum concentrations ranging from 2 × 10-2 to 1.6 × 10-4. Identification of Anti-Human CD4 Hybridomas
A transgenic mouse homozygous for the human heavy chain transgene HC2 and human ĸ light chain transgene KCo4 were immunized with 20 μg of recombinant human CD4 in complete
Freund's adjuvant on day 0. Thereafter, animals were injected with CD4 in incomplete Freund's adjuvant at approximately weekly intervals. Fig. 73 shows human antibody response to human CD4 in serum of the transgenic mouse. Serum samples were diluted 1:50, and antigen-specific ELISAs were performed on human CD4 coated plates. Each line represents individual sample determinations. Solid circles represent IgM, open squares represent IgG.
A mouse of line #7494 (0012;HC1-26+;JHD++;JKD++;KC2-1610++) was immunized on days 0, 13, 20, 28, 33, and 47 with human CD4, and produced anti-human CD4 antibodies comprised of human ĸ and human μ or 7.
By day 28, human μ and human ĸ were found present in the serum. By day 47, the serum response against human CD4 comprised both human μ and human 7, as well as human ĸ. On day 50, splenocytes were fused with P3X63-Ag8.653 mouse myeloma cells and cultured. Forty-four out of 700 wells (6.3%)
contained human 7 and/or ĸ anti-human CD4 monoclonal
antibodies. Three of these wells were confirmed to contain human 7 anti-CD4 monoclonal antibodies, but lacked human ĸ chains (presumably expressing mouse λ). Nine of the primary wells contained fully human IgMĸ anti-CD4 monoclonal
antibodies, and were selected for further characterization. One such hybridoma expressing fully human IgMĸ anti-CD4
monoclonal antibodies was designated 2C11-8.
Primary hybridomas were cloned by limiting dilution and assessed for secretion of human μ and ĸ monoclonal antibodies reactive against CD4. Five of the nine hybridomas remained positive in the CD4 ELISA. The specificity of these human IgMĸ monoclonal antibodies for human CD4 was demonstrated by their lack of reactivity with other antigens including ovalbumin, bovine serum albumin, human serum albumin, keyhole limpet hemacyanin, and carcinoembryonic antigen. To determine whether these monoclonal antibodies could recognize CD4 on the surface of cells (i.e., native CD4), supernatants from these five clones were also tested for reactivity with a CD4+ T cell line, Sup T1. Four of the five human IgMĸ monoclonal
antibodies reacted with these CD4+ cells. To further confirm the specificity of these IgMĸ monoclonal antibodies, freshly isolated human peripheral blood lymphocytes (PBL) were stained with these antibodies. Supernatants from clones derived from four of the five primary hybrids bound only to CD4+ lymphocytes and not to CD8+ lymphocytes (Figure 72).
Fig. 72 shows reactivity of IgMĸ anti-CD4
monoclonal antibody with human PBL. Human PBL were incubated with supernatant from each clone or with an isotype matched negative control monoclonal antibody, followed by either a mouse anti-human CD4 monoclonal antibody conjugated to PE (top row) or a mouse anti-human CD8 Ab conjugated to FITC (bottom row). Any bound human IgMĸ was detected with a mouse anti-human μ conjugated to FITC or to PE, respectively.
Representative results for one of the clones, 2C11-8 (right side) and for the control IgMĸ (left side) are shown. As expected, the negative control IgMĸ did not react with T cells and the goat anti-human μ reacted with approximately 10% of PBL, which were presumably human B cells.
Good growth and high levels of IgMĸ anti-CD4 monoclonal antibody production are important factors in
choosing a clonal hybridoma cell line for development. Data from one of the hybridomas, 2C11-8, shows that up to 5
pg/cell/d can be produced (Figure 74). Similar results were seen with a second clone. As is commonly observed, production increases dramatically as cells enter stationary phase growth. Fig. 74 shows cell growth and human IgMĸ anti-CD4 monoclonal antibody secretion in small scale cultures. Replicate cultures were seeded at 2×105 cells/ml in a total volume of 2 ml. Every twenty-four hours thereafter for four days, cultures were harvested. Cell growth was determined by counting viable cells and IgMĸ production was quantitated by an ELISA for total human μ (top panel). The production per cell per day was calculated by dividing the amount of IgMĸ by the cell number (bottom panel). Fig. 75 shows epitope mapping of a human IgMĸ anti-CD4 monoclonal antibody. Competition binding flow
cytometric experiments were used to localize the epitope recognized by the IgM/c anti-CD4 monoclonal antibody, 2C11-8. For these studies, the mouse anti-CD4 monoclonal antibodies,
Leu3a and RPA-T4, which bind to unique, nonoverlapping epitopes on CD4 were used. PE fluorescence of CD4+ cells preincubated with decreasing concentrations of either RPA-TA or Leu-3a followed by staining with 2C11-8 detected with PE-conjugated goat anti-human IgM. IThere was concentration-dependent competition for the binding of the human IgMĸ anti-CD4
monoclonal antibody 2C11-8 by Leu3a but not by RPA-T4 (Figure 75). Thus, the epitope recognized by 2C11-8 was similar to or identical with that recognized by monoclonal antibody Leu3a, but distinct from that recognized by RPA-T4.
In summary, we have produced several hybridoma clones that secrete human IgMĸ monoclonal antibodies that specifically react with native human CD4 and can be used to discriminate human PBLs into CD4+ and CD4- subpopulations. At least one of these antibodies binds at or near the epitope defined by monoclonal antibody Leu3a. Monoclonal antibodies directed to this epitope have been shown to inhibit a mixed leukocyte response (Engleman et al., J. Exp. Med. (1981)
153:193). A chimeric version of monoclonal antibody Leu3a has shown some clinical efficacy in patients with mycosis fungoides (Knox et al. (1991) Blood 77:20).
The association and dissociation rates of the immunizing human CD4 antigen for the monoclonal antibodies secreted by two of the hybridomas, 4E4.2 and 2C5.1, were determined. The experimentally-derived binding constants (Ka) were approximately 9 × 107 M-1 and 8 × 107 M-1 for antibodies 4E4.2 and 2C5.1, respectively. These Ka values fall within the range of murine IgG anti-human CD4 antibodies that have been used in clinical trials by others (Chen et al. (1993) Int.
Immunol. 6: 647).
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. It will be apparent that certain changes and modifications may be practiced within the scope of the claims.
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. Commonly assigned applications U.S.S.N. 08/209,741 filed 9 March 1994, U.S.S.N. 08/165,699 filed 10 December 1993 and U.S.S.N. 08/161,739 filed 03 December 1993, which is a continuation-in-part of 08/155,301 filed 18 November 1993, WO92/03918, USSN 07/810,279 filed 17 December 1991, USSN
07/853,408 filed 18 March 1992, USSN 07/904,068 filed 23 June 1992, USSN 07/990,860 filed 16 December 1992, W093/12227, and USSN 08/053,131 filed 26 April 1993 are each incorporated herein by reference.

Claims

WHAT IS CLAIMED IS:
1. A transgenic non-human animal
comprising: a homozygous pair of functionally disrupted
endogenous heavy chain alleles, a homozygous pair of
functionally disrupted endogenous light chain alleles, at least one copy of a heterologous immunoglobulin heavy chain
transgene, and at least one copy of a heterologous
immunoglobulin heavy chain transgene, and wherein said animal makes an antibody response following immunization with an antigen.
2. A transgenic non-human animal of Claim 1, wherein said functionally disrupted endogenous heavy chain allele is a JH region homologous recombination knockout, said functionally disrupted endogenous light chain allele is a Jκ region homologous recombination knockout, said heterologous immunoglobulin heavy chain transgene is the HC1 or HC2 human minigene transgene, said heterologous light chain transgene is the KC2 or KC1e human ĸ transgene, and wherein said antigen is a human antigen.
3. A transgenic non-human animal of Claim
1, wherein the antibody response comprises a population of antibodies which comprise human μ chain-containing
immunoglobulins and human 7 chain-containing immunoglobulins.
4. A transgenic non-human animal of Claim
2, wherein the heterologous antibodies comprise a population of heterologous immunoglobulins which bind specifically to human CD4 with an dissociation constant of approximately 8 × 107 M-1.
5. A transgenic animal of claim 2 wherein the animal comprises a transgenic mouse having a genotype selected from the group consisting of:
HC1-26+;JHD++;JKD++;KC2-1610++;
HC1-26+;JHD++;JKD++;KC2-1610+;
HC1-26+;JHD++;JKD++;KC1e-1527+; and
HC1-26+;JHD++;JKD++;KC1e-1399+.
6. A transgenic animal of claim 5 wherein the animal comprises a transgenic mouse having a genotype selected from the group consisting of: HC1-26+;JHD++;
JKD++;KC1e-1527+ and HC1-26+;JHD++; JKD++;KC1e-1399+, wherein the antibody response comprises a population of antibodies which comprise human μ chain-containing immunoglobulins and human γ chain-containing immunoglobulins.
7. A transgenic mouse comprising a genome comprising: (1) a homozygous functionally disrupted endogenous heavy chain locus comprising at least one murine constant region gene comprising a functional switch recombination sequence and capable of trans-switching, and (2) a human heavy chain transgene capable of rearranging to encode a functional human heavy chain variable region and containing a functional switch recombination sequence capable of undergoing trans-switching.
8. A transgenic mouse of claim 7, further comprising a human light chain transgene capable of rearranging to encode a functional human light chain variable region and expressing a human sequence light chain.
9. A transgenic mouse of claim 7, further comprising a homozygous functionally disrupted endogenous light chain locus.
10. A transgenic mouse of claim 9, further comprising a serum comprising an antibody comprising a chimeric heavy chain composed of a human sequence variable region encoded by a human transgene and a murine constant region sequence encoded by an endogenous murine heavy chain constant region gene.
11. A transgenic mouse comprising a serum having a detectable amount of a chimeric heavy chain encoded by a sequence produced by trans-switching between a human transgene and an endogenous murine heavy chain constant region gene.
12. A transgenic mouse comprising B cells which produce a human sequence heavy chain at a first timepoint and trans-switch to produce a chimeric heavy chain composed of a human variable region and a murine constant region at a second timepoint.
13. A transgenic mouse comprising B cells which produce a chimeric antibody comprising a chimeric heavy chain comprising a human sequence heavy chain variable region and a murine sequence heavy chain constant region.
14. A transgenic mouse of claim 13, wherein said chimeric antibody comprises a human sequence light chain.
15. A transgenic mouse of claim 14, wherein the chimeric antibody binds to a predetermined antigen (e.g., the immunogen) with an affinity of about at least 1 × 107 M-1.
16. A transgenic mouse of claim 15, wherein the predetermined antigen is human CD4 or human CEA.
17. A transgenic mouse having a genome comprising a human heavy chain transgene comprising two human VH gene segments, eight human D gene segments, six human JH gene segments, a human J-μ enhancer, a human μ switch region, a complete human μ CH gene, a human sterile transcript promoter, a human 7 switch region, a complete human γ CH gene, and a heavy chain 3' enhancer, and wherein said unrearranged human heavy chain transgene lacks mouse VH gene segments, mouse D gene segments, mouse JH gene segments, mouse CH genes, mouse switch regions, and a mouse heavy chain enhancer, and wherein B lymphocytes of said transgenic mouse rearrange said
unrearranged human heavy chain transgene by V-D-J joining to produce a V-D-J gene joined in-frame encoding a heavy chain variable region expressed in polypeptide linkage to the constant region encoded by said complete human μ cc complete human γ CH gene on said transgene.
18. A transgenic mouse of claim 17, wherein said human heavy chain transgene comprises a 5.3 kb HindIII fragment of a human heavy chain gene locus containing the γ1 switch region and the first exon of the preswitch sterile transcript, and wherein said B lymphocytes rearrange said human heavy chain transgene forming a V-D-J gene joined in-frame encoding a heavy chain variable region which is expressed as a human A or human chain in B lymphocytes of said transgenic mouse.
19. A transgenic mouse of claim 18, wherein said transgene further comprises a 0.7 kb Xbal/HindIII fragment of a human heavy chain gene locus, said 0.7 kb
Xbal/HindIII fragment consisting essentially of sequences immediately upstream of, and adjacent to, said 5.3 kb γ1 fragment and further comprising a neighboring upstream 3.1 kb Xbal fragment of said human heavy chain gene locus.
20. A transgenic mouse of claim 19, wherein said human heavy chain transgene comprises a human γ1 constant region including the associated switch region and sterile transcript associated exons, together with
approximately 4 kb flanking sequences upstream of the sterile transcript initiation site, and a rat heavy chain 3' enhancer that can be PCR amplified with the following oligonucleotide primers: 5' CAG GAT CCA GAT ATC AGT ACC TGA AAC AGG GCT TGC 31 51 GAG CAT GCA CAG GAC CTG GAG CAC ACA CAG CCT TCC 3'.
21. A transgenic mouse of claim 20, wherein said human heavy chain transgene comprises a NotI insert of pHC1.
22. A transgenic mouse of claim 21, wherein said transgenic mouse comprises one intact germline copy of said NotI insert of pHC1 and wherein said transgenic mouse expresses both human μ and human γ1 chains in serum.
23. A transgenic mouse of claim 22, wherein said human heavy chain transgene undergoes isotype switching whereby said V-D-J gene joined in-frame encodes a human heavy chain variable region which is initially expressed in peptide linkage to a human μ constant region and
subsequently expressed in peptide linkage to a human γ constant region in B lymphocytes of said transgenic mouse.
24. A transgenic mouse comprising an intact integrated germline copy of a NotI insert of pHC1 or pIGM1, wherein said transgenic mouse expresses human μ and human γ1 chains in serum, each human μ or human γ1 chain comprising a variable region consisting essentially of a polypeptide sequence encoded by a human VH gene segment, a human D gene segment, and a human JH gene segment, joined in-frame as a VDJ gene.
25. A transgenic mouse of claim 17, wherein said transgenic mouse further comprises a functionally disrupted endogenous heavy chain locus which lacks mouse JH gene segments.
26. A transgenic mouse having an intact integrated germline copy of a human heavy chain transgene consisting essentially of a NotI insert of pHC1, wherein said transgenic mouse expresses, in its serum, immunoglobulin chains encoded by said human heavy chain transgene and comprising human or human γ1 constant regions.
27. A method for producing an antibody comprising a human immunoglobulin in serum of a transgenic mouse, said method comprising the step of immunizing with a predetermined antigen a transgenic mouse of claim 17 or claim 25, and collecting serum from said animal after a suitable period for a humoral immune response.
28. A hybridoma comprising a B cell of an transgenic mouse of claim 17 or claim 25 which has been immunized with a predetermined antigen, fused with a second cell capable of immortalizing said B cell, wherein the
hybridoma produces a monoclonal antibody comprising a human heavy chain and wherein said monoclonal antibody binds to said predetermined antigen.
29. A hybridoma of Claim 28, wherein the predetermined antigen is a human antigen.
30. A hybridoma of Claim 29, wherein the human antigen is CEA, CD4, or NCA-2.
31. A hybridoma of Claim 28, wherein the monoclonal antibody binds to a human antigen with an affinity of at least 1 × 107 M-1.
32. A hybridoma of Claim 28, wherein the hybridoma comprises a functionally disrupted murine
immunoglobulin allele.
33. A hybridoma of Claim 31, wherein the monoclonal antibody binds human CD4 with an affinity of approximately 8 × 107 M-1.
34. A human monoclonal antibody produced by a hybridoma of Claim 28.
35. A human monoclonal antibody of Claim 34, wherein said human antigen is CEA, CD4, or NCA-2.
36. An immunoglobulin heavy chain minilocus 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)]g wherein x, y, z, m, n, p, and q are integers and x is 2-100, n is 2-10, y is 2-8, p is 1-10, z is 1-50, q is 0-50, and m is 0-10.
37. An immunoglobulin heavy chain
transgene of claim 36, wherein said transgene is replicated in a mammalian genome.
38. A transgenic mouse of Claim 17 or Claim 25, wherein said heavy chain transgene is a minilocus.
PCT/US1994/004580 1990-08-29 1994-04-25 Transgenic non-human animals capable of producing heterologous antibodies WO1994025585A1 (en)

Priority Applications (8)

Application Number Priority Date Filing Date Title
AU68194/94A AU6819494A (en) 1993-04-26 1994-04-25 Transgenic non-human animals capable of producing heterologous antibodies
CA2161351A CA2161351C (en) 1993-04-26 1994-04-25 Transgenic non-human animals capable of producing heterologous antibodies
EP94916581A EP0754225A4 (en) 1993-04-26 1994-04-25 Transgenic non-human animals capable of producing heterologous antibodies
JP6524481A JPH08509612A (en) 1993-04-26 1994-04-25 Transgenic non-human animal capable of producing heterologous antibody
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 (12)

Application Number Priority Date Filing Date Title
US08/053,131 US5661016A (en) 1990-08-29 1993-04-26 Transgenic non-human animals capable of producing heterologous antibodies of various isotypes
US08/053,131 1993-04-26
US08/096,762 1993-07-22
US08/096,762 US5814318A (en) 1990-08-29 1993-07-22 Transgenic non-human animals for producing heterologous antibodies
US15530193A 1993-11-18 1993-11-18
US08/155,301 1993-11-18
US16173993A 1993-12-03 1993-12-03
US08/161,739 1993-12-03
US16569993A 1993-12-10 1993-12-10
US08/165,699 1993-12-10
US20974194A 1994-03-09 1994-03-09
US08/209,741 1994-03-09

Publications (1)

Publication Number Publication Date
WO1994025585A1 true WO1994025585A1 (en) 1994-11-10

Family

ID=27556670

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1994/004580 WO1994025585A1 (en) 1990-08-29 1994-04-25 Transgenic non-human animals capable of producing heterologous antibodies

Country Status (5)

Country Link
EP (1) EP0754225A4 (en)
JP (3) JPH08509612A (en)
AU (1) AU6819494A (en)
CA (1) CA2161351C (en)
WO (1) WO1994025585A1 (en)

Cited By (703)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1997013852A1 (en) 1995-10-10 1997-04-17 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
WO1997042329A1 (en) * 1996-05-04 1997-11-13 Zeneca Limited Monoclonal antibody to cea, conjugates comprising said antibody, and their therapeutic use in an adept system
EP0822830A1 (en) * 1995-04-27 1998-02-11 Abgenix, Inc. Human antibodies derived from immunized xenomice
EP0823941A1 (en) * 1995-04-28 1998-02-18 Abgenix, Inc. Human antibodies derived from immunized xenomice
WO1998024893A2 (en) * 1996-12-03 1998-06-11 Abgenix, Inc. TRANSGENIC MAMMALS HAVING HUMAN IG LOCI INCLUDING PLURAL VH AND Vλ REGIONS AND ANTIBODIES PRODUCED THEREFROM
EP0871485A1 (en) * 1995-07-21 1998-10-21 University Of Nebraska Board Of Regents COMPOSITIONS AND METHODS FOR CATALYZING HYDROLYSIS OF HIV gp120
WO1998054348A1 (en) * 1997-05-31 1998-12-03 Babraham Institute Telomere-associated chromosome fragmentation
US5914256A (en) * 1995-06-07 1999-06-22 Wohlstadter Jacob N Method for promoting enzyme diversity
US5919681A (en) * 1995-06-07 1999-07-06 Wohlstadter Jacob N Method for promoting enzyme diversity
US5922584A (en) * 1995-06-07 1999-07-13 Wohlstadter Jacob N Method for promoting enzyme diversity
EP0942959A1 (en) * 1996-12-02 1999-09-22 GenPharm International Transgenic non-human animals capable of producing heterologous antibodies
GB2344344A (en) * 1998-11-03 2000-06-07 Babraham Inst Transgenic mice containing Human Ig lambda locus
US6080910A (en) * 1997-02-20 2000-06-27 Case Western Reserve University Transgenic knockout animals lacking IgG3
WO2001018200A1 (en) 1999-09-06 2001-03-15 Chugai Seiyaku Kabushiki Kaisha Tsg-like gene
WO2001019394A2 (en) * 1999-09-15 2001-03-22 Therapeutic Human Polyclonals, Inc. Immunotherapy with substantially human polyclonal antibody preparations purified from genetically engineered birds
WO2003040169A2 (en) 2001-11-07 2003-05-15 Celldex Therapeutics , Inc. Human monoclonal antibodies to dendritic cells
WO2003068259A1 (en) 2002-02-14 2003-08-21 Chugai Seiyaku Kabushiki Kaisha Antibody-containing solution pharmaceuticals
WO2003083116A1 (en) 2002-03-29 2003-10-09 Chugai Seiyaku Kabushiki Kaisha Emthod of screening transporter inhibitor
WO2003104276A2 (en) 2002-06-06 2003-12-18 Oncotherapy Science, Inc. Genes and polypeptides relating to hepatocellular or colorectal carcinoma
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
WO2004038018A1 (en) 2002-10-22 2004-05-06 Eisai Co., Ltd. Gene expressed specifically in dopamine-producing neuron precursor cells after termination of division
WO2004039981A1 (en) 2002-10-30 2004-05-13 Chugai Seiyaku Kabushiki Kaisha Membrane protein originating in mast cells
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
WO2004106515A1 (en) 2003-05-28 2004-12-09 Scimedia Ltd. Anti-bambi antibody and diagnostic or remedy for colon cancer and liver cancer containing the same
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
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
WO2005056605A1 (en) 2003-12-12 2005-06-23 Chugai Seiyaku Kabushiki Kaisha Modified antibodies recognizing trimer receptor or higher
WO2005092926A2 (en) 2004-03-19 2005-10-06 Amgen Inc. Reducing the risk of human and anti-human antibodies through v gene manipulation
WO2005100402A1 (en) 2004-04-13 2005-10-27 F.Hoffmann-La Roche Ag Anti-p-selectin antibodies
AU2003227322B2 (en) * 1995-04-27 2005-12-01 Amgen Fremont Inc. Human Antibodies Derived From Immunized Xenomice
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
WO2006009241A1 (en) 2004-07-22 2006-01-26 Eisai Co., Ltd. Lrp4/CORIN DOPAMINE-PRODUCING NEURON PRECURSOR CELL MARKER
US7041871B1 (en) 1995-10-10 2006-05-09 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
WO2006055638A2 (en) 2004-11-17 2006-05-26 Abgenix, Inc. Fully human monoclonal antibodies to il-13
WO2006068975A2 (en) 2004-12-20 2006-06-29 Abgenix, Inc. Binding proteins specific for human matriptase
US7109003B2 (en) 1998-12-23 2006-09-19 Abgenix, Inc. Methods for expressing and recovering human monoclonal antibodies to CTLA-4
WO2006098464A1 (en) 2005-03-14 2006-09-21 Link Genomics, Inc. Method for diagnosis of prostate cancer
WO2006132363A1 (en) 2005-06-10 2006-12-14 Chugai Seiyaku Kabushiki Kaisha Stabilizer for protein preparation comprising meglumine and use thereof
EP1743947A2 (en) 2002-09-30 2007-01-17 Oncotherapy Science, Inc. Method for diagnosing non-small cell lung cancers
US7192582B2 (en) 2001-02-12 2007-03-20 Medarex, Inc. Human monoclonal antibodies to FC alpha receptor (CD89)
WO2007038637A2 (en) 2005-09-26 2007-04-05 Medarex, Inc. Human monoclonal antibodies to cd70
WO2007037430A1 (en) 2005-09-29 2007-04-05 Eisai R & D Management Co., Ltd. T-cell adhesion molecule and antibody directed against the molecule
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
WO2007058194A1 (en) 2005-11-15 2007-05-24 National Hospital Organization Inhibitor of cytotoxic t cell induction
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
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
WO2007077934A1 (en) 2005-12-28 2007-07-12 Asubio Pharma Co., Ltd. Anti-periostin antibody and pharmaceutical composition for preventing or treating periostin-related disease containing the same
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
US7282568B2 (en) 2002-12-16 2007-10-16 Medarex, Inc. Human monoclonal antibodies against interleukin 8 (IL-8)
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
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
WO2008020586A1 (en) 2006-08-14 2008-02-21 Forerunner Pharma Research Co., Ltd. Diagnosis and treatment of cancer using anti-desmoglein-3 antibody
EP1895001A1 (en) 1998-11-04 2008-03-05 Chugai Seiyaku Kabushiki Kaisha Novel trypsin family serine proteases
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
WO2008047914A1 (en) 2006-10-20 2008-04-24 Forerunner Pharma Research Co., Ltd. Cancer therapeutic agent 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
WO2008047925A1 (en) 2006-10-20 2008-04-24 Forerunner Pharma Research Co., Ltd. Pharmaceutical composition comprising anti-hb-egf antibody as active ingredient
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
WO2008059959A1 (en) 2006-11-17 2008-05-22 The Research Foundation For Microbial Diseases Of Osaka University Nerve elongation promoter and elongation inhibitor
WO2008070569A2 (en) 2006-12-01 2008-06-12 Medarex, Inc. Human antibodies that bind cd22 and uses thereof
US7387776B2 (en) 2002-01-09 2008-06-17 Medarex, Inc. Human monoclonal antibodies against CD30
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
US7396914B2 (en) 2003-08-04 2008-07-08 University Of Massachusetts SARS nucleic acids, proteins, antibodies, 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
WO2008096817A1 (en) 2007-02-09 2008-08-14 Eisai R & D Management Co., Ltd. Gaba neuron progenitor cell marker 65b13
WO2008099920A1 (en) 2007-02-15 2008-08-21 Kyushu University, National University Corporation Therapeutic agent for interstitial pulmonary disease comprising anti-hmgb-1 antibody
EP1961818A2 (en) 1999-04-09 2008-08-27 Chugai Seiyaku Kabushiki Kaisha Novel fetal genes
WO2008105560A1 (en) 2007-02-27 2008-09-04 Forerunner Pharma Research Co., Ltd. Pharmaceutical composition comprising anti-grp78 antibody as active ingredient
WO2008108918A1 (en) 2007-02-21 2008-09-12 University Of Massachusetts Human antibodies against hepatitis c virus (hcv) uses thereof
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
US7435871B2 (en) 2001-11-30 2008-10-14 Amgen Fremont Inc. Transgenic animals bearing human Igλ light chain genes
US7438907B2 (en) 2002-11-15 2008-10-21 Genmab A/S Human monoclonal antibodies against CD25
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
WO2009001940A1 (en) 2007-06-27 2008-12-31 Asubio Pharma Co., Ltd. Cancer remedy containing antibody against peptide encoded by exon-17 of periostin
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
EP2011885A2 (en) 2005-02-10 2009-01-07 Oncotherapy Science, Inc. Method of diagnosing bladder cancer
WO2009008414A1 (en) 2007-07-10 2009-01-15 Shionogi & Co., Ltd. Monoclonal antibody having neutralizing activity against mmp13
EP2019139A1 (en) 2000-01-24 2009-01-28 Sugiyama, Haruo WT1 interacting protein WTIP
WO2009014263A1 (en) 2007-07-26 2009-01-29 Osaka University Agent for treatment of ophthalmia containing interleukin-6 receptor inhibitor as active ingredient
WO2009025116A1 (en) 2007-08-20 2009-02-26 Oncotherapy Science, Inc. Cdh3 peptide and medicinal agent comprising the same
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
WO2009025117A1 (en) 2007-08-20 2009-02-26 Oncotherapy Science, Inc. Cdca1 peptide and pharmaceutical agent comprising the same
EP2030985A1 (en) 1999-09-21 2009-03-04 Chugai Seiyaku Kabushiki Kaisha Transporter genes OATP-B, C, D, and E
US7501496B1 (en) 2004-09-17 2009-03-10 Roche Palo Alto Llc Anti-OX40L 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
WO2009040134A1 (en) 2007-09-26 2009-04-02 U3 Pharma Gmbh Heparin-binding epidermal growth factor-like growth factor antigen binding proteins
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
WO2009051220A1 (en) 2007-10-19 2009-04-23 Immunas Pharma, Inc. Antibody capable of specifically binding to aβ oligomer, and use thereof
WO2009051108A1 (en) 2007-10-15 2009-04-23 Chugai Seiyaku Kabushiki Kaisha Method for production of antibody
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
WO2009072598A1 (en) 2007-12-05 2009-06-11 Chugai Seiyaku Kabushiki Kaisha Therapeutic agent for pruritus
WO2009072604A1 (en) 2007-12-05 2009-06-11 Chugai Seiyaku Kabushiki Kaisha Anti-nr10 antibody and use thereof
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
WO2009084659A1 (en) 2007-12-27 2009-07-09 Chugai Seiyaku Kabushiki Kaisha Solution preparation containing antibody at high concentration
WO2009087978A1 (en) 2008-01-11 2009-07-16 The University Of Tokyo Anti-cldn6 antibody
WO2009088032A1 (en) 2008-01-10 2009-07-16 Shionogi & Co., Ltd. Antibody directed against pcrv
WO2009099176A1 (en) 2008-02-08 2009-08-13 Immunas Pharma, Inc. Antibody capable of binding specifically to aβ-oligomer, and use thereof
EP2090657A2 (en) 2000-08-07 2009-08-19 Centocor Ortho Biotech Inc. Anti-IL-12 antibodies, compositions, methods and uses
EP2090587A1 (en) 2002-09-30 2009-08-19 Oncotherapy Science, Inc. Genes and polypeptides relating to prostate cancers
WO2009122667A1 (en) 2008-04-04 2009-10-08 中外製薬株式会社 Therapeutic for hepatic cancer
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
US7625549B2 (en) 2004-03-19 2009-12-01 Amgen Fremont Inc. Determining the risk of human anti-human antibodies in transgenic mice
US7625559B2 (en) 2004-02-06 2009-12-01 University Of Massachusetts Antibodies against Clostridium difficile toxins and uses thereof
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
EP2135953A1 (en) 1999-06-02 2009-12-23 Chugai Seiyaku Kabushiki Kaisha Novel hemopoietin receptor protein, Nr. 10
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
US7642341B2 (en) 2003-12-18 2010-01-05 Merck Serono S.A. Angiogenesis inhibiting molecules, their selection, production and their use in the treatment of cancer
WO2010015608A1 (en) 2008-08-05 2010-02-11 Novartis Ag Compositions and methods for antibodies targeting complement protein c5
WO2010016590A1 (en) 2008-08-07 2010-02-11 国立大学法人 長崎大学 Therapeutic or prophylactic agent for generalized pain syndrome
EP2159230A1 (en) 2000-08-07 2010-03-03 Centocor Ortho Biotech Inc. Anti-TNF antibodies, compositions, methods and uses
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
EP2172480A2 (en) 2005-02-21 2010-04-07 Chugai Seiyaku Kabushiki Kaisha Methods for producing proteins using hamster IGF-1
US7700742B2 (en) 2001-01-05 2010-04-20 Amgen Fremont Antibodies to insulin-like growth factor I receptor
WO2010054403A1 (en) 2008-11-10 2010-05-14 Alexion Pharmaceuticals, Inc. Methods and compositions for treating complement-associated disorders
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
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
WO2010072740A2 (en) 2008-12-23 2010-07-01 Astrazeneca Ab TARGETED BINDING AGENTS DIRECTED TO α5β1 AND USES THEREOF
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
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
WO2010073972A1 (en) 2008-12-22 2010-07-01 エーザイ・アール・アンド・ディー・マネジメント株式会社 Method for obtaining pancreatic progenitor cell using neph3
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
EP2216046A2 (en) 2004-07-09 2010-08-11 Chugai Seiyaku Kabushiki Kaisha Anti-glypican 3 antibody
EP2216343A1 (en) 2000-05-18 2010-08-11 Japan Tobacco, Inc. Human monoclonal antibody against a costimulatory signal transduction molecule AILIM and pharmaceutical use thereof
US7790863B2 (en) 2003-11-19 2010-09-07 Merck Serono S.A. Angiogenesis inhibiting molecules and their use in the treatment and diagnosis of cancer
US7790160B2 (en) 2004-10-01 2010-09-07 Medarex, Inc. Method of treating CD30 positive lymphomas
WO2010102175A1 (en) 2009-03-05 2010-09-10 Medarex, Inc. Fully human antibodies specific to cadm1
EP2233572A2 (en) 2002-06-06 2010-09-29 Oncotherapy Science, Inc. Genes and polypeptides relating to human colon cancers
WO2010110346A1 (en) 2009-03-24 2010-09-30 独立行政法人理化学研究所 Leukemia stem cell markers
US7807798B2 (en) 1997-05-05 2010-10-05 Amgen Fremont Inc. Human monoclonal antibodies to epidermal growth factor receptor
WO2010112034A2 (en) 2009-04-02 2010-10-07 Aarhus Universitet Compositions and methods for treatment and diagnosis of synucleinopathies
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
WO2010119704A1 (en) 2009-04-17 2010-10-21 Immunas Pharma, Inc. Antibodies that specifically bind to a beta oligomers and use thereof
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
WO2010126137A1 (en) 2009-05-01 2010-11-04 国立大学法人 東京大学 Anti-cadherin antibody
WO2010125003A1 (en) 2009-04-27 2010-11-04 Novartis Ag Compositions and methods for increasing muscle growth
WO2010128407A2 (en) 2009-05-05 2010-11-11 Novimmune S.A. Anti-il-17f antibodies and methods of use thereof
WO2010128398A1 (en) 2009-05-04 2010-11-11 Pangenetics 110 B.V. Antibodies against nerve growth factor (ngf) with enhanced in vivo stability
EP2253646A1 (en) 2000-08-07 2010-11-24 Centocor Ortho Biotech Inc. Anti-dual integrin antibody and compositions and conjugates comprising said antibody
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
US7850962B2 (en) 2004-04-20 2010-12-14 Genmab A/S Human monoclonal antibodies against CD20
EP2261230A1 (en) 2002-09-11 2010-12-15 Chugai Seiyaku Kabushiki Kaisha Protein purification method
WO2010151632A1 (en) 2009-06-25 2010-12-29 Bristol-Myers Squibb Company Protein purifacation by caprylic acid (octanoic acid ) precipitation
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
WO2011016238A1 (en) 2009-08-06 2011-02-10 Immunas Pharma, Inc. Antibodies that specifically bind to a beta oligomers and use thereof
WO2011016239A1 (en) 2009-08-06 2011-02-10 Immunas Pharma, Inc. Antibodies that specifically bind to a beta oligomers and use thereof
EP2284194A1 (en) 2004-12-21 2011-02-16 AstraZeneca AB Antibodies directed to angiopoietin-2 and uses thereof
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
EP2289944A2 (en) 2003-10-10 2011-03-02 Chugai Seiyaku Kabushiki Kaisha Bispecific antibody substituting for functional proteins
WO2011037160A1 (en) 2009-09-24 2011-03-31 中外製薬株式会社 Antibody capable of recognizing hla class i
WO2011037983A1 (en) 2009-09-23 2011-03-31 Medarex, Inc. Cation exchange chromatography
US7931897B2 (en) 2001-02-07 2011-04-26 Chugai Seiyaku Kabushiki Kaisha Therapeutic agent for hematopoietic tumors
EP2316856A1 (en) 2002-10-17 2011-05-04 Genmab A/S Human monoclonal antibodies against CD20
EP2316922A1 (en) 2002-05-24 2011-05-04 Schering Corporation Neutralizing human anti-IGFR antibody
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
WO2011057188A1 (en) 2009-11-06 2011-05-12 Idexx Laboratories, Inc. Canine anti-cd20 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
US7955597B2 (en) 2001-11-14 2011-06-07 Centocor, Inc. Anti-IL-6 antibodies, compositions, methods and uses
WO2011067711A2 (en) 2009-12-01 2011-06-09 Compugen Ltd Novel heparanase splice variant
EP2336780A1 (en) 2005-07-27 2011-06-22 Oncotherapy Science, Inc. Genes and polypeptides relating to prostate cancers
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
WO2011093097A1 (en) 2010-01-29 2011-08-04 株式会社未来創薬研究所 Anti-dll3 antibody
EP2357202A1 (en) 2006-04-10 2011-08-17 AstraZeneca AB Targeted binding agents directed to Upar and uses thereof
WO2011100403A1 (en) 2010-02-10 2011-08-18 Immunogen, Inc Cd20 antibodies 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
US8007797B2 (en) 2006-09-28 2011-08-30 Merck Serono S.A. Junctional adhesion molecule-C (JAM-C) binding compounds and methods of their use
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
WO2011108714A1 (en) 2010-03-04 2011-09-09 中外製薬株式会社 Antibody constant region variant
WO2011110642A2 (en) 2010-03-10 2011-09-15 Genmab A/S Monoclonal antibodies against c-met
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
WO2011130434A2 (en) 2010-04-13 2011-10-20 Celldex Therapeutics Inc. Antibodies that bind human cd27 and uses thereof
WO2011131746A2 (en) 2010-04-20 2011-10-27 Genmab A/S Heterodimeric antibody fc-containing proteins and methods for production thereof
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
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
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
WO2011149051A1 (en) 2010-05-28 2011-12-01 中外製薬株式会社 Antitumor t cell response enhancer
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
WO2011147986A1 (en) 2010-05-27 2011-12-01 Genmab A/S Monoclonal antibodies against her2
WO2011147982A2 (en) 2010-05-27 2011-12-01 Genmab A/S Monoclonal antibodies against her2 epitope
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
WO2011154453A1 (en) 2010-06-09 2011-12-15 Genmab A/S Antibodies against human cd38
WO2011157741A2 (en) 2010-06-15 2011-12-22 Genmab A/S Human antibody drug conjugates against tissue factor
EP2404616A2 (en) 2005-12-13 2012-01-11 AstraZeneca AB Binding proteins specific for insulin-like growth factors and uses thereof
WO2012018404A2 (en) 2010-08-06 2012-02-09 U3 Pharma Gmbh Use of her3 binding agents in prostate treatment
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
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
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
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
US8173127B2 (en) 1997-04-09 2012-05-08 Intellect Neurosciences, Inc. Specific antibodies to amyloid beta peptide, pharmaceutical compositions and methods of use thereof
WO2012061120A1 (en) 2010-10-25 2012-05-10 Regents Of The University Of Minnesota Therapeutic composition for treatment of glioblastoma
EP2452694A1 (en) 2005-06-30 2012-05-16 Janssen Biotech, Inc. Anti-IL-23 antibodies, compositions, methods and uses
AU2008200005B2 (en) * 1996-12-03 2012-05-17 Amgen Fremont Inc. Transgenic Mammals Having Human Ig Loci Including Plural Vh and Vk Regions and Antibodies Produced Therefrom
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
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
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
WO2012069466A1 (en) 2010-11-24 2012-05-31 Novartis Ag Multispecific molecules
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
US8207303B2 (en) 2005-02-18 2012-06-26 Medarex, Inc. Monoclonal antibodies against CD30 lacking in fucosyl residues
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
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
WO2012118042A1 (en) 2011-02-28 2012-09-07 独立行政法人国立循環器病研究センター Medicinal agent for inhibiting metastasis of malignant tumor
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
WO2012136552A1 (en) 2011-04-08 2012-10-11 H. Lundbeck A/S ANTIBODIES SPECIFIC TO PYROGLUTAMATED Αβ
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
WO2012143524A2 (en) 2011-04-20 2012-10-26 Genmab A/S Bispecific antibodies against her2 and cd3
WO2012143523A1 (en) 2011-04-20 2012-10-26 Genmab A/S Bispecifc antibodies against her2
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
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
EP2532679A1 (en) 2005-10-21 2012-12-12 Novartis AG Human antibodies against il13 and therapeutic uses
EP2535355A2 (en) 2005-03-23 2012-12-19 Genmab A/S Antibodies against CD38 for treatment of multiple myeloma
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
WO2013004842A2 (en) 2011-07-06 2013-01-10 Genmab A/S Antibody variants and uses thereof
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
WO2013004841A1 (en) 2011-07-06 2013-01-10 Genmab A/S Modulation of complement-dependent cytotoxicity through modifications of the c-terminus of antibody heavy chains
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
WO2013017691A1 (en) 2011-08-04 2013-02-07 Medizinische Universität Innsbruck Cahgt1p inhibitors for use in the treatment of candidiasis
WO2013017656A1 (en) 2011-08-02 2013-02-07 Medizinische Universität Wien Antagonists of ribonucleases for treating obesity
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
EP2570137A2 (en) 2007-11-07 2013-03-20 Celldex Therapeutics, Inc. Antibodies that bind human dendritic and epithelial cell 205 (DEC-205)
WO2013047752A1 (en) 2011-09-30 2013-04-04 中外製薬株式会社 Antigen-binding molecule for promoting loss of antigens
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
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
EP2586796A1 (en) 2007-10-12 2013-05-01 Novartis AG Compositions and methods for use for antibodies against sclerostin
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
WO2013060867A2 (en) 2011-10-27 2013-05-02 Genmab A/S Production of heterodimeric proteins
WO2013067057A1 (en) 2011-11-01 2013-05-10 Bionomics, Inc. Anti-gpr49 antibodies
WO2013067060A1 (en) 2011-11-01 2013-05-10 Bionomics, Inc. Anti-gpr49 antibodies
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
WO2013075048A1 (en) 2011-11-16 2013-05-23 Amgen Inc. Methods of treating epidermal growth factor deletion mutant viii related disorders
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
WO2013084147A2 (en) 2011-12-05 2013-06-13 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
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
EP2615175A1 (en) 2007-05-31 2013-07-17 Genmab A/S Transgenic animals producing monovalent human antibodies and antibodies obtainable from these animals
EP2626372A1 (en) 2007-03-29 2013-08-14 Genmab A/S Bispecific antibodies and methods for production 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
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
US8552154B2 (en) 2008-09-26 2013-10-08 Emory University Anti-PD-L1 antibodies and uses therefor
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
US8557239B2 (en) 2009-09-14 2013-10-15 Abbvie Inc. Methods for treating psoriasis using antibodies that bind to the P40 subunit of IL-12 and/or IL-23
WO2013166448A1 (en) 2012-05-03 2013-11-07 Amgen Inc. Stable formulations containing anti-pcsk9 antibodies
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
WO2014006217A1 (en) 2012-07-06 2014-01-09 Genmab B.V. Dimeric protein with triple mutations
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
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
US20140017238A1 (en) 2001-02-16 2014-01-16 Regeneron Pharmaceuticals, Inc. Methods of Modifying Eukaryotic Cells
EP2692737A1 (en) 2007-04-20 2014-02-05 Biotie Therapies Corp. Fully human anti-vap-1 monoclonal antibodies
US8653020B2 (en) 2008-01-25 2014-02-18 Aarhus Universitet Selective exosite inhibition of PAPP-A activity against IGFBP-4
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
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
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
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
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
EP2740743A2 (en) 2004-06-01 2014-06-11 Domantis Limited Bispecific fusion antibodies with enhanced serum half-life
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
US8765918B2 (en) 1999-03-25 2014-07-01 Abbott Gmbh & Co., Kg Human antibodies that bind human interleukin-12
WO2014104165A1 (en) 2012-12-27 2014-07-03 中外製薬株式会社 Heterodimerized polypeptide
WO2014108198A1 (en) 2013-01-10 2014-07-17 Genmab B.V. Human igg1 fc region variants and uses thereof
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
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
WO2014122613A1 (en) 2013-02-08 2014-08-14 Novartis Ag Anti-il-17a antibodies and their use in treating autoimmune and inflammatory disorders
EP2769993A1 (en) 2007-12-14 2014-08-27 Novo Nordisk A/S Antibodies against human NKG2D and uses thereof
EP2769990A2 (en) 2004-12-02 2014-08-27 Domantis Limited Bispecific domain antibodies targeting serum albumin and GLP-1 or PYY
WO2014136910A1 (en) 2013-03-08 2014-09-12 国立大学法人大阪大学 ANTIBODY TO PEPTIDE ENCODED BY Exon-21 OF PERIOSTIN, AND PHARMACEUTICAL COMPOSITION FOR PREVENTION OR TREATMENT OF INFLAMMATORY DISEASE CONTAINING SAID ANTIBODY
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
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
WO2014185550A1 (en) 2013-05-16 2014-11-20 Kyoto University Method for determining prognosis of cancer
US8906635B2 (en) 2011-02-28 2014-12-09 Northshore University Healthsystem Methods of diagnosing Clostridium difficile infection
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
US8921526B2 (en) 2013-03-14 2014-12-30 Abbvie, Inc. Mutated anti-TNFα antibodies and methods of their 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
US8945545B2 (en) 2008-03-18 2015-02-03 Abbvie Inc. Methods of treating psoriasis by administration of antibodies to the p40 subunit of IL-12 and/or IL-23
US8946395B1 (en) 2013-10-18 2015-02-03 Abbvie Inc. Purification of proteins using hydrophobic interaction chromatography
WO2015022658A2 (en) 2013-08-14 2015-02-19 Novartis Ag Methods of treating sporadic inclusion body myositis
EP2842968A1 (en) 2005-04-29 2015-03-04 Janssen Biotech, Inc. Anti-IL-6 antibodies, compositions, methods and uses
WO2015033831A1 (en) 2013-09-04 2015-03-12 国立大学法人 大阪大学 Dpp-4-targeting vaccine for treating diabetes
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
EP2853544A1 (en) 2007-11-15 2015-04-01 Chugai Seiyaku Kabushiki Kaisha Monoclonal antibody capable of binding to anexelekto, and use thereof
US9017687B1 (en) 2013-10-18 2015-04-28 Abbvie, Inc. Low acidic species compositions and methods for producing and using the same using displacement chromatography
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
US9045541B2 (en) 2012-02-06 2015-06-02 Inhibrx Llc CD47 antibodies and methods of use thereof
US9051368B2 (en) 2007-01-16 2015-06-09 Abbvie, Inc. Methods for treating psoriasis by administering an antibody which binds an epitope of the p40 subunit of IL-12 and/or IL-23
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
US9062106B2 (en) 2011-04-27 2015-06-23 Abbvie Inc. Methods for controlling the galactosylation profile of recombinantly-expressed proteins
US9062111B2 (en) 2005-12-07 2015-06-23 Medarex, L.L.C. CTLA-4 antibody dosage escalation regimens
US9067990B2 (en) 2013-03-14 2015-06-30 Abbvie, Inc. Protein purification using displacement chromatography
WO2015099165A1 (en) 2013-12-27 2015-07-02 中外製薬株式会社 Method for purifying antibody having low isoelectric point
WO2015097536A2 (en) 2013-12-24 2015-07-02 Janssen Pharmaceutical Nv Anti-vista antibodies and fragments
WO2015099127A1 (en) 2013-12-27 2015-07-02 中外製薬株式会社 Fgfr gatekeeper mutant gene and drug targeting same
WO2015099167A1 (en) 2013-12-27 2015-07-02 国立大学法人大阪大学 Vaccine targeting il-17a
US9085618B2 (en) 2013-10-18 2015-07-21 Abbvie, Inc. Low acidic species compositions and methods for producing and using the same
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
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
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
US9150658B2 (en) 2008-12-09 2015-10-06 Genmab A/S Human antibodies against tissue factor and methods of use thereof
EP2927244A1 (en) 2008-09-19 2015-10-07 MedImmune, LLC Antibodies directed to DLL4 and uses thereof
WO2015150900A2 (en) 2014-03-31 2015-10-08 Debiopharm International Sa Fgfr fusions
WO2015162590A1 (en) 2014-04-24 2015-10-29 Novartis Ag Methods of improving or accelerating physical recovery after surgery for hip fracture
DE202008018562U1 (en) 2007-08-23 2015-11-02 Amgen Inc. Antigen-binding Proteins Against Proprotein Convertase Subtilisin Kexin Type 9 (PCSK9)
US9181337B2 (en) 2013-10-18 2015-11-10 Abbvie, Inc. Modulated lysine variant species compositions and methods for producing and using the same
US9181572B2 (en) 2012-04-20 2015-11-10 Abbvie, Inc. Methods to modulate lysine variant distribution
US9193787B2 (en) 2012-04-20 2015-11-24 Abbvie Inc. Human antibodies that bind human TNF-alpha and methods of preparing the same
US9206390B2 (en) 2012-09-02 2015-12-08 Abbvie, Inc. Methods to control protein heterogeneity
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
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
WO2015198243A2 (en) 2014-06-25 2015-12-30 Novartis Ag Compositions and methods for long acting proteins
US9234033B2 (en) 2012-09-02 2016-01-12 Abbvie, Inc. Methods to control protein heterogeneity
WO2016005593A1 (en) 2014-07-11 2016-01-14 Genmab A/S Antibodies binding axl
WO2016007414A1 (en) 2014-07-08 2016-01-14 New York University Tau imaging ligands and their uses in the diagnosis and treatment of tauopathy
US9249182B2 (en) 2012-05-24 2016-02-02 Abbvie, Inc. Purification of antibodies using hydrophobic interaction chromatography
WO2016016859A1 (en) 2014-07-31 2016-02-04 Amgen Research (Munich) Gmbh Optimized cross-species specific bispecific single chain antibody constructs
WO2016016415A1 (en) 2014-07-31 2016-02-04 Amgen Research (Munich) Gmbh Bispecific single chain antibody construct with enhanced tissue distribution
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
WO2016016412A1 (en) 2014-07-31 2016-02-04 Amgen Research (Munich) Gmbh Antibody constructs for cdh19 and cd3
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)
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
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
US9334319B2 (en) 2012-04-20 2016-05-10 Abbvie Inc. Low acidic species compositions
WO2016073894A1 (en) 2014-11-07 2016-05-12 Eleven Biotherapeutics, Inc. Therapeutic agents with increased ocular retention
WO2016073685A1 (en) 2014-11-05 2016-05-12 Annexon, Inc. Humanized anti-complement factor c1q antibodies and uses thereof
WO2016073401A1 (en) 2014-11-03 2016-05-12 Bristol-Myers Squibb Company Use of caprylic acid precipitation for protein purification
US9346873B2 (en) 2008-09-30 2016-05-24 Ablexis, Llc Non-human mammals for the production of chimeric antibodies
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
WO2016097865A1 (en) 2014-12-19 2016-06-23 Regenesance B.V. Antibodies that bind human c6 and uses thereof
US9428574B2 (en) 2011-06-30 2016-08-30 Compugen Ltd. Polypeptides and uses thereof for treatment of autoimmune disorders and infection
WO2016149088A1 (en) 2015-03-13 2016-09-22 Bristol-Myers Squibb Company Use of alkaline washes during chromatography to remove impurities
WO2016153978A1 (en) 2015-03-20 2016-09-29 Bristol-Myers Squibb Company Use of dextran to enhance protein purification by affinity chromatography
WO2016153983A1 (en) 2015-03-20 2016-09-29 Bristol-Myers Squibb Company Use of dextran for protein purification
KR20160116056A (en) 2008-08-14 2016-10-06 테바 파마슈티컬즈 오스트레일리아 피티와이 엘티디 Anti-il-12/il-23 antibodies
WO2016166014A1 (en) 2015-04-17 2016-10-20 F. Hoffmann-La Roche Ag Combination therapy with coagulation factors and multispecific antibodies
WO2016166360A1 (en) 2015-04-17 2016-10-20 Bayer Pharma Aktiengesellschaft Bispecific antibody constructs for cdh3 and cd3
US9499614B2 (en) 2013-03-14 2016-11-22 Abbvie Inc. Methods for modulating protein glycosylation profiles of recombinant protein therapeutics using monosaccharides and oligosaccharides
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
US9516868B2 (en) 2010-08-02 2016-12-13 Regeneron Pharmaceuticals, Inc. Mice that make VL binding proteins
WO2016207717A1 (en) 2015-06-24 2016-12-29 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
US9546214B2 (en) 2014-04-04 2017-01-17 Bionomics, Inc. Humanized antibodies that bind LGR5
WO2017009258A1 (en) 2015-07-10 2017-01-19 Genmab A/S Axl-specific antibody-drug conjugates for cancer treatment
US9550826B2 (en) 2013-11-15 2017-01-24 Abbvie Inc. Glycoengineered binding protein compositions
EP3128009A1 (en) * 2011-09-19 2017-02-08 Kymab Limited Antibodies, variable domains&chains tailored for human use
WO2017021349A1 (en) 2015-07-31 2017-02-09 Amgen Research (Munich) Gmbh Bispecific antibody constructs binding dll3 and cd3
WO2017021370A1 (en) 2015-07-31 2017-02-09 Amgen Research (Munich) Gmbh Bispecific antibody constructs binding egfrviii and cd3
WO2017021362A1 (en) 2015-07-31 2017-02-09 Amgen Research (Munich) Gmbh Antibody constructs for flt3 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
WO2017021354A1 (en) 2015-07-31 2017-02-09 Amgen Research (Munich) Gmbh Antibody constructs for cd70 and cd3
WO2017027691A1 (en) 2015-08-13 2017-02-16 New York University Antibody-based molecules selective for the {p}ser404 epitope of tau and their uses in the diagnosis and treatment of tauopathy
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
US9598667B2 (en) 2013-10-04 2017-03-21 Abbvie Inc. Use of metal ions for modulation of protein glycosylation profiles of recombinant proteins
US9617336B2 (en) 2012-02-01 2017-04-11 Compugen Ltd C10RF32 antibodies, and uses thereof for treatment of cancer
EP3153524A1 (en) 2008-12-03 2017-04-12 Genmab A/S Antibody variants having modifications in the constant region
EP3168232A1 (en) 2009-11-13 2017-05-17 Dana-Farber Cancer Institute, Inc. Compositions, kits, and methods for the diagnosis, prognosis, monitoring, treatment and modulation of post-transplant lymphoproliferative disorders and hypoxia associated angiogenesis disorders using galectin-1
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
WO2017086419A1 (en) 2015-11-18 2017-05-26 中外製薬株式会社 Method for enhancing humoral immune response
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
US9693539B2 (en) 2007-08-10 2017-07-04 E. R. Squibb & Sons, L.L.C. HCO32 and HCO27 and related examples
US9708410B2 (en) 2003-05-30 2017-07-18 Janssen Biotech, Inc. Anti-tissue factor antibodies and compositions
WO2017125897A1 (en) 2016-01-21 2017-07-27 Novartis Ag Multispecific molecules targeting cll-1
WO2017137830A1 (en) 2016-02-12 2017-08-17 Janssen Pharmaceutica Nv Anti-vista (b7h5) antibodies
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
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
WO2017172771A2 (en) 2016-03-29 2017-10-05 Janssen Biotech, Inc. Method of treating psoriasis with increased interval dosing of anti-il12/23 antibody
US9783593B2 (en) 2013-05-02 2017-10-10 Kymab Limited Antibodies, variable domains and chains tailored for human use
WO2017175058A1 (en) 2016-04-07 2017-10-12 Janssen Pharmaceutica Nv Anti-vista antibodies and fragments, uses thereof, and methods of identifying same
US9788534B2 (en) 2013-03-18 2017-10-17 Kymab Limited Animal models and therapeutic molecules
US9796788B2 (en) 2010-02-08 2017-10-24 Regeneron Pharmaceuticals, Inc. Mice expressing a limited immunoglobulin light chain repertoire
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
WO2017184619A2 (en) 2016-04-18 2017-10-26 Celldex Therapeutics, Inc. Agonistic antibodies that bind human cd40 and uses thereof
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
WO2017196663A1 (en) 2016-05-09 2017-11-16 Bristol-Myers Squibb Company Tl1a 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)
EP3263585A2 (en) 2007-08-20 2018-01-03 Oncotherapy Science, Inc. Foxm1 peptide and medicinal agent comprising the same
WO2018011073A1 (en) 2016-07-12 2018-01-18 H. Lundbeck A/S Antibodies specific for hyperphosphorylated tau and methods of use thereof
WO2018013818A2 (en) 2016-07-14 2018-01-18 Bristol-Myers Squibb Company Antibodies against tim3 and uses thereof
EP3279215A1 (en) 2009-11-24 2018-02-07 MedImmune Limited Targeted binding agents against b7-h1
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
US9896516B2 (en) 2012-03-28 2018-02-20 Kymab Limited Animal models and therapeutic molecules
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
US9924705B2 (en) 2012-03-28 2018-03-27 Kymab Limited Animal models and therapeutic molecules
US9932398B2 (en) 2011-10-17 2018-04-03 Regeneron Pharmaceuticals, Inc. Restricted immunoglobulin heavy chain mice
US9930871B2 (en) 2013-02-20 2018-04-03 Regeneron Pharmaceuticals, Inc. Non-human animals with modified immunoglobulin heavy chain sequences
WO2018064436A1 (en) 2016-09-30 2018-04-05 Janssen Biotech, Inc. Safe and effective method of treating psoriasis with anti-il23 specific antibody
WO2018071822A2 (en) 2016-10-13 2018-04-19 Massachusetts Institute Of Technology Antibodies that bind zika virus envelope protein and uses thereof
US9963716B2 (en) 2011-09-26 2018-05-08 Kymab Limited Chimaeric surrogate light chains (SLC) comprising human VpreB
US9969814B2 (en) 2010-02-08 2018-05-15 Regeneron Pharmaceuticals, Inc. Methods for making fully human bispecific antibodies using a common light chain
WO2018087720A1 (en) 2016-11-14 2018-05-17 Novartis Ag Compositions, methods, and therapeutic uses related to fusogenic protein minion
WO2018091444A1 (en) 2016-11-15 2018-05-24 H. Lundbeck A/S Agents, uses and methods for the treatment of synucleinopathy
WO2018093841A1 (en) 2016-11-16 2018-05-24 Janssen Biotech, Inc. Method of treating psoriasis with anti-il-23 specific antibody
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
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
WO2018109058A1 (en) 2016-12-16 2018-06-21 H. Lundbeck A/S Agents, uses and methods
WO2018115225A1 (en) 2016-12-22 2018-06-28 H. Lundbeck A/S Monoclonal anti-alpha-synuclein antibodies for preventing tau aggregation
WO2018119380A2 (en) 2016-12-23 2018-06-28 Bristol-Myers Squibb Company Design of therapeutic immunoglobulin g4 for improved bioanalytical and bioprocessing properties
WO2018127519A1 (en) 2017-01-04 2018-07-12 H. Lundbeck A/S Antibodies specific for hyperphosphorylated tau for the treatment of ocular diseases
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
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
US10064398B2 (en) 2009-07-08 2018-09-04 Kymab Limited Animal models and therapeutic molecules
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
US10130081B2 (en) 2011-08-05 2018-11-20 Regeneron Pharmaceuticals, Inc. Humanized universal light chain mice
US10143186B2 (en) 2010-02-08 2018-12-04 Regeneron Pharmaceuticals, Inc. Common light chain mouse
US10149461B2 (en) 2008-10-27 2018-12-11 Revivicor, Inc. Immunocompromised ungulates
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
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
WO2019016247A2 (en) 2017-07-20 2019-01-24 H. Lundbeck A/S Agents, uses and methods for treatment
US10196439B2 (en) 2015-07-13 2019-02-05 H. Lundbeck A/S Antibodies specific for hyperphosphorylated tau and methods of use thereof
WO2019058345A2 (en) 2017-09-25 2019-03-28 Janssen Biotech, Inc. Safe and effective method of treating lupus with anti-il12/il23 antibody
US10251377B2 (en) 2012-03-28 2019-04-09 Kymab Limited Transgenic non-human vertebrate for the expression of class-switched, fully human, antibodies
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
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
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
WO2019118426A1 (en) 2017-12-11 2019-06-20 Amgen Inc. Continuous manufacturing process for bispecific antibody products
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
US10344299B2 (en) 2000-10-31 2019-07-09 Regeneron Pharmaceuticals, Inc. Compositions and methods for modifying cells
WO2019140229A1 (en) 2018-01-12 2019-07-18 Bristol-Myers Squibb Company Antibodies against tim3 and uses thereof
US10358482B2 (en) 2015-07-13 2019-07-23 H. Lundbeck A/S Antibodies for the treatment of synucleinopathy
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
WO2019151418A1 (en) 2018-01-31 2019-08-08 元一 加藤 Therapeutic agent for asthma containing il-6 inhibitor
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
WO2019171252A1 (en) 2018-03-05 2019-09-12 Janssen Biotech, Inc. Methods of treating crohn's disease with anti-il23 specific antibody
US10428147B2 (en) 2015-07-13 2019-10-01 H. Lundbeck A/S Anti-sortilin antibodies, uses and methods for treatment
WO2019191416A1 (en) 2018-03-29 2019-10-03 Bristol-Myers Squibb Company Methods of purifying monomeric monoclonal antibodies
WO2019217455A1 (en) 2018-05-07 2019-11-14 Genmab A/S Methods of treating cancer with a combination of an anti-pd-1 antibody and an anti-tissue factor antibody-drug conjugate
WO2019217457A1 (en) 2018-05-07 2019-11-14 Genmab A/S Methods of treating cancer with a combination of an anti-pd-1 antibody and an anti-tissue factor antibody-drug conjugate
WO2019215701A1 (en) 2018-05-11 2019-11-14 Janssen Biotech, Inc. Methods of treating depression using il-23 antibodies
EP3569245A1 (en) 2006-09-26 2019-11-20 Genmab A/S Combination treatment of cd38-expressing tumors
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
EP3584260A1 (en) 2015-04-28 2019-12-25 Mitsubishi Tanabe Pharma Corporation Rgma binding protein and use thereof
WO2020013238A1 (en) 2018-07-10 2020-01-16 田辺三菱製薬株式会社 Prevention or treatment method for peripheral neuropathy or pain accompanying disease in which peripheral neuropathy or astrocyte disorder is recognized
WO2020016838A2 (en) 2018-07-18 2020-01-23 Janssen Biotech, Inc. Sustained response predictors after treatment with anti-il23 specific antibody
EP3604523A1 (en) 2001-12-28 2020-02-05 Chugai Seiyaku Kabushiki Kaisha Method for stabilizing proteins
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
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
WO2020065532A1 (en) 2018-09-24 2020-04-02 Janssen Biotech, Inc. Safe and effective method of treating ulcerative colitis with anti-il12/il23 antibody
EP3632462A1 (en) 2012-07-06 2020-04-08 Genmab B.V. Dimeric protein with triple mutations
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
WO2020092210A1 (en) 2018-10-30 2020-05-07 Genmab A/S Methods of treating cancer with a combination of an anti-vegf antibody and an anti-tissue factor antibody-drug conjugate
WO2020102501A1 (en) 2018-11-16 2020-05-22 Bristol-Myers Squibb Company Anti-nkg2a antibodies and uses thereof
US10662256B2 (en) 2010-07-26 2020-05-26 Trianni, Inc. Transgenic mammals and methods of use thereof
WO2020104943A2 (en) 2018-11-20 2020-05-28 Janssen Biotech, Inc. Safe and effective method of treating psoriasis with anti-il-23 specific antibody
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
WO2020128864A1 (en) 2018-12-18 2020-06-25 Janssen Biotech, Inc. Safe and effective method of treating lupus with anti-il12/il23 antibody
WO2020148651A1 (en) 2019-01-15 2020-07-23 Janssen Biotech, Inc. Anti-tnf antibody compositions and methods for the treatment of juvenile idiopathic arthritis
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
WO2020152544A1 (en) 2019-01-23 2020-07-30 Janssen Biotech, Inc. Anti-tnf antibody compositions for use in methods for the treatment of psoriatic arthritis
WO2020162452A1 (en) 2019-02-04 2020-08-13 国立大学法人愛媛大学 Car library and production method for scfv
US10745487B2 (en) 2016-03-22 2020-08-18 Bionomics Limited Method of treating cancer by administering an anti-LGR5 monoclonal antibody
US10745467B2 (en) 2010-03-26 2020-08-18 The Trustees Of Dartmouth College VISTA-Ig for treatment of autoimmune, allergic and inflammatory disorders
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
WO2020183418A1 (en) 2019-03-14 2020-09-17 Janssen Biotech, Inc. Manufacturing methods for producing anti-il12/il23 antibody compositions
WO2020183269A1 (en) 2019-03-14 2020-09-17 Janssen Biotech, Inc. Manufacturing methods for producing anti-tnf antibody compositions
WO2020183270A1 (en) 2019-03-14 2020-09-17 Janssen Biotech, Inc. Methods for producing anti-tnf antibody compositions
WO2020183271A1 (en) 2019-03-14 2020-09-17 Janssen Biotech, Inc. Methods for producing anti-tnf antibody compositions
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
WO2020188466A1 (en) 2019-03-18 2020-09-24 Janssen Biotech, Inc. Method of treating psoriasis in pediatric subjects with anti-il12/il23 antibody
US10787522B2 (en) 2014-03-21 2020-09-29 Regeneron Pharmaceuticals, Inc. VL antigen binding proteins exhibiting distinct binding characteristics
US10793829B2 (en) 2010-07-26 2020-10-06 Trianni, Inc. Transgenic mammals and methods of use thereof
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
US10813346B2 (en) 2015-12-03 2020-10-27 Trianni, Inc. Enhanced immunoglobulin diversity
WO2020245677A1 (en) 2019-06-03 2020-12-10 Janssen Biotech, Inc. Anti-tnf antibodies, compositions, and methods for the treatment of active ankylosing spondylitis
WO2020245676A1 (en) 2019-06-03 2020-12-10 Janssen Biotech, Inc. Anti-tnf antibody compositions, and methods for the treatment of psoriatic arthritis
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
US10870701B2 (en) 2016-03-15 2020-12-22 Generon (Shanghai) Corporation Ltd. Multispecific fab fusion proteins and use thereof
US10881085B2 (en) 2014-03-21 2021-01-05 Regeneron Pharmaceuticals, Inc. Non-human animals that make single domain binding proteins
US10881084B2 (en) 2010-07-26 2021-01-05 Trianni, Inc Transgenic animals and methods of use
US10894818B2 (en) 2014-10-03 2021-01-19 Massachusetts Institute Of Technology Antibodies that bind Ebola glycoprotein and uses thereof
WO2021021676A1 (en) 2019-07-26 2021-02-04 Amgen Inc. Anti-il13 antigen binding proteins
WO2021028752A1 (en) 2019-08-15 2021-02-18 Janssen Biotech, Inc. Anti-tfn antibodies for treating type i diabetes
US10933115B2 (en) 2012-06-22 2021-03-02 The Trustees Of Dartmouth College VISTA antagonist and methods of use
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
CN112626117A (en) * 2008-12-18 2021-04-09 伊拉兹马斯大学鹿特丹医学中心 Non-human transgenic animals expressing humanized antibodies and uses thereof
US10988528B2 (en) 2015-08-13 2021-04-27 New York University Antibody-based molecules specific for the truncated ASP421 epitope of Tau and their uses in the diagnosis and treatment of tauopathy
WO2021089794A1 (en) 2019-11-07 2021-05-14 Genmab A/S Methods of treating cancer with a combination of a platinum-based agent and an anti-tissue factor antibody-drug conjugate
WO2021091815A1 (en) 2019-11-04 2021-05-14 Seagen Inc. Anti-cd30 antibody-drug conjugates and their use for the treatment of hiv infection
WO2021090272A1 (en) 2019-11-07 2021-05-14 Genmab A/S Methods of treating cancer with a combination of an anti-pd-1 antibody and an anti-tissue factor antibody-drug conjugate
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
US11014982B2 (en) 2017-02-07 2021-05-25 Janssen Biotech, Inc. Anti-TNF antibodies, compositions, and methods for the treatment of active ankylosing spondylitis
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
US11041020B2 (en) 2017-01-30 2021-06-22 Janssen Biotech, Inc. Methods for the treatment of active Psoriatic Arthritis
WO2021126435A1 (en) 2019-12-20 2021-06-24 Novarock Biotherapeutics, Ltd. Anti-interleukin-23 p19 antibodies and methods of use thereof
WO2021127528A1 (en) 2019-12-20 2021-06-24 Amgen Inc. Mesothelin-targeted cd40 agonistic multispecific antibody constructs for the treatment of solid tumors
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
WO2021132673A1 (en) 2019-12-26 2021-07-01 国立大学法人大阪大学 Agent for preventing or treating acute-phase neuromyelitis optica
US11053288B2 (en) 2016-02-04 2021-07-06 Trianni, Inc. Enhanced production of immunoglobulins
WO2021145432A1 (en) 2020-01-15 2021-07-22 国立大学法人大阪大学 Agent for prevention or treatment of diabetic autonomic neuropathy
WO2021145435A1 (en) 2020-01-15 2021-07-22 国立大学法人大阪大学 Prophylactic or therapeutic agent for dementia
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
US11111314B2 (en) 2015-03-19 2021-09-07 Regeneron Pharmaceuticals, Inc. Non-human animals that select for light chain variable regions that bind antigen
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
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
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
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
WO2021236638A1 (en) 2020-05-19 2021-11-25 Amgen Inc. Mageb2 binding constructs
WO2021243320A2 (en) 2020-05-29 2021-12-02 Amgen Inc. Adverse effects-mitigating administration of a bispecific antibody construct binding to cd33 and cd3
WO2021261546A1 (en) 2020-06-24 2021-12-30 国立大学法人 東京大学 Photosensitizing dye
WO2022002940A1 (en) 2020-06-29 2022-01-06 Genmab A/S Anti-tissue factor antibody-drug conjugates and their use in the treatment of cancer
WO2022014703A1 (en) 2020-07-17 2022-01-20 田辺三菱製薬株式会社 Agent for preventing or treating muscular disease
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
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
WO2022051591A2 (en) 2020-09-04 2022-03-10 Novarock Biotherapeutics, Ltd. Nectin-4 antibodies and uses thereof
WO2022074206A1 (en) 2020-10-08 2022-04-14 Affimed Gmbh Trispecific binders
WO2022096700A1 (en) 2020-11-06 2022-05-12 Amgen Research (Munich) Gmbh Polypeptide constructs selectively binding to cldn6 and cd3
WO2022097065A2 (en) 2020-11-06 2022-05-12 Novartis Ag ANTIBODY Fc VARIANTS
WO2022096704A1 (en) 2020-11-06 2022-05-12 Amgen Inc. Antigen binding domain with reduced clipping rate
WO2022096698A1 (en) 2020-11-06 2022-05-12 Amgen Inc. Polypeptide constructs binding to cd3
WO2022096716A2 (en) 2020-11-06 2022-05-12 Amgen Inc. Multitargeting bispecific antigen-binding molecules of increased selectivity
WO2022108931A2 (en) 2020-11-17 2022-05-27 Seagen Inc. Methods of treating cancer with a combination of tucatinib and an anti-pd-1/anti-pd-l1 antibody
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
WO2022176970A1 (en) 2021-02-18 2022-08-25 田辺三菱製薬株式会社 Novel anti-pad4 antibody
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
WO2022190033A1 (en) 2021-03-12 2022-09-15 Janssen Biotech, Inc. Safe and effective method of treating psoriatic arthritis with anti-il23 specific antibody
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
WO2022190034A1 (en) 2021-03-12 2022-09-15 Janssen Biotech, Inc. Method of treating psoriatic arthritis patients with inadequate response to tnf therapy with anti-il23 specific antibody
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
WO2023281463A1 (en) 2021-07-09 2023-01-12 Janssen Biotech, Inc. Manufacturing methods for producing anti-tnf antibody compositions
WO2023281462A1 (en) 2021-07-09 2023-01-12 Janssen Biotech, Inc. Manufacturing methods for producing anti-tnf antibody compositions
WO2023281466A1 (en) 2021-07-09 2023-01-12 Janssen Biotech, Inc. Manufacturing methods for producing anti-il12/il23 antibody compositions
EP4119579A1 (en) 2007-05-31 2023-01-18 Genmab A/S Stable igg4 antibodies
US11559050B2 (en) 2012-06-12 2023-01-24 Regeneron Pharmaceuticals, Inc. Humanized non-human animals with restricted immunoglobulin heavy chain loci
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
US11572405B2 (en) 2018-01-12 2023-02-07 Bristol-Myers Squibb Company Combination therapy with anti-IL-8 antibodies and anti-PD-1 antibodies for treating cancer
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
WO2023076989A1 (en) 2021-10-29 2023-05-04 Seagen Inc. Methods of treating cancer with a combination of an anti-pd-1 antibody and an anti-cd30 antibody-drug conjugate
WO2023073615A1 (en) 2021-10-29 2023-05-04 Janssen Biotech, Inc. Methods of treating crohn's disease with anti-il23 specific antibody
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
WO2023084488A1 (en) 2021-11-15 2023-05-19 Janssen Biotech, Inc. Methods of treating crohn's disease with anti-il23 specific antibody
WO2023095000A1 (en) 2021-11-23 2023-06-01 Janssen Biotech, Inc. Method of treating ulcerative colitis with anti-il23 specific antibody
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
US11745165B2 (en) 2017-08-18 2023-09-05 The Johns Hopkins University Supramolecular filamentous assemblies for protein purification
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)
WO2023187707A1 (en) 2022-03-30 2023-10-05 Janssen Biotech, Inc. Method of treating mild to moderate psoriasis with il-23 specific antibody
US11780911B2 (en) 2019-05-23 2023-10-10 Janssen Biotech, Inc. Method of treating inflammatory bowel disease with a combination therapy of antibodies to IL-23 and TNF alpha
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
WO2023213960A1 (en) 2022-05-06 2023-11-09 Genmab A/S Methods of treating cancer with anti-tissue factor antibody-drug conjugates
WO2023218027A1 (en) 2022-05-12 2023-11-16 Amgen Research (Munich) Gmbh Multichain multitargeting bispecific antigen-binding molecules of increased selectivity
WO2023223265A1 (en) 2022-05-18 2023-11-23 Janssen Biotech, Inc. Method for evaluating and treating psoriatic arthritis with il23 antibody
WO2024013723A1 (en) 2022-07-15 2024-01-18 Pheon Therapeutics Ltd Antibody drug conjugates that bind cdcp1 and uses thereof
WO2024059675A2 (en) 2022-09-14 2024-03-21 Amgen Inc. Bispecific molecule stabilizing composition

Families Citing this family (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6586251B2 (en) * 2000-10-31 2003-07-01 Regeneron Pharmaceuticals, Inc. Methods of modifying eukaryotic cells
EP1946768A4 (en) 2005-10-28 2009-11-11 Meiji Seika Kaisha Outer coat protein pa5158 of pseudomonas aeruginosa
WO2007114340A1 (en) 2006-03-30 2007-10-11 Meiji Seika Kaisha, Ltd. Pseudomonas aeruginosa outer membrane protein pa0427
EP2236515A4 (en) 2007-12-25 2012-10-31 Meiji Seika Kaisha Component protein pa1698 for type-iii secretion system of pseudomonas aeruginosa
US20110028697A1 (en) 2008-03-27 2011-02-03 Takara Bio Inc. Prophylactic/therapeutic agent for infectious disease
CN102959097B (en) 2010-07-08 2014-07-16 本田技研工业株式会社 High frequency heating coil
WO2012043747A1 (en) 2010-09-30 2012-04-05 独立行政法人理化学研究所 Glioma treatment method, glioma examination method, method of delivering a desired material to a glioma, and drug used in said methods
EP2659910B1 (en) 2010-12-27 2016-11-09 National University Corporation Nagoya University Screening method for a compound capable of suppressing receptor tyrosine kinase-mediated pro-survival signaling in a cancer cell
US20140242085A1 (en) 2011-08-12 2014-08-28 Medical & Biological Laboratories Co., Ltd. Methods and composition for testing, preventing, and treating aspergillus fumigatus infection
CN104470950B (en) 2012-05-11 2017-04-26 公益财团法人微生物化学研究会 Anti-cxadr antibody
CN104582476B (en) * 2012-06-05 2017-03-08 瑞泽恩制药公司 The method preparing complete people bi-specific antibody using common light chain
AU2013297478B2 (en) 2012-07-30 2018-12-20 Medical & Biological Laboratories Co., Ltd. Monoclonal antibody against human midkine
WO2014042251A1 (en) 2012-09-13 2014-03-20 中外製薬株式会社 Gene knock-in non-human animal
JP6357113B2 (en) 2013-02-08 2018-07-11 株式会社医学生物学研究所 Antibodies against human NRG1 protein
WO2014133093A1 (en) 2013-02-28 2014-09-04 独立行政法人国立がん研究センター Antibody against insoluble fibrin
WO2014174596A1 (en) 2013-04-23 2014-10-30 株式会社医学生物学研究所 Functional monoclonal antibody against heparin-binding epidermal growth factor-like growth factor
WO2015068781A1 (en) 2013-11-06 2015-05-14 国立大学法人大阪大学 Antibody having broad neutralizing activity in group 1 influenza a virus

Citations (2)

* 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
WO1991010741A1 (en) * 1990-01-12 1991-07-25 Cell Genesys, Inc. Generation of xenogeneic antibodies

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB8823869D0 (en) * 1988-10-12 1988-11-16 Medical Res Council Production of antibodies
ES2108048T3 (en) * 1990-08-29 1997-12-16 Genpharm Int PRODUCTION AND USE OF LOWER TRANSGENIC ANIMALS CAPABLE OF PRODUCING HETEROLOGICAL ANTIBODIES.
EP0746609A4 (en) * 1991-12-17 1997-12-17 Genpharm Int Transgenic non-human animals capable of producing heterologous antibodies

Patent Citations (2)

* 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
WO1991010741A1 (en) * 1990-01-12 1991-07-25 Cell Genesys, Inc. Generation of xenogeneic antibodies

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES, Volume 86, issued September 1989, M. BRUGGEMANN et al., "A Repetoire of Monoclonal Antibodies with Human Heavy Chains from Transgenic Mice", pages 6709-6713. *
See also references of EP0754225A4 *

Cited By (1206)

* 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
US8158419B2 (en) 1994-03-09 2012-04-17 Medarex, Inc. Transgenic non-human animals for producing chimeric antibodies
US8293480B2 (en) 1994-03-09 2012-10-23 Genpharm International Transgenic non-human animals for producing chimeric antibodies
JP2009039127A (en) * 1995-04-27 2009-02-26 Amgen Fremont Inc Human antibody derived from immunized xenomouse
EP1709970A1 (en) * 1995-04-27 2006-10-11 Abgenix, Inc. Human antibodies against EGFR, derived from immunized xenomice
JP2010057507A (en) * 1995-04-27 2010-03-18 Amgen Fremont Inc Human antibody derived from immunized xenomouse
AU2003227322B2 (en) * 1995-04-27 2005-12-01 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
AU2008202860B2 (en) * 1995-04-27 2012-03-01 Amgen Fremont Inc. Human Antibodies Derived From Immunized Xenomice
JP2009136290A (en) * 1995-04-27 2009-06-25 Amgen Fremont Inc Human antibody derived from immunized xenomouce
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
EP0823941A4 (en) * 1995-04-28 2001-09-19 Abgenix Inc Human antibodies derived from immunized xenomice
EP0823941A1 (en) * 1995-04-28 1998-02-18 Abgenix, Inc. Human antibodies derived from immunized xenomice
US5922584A (en) * 1995-06-07 1999-07-13 Wohlstadter Jacob N Method for promoting enzyme diversity
US6242236B1 (en) 1995-06-07 2001-06-05 Meso Scale Technology, Inc. Method of promoting enzyme diversity
US5919681A (en) * 1995-06-07 1999-07-06 Wohlstadter Jacob N Method for promoting enzyme diversity
US5914256A (en) * 1995-06-07 1999-06-22 Wohlstadter Jacob N Method for promoting enzyme diversity
EP0871485B1 (en) * 1995-07-21 2007-02-21 University Of Nebraska Board Of Regents COMPOSITIONS AND METHODS FOR CATALYZING HYDROLYSIS OF HIV gp120
EP0871485A1 (en) * 1995-07-21 1998-10-21 University Of Nebraska Board Of Regents COMPOSITIONS AND METHODS FOR CATALYZING HYDROLYSIS OF HIV gp120
JP2007332152A (en) * 1995-10-10 2007-12-27 Genpharm Internatl Inc Transgenic non-human animal capable of producing heterologous antibody
EP2186888A1 (en) * 1995-10-10 2010-05-19 GenPharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
EP0854917A4 (en) * 1995-10-10 2002-07-24 Genpharm Int Transgenic non-human animals capable of producing heterologous antibodies
JP2009201524A (en) * 1995-10-10 2009-09-10 Genpharm Internatl Inc Transgenic non-human animal producing heterologous antibody
US7041871B1 (en) 1995-10-10 2006-05-09 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
JP2008189691A (en) * 1995-10-10 2008-08-21 Genpharm Internatl Inc Transgenic non-human animal producing heterologous antibody
EP1813672A2 (en) * 1995-10-10 2007-08-01 GenPharm International, Inc. 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
WO1997013852A1 (en) 1995-10-10 1997-04-17 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
JP2008239630A (en) * 1995-10-10 2008-10-09 Genpharm Internatl Inc Transgenic non-human animal producing heterologous antibody
EP0854917A1 (en) * 1995-10-10 1998-07-29 GenPharm International, Inc. 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
CN102827283A (en) * 1995-10-10 2012-12-19 真药物国际公司 Transgenic non-human animals capable of producing heterologous antibodies
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
US6903203B2 (en) 1996-05-04 2005-06-07 Astrazeneca Uk Ltd. Monoclonal antibody to CEA, conjugates comprising said antibody and their therapeutic use
WO1997042329A1 (en) * 1996-05-04 1997-11-13 Zeneca Limited Monoclonal antibody to cea, conjugates comprising said antibody, and their therapeutic use in an adept system
US6277599B1 (en) 1996-05-04 2001-08-21 Zeneca Limited Anti-CEA antibody designated 806.077, hybridoma and method of manufacture
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
EP0942959A1 (en) * 1996-12-02 1999-09-22 GenPharm International Transgenic non-human animals capable of producing heterologous antibodies
AU747370B2 (en) * 1996-12-02 2002-05-16 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibo dies
EP0942959A4 (en) * 1996-12-02 2005-04-13 Genpharm Internat Transgenic non-human animals capable of producing heterologous antibodies
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
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
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
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
WO1998024893A3 (en) * 1996-12-03 1998-08-20 Abgenix Inc TRANSGENIC MAMMALS HAVING HUMAN IG LOCI INCLUDING PLURAL VH AND Vλ 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
WO1998024893A2 (en) * 1996-12-03 1998-06-11 Abgenix, Inc. TRANSGENIC MAMMALS HAVING HUMAN IG LOCI INCLUDING PLURAL VH AND Vλ REGIONS AND ANTIBODIES PRODUCED THEREFROM
AU2008200005B2 (en) * 1996-12-03 2012-05-17 Amgen Fremont Inc. Transgenic Mammals Having Human Ig Loci Including Plural Vh and Vk Regions and Antibodies Produced Therefrom
US6080910A (en) * 1997-02-20 2000-06-27 Case Western Reserve University Transgenic knockout animals lacking IgG3
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
US8173127B2 (en) 1997-04-09 2012-05-08 Intellect Neurosciences, Inc. Specific antibodies to amyloid beta peptide, pharmaceutical compositions and methods of use thereof
US7807798B2 (en) 1997-05-05 2010-10-05 Amgen Fremont Inc. Human monoclonal antibodies to epidermal growth factor receptor
EP2332993A1 (en) 1997-05-05 2011-06-15 Amgen Fremont Inc. Human monoclonal antibody to the human epidermal growth factor receptor
US8227580B2 (en) 1997-05-05 2012-07-24 Amgen Inc Human monoclonal antibodies to 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
GB2344344A (en) * 1998-11-03 2000-06-07 Babraham Inst Transgenic mice containing Human Ig lambda locus
GB2344344B (en) * 1998-11-03 2001-08-15 Babraham Inst Murine expression of Human Ig locus
EP1895002A1 (en) 1998-11-04 2008-03-05 Chugai Seiyaku Kabushiki Kaisha Novel trypsin family serine proteases
EP1895001A1 (en) 1998-11-04 2008-03-05 Chugai Seiyaku Kabushiki Kaisha Novel trypsin family serine proteases
US7132281B2 (en) 1998-12-23 2006-11-07 Amgen Fremont Inc. Methods and host cells for producing 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
US7807797B2 (en) 1998-12-23 2010-10-05 Amgen Fremont Inc. Human monoclonal antibodies to CTLA-4
US9963508B2 (en) 1998-12-23 2018-05-08 Amgen Fremont Inc. Human monoclonal antibodies to CTLA-4
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
US7109003B2 (en) 1998-12-23 2006-09-19 Abgenix, Inc. Methods for expressing and recovering human monoclonal antibodies to CTLA-4
US6682736B1 (en) 1998-12-23 2004-01-27 Abgenix, 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
US7824679B2 (en) 1998-12-23 2010-11-02 Amgen Fremont Inc. Human monoclonal antibodies to CTLA-4
US8883984B2 (en) 1998-12-23 2014-11-11 Amgen Fremont Inc. Human monoclonal antibodies to CTLA-4
US8865174B2 (en) 1999-03-25 2014-10-21 Abbvie Inc. Methods of treatment using human antibodies that bind IL-12
US9035030B2 (en) 1999-03-25 2015-05-19 AbbVie Deutschland GmbH & Co. KG Human antibodies that bind the P40 subunit of human IL-12 and methods for using the same
US8765918B2 (en) 1999-03-25 2014-07-01 Abbott Gmbh & Co., Kg Human antibodies that bind human interleukin-12
EP1961818A2 (en) 1999-04-09 2008-08-27 Chugai Seiyaku Kabushiki Kaisha Novel fetal genes
EP2135953A1 (en) 1999-06-02 2009-12-23 Chugai Seiyaku Kabushiki Kaisha Novel hemopoietin receptor protein, Nr. 10
EP2325316A1 (en) 1999-06-02 2011-05-25 Chugai Seiyaku Kabushiki Kaisha Novel hemopoietin receptor protein, nr10
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
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
US6984720B1 (en) 1999-08-24 2006-01-10 Medarex, Inc. Human CTLA-4 antibodies
EP3214175A1 (en) 1999-08-24 2017-09-06 E. R. Squibb & Sons, L.L.C. Human ctla-4 antibodies and their uses
EP2829609A1 (en) 1999-08-24 2015-01-28 E. R. Squibb & Sons, L.L.C. Human CTLA-4 antibodies and their uses
US7605238B2 (en) 1999-08-24 2009-10-20 Medarex, Inc. Human CTLA-4 antibodies and their uses
WO2001018200A1 (en) 1999-09-06 2001-03-15 Chugai Seiyaku Kabushiki Kaisha Tsg-like gene
WO2001019394A3 (en) * 1999-09-15 2001-10-04 Therapeutic Human Polyclonals Immunotherapy with substantially human polyclonal antibody preparations purified from genetically engineered birds
WO2001019394A2 (en) * 1999-09-15 2001-03-22 Therapeutic Human Polyclonals, Inc. Immunotherapy with substantially human polyclonal antibody preparations purified from genetically engineered birds
EP2316848A1 (en) 1999-09-21 2011-05-04 Chugai Seiyaku Kabushiki Kaisha Transporter genes OATP-B, C, D and E
EP2030985A1 (en) 1999-09-21 2009-03-04 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
EP2019139A1 (en) 2000-01-24 2009-01-28 Sugiyama, Haruo WT1 interacting protein WTIP
EP1987842A1 (en) 2000-04-28 2008-11-05 Chugai Seiyaku Kabushiki Kaisha Cell proliferation inhibitor
EP2216343A1 (en) 2000-05-18 2010-08-11 Japan Tobacco, Inc. Human monoclonal antibody against a costimulatory signal transduction molecule AILIM and pharmaceutical use thereof
EP2295467A1 (en) 2000-05-18 2011-03-16 Japan Tobacco Inc Human monoclonal antibody against a costimulatory signal transduction molecule AILIM and pharmaceutical use thereof
EP2330129A2 (en) 2000-08-07 2011-06-08 Centocor Ortho Biotech Inc. Anti-TNF antibodies, compositions, methods and uses
EP2253646A1 (en) 2000-08-07 2010-11-24 Centocor Ortho Biotech Inc. Anti-dual integrin antibody and compositions and conjugates comprising said antibody
EP3597752A1 (en) 2000-08-07 2020-01-22 Janssen Biotech, Inc. Anti-il-12 antibodies, compositions, methods and uses
EP2305817A2 (en) 2000-08-07 2011-04-06 Centocor Ortho Biotech Inc. Anti-IL-12 antibodies, compositions, methods and uses
EP2090657A2 (en) 2000-08-07 2009-08-19 Centocor Ortho Biotech Inc. Anti-IL-12 antibodies, compositions, methods and uses
EP2159230A1 (en) 2000-08-07 2010-03-03 Centocor Ortho Biotech Inc. Anti-TNF antibodies, compositions, methods and uses
EP3118318A1 (en) 2000-08-07 2017-01-18 Janssen Biotech, Inc. Anti-tnf antibodies, compositions, methods and uses
US10344299B2 (en) 2000-10-31 2019-07-09 Regeneron Pharmaceuticals, Inc. Compositions and methods for modifying cells
US9376699B2 (en) 2000-10-31 2016-06-28 Regeneron Pharmaceuticals, Inc. Methods of producing hybrid antibodies
US10227625B2 (en) 2000-10-31 2019-03-12 Regeneron Pharmaceuticals, Inc. Methods of modifying eukaryotic cells
US9708635B2 (en) 2000-10-31 2017-07-18 Regeneron Pharmaceuticals, Inc. Methods of making a nucleic acid encoding a human variable region
US10584364B2 (en) 2000-12-07 2020-03-10 Rgeneron Pharmaceuticals, Inc. Mice that produce hybrid antibodies
US9234041B2 (en) 2001-01-05 2016-01-12 Pfizer Inc. Antibodies to insulin-like growth factor I receptor
US7982024B2 (en) 2001-01-05 2011-07-19 Amgen Fremont Inc. Antibodies to insulin-like growth factor I receptor
US7700742B2 (en) 2001-01-05 2010-04-20 Amgen Fremont Antibodies to insulin-like growth factor I receptor
US7815907B2 (en) 2001-01-05 2010-10-19 Amgen Fremont Inc. Antibodies to insulin-like growth factor I receptor
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
US7192582B2 (en) 2001-02-12 2007-03-20 Medarex, Inc. Human monoclonal antibodies to FC alpha receptor (CD89)
US10378038B2 (en) 2001-02-16 2019-08-13 Regeneron Pharmaceuticals, Inc. Mice that produce hybrid antibodies
US10378039B2 (en) 2001-02-16 2019-08-13 Regeneron Pharmaceuticals, Inc. Mouse embryonic stem cells comprising a hybrid heavy chain immunoglobulin locus
US10526630B2 (en) 2001-02-16 2020-01-07 Regeneron Pharmaceuticals, Inc. Genetically modified mice that produce hybrid antibodies
EP2264163B1 (en) * 2001-02-16 2015-10-14 Regeneron Pharmaceuticals, Inc. Methods of modifying eukaryotic cells
US10378040B2 (en) 2001-02-16 2019-08-13 Regeneron Pharmaceuticals, Inc. Mice that produce hybrid antibodies
US10378037B2 (en) 2001-02-16 2019-08-13 Regeneron Pharmaceuticals, Inc. Methods of making a nucleic acid encoding a human variable region
US9388446B2 (en) 2001-02-16 2016-07-12 Regeneron Pharmaceuticals, Inc. Methods of producing hybrid antibodies
US20140017238A1 (en) 2001-02-16 2014-01-16 Regeneron Pharmaceuticals, Inc. Methods of Modifying Eukaryotic Cells
US10640800B2 (en) 2001-02-16 2020-05-05 Regeneron Pharmaceuticals, Inc. Mice that produce hybrid antibodies
US9353394B2 (en) 2001-02-16 2016-05-31 Regeneron Pharmaceuticals, Inc. Methods of producing hybrid antibodies
US9371553B2 (en) 2001-02-16 2016-06-21 Regeneron Pharmaceuticals, Inc. Genetically modified mice that produce hybrid antibodies
US9528136B2 (en) 2001-02-16 2016-12-27 Regeneron Pharmaceuticals, Inc. Methods of modifying eukaryotic cells
EP2298812A2 (en) 2001-04-02 2011-03-23 Chugai Seiyaku Kabushiki Kaisha Remedies for juvenile chronic arthritis and related diseases
EP3640261A1 (en) 2001-04-02 2020-04-22 Chugai Seiyaku Kabushiki Kaisha Therapeutic agent for chronic arthritides diseases of childhood-related diseases
EP1972638A1 (en) 2001-04-02 2008-09-24 Chugai Seiyaku Kabushiki Kaisha Remedies for juvenile chronic arthritis and related diseases
WO2003040169A2 (en) 2001-11-07 2003-05-15 Celldex Therapeutics , Inc. Human monoclonal antibodies to dendritic cells
US7955597B2 (en) 2001-11-14 2011-06-07 Centocor, Inc. Anti-IL-6 antibodies, compositions, methods and uses
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
EP3604523A1 (en) 2001-12-28 2020-02-05 Chugai Seiyaku Kabushiki Kaisha Method for stabilizing proteins
EP3960855A1 (en) 2001-12-28 2022-03-02 Chugai Seiyaku Kabushiki Kaisha Method for stabilizing proteins
US7387776B2 (en) 2002-01-09 2008-06-17 Medarex, Inc. Human monoclonal antibodies against CD30
US8088377B2 (en) 2002-01-09 2012-01-03 Medarex, Inc. Human monoclonal antibodies against CD30
EP2311489A2 (en) 2002-02-14 2011-04-20 Chugai Seiyaku Kabushiki Kaisha Formulation of antibody-containing solutions comprising a sugar as a stabilizer
WO2003068259A1 (en) 2002-02-14 2003-08-21 Chugai Seiyaku Kabushiki Kaisha Antibody-containing solution pharmaceuticals
US8921527B2 (en) 2002-02-14 2014-12-30 Chugai Seiyaku Kabushiki Kaisha Antibody-containing solution formulations
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
EP2110434A1 (en) 2002-02-25 2009-10-21 Genentech, Inc. Type-1 cytokine receptor GLM-R
WO2003083116A1 (en) 2002-03-29 2003-10-09 Chugai Seiyaku Kabushiki Kaisha Emthod of screening transporter inhibitor
US7452535B2 (en) 2002-04-12 2008-11-18 Medarex, Inc. Methods of treatment using CTLA-4 antibodies
EP2316921A1 (en) 2002-05-24 2011-05-04 Schering Corporation Neutralizing human anti-IGFR antibody
EP2316922A1 (en) 2002-05-24 2011-05-04 Schering Corporation Neutralizing human anti-IGFR antibody
EP2233572A2 (en) 2002-06-06 2010-09-29 Oncotherapy Science, Inc. Genes and polypeptides relating to human colon cancers
WO2003104276A2 (en) 2002-06-06 2003-12-18 Oncotherapy Science, Inc. Genes and polypeptides relating to hepatocellular or colorectal carcinoma
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
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
EP2264186A1 (en) 2002-09-30 2010-12-22 Oncotherapy Science, Inc. Method for diagnosing non-small cell lung cancers
EP1743947A2 (en) 2002-09-30 2007-01-17 Oncotherapy Science, Inc. Method for diagnosing non-small cell lung cancers
EP2278028A1 (en) 2002-09-30 2011-01-26 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
EP2270215A2 (en) 2002-09-30 2011-01-05 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
EP2090587A1 (en) 2002-09-30 2009-08-19 Oncotherapy Science, Inc. Genes and polypeptides relating to prostate cancers
EP2270213A2 (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
EP2270217A2 (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
EP2270212A2 (en) 2002-09-30 2011-01-05 Oncotherapy Science, Inc. Method for diagnosing non-small cell lung cancers
EP2270224A2 (en) 2002-09-30 2011-01-05 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
EP2270219A2 (en) 2002-09-30 2011-01-05 Oncotherapy Science, Inc. Method for diagnosing non-small cell lung cancers
EP2270225A2 (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
EP2270222A2 (en) 2002-09-30 2011-01-05 Oncotherapy Science, Inc. Method for diagnosing non-small cell lung cancers
EP2270218A2 (en) 2002-09-30 2011-01-05 Oncotherapy Science, Inc. Method for diagnosing non-small cell lung cancers
EP2270210A2 (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
EP2270498A2 (en) 2002-09-30 2011-01-05 Oncotherapy Science, Inc. Method for diagnosing non-small cell lung cancers
EP2270221A2 (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
EP2270220A2 (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
EP2264457A1 (en) 2002-09-30 2010-12-22 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
EP2264190A1 (en) 2002-09-30 2010-12-22 Oncotherapy Science, Inc. Method for diagnosing non-small cell lung cancers
US8529902B2 (en) 2002-10-17 2013-09-10 Genmab A/S Human monoclonal antibodies against CD20
EP3284753A2 (en) 2002-10-17 2018-02-21 Genmab A/S Human monoclonal antibodies against cd20
EP2330130A1 (en) 2002-10-17 2011-06-08 Genmab A/S Human monoclonal antibodies against CD20
EP2316856A1 (en) 2002-10-17 2011-05-04 Genmab A/S Human monoclonal antibodies against CD20
WO2004038018A1 (en) 2002-10-22 2004-05-06 Eisai Co., Ltd. Gene expressed specifically in dopamine-producing neuron precursor cells after termination of division
WO2004039981A1 (en) 2002-10-30 2004-05-13 Chugai Seiyaku Kabushiki Kaisha Membrane protein originating in mast cells
US7438907B2 (en) 2002-11-15 2008-10-21 Genmab A/S Human monoclonal antibodies against CD25
US9598493B2 (en) 2002-11-15 2017-03-21 Genmab A/S Human monoclonal antibodies against CD25
US10703818B2 (en) 2002-11-15 2020-07-07 Genmab A/S Human monoclonal antibodies against CD25
US8961968B2 (en) 2002-11-15 2015-02-24 Genmab A/S Human monoclonal antibodies against CD25
US8182812B2 (en) 2002-11-15 2012-05-22 Genmab A/S Human monoclonal antibodies against CD25
WO2004050683A2 (en) 2002-12-02 2004-06-17 Abgenix, Inc. Antibodies directed to tumor necrosis factor and uses thereof
US8603469B2 (en) 2002-12-16 2013-12-10 Genmab A/S Methods of treating cancer with human monoclonal antibodies against interleukin 8
US8105588B2 (en) 2002-12-16 2012-01-31 Genmab A/S Human monoclonal antibodies against interleukin 8 (IL-8)
US7622559B2 (en) 2002-12-16 2009-11-24 Genmab A/S Human monoclonal antibodies against interleukin 8 (IL-8)
US10253093B2 (en) 2002-12-16 2019-04-09 Cormorant Pharmaceuticals Ab Human monoclonal antibodies against interleukin 8 (IL-8)
US11339215B2 (en) 2002-12-16 2022-05-24 Cormorant Pharmaceuticals Ab Methods of treating cancer with human monoclonal antibodies against interleukin 8 (IL-8)
US7282568B2 (en) 2002-12-16 2007-10-16 Medarex, Inc. Human monoclonal antibodies against interleukin 8 (IL-8)
US10066012B2 (en) 2002-12-16 2018-09-04 Cormorant Pharmaceuticals Ab Human monoclonal antibodies against interleukin 8 (IL-8)
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
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
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
WO2004106515A1 (en) 2003-05-28 2004-12-09 Scimedia Ltd. Anti-bambi antibody and diagnostic or remedy for colon cancer and liver cancer containing the same
US9708410B2 (en) 2003-05-30 2017-07-18 Janssen Biotech, Inc. Anti-tissue factor antibodies and compositions
EP2508608A1 (en) 2003-06-09 2012-10-10 Alnylam Pharmaceuticals Inc. Method of treating neurodegenerative disease
WO2005017155A1 (en) 2003-06-18 2005-02-24 Chugai Seiyaku Kabushiki Kaisha Fucose transporter
EP2228445A1 (en) 2003-06-18 2010-09-15 Chugai Seiyaku Kabushiki Kaisha Fucose Transporter
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
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
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
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
US7396914B2 (en) 2003-08-04 2008-07-08 University Of Massachusetts SARS nucleic acids, proteins, antibodies, and uses thereof
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
WO2005035754A1 (en) 2003-10-14 2005-04-21 Chugai Seiyaku Kabushiki Kaisha Double specific antibodies substituting for functional protein
EP2311945A1 (en) 2003-10-14 2011-04-20 Chugai Seiyaku Kabushiki Kaisha Bispecific antibodies substituting for functional proteins
US7790863B2 (en) 2003-11-19 2010-09-07 Merck Serono S.A. Angiogenesis inhibiting molecules and their use in the treatment and diagnosis of cancer
WO2005054467A1 (en) 2003-12-03 2005-06-16 Chugai Seiyaku Kabushiki Kaisha EXPRESSION SYSTEM WITH THE USE OF MAMMALIAN β-ACTIN PROMOTER
EP2865687A1 (en) 2003-12-10 2015-04-29 E. R. Squibb & Sons, L.L.C. IP-10 antibodies and their uses
EP2418220A2 (en) 2003-12-10 2012-02-15 Medarex, Inc. Interferon alpha antibodies and their uses
EP2383295A1 (en) 2003-12-10 2011-11-02 Medarex, Inc. 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
US8093010B2 (en) 2003-12-18 2012-01-10 Merck Serono S.A. Angiogenesis inhibiting molecules, their selection, production and their use in the treatment of cancer
US7642341B2 (en) 2003-12-18 2010-01-05 Merck Serono S.A. Angiogenesis inhibiting molecules, their selection, production and their use in the treatment of cancer
US8609111B2 (en) 2004-02-06 2013-12-17 University Of Massachusetts Antibodies against clostridium difficile toxins and uses thereof
US7625559B2 (en) 2004-02-06 2009-12-01 University Of Massachusetts Antibodies against Clostridium difficile toxins and uses thereof
US8236311B2 (en) 2004-02-06 2012-08-07 University Of Massachusetts Antibodies against clostridium difficile toxins and uses thereof
EP2857418A1 (en) 2004-02-06 2015-04-08 University of Massachusetts Antibodies against Clostridium difficile toxins and uses thereof
EP2270045A1 (en) 2004-02-06 2011-01-05 University of Massachusetts Antibodies against Clostridium difficile toxins and uses thereof
US9217029B2 (en) 2004-02-06 2015-12-22 University Of Massachusetts Antibodies against clostridium difficile toxins and uses thereof
US8257709B2 (en) 2004-02-06 2012-09-04 University Of Massachusetts Antibodies against Clostridium difficile toxins and uses thereof
US7625549B2 (en) 2004-03-19 2009-12-01 Amgen Fremont Inc. Determining the risk of human anti-human antibodies in transgenic mice
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
US8597615B2 (en) 2004-03-19 2013-12-03 Amgen Fremont Inc. Methods of monitoring a human anti-human antibody response and inhibitors thereof
EP2418224A2 (en) 2004-03-19 2012-02-15 Amgen Inc. Reducing the risk of human and anti-human antibodies through V gene manipulation
EP2343384A2 (en) 2004-03-23 2011-07-13 Oncotherapy Science, Inc. Method for diagnosing non-small cell lung cancer
WO2005100402A1 (en) 2004-04-13 2005-10-27 F.Hoffmann-La Roche Ag Anti-p-selectin antibodies
EP2067789A1 (en) 2004-04-13 2009-06-10 F. Hoffmann-La Roche Ag Anti-P selectin antibodies
EP2357201A1 (en) 2004-04-13 2011-08-17 F. Hoffmann-La Roche AG Anti-P-selectin antibodies
EP2360186A1 (en) 2004-04-13 2011-08-24 F. Hoffmann-La Roche AG Anti-P-selectin antibodies
EP2374817A1 (en) 2004-04-13 2011-10-12 F. Hoffmann-La Roche AG Anti-P-selectin antibodies
US7850962B2 (en) 2004-04-20 2010-12-14 Genmab A/S Human monoclonal antibodies against CD20
EP2740743A2 (en) 2004-06-01 2014-06-11 Domantis Limited Bispecific fusion antibodies with enhanced serum half-life
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
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
EP2314317A2 (en) 2004-07-09 2011-04-27 Chugai Seiyaku Kabushiki Kaisha Anti-glypican 3 antibody
WO2006009241A1 (en) 2004-07-22 2006-01-26 Eisai Co., Ltd. Lrp4/CORIN DOPAMINE-PRODUCING NEURON PRECURSOR CELL MARKER
US9102733B2 (en) 2004-09-17 2015-08-11 Roche Palo Alto Llc Anti-OX40L antibodies
US7868141B2 (en) 2004-09-17 2011-01-11 Roche Palo Alto Llc Anti-OX40L antibodies
US7501496B1 (en) 2004-09-17 2009-03-10 Roche Palo Alto Llc Anti-OX40L antibodies
US7790160B2 (en) 2004-10-01 2010-09-07 Medarex, Inc. Method of treating CD30 positive lymphomas
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
WO2006055638A2 (en) 2004-11-17 2006-05-26 Abgenix, Inc. Fully human monoclonal antibodies to il-13
EP2769990A2 (en) 2004-12-02 2014-08-27 Domantis Limited Bispecific domain antibodies targeting serum albumin and GLP-1 or PYY
WO2006068975A2 (en) 2004-12-20 2006-06-29 Abgenix, Inc. Binding proteins specific for human matriptase
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
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
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
EP2361933A2 (en) 2005-01-26 2011-08-31 Amgen Fremont Inc. Antibodies against interleukin-1 beta
EP2011885A2 (en) 2005-02-10 2009-01-07 Oncotherapy Science, Inc. Method of diagnosing bladder cancer
EP2295601A1 (en) 2005-02-10 2011-03-16 Oncotherapy Science, Inc. Method of diagnosing bladder cancer
US8491898B2 (en) 2005-02-18 2013-07-23 Medarex, L.L.C. Monoclonal antibodies against CD30 lacking in fucosyl residues
US8207303B2 (en) 2005-02-18 2012-06-26 Medarex, Inc. Monoclonal antibodies against CD30 lacking in fucosyl residues
EP2172480A2 (en) 2005-02-21 2010-04-07 Chugai Seiyaku Kabushiki Kaisha Methods for producing proteins using hamster IGF-1
WO2006098464A1 (en) 2005-03-14 2006-09-21 Link Genomics, Inc. Method for diagnosis of prostate cancer
EP3312196A1 (en) 2005-03-23 2018-04-25 Genmab A/S Antibodies against cd38 for treatment of multiple myeloma
EP3153525A1 (en) 2005-03-23 2017-04-12 Genmab A/S Antibodies against cd38 for treatment of multiple myeloma
EP2535355A2 (en) 2005-03-23 2012-12-19 Genmab A/S Antibodies against CD38 for treatment of multiple myeloma
EP2551282A2 (en) 2005-03-23 2013-01-30 Genmab A/S Antibodies against CD38 for treatment of multiple myeloma
EP2567976A2 (en) 2005-03-23 2013-03-13 Genmab A/S Antibodies against CD38 for treatment of multiple myeloma
EP2824183A1 (en) 2005-04-08 2015-01-14 Chugai Seiyaku Kabushiki Kaisha Antibody substituting for function of blood coagulation factor VIII
EP2842968A1 (en) 2005-04-29 2015-03-04 Janssen Biotech, Inc. Anti-IL-6 antibodies, compositions, methods and uses
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
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
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
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
WO2006132363A1 (en) 2005-06-10 2006-12-14 Chugai Seiyaku Kabushiki Kaisha Stabilizer for protein preparation comprising meglumine and use thereof
EP2452694A1 (en) 2005-06-30 2012-05-16 Janssen Biotech, Inc. Anti-IL-23 antibodies, compositions, methods and uses
EP3501537A1 (en) 2005-06-30 2019-06-26 Janssen Biotech, Inc. Anti-il23 antibodies, compositions, methods and uses
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)
EP2336780A1 (en) 2005-07-27 2011-06-22 Oncotherapy Science, Inc. Genes and polypeptides relating to prostate cancers
US9309315B2 (en) 2005-08-18 2016-04-12 Genmab A/S Therapy with CD4 binding peptides and radiation
WO2007038637A2 (en) 2005-09-26 2007-04-05 Medarex, Inc. Human monoclonal antibodies to cd70
WO2007037430A1 (en) 2005-09-29 2007-04-05 Eisai R & D Management Co., Ltd. T-cell adhesion molecule and antibody directed against the molecule
WO2007043641A1 (en) 2005-10-14 2007-04-19 Fukuoka University Inhibitor of transplanted islet dysfunction in islet transplantation
EP2532679A1 (en) 2005-10-21 2012-12-12 Novartis AG Human antibodies against il13 and therapeutic uses
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
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
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
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
EP2404616A2 (en) 2005-12-13 2012-01-11 AstraZeneca AB Binding proteins specific for insulin-like growth factors and uses thereof
WO2007077934A1 (en) 2005-12-28 2007-07-12 Asubio Pharma Co., Ltd. Anti-periostin antibody and pharmaceutical composition for preventing or treating periostin-related disease containing the same
WO2007074880A1 (en) 2005-12-28 2007-07-05 Chugai Seiyaku Kabushiki Kaisha Antibody-containing stabilizing preparation
EP2993187A2 (en) 2005-12-30 2016-03-09 U3 Pharma GmbH 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
WO2007077028A2 (en) 2005-12-30 2007-07-12 U3 Pharma Ag 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
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
EP3135298A1 (en) 2006-01-27 2017-03-01 Keio University Remedy for disease associated with choroidal angiogenesis
WO2007086490A1 (en) 2006-01-27 2007-08-02 Keio University Remedy for disease associated with choroidal angiogenesis
WO2007117410A2 (en) 2006-03-31 2007-10-18 Medarex, Inc. Transgenic animals expressing chimeric antibodies for use in preparing human antibodies
US7910798B2 (en) 2006-03-31 2011-03-22 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
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
EP2505058A1 (en) 2006-03-31 2012-10-03 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
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
EP2357202A1 (en) 2006-04-10 2011-08-17 AstraZeneca AB Targeted binding agents directed to Upar and uses thereof
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
EP2371393A1 (en) 2006-06-08 2011-10-05 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
WO2008010556A1 (en) 2006-07-21 2008-01-24 Chugai Seiyaku Kabushiki Kaisha Remedy for renal disease
EP2574625A1 (en) 2006-07-21 2013-04-03 Chugai Seiyaku Kabushiki Kaisha Remedy for renal disease
US8894998B2 (en) 2006-08-03 2014-11-25 Medimmune Limited Antibodies directed to αVβ6 and uses thereof
EP2420513A1 (en) 2006-08-03 2012-02-22 MedImmune Limited Targeted binding agents directed to PDGFR-alpha 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
EP2548576A1 (en) 2006-08-14 2013-01-23 Forerunner Pharma Research Co., Ltd. Diagnosis of cancer using anti-desmoglein-3 antibodies
EP3111955A1 (en) 2006-08-14 2017-01-04 Chugai Seiyaku Kabushiki Kaisha Diagnosis and treatment of cancer using anti-desmoglein-3 antibody
WO2008020586A1 (en) 2006-08-14 2008-02-21 Forerunner Pharma Research Co., Ltd. Diagnosis and treatment of cancer using anti-desmoglein-3 antibody
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
EP3753576A1 (en) 2006-09-26 2020-12-23 Genmab A/S Combination treatment of cd38-expressing tumors
EP3569245A1 (en) 2006-09-26 2019-11-20 Genmab A/S Combination treatment of cd38-expressing tumors
US8007797B2 (en) 2006-09-28 2011-08-30 Merck Serono S.A. Junctional adhesion molecule-C (JAM-C) binding compounds and methods of their use
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
WO2008059959A1 (en) 2006-11-17 2008-05-22 The Research Foundation For Microbial Diseases Of Osaka University Nerve elongation promoter and elongation inhibitor
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
WO2008074004A2 (en) 2006-12-14 2008-06-19 Medarex, Inc. Human antibodies that bind cd70 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
WO2008081942A1 (en) 2007-01-05 2008-07-10 The University Of Tokyo Diagnosis and treatment of cancer by using anti-prg-3 antibody
US9051368B2 (en) 2007-01-16 2015-06-09 Abbvie, Inc. Methods for treating psoriasis by administering an antibody which binds an epitope of the p40 subunit of IL-12 and/or IL-23
WO2008090901A1 (en) 2007-01-23 2008-07-31 Shinshu University Chronic rejection inhibitor
EP3246407A1 (en) 2007-02-09 2017-11-22 Eisai R&D Management Co., Ltd. Gaba neuron progenitor cell marker 65b13
WO2008096817A1 (en) 2007-02-09 2008-08-14 Eisai R & D Management Co., Ltd. Gaba neuron progenitor cell marker 65b13
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
EP2716301A2 (en) 2007-02-16 2014-04-09 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
WO2008108918A1 (en) 2007-02-21 2008-09-12 University Of Massachusetts Human antibodies against hepatitis c virus (hcv) uses thereof
US8551484B2 (en) 2007-02-21 2013-10-08 University Of Massachusetts Human antibodies against hepatitis C virus (HCV) 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
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
EP2626372A1 (en) 2007-03-29 2013-08-14 Genmab A/S Bispecific antibodies and methods for production thereof
EP2692737A1 (en) 2007-04-20 2014-02-05 Biotie Therapies Corp. Fully human anti-vap-1 monoclonal antibodies
US7875705B2 (en) 2007-05-28 2011-01-25 The University Of Tokyo Tumor diagnostic agent used in PET comprising anti-ROBO1 antibody
EP2615175A1 (en) 2007-05-31 2013-07-17 Genmab A/S Transgenic animals producing monovalent human antibodies and antibodies obtainable from these animals
EP4119579A1 (en) 2007-05-31 2023-01-18 Genmab A/S Stable igg4 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
WO2009001940A1 (en) 2007-06-27 2008-12-31 Asubio Pharma Co., Ltd. Cancer remedy containing antibody against peptide encoded by exon-17 of periostin
WO2009008414A1 (en) 2007-07-10 2009-01-15 Shionogi & Co., Ltd. Monoclonal antibody having neutralizing activity against mmp13
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
WO2009054873A2 (en) 2007-08-02 2009-04-30 Novimmune S.A. Anti-rantes antibodies and methods of use thereof
US11384363B2 (en) 2007-08-10 2022-07-12 E.R. Squibb & Sons, L.L.C. HCO32 and HCO27 and related examples
US9693539B2 (en) 2007-08-10 2017-07-04 E. R. Squibb & Sons, L.L.C. HCO32 and HCO27 and related examples
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
WO2009025116A1 (en) 2007-08-20 2009-02-26 Oncotherapy Science, Inc. Cdh3 peptide and medicinal agent comprising the same
EP4032899A1 (en) 2007-08-20 2022-07-27 Oncotherapy Science, Inc. Foxm1 peptide and medicinal agent comprising the same
EP3263585A2 (en) 2007-08-20 2018-01-03 Oncotherapy Science, Inc. Foxm1 peptide and medicinal agent comprising the same
WO2009025117A1 (en) 2007-08-20 2009-02-26 Oncotherapy Science, Inc. Cdca1 peptide and pharmaceutical agent comprising the same
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
DE202008018562U1 (en) 2007-08-23 2015-11-02 Amgen Inc. Antigen-binding Proteins Against 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)
EP3666797A1 (en) 2007-08-23 2020-06-17 Amgen, Inc Antigen binding proteins to proprotein convertase subtilisin kexin type 9 (pcsk9)
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
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
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
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
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
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
WO2009040134A1 (en) 2007-09-26 2009-04-02 U3 Pharma Gmbh Heparin-binding epidermal growth factor-like growth factor antigen binding proteins
EP2497783A2 (en) 2007-09-26 2012-09-12 U3 Pharma GmbH Heparin-binding epidermal growth factor-like growth factor antigen binding proteins
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
WO2009051220A1 (en) 2007-10-19 2009-04-23 Immunas Pharma, Inc. Antibody capable of specifically binding to aβ oligomer, and use thereof
EP2567709A2 (en) 2007-11-02 2013-03-13 Novartis AG Molecules and methods for modulating low-density-lipoprotein receptor-related protein 6 (LRP6)
EP3305324A1 (en) 2007-11-02 2018-04-11 Novartis AG Molecules and methods for modulating low-density-lipoprotein receptor-related protein 6 (lrp6)
EP2570137A2 (en) 2007-11-07 2013-03-20 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
EP3095862A1 (en) 2007-12-05 2016-11-23 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
WO2009072604A1 (en) 2007-12-05 2009-06-11 Chugai Seiyaku Kabushiki Kaisha Anti-nr10 antibody and use thereof
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
WO2009084659A1 (en) 2007-12-27 2009-07-09 Chugai Seiyaku Kabushiki Kaisha Solution preparation containing antibody at high concentration
EP2599792A1 (en) 2008-01-10 2013-06-05 Shionogi&Co., Ltd. Antibody directed against PcrV
WO2009088032A1 (en) 2008-01-10 2009-07-16 Shionogi & Co., Ltd. Antibody directed against pcrv
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
EP3064512A2 (en) 2008-01-11 2016-09-07 The University of Tokyo Anti-cldn6 antibody
US8653020B2 (en) 2008-01-25 2014-02-18 Aarhus Universitet Selective exosite inhibition of PAPP-A activity against IGFBP-4
EP2650017A2 (en) 2008-02-05 2013-10-16 Bristol-Myers Squibb Company Alpha 5 - beta 1 antibodies and their uses
EP2641612A1 (en) 2008-02-05 2013-09-25 Bristol-Myers Squibb Company Alpha 5 - beta 1 antibodies and their uses
WO2009099176A1 (en) 2008-02-08 2009-08-13 Immunas Pharma, Inc. Antibody capable of binding specifically to aβ-oligomer, and use thereof
EP3121277A1 (en) 2008-02-08 2017-01-25 Immunas Pharma, Inc. Antibody capable of binding specifically to ab-oligomer, and use thereof
US8945545B2 (en) 2008-03-18 2015-02-03 Abbvie Inc. Methods of treating psoriasis by administration of antibodies to the p40 subunit of IL-12 and/or IL-23
WO2009122667A1 (en) 2008-04-04 2009-10-08 中外製薬株式会社 Therapeutic for hepatic cancer
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
US8575314B2 (en) 2008-06-20 2013-11-05 National University Corporation Okayama University Antibody against oxidized LDL/β2GPI complex and use of the same
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
EP2837388A1 (en) 2008-08-05 2015-02-18 Novartis AG Compositions and methods for antibodies targeting complement protein C5
EP2815766A1 (en) 2008-08-05 2014-12-24 Novartis AG Compositions and methods for antibodies targeting complement protein C5
WO2010016590A1 (en) 2008-08-07 2010-02-11 国立大学法人 長崎大学 Therapeutic or prophylactic agent for generalized pain syndrome
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
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
KR20160116056A (en) 2008-08-14 2016-10-06 테바 파마슈티컬즈 오스트레일리아 피티와이 엘티디 Anti-il-12/il-23 antibodies
EP2927244A1 (en) 2008-09-19 2015-10-07 MedImmune, LLC Antibodies directed to DLL4 and uses thereof
US10370448B2 (en) 2008-09-26 2019-08-06 Emory University Human anti-PD-1, PD-L1, and PD-L2 antibodies and uses therefor
US10011656B2 (en) 2008-09-26 2018-07-03 Emory University Human anti-PD-1, PD-L1, and PD-L2 antibodies and uses therefor
US9102727B2 (en) 2008-09-26 2015-08-11 Emory University Human anti-PD-1 antibodies and uses therefor
US11261251B2 (en) 2008-09-26 2022-03-01 Dana-Farber Cancer Institute, Inc. Human anti-PD-1, PD-L1, and PD-L2 antibodies and uses therefor
US8552154B2 (en) 2008-09-26 2013-10-08 Emory University Anti-PD-L1 antibodies and uses therefor
EP3530672A1 (en) 2008-09-26 2019-08-28 Dana Farber Cancer Institute, Inc. Human anti-pd-1, pd-l1, and pd-l2 antibodies 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
EP3121197A1 (en) 2008-11-10 2017-01-25 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
EP3101031A1 (en) 2008-11-10 2016-12-07 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
EP2894165A1 (en) 2008-11-10 2015-07-15 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
EP3153524A1 (en) 2008-12-03 2017-04-12 Genmab A/S Antibody variants having modifications in the constant region
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
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
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
US9150658B2 (en) 2008-12-09 2015-10-06 Genmab A/S Human antibodies against tissue factor and methods of use thereof
EP3159358A1 (en) 2008-12-09 2017-04-26 Genmab A/S Human antibodies against human tissue factor
EP4279140A2 (en) 2008-12-09 2023-11-22 Genmab A/S Human antibodies against tissue factor
US9714297B2 (en) 2008-12-09 2017-07-25 Genmab A/S Human antibodies against tissue factor and methods of use thereof
CN112680475A (en) * 2008-12-18 2021-04-20 伊拉兹马斯大学鹿特丹医学中心 Non-human transgenic animals expressing humanized antibodies and uses thereof
CN112626117A (en) * 2008-12-18 2021-04-09 伊拉兹马斯大学鹿特丹医学中心 Non-human transgenic animals expressing humanized antibodies and uses thereof
WO2010073972A1 (en) 2008-12-22 2010-07-01 エーザイ・アール・アンド・ディー・マネジメント株式会社 Method for obtaining pancreatic progenitor cell using neph3
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
WO2010102175A1 (en) 2009-03-05 2010-09-10 Medarex, Inc. Fully human antibodies specific to cadm1
WO2010110346A1 (en) 2009-03-24 2010-09-30 独立行政法人理化学研究所 Leukemia stem cell markers
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
WO2010112034A2 (en) 2009-04-02 2010-10-07 Aarhus Universitet Compositions and methods for treatment and diagnosis of synucleinopathies
WO2010117325A1 (en) 2009-04-08 2010-10-14 Olle Hernell New methods for treatment of inflammatory diseases
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
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
WO2010119704A1 (en) 2009-04-17 2010-10-21 Immunas Pharma, Inc. Antibodies that specifically bind to a beta oligomers and use thereof
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
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
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
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
US10165763B2 (en) 2009-07-08 2019-01-01 Kymab Limited Animal models and therapeutic molecules
US11606941B2 (en) 2009-07-08 2023-03-21 Kymab Limited Animal models and therapeutic molecules
US10064398B2 (en) 2009-07-08 2018-09-04 Kymab Limited Animal models and therapeutic molecules
US11564380B2 (en) 2009-07-08 2023-01-31 Kymab Limited Animal models and therapeutic molecules
US11812731B2 (en) 2009-07-08 2023-11-14 Kymab Ltd. Animal models and therapeutic molecules
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
WO2011016238A1 (en) 2009-08-06 2011-02-10 Immunas Pharma, Inc. Antibodies that specifically bind to a beta oligomers and use thereof
WO2011016239A1 (en) 2009-08-06 2011-02-10 Immunas Pharma, Inc. Antibodies that specifically bind to a beta oligomers and use thereof
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
US8557239B2 (en) 2009-09-14 2013-10-15 Abbvie Inc. Methods for treating psoriasis using antibodies that bind to the P40 subunit of IL-12 and/or IL-23
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
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
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
EP3168232A1 (en) 2009-11-13 2017-05-17 Dana-Farber Cancer Institute, Inc. Compositions, kits, and methods for the diagnosis, prognosis, monitoring, treatment and modulation of post-transplant lymphoproliferative disorders and hypoxia associated angiogenesis disorders using galectin-1
EP2719708A2 (en) 2009-11-13 2014-04-16 U3 Pharma GmbH Material and methods for treating or preventing HER-3 associated diseases
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
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
WO2011062926A2 (en) 2009-11-17 2011-05-26 Medarex, Inc. 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
WO2011085343A1 (en) 2010-01-11 2011-07-14 Alexion Pharmaceuticals, Inc Biomarkers of immunomodulatory effects in humans treated with anti-cd200 antibodies
EP3892292A2 (en) 2010-01-20 2021-10-13 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
EP3378486A2 (en) 2010-01-20 2018-09-26 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
US10143186B2 (en) 2010-02-08 2018-12-04 Regeneron Pharmaceuticals, Inc. Common light chain mouse
US9796788B2 (en) 2010-02-08 2017-10-24 Regeneron Pharmaceuticals, Inc. Mice expressing a limited immunoglobulin light chain repertoire
US10412940B2 (en) 2010-02-08 2019-09-17 Regeneron Pharmaceuticals, Inc. Mice expressing a limited immunoglobulin light chain repertoire
US10986820B2 (en) 2010-02-08 2021-04-27 Regeneron Pharmaceuticals, Inc. Common light chain mouse
US11026407B2 (en) 2010-02-08 2021-06-08 Regeneran Pharmaceuticals, Inc. Mice expressing a limited immunoglobulin light chain repertoire
US9969814B2 (en) 2010-02-08 2018-05-15 Regeneron Pharmaceuticals, Inc. Methods for making fully human bispecific antibodies using a common light chain
US10167344B2 (en) 2010-02-08 2019-01-01 Regeneron Pharmaceuticals, Inc. Mice expressing a limited immunoglobulin light chain repertoire
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
WO2011100403A1 (en) 2010-02-10 2011-08-18 Immunogen, Inc Cd20 antibodies and uses thereof
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
WO2011108714A1 (en) 2010-03-04 2011-09-09 中外製薬株式会社 Antibody constant region variant
EP3904391A1 (en) 2010-03-10 2021-11-03 Genmab A/S Monoclonal antibodies against c-met
US9068011B2 (en) 2010-03-10 2015-06-30 Genmab A+S Monoclonal antibodies against c-Met
WO2011110642A2 (en) 2010-03-10 2011-09-15 Genmab A/S Monoclonal antibodies against c-met
US11512140B2 (en) 2010-03-10 2022-11-29 Genmab A/S Monoclonal antibodies against c-Met
EP3511342A1 (en) 2010-03-10 2019-07-17 Genmab A/S Monoclonal antibodies against c-met
WO2011116090A1 (en) 2010-03-17 2011-09-22 Abbott Research B.V. Anti-nerve growth factor (ngf) antibody compositions
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
US10745467B2 (en) 2010-03-26 2020-08-18 The Trustees Of Dartmouth College VISTA-Ig for treatment of autoimmune, allergic and inflammatory disorders
US10604587B2 (en) 2010-03-31 2020-03-31 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
US11104743B2 (en) 2010-03-31 2021-08-31 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
US11220555B2 (en) 2010-03-31 2022-01-11 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
US10829564B2 (en) 2010-03-31 2020-11-10 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
US9580491B2 (en) 2010-03-31 2017-02-28 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
US10662255B2 (en) 2010-03-31 2020-05-26 Ablexis, Llc Genetic engineering of non-human animals for the production of chimeric antibodies
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
WO2011130434A2 (en) 2010-04-13 2011-10-20 Celldex Therapeutics Inc. Antibodies that bind human cd27 and uses thereof
EP3165540A1 (en) 2010-04-13 2017-05-10 Celldex Therapeutics, Inc. Antibodies that bind human cd27 and uses thereof
WO2011131746A2 (en) 2010-04-20 2011-10-27 Genmab A/S Heterodimeric antibody fc-containing proteins and methods for production thereof
US11407821B2 (en) 2010-04-30 2022-08-09 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
US9434784B1 (en) 2010-04-30 2016-09-06 Alexion Pharmaceuticals, Inc. Nucleic acids encodng anti-C5A antibodies
US9371378B1 (en) 2010-04-30 2016-06-21 Alexion Pharmaceuticals, Inc. Anti-C5a antibodies
US10450370B2 (en) 2010-04-30 2019-10-22 Alexion Pharmaceuticals, Inc. Anti-C5a antibodies
US9963503B2 (en) 2010-04-30 2018-05-08 Alexion Pharmaceuticals, Inc. Methods of producing anti-C5a antibodies
EP2824111A2 (en) 2010-04-30 2015-01-14 Alexion Pharmaceuticals, Inc. Anti-C5A Antibodies and Methods for Using the 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
US9309310B2 (en) 2010-04-30 2016-04-12 Alexion Pharmaceuticals, Inc. Nucleic acids encoding anti-C5a antibodies
WO2011140254A1 (en) 2010-05-04 2011-11-10 Adimab, Llc Antibodies against epidermal growth factor receptor (egfr) and uses thereof
WO2011140151A1 (en) 2010-05-04 2011-11-10 Dyax Corp. Antibodies against epidermal growth factor receptor (egfr)
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
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
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
WO2011147986A1 (en) 2010-05-27 2011-12-01 Genmab A/S Monoclonal antibodies against her2
EP3539988A2 (en) 2010-05-27 2019-09-18 Genmab A/S Monoclonal antibodies against her2
WO2011147982A2 (en) 2010-05-27 2011-12-01 Genmab A/S Monoclonal antibodies against her2 epitope
WO2011149051A1 (en) 2010-05-28 2011-12-01 中外製薬株式会社 Antitumor t cell response enhancer
EP4115906A1 (en) 2010-05-28 2023-01-11 Chugai Seiyaku Kabushiki Kaisha Antitumor t cell response enhancer
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
WO2011154453A1 (en) 2010-06-09 2011-12-15 Genmab A/S Antibodies against human cd38
EP3613774A1 (en) 2010-06-09 2020-02-26 Genmab A/S Antibodies against human cd38
US9492565B2 (en) 2010-06-15 2016-11-15 Genmab A/S Human antibody drug conjugates against tissue factor
US9168314B2 (en) 2010-06-15 2015-10-27 Genmab A/S Human antibody drug conjugates against tissue factor
EP3281956A2 (en) 2010-06-15 2018-02-14 Genmab A/S Human antibody drug conjugates against tissue factor
WO2011157741A2 (en) 2010-06-15 2011-12-22 Genmab A/S Human antibody drug conjugates against tissue factor
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
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
US10793829B2 (en) 2010-07-26 2020-10-06 Trianni, Inc. Transgenic mammals and methods of use thereof
US10662256B2 (en) 2010-07-26 2020-05-26 Trianni, Inc. Transgenic mammals and methods of use thereof
US10881084B2 (en) 2010-07-26 2021-01-05 Trianni, Inc Transgenic animals and methods of use
US10954310B2 (en) 2010-08-02 2021-03-23 Regeneran Pharmaceuticals, Inc. Mice that make VL binding proteins
US9516868B2 (en) 2010-08-02 2016-12-13 Regeneron Pharmaceuticals, Inc. Mice that make VL binding proteins
WO2012018404A2 (en) 2010-08-06 2012-02-09 U3 Pharma Gmbh Use of her3 binding agents in prostate treatment
WO2012022814A1 (en) 2010-08-20 2012-02-23 Novartis Ag Antibodies for epidermal growth factor receptor 3 (her3)
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
WO2012035518A1 (en) 2010-09-17 2012-03-22 Compugen Ltd. Compositions and methods for treatment of drug resistant multiple myeloma
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
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
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
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
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
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
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
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
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
WO2012073985A1 (en) 2010-11-30 2012-06-07 中外製薬株式会社 Cytotoxicity-inducing therapeutic agent
EP4303237A2 (en) 2010-11-30 2024-01-10 Chugai Seiyaku Kabushiki Kaisha Cytotoxicity-inducing therapeutic agent
EP4279511A2 (en) 2010-11-30 2023-11-22 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
EP3434767A1 (en) 2010-11-30 2019-01-30 Chugai Seiyaku Kabushiki Kaisha Cytotoxicity-inducing therapeutic agent
EP4303236A2 (en) 2010-11-30 2024-01-10 Chugai Seiyaku Kabushiki Kaisha Cytotoxicity-inducing therapeutic agent
EP4279513A2 (en) 2010-11-30 2023-11-22 Chugai Seiyaku Kabushiki Kaisha Cytotoxicity-inducing therapeutic agent
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
WO2012118042A1 (en) 2011-02-28 2012-09-07 独立行政法人国立循環器病研究センター Medicinal agent for inhibiting metastasis of malignant tumor
EP3189835A1 (en) 2011-02-28 2017-07-12 National Cerebral and Cardiovascular Center Medical agent for suppressing malignant tumor metastasis
US8906635B2 (en) 2011-02-28 2014-12-09 Northshore University Healthsystem Methods of diagnosing Clostridium difficile infection
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
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
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
WO2012136552A1 (en) 2011-04-08 2012-10-11 H. Lundbeck A/S ANTIBODIES SPECIFIC TO PYROGLUTAMATED Αβ
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
WO2012143524A2 (en) 2011-04-20 2012-10-26 Genmab A/S Bispecific antibodies against her2 and cd3
WO2012143523A1 (en) 2011-04-20 2012-10-26 Genmab A/S Bispecifc antibodies against her2
US9255143B2 (en) 2011-04-27 2016-02-09 Abbvie Inc. Methods for controlling the galactosylation profile of recombinantly-expressed proteins
US9365645B1 (en) 2011-04-27 2016-06-14 Abbvie, Inc. Methods for controlling the galactosylation profile of recombinantly-expressed proteins
US9505834B2 (en) 2011-04-27 2016-11-29 Abbvie Inc. Methods for controlling the galactosylation profile of recombinantly-expressed proteins
US9090688B2 (en) 2011-04-27 2015-07-28 Abbvie Inc. Methods for controlling the galactosylation profile of recombinantly-expressed proteins
US9062106B2 (en) 2011-04-27 2015-06-23 Abbvie Inc. Methods for controlling the galactosylation profile of recombinantly-expressed proteins
WO2012154999A1 (en) 2011-05-10 2012-11-15 Amgen Inc. Methods of treating or preventing cholesterol related disorders
EP3597218A1 (en) 2011-05-10 2020-01-22 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
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
US9428574B2 (en) 2011-06-30 2016-08-30 Compugen Ltd. Polypeptides and uses thereof for treatment of autoimmune disorders and infection
EP4011913A1 (en) 2011-06-30 2022-06-15 Chugai Seiyaku Kabushiki Kaisha Heterodimerized polypeptide
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
WO2013004841A1 (en) 2011-07-06 2013-01-10 Genmab A/S Modulation of complement-dependent cytotoxicity through modifications of the c-terminus of antibody heavy chains
EP3970746A2 (en) 2011-07-06 2022-03-23 Genmab B.V. Polypeptide variants and uses thereof
WO2013004842A2 (en) 2011-07-06 2013-01-10 Genmab A/S Antibody variants 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
US11357217B2 (en) 2011-08-05 2022-06-14 Regeneron Pharmaceuticals, Inc. Humanized universal light chain mice
US10130081B2 (en) 2011-08-05 2018-11-20 Regeneron Pharmaceuticals, Inc. Humanized universal light chain mice
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
EP3128009A1 (en) * 2011-09-19 2017-02-08 Kymab Limited Antibodies, variable domains&chains tailored for human use
US11051497B2 (en) 2011-09-19 2021-07-06 Kymab Limited Manipulation of immunoglobulin gene diversity and multi-antibody therapeutics
EP3128009B1 (en) 2011-09-19 2020-07-29 Kymab Limited Antibodies, variable domains & chains tailored for human use
US9963716B2 (en) 2011-09-26 2018-05-08 Kymab Limited Chimaeric surrogate light chains (SLC) comprising human VpreB
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
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
EP3680251A1 (en) 2011-09-30 2020-07-15 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecules for promoting elimination 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
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
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
US11261248B2 (en) 2011-10-17 2022-03-01 Regeneron Pharmaceuticals, Inc. Restricted immunoglobulin heavy chain mice
US10246509B2 (en) 2011-10-17 2019-04-02 Regeneron Pharmaceuticals, Inc. Restricted immunoglobulin heavy chain mice
US9932398B2 (en) 2011-10-17 2018-04-03 Regeneron Pharmaceuticals, Inc. Restricted immunoglobulin heavy chain mice
WO2013060867A2 (en) 2011-10-27 2013-05-02 Genmab A/S Production of heterodimeric proteins
EP3603671A2 (en) 2011-10-28 2020-02-05 Chugai Seiyaku Kabushiki Kaisha Cancer stem cell-specific molecule
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
WO2013067054A1 (en) 2011-11-01 2013-05-10 Bionomics, Inc. Antibodies and methods of treating cancer
US9220774B2 (en) 2011-11-01 2015-12-29 Bionomics Inc. Methods of treating cancer by administering anti-GPR49 antibodies
WO2013067055A1 (en) 2011-11-01 2013-05-10 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)
US9221907B2 (en) 2011-11-01 2015-12-29 Bionomics Inc. Anti-GPR49 monoclonal antibodies
WO2013067060A1 (en) 2011-11-01 2013-05-10 Bionomics, Inc. Anti-gpr49 antibodies
US10598653B2 (en) 2011-11-01 2020-03-24 Bionomics Inc. Methods of blocking cancer stem cell growth
WO2013067057A1 (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
WO2013075048A1 (en) 2011-11-16 2013-05-23 Amgen Inc. Methods of treating epidermal growth factor deletion mutant viii related disorders
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
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)
EP3590538A1 (en) 2011-12-05 2020-01-08 Novartis AG Antibodies for epidermal growth factor receptor 3 (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
US9663575B2 (en) 2012-02-06 2017-05-30 Inhibrx, Lp CD47 antibodies and methods of use thereof
US9045541B2 (en) 2012-02-06 2015-06-02 Inhibrx Llc CD47 antibodies and methods of use thereof
EP3578569A1 (en) 2012-02-06 2019-12-11 Inhibrx, Inc. 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
EP3738980A1 (en) 2012-02-24 2020-11-18 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule for promoting disappearance of antigen via fc gamma riib
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
US9896516B2 (en) 2012-03-28 2018-02-20 Kymab Limited Animal models and therapeutic molecules
US9938358B2 (en) 2012-03-28 2018-04-10 Kymab Limited Animal models and therapeutic molecules
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
US10251377B2 (en) 2012-03-28 2019-04-09 Kymab Limited Transgenic non-human vertebrate for the expression of class-switched, fully human, antibodies
US9924705B2 (en) 2012-03-28 2018-03-27 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
US9683033B2 (en) 2012-04-20 2017-06-20 Abbvie, Inc. Cell culture methods to reduce acidic species
US9193787B2 (en) 2012-04-20 2015-11-24 Abbvie Inc. Human antibodies that bind human TNF-alpha and methods of preparing the same
US9708400B2 (en) 2012-04-20 2017-07-18 Abbvie, Inc. Methods to modulate lysine variant distribution
US9181572B2 (en) 2012-04-20 2015-11-10 Abbvie, Inc. Methods to modulate lysine variant distribution
US9334319B2 (en) 2012-04-20 2016-05-10 Abbvie Inc. Low acidic species compositions
US9957318B2 (en) 2012-04-20 2018-05-01 Abbvie Inc. Protein purification methods to reduce acidic species
US9505833B2 (en) 2012-04-20 2016-11-29 Abbvie Inc. Human antibodies that bind human TNF-alpha and methods of preparing the same
US9346879B2 (en) 2012-04-20 2016-05-24 Abbvie Inc. Protein purification methods to reduce acidic species
US9359434B2 (en) 2012-04-20 2016-06-07 Abbvie, Inc. Cell culture methods to reduce acidic species
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
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
US9249182B2 (en) 2012-05-24 2016-02-02 Abbvie, Inc. Purification of antibodies using hydrophobic interaction chromatography
WO2013180201A1 (en) 2012-05-30 2013-12-05 中外製薬株式会社 Antigen-binding molecule for eliminating aggregated antigens
EP3892638A1 (en) 2012-05-30 2021-10-13 Chugai Seiyaku Kabushiki Kaisha 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
EP3795215A1 (en) 2012-05-30 2021-03-24 Chugai Seiyaku Kabushiki Kaisha 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
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
US11666040B2 (en) 2012-06-12 2023-06-06 Regeneron Pharmaceuticals, Inc. Humanized non-human animals with restricted immunoglobulin heavy chain loci
US11559050B2 (en) 2012-06-12 2023-01-24 Regeneron Pharmaceuticals, Inc. Humanized non-human animals with restricted immunoglobulin heavy chain loci
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
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
US11180557B2 (en) 2012-06-22 2021-11-23 King's College London Vista modulators for diagnosis and treatment of cancer
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
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
EP3632462A1 (en) 2012-07-06 2020-04-08 Genmab B.V. Dimeric protein with triple mutations
WO2014006217A1 (en) 2012-07-06 2014-01-09 Genmab B.V. Dimeric protein with triple mutations
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
EP3721900A1 (en) 2012-08-24 2020-10-14 Chugai Seiyaku Kabushiki Kaisha Fcgammariib-specific fc region variant
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
US9206390B2 (en) 2012-09-02 2015-12-08 Abbvie, Inc. Methods to control protein heterogeneity
US9512214B2 (en) 2012-09-02 2016-12-06 Abbvie, Inc. Methods to control protein heterogeneity
US9290568B2 (en) 2012-09-02 2016-03-22 Abbvie, Inc. Methods to control protein heterogeneity
US9234033B2 (en) 2012-09-02 2016-01-12 Abbvie, Inc. Methods to control protein heterogeneity
EP3725805A1 (en) 2012-09-07 2020-10-21 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
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
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
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
US9593165B2 (en) 2012-11-08 2017-03-14 University Of Miyazaki Antibody capable of specifically recognizing transferrin receptor
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
EP3738979A1 (en) 2013-01-10 2020-11-18 Genmab A/S Inert format
WO2014108198A1 (en) 2013-01-10 2014-07-17 Genmab B.V. Human igg1 fc region variants and uses thereof
WO2014108483A1 (en) 2013-01-10 2014-07-17 Genmab B.V. Inert format
WO2014114801A1 (en) 2013-01-25 2014-07-31 Amgen Inc. Antibodies targeting cdh19 for melanoma
US11498964B2 (en) 2013-01-25 2022-11-15 Amgen Research (Munich) Gmbh Antibody constructs for CDH19 and CD3
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
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
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
WO2014122613A1 (en) 2013-02-08 2014-08-14 Novartis Ag Anti-il-17a antibodies and their use in treating autoimmune and inflammatory disorders
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
US9930871B2 (en) 2013-02-20 2018-04-03 Regeneron Pharmaceuticals, Inc. Non-human animals with modified immunoglobulin heavy chain sequences
WO2014136910A1 (en) 2013-03-08 2014-09-12 国立大学法人大阪大学 ANTIBODY TO PEPTIDE ENCODED BY Exon-21 OF PERIOSTIN, AND PHARMACEUTICAL COMPOSITION FOR PREVENTION OR TREATMENT OF INFLAMMATORY DISEASE CONTAINING SAID ANTIBODY
US9708399B2 (en) 2013-03-14 2017-07-18 Abbvie, Inc. Protein purification using displacement chromatography
US9499614B2 (en) 2013-03-14 2016-11-22 Abbvie Inc. Methods for modulating protein glycosylation profiles of recombinant protein therapeutics using monosaccharides and oligosaccharides
EP3611189A1 (en) 2013-03-14 2020-02-19 Novartis AG Antibodies against notch 3
US8921526B2 (en) 2013-03-14 2014-12-30 Abbvie, Inc. Mutated anti-TNFα antibodies and methods of their use
WO2014159239A2 (en) 2013-03-14 2014-10-02 Novartis Ag Antibodies against notch 3
US9067990B2 (en) 2013-03-14 2015-06-30 Abbvie, Inc. Protein purification using displacement chromatography
WO2014140368A1 (en) 2013-03-15 2014-09-18 Amgen Research (Munich) Gmbh Antibody constructs for influenza m2 and cd3
EP3653642A1 (en) 2013-03-15 2020-05-20 Amgen Research (Munich) GmbH Single chain binding molecules comprising n-terminal abp
WO2014140358A1 (en) 2013-03-15 2014-09-18 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
US9788534B2 (en) 2013-03-18 2017-10-17 Kymab Limited Animal models and therapeutic molecules
US10226033B2 (en) 2013-03-18 2019-03-12 Kymab Limited Animal models and therapeutic molecules
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
US10730930B2 (en) 2013-05-02 2020-08-04 Kymab Limited Antibodies, variable domains and chains tailored for human use
US11820810B2 (en) 2013-05-02 2023-11-21 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
EP3705494A2 (en) 2013-08-14 2020-09-09 Sachdev Sidhu Antibodies against frizzled proteins and methods of use 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
WO2015022658A2 (en) 2013-08-14 2015-02-19 Novartis Ag Methods of treating sporadic inclusion body myositis
WO2015033831A1 (en) 2013-09-04 2015-03-12 国立大学法人 大阪大学 Dpp-4-targeting vaccine for treating diabetes
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
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
US9598667B2 (en) 2013-10-04 2017-03-21 Abbvie Inc. Use of metal ions for modulation of protein glycosylation profiles of recombinant proteins
US9181337B2 (en) 2013-10-18 2015-11-10 Abbvie, Inc. Modulated lysine variant species compositions and methods for producing and using the same
US8946395B1 (en) 2013-10-18 2015-02-03 Abbvie Inc. Purification of proteins using hydrophobic interaction chromatography
US9085618B2 (en) 2013-10-18 2015-07-21 Abbvie, Inc. Low acidic species compositions and methods for producing and using the same
US9688752B2 (en) 2013-10-18 2017-06-27 Abbvie Inc. Low acidic species compositions and methods for producing and using the same using displacement chromatography
US9522953B2 (en) 2013-10-18 2016-12-20 Abbvie, Inc. Low acidic species compositions and methods for producing and using the same
US9017687B1 (en) 2013-10-18 2015-04-28 Abbvie, Inc. Low acidic species compositions and methods for producing and using the same using displacement chromatography
US9315574B2 (en) 2013-10-18 2016-04-19 Abbvie, Inc. Low acidic species compositions and methods for producing and using the same
US9266949B2 (en) 2013-10-18 2016-02-23 Abbvie, Inc. Low acidic species compositions and methods for producing and using the same
US9499616B2 (en) 2013-10-18 2016-11-22 Abbvie Inc. Modulated lysine variant species compositions and methods for producing and using the same
US9200070B2 (en) 2013-10-18 2015-12-01 Abbvie, Inc. Low acidic species compositions and methods for producing and using the same
US9200069B2 (en) 2013-10-18 2015-12-01 Abbvie, Inc. Low acidic species compositions and methods for producing and using the same
WO2015068847A1 (en) 2013-11-11 2015-05-14 中外製薬株式会社 Antigen-binding molecule containing modified antibody variable region
US9550826B2 (en) 2013-11-15 2017-01-24 Abbvie Inc. Glycoengineered binding protein compositions
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
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
US11014987B2 (en) 2013-12-24 2021-05-25 Janssen Pharmaceutics Nv Anti-vista antibodies and fragments, uses thereof, and methods of identifying same
US11242392B2 (en) 2013-12-24 2022-02-08 Janssen Pharmaceutica Nv Anti-vista antibodies and fragments
WO2015097536A2 (en) 2013-12-24 2015-07-02 Janssen Pharmaceutical Nv Anti-vista antibodies and fragments
EP4043493A1 (en) 2013-12-24 2022-08-17 Janssen Pharmaceutica NV Anti-vista antibodies and fragments
EP3712174A1 (en) 2013-12-24 2020-09-23 Janssen Pharmaceutica NV Anti-vista antibodies and fragments
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
WO2015099167A1 (en) 2013-12-27 2015-07-02 国立大学法人大阪大学 Vaccine targeting il-17a
WO2015099165A1 (en) 2013-12-27 2015-07-02 中外製薬株式会社 Method for purifying antibody having low isoelectric point
WO2015121383A1 (en) 2014-02-12 2015-08-20 Michael Uhlin Bispecific antibodies for use in stem cell transplantation
US10787522B2 (en) 2014-03-21 2020-09-29 Regeneron Pharmaceuticals, Inc. VL antigen binding proteins exhibiting distinct binding characteristics
US10881085B2 (en) 2014-03-21 2021-01-05 Regeneron Pharmaceuticals, Inc. Non-human animals that make single domain binding proteins
EP3572093A1 (en) 2014-03-31 2019-11-27 Debiopharm International SA Fgfr fusions
WO2015150900A2 (en) 2014-03-31 2015-10-08 Debiopharm International Sa Fgfr fusions
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
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
EP3610924A1 (en) 2014-06-06 2020-02-19 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
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
WO2016007414A1 (en) 2014-07-08 2016-01-14 New York University Tau imaging ligands and their uses in the diagnosis and treatment of tauopathy
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
WO2016005593A1 (en) 2014-07-11 2016-01-14 Genmab A/S Antibodies binding axl
EP3763738A1 (en) 2014-07-11 2021-01-13 Genmab A/S Antibodies binding axl
US11661462B2 (en) 2014-07-31 2023-05-30 Amgen Research (Munich) Gmbh Optimized cross-species specific bispecific single chain antibody contructs
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
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
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
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
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
US10894818B2 (en) 2014-10-03 2021-01-19 Massachusetts Institute Of Technology Antibodies that bind Ebola glycoprotein and uses thereof
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
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
WO2016073685A1 (en) 2014-11-05 2016-05-12 Annexon, Inc. Humanized anti-complement factor c1q antibodies and uses thereof
EP4295911A2 (en) 2014-11-05 2023-12-27 Annexon, Inc. Humanized anti-complement factor c1q antibodies and uses thereof
EP4268843A2 (en) 2014-11-07 2023-11-01 F. Hoffmann-La Roche Ltd Improved il-6 antibodies
US11142571B2 (en) 2014-11-07 2021-10-12 Sesen Bio, Inc. IL-6 antibodies
WO2016073894A1 (en) 2014-11-07 2016-05-12 Eleven Biotherapeutics, Inc. Therapeutic agents with increased ocular retention
EP3632931A1 (en) 2014-11-07 2020-04-08 Sesen Bio, Inc. Improved il-6 antibodies
EP3725808A1 (en) 2014-11-21 2020-10-21 Bristol-Myers Squibb Company Antibodies against cd73 and uses thereof
WO2016081748A2 (en) 2014-11-21 2016-05-26 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
EP3945096A1 (en) 2014-12-19 2022-02-02 Regenesance B.V. Antibodies that bind human c6 and uses thereof
WO2016097865A1 (en) 2014-12-19 2016-06-23 Regenesance B.V. Antibodies that bind human c6 and uses thereof
EP4249066A2 (en) 2014-12-23 2023-09-27 Bristol-Myers Squibb Company Antibodies to tigit
EP4269440A2 (en) 2015-02-27 2023-11-01 Chugai Seiyaku Kabushiki Kaisha Composition for treating il-6-related diseases
WO2016149088A1 (en) 2015-03-13 2016-09-22 Bristol-Myers Squibb Company Use of alkaline washes during chromatography to remove impurities
EP4194071A1 (en) 2015-03-13 2023-06-14 Bristol-Myers Squibb Company Use of alkaline washes during chromatography to remove impurities
US11111314B2 (en) 2015-03-19 2021-09-07 Regeneron Pharmaceuticals, Inc. Non-human animals that select for light chain variable regions that bind antigen
WO2016153983A1 (en) 2015-03-20 2016-09-29 Bristol-Myers Squibb Company Use of dextran for protein purification
WO2016153978A1 (en) 2015-03-20 2016-09-29 Bristol-Myers Squibb Company Use of dextran to enhance protein purification by affinity chromatography
WO2016166014A1 (en) 2015-04-17 2016-10-20 F. Hoffmann-La Roche Ag Combination therapy with coagulation factors and multispecific antibodies
US11028171B2 (en) 2015-04-17 2021-06-08 Amgen Research (Munich) Gmbh Bispecific antibody constructs for CDH3 and CD3
WO2016166360A1 (en) 2015-04-17 2016-10-20 Bayer Pharma Aktiengesellschaft Bispecific antibody constructs for cdh3 and cd3
US11926666B2 (en) 2015-04-17 2024-03-12 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
EP3584260A1 (en) 2015-04-28 2019-12-25 Mitsubishi Tanabe Pharma Corporation Rgma binding protein and use thereof
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
EP3722314A1 (en) 2015-06-24 2020-10-14 Janssen Pharmaceutica NV Anti-vista antibodies and fragments
WO2016207717A1 (en) 2015-06-24 2016-12-29 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
EP3730520A1 (en) 2015-07-10 2020-10-28 Genmab A/S Axl-specific antibody-drug conjugates for cancer treatment
WO2017009258A1 (en) 2015-07-10 2017-01-19 Genmab A/S Axl-specific antibody-drug conjugates for cancer treatment
US11542323B2 (en) 2015-07-13 2023-01-03 H. Lundbeck A/S Agents, uses and methods for the treatment of synucleinopathy
US11548950B2 (en) 2015-07-13 2023-01-10 H. Lundbeck A/S Agent, uses and methods for treatment
US10479835B2 (en) 2015-07-13 2019-11-19 H. Lundbeck A/S Agent, uses and methods for treatment
US10934348B2 (en) 2015-07-13 2021-03-02 H. Lundbeck A/S Antibodies specific for hyperphosphorylated tau and methods of use thereof
US10889650B2 (en) 2015-07-13 2021-01-12 H. Lundbeck A/S Agent, uses and methods for treatment
US10428147B2 (en) 2015-07-13 2019-10-01 H. Lundbeck A/S Anti-sortilin antibodies, uses and methods for treatment
US10562962B2 (en) 2015-07-13 2020-02-18 H. Lundbeck A/S Antibodies specific for hyperphosphorylated tau and methods of use thereof
US11421024B2 (en) 2015-07-13 2022-08-23 H. Lundbeck A/S Agents, uses and methods for the treatment of synucleinopathy
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
US10364285B2 (en) 2015-07-13 2019-07-30 H. Lundbeck A/S Antibodies for the treatment of synucleinopathy
US10358484B2 (en) 2015-07-13 2019-07-23 H. Lundbeck A/S Antibodies for the treatment of synucleinopathy
US10800836B2 (en) 2015-07-13 2020-10-13 H. Lundbeck A/S Agents, uses and methods for the treatment of synucleinopathy
US10358483B2 (en) 2015-07-13 2019-07-23 H. Lundbeck A/S Antibodies for the treatment of synucleinopathy
US10358482B2 (en) 2015-07-13 2019-07-23 H. Lundbeck A/S Antibodies for the treatment of synucleinopathy
US11524995B2 (en) 2015-07-13 2022-12-13 H. Lundbeck A/S Agents, uses and methods for the treatment of synucleinopathy
US11591396B2 (en) 2015-07-31 2023-02-28 Amgen Research (Munich) Gmbh Antibody constructs for DLL3 and CD3
EP4219562A2 (en) 2015-07-31 2023-08-02 Amgen Research (Munich) GmbH Antibody constructs for flt3 and cd3
WO2017021356A1 (en) 2015-07-31 2017-02-09 Amgen Research (Munich) Gmbh Bispecific antibody constructs binding mesothelin and cd3
WO2017021349A1 (en) 2015-07-31 2017-02-09 Amgen Research (Munich) Gmbh Bispecific antibody constructs binding dll3 and cd3
EP4327885A2 (en) 2015-07-31 2024-02-28 Amgen Research (Munich) GmbH Antibody constructs for msln and cd3
US11884720B2 (en) 2015-07-31 2024-01-30 Amgen Research (Munich) Gmbh Antibody constructs for MSLN and CD3
WO2017021370A1 (en) 2015-07-31 2017-02-09 Amgen Research (Munich) Gmbh Bispecific antibody constructs binding egfrviii and cd3
EP3865514A1 (en) 2015-07-31 2021-08-18 Amgen Research (Munich) GmbH Bispecific antibody constructs binding dll3 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
US10851170B2 (en) 2015-07-31 2020-12-01 Amgen Research (Munich) Gmbh Antibody constructs for CD70 and CD3
WO2017021354A1 (en) 2015-07-31 2017-02-09 Amgen Research (Munich) Gmbh Antibody constructs for cd70 and cd3
US10519241B2 (en) 2015-07-31 2019-12-31 Amgen Research (Munich) Gmbh Antibody constructs for EGFRVIII and CD3
US10683351B2 (en) 2015-07-31 2020-06-16 Amgen Research (Munich) Gmbh Antibody constructs for DLL3 and CD3
US10294300B2 (en) 2015-07-31 2019-05-21 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
EP3912999A1 (en) 2015-07-31 2021-11-24 Amgen Research (Munich) GmbH Bispecific antibody constructs binding egfrviii and cd3
US11447567B2 (en) 2015-07-31 2022-09-20 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
US10988528B2 (en) 2015-08-13 2021-04-27 New York University Antibody-based molecules specific for the truncated ASP421 epitope of Tau and their uses in the diagnosis and treatment of tauopathy
WO2017027691A1 (en) 2015-08-13 2017-02-16 New York University Antibody-based molecules selective for the {p}ser404 epitope of tau and their uses in the diagnosis and treatment of tauopathy
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
WO2017086419A1 (en) 2015-11-18 2017-05-26 中外製薬株式会社 Method for enhancing humoral immune response
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
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
US10813346B2 (en) 2015-12-03 2020-10-27 Trianni, Inc. Enhanced immunoglobulin diversity
US11889821B2 (en) 2015-12-03 2024-02-06 Trianni, Inc. Enhanced immunoglobulin diversity
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
WO2017121877A1 (en) 2016-01-13 2017-07-20 Genmab A/S Axl-specific antibody-drug conjugates for cancer treatment
EP3851457A1 (en) 2016-01-21 2021-07-21 Novartis AG Multispecific molecules targeting cll-1
WO2017125897A1 (en) 2016-01-21 2017-07-27 Novartis Ag Multispecific molecules targeting cll-1
EP4206228A1 (en) 2016-02-03 2023-07-05 Amgen Research (Munich) GmbH Psma and cd3 bispecific t cell engaging constructs
US10301391B2 (en) 2016-02-03 2019-05-28 Amgen Research (Munich) Gmbh BCMA and CD3 bispecific T cell engaging antibody constructs
US11434302B2 (en) 2016-02-03 2022-09-06 Amgen Research (Munich) Gmbh 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
US11352433B2 (en) 2016-02-03 2022-06-07 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
US11053288B2 (en) 2016-02-04 2021-07-06 Trianni, Inc. Enhanced production of immunoglobulins
WO2017137830A1 (en) 2016-02-12 2017-08-17 Janssen Pharmaceutica Nv Anti-vista (b7h5) antibodies
US10899836B2 (en) 2016-02-12 2021-01-26 Janssen Pharmaceutica Nv Method of identifying anti-VISTA antibodies
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
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
US10870701B2 (en) 2016-03-15 2020-12-22 Generon (Shanghai) Corporation Ltd. Multispecific fab fusion proteins and use thereof
WO2017160754A1 (en) 2016-03-15 2017-09-21 Mersana Therapeutics,Inc. Napi2b-targeted antibody-drug conjugates and methods of use thereof
EP4302782A2 (en) 2016-03-15 2024-01-10 Mersana Therapeutics, Inc. Napi2b-targeted antibody-drug conjugates and methods of use thereof
US10745487B2 (en) 2016-03-22 2020-08-18 Bionomics Limited Method of treating cancer by administering an anti-LGR5 monoclonal antibody
WO2017172771A2 (en) 2016-03-29 2017-10-05 Janssen Biotech, Inc. Method of treating psoriasis with increased interval dosing of anti-il12/23 antibody
WO2017175058A1 (en) 2016-04-07 2017-10-12 Janssen Pharmaceutica Nv Anti-vista antibodies and fragments, uses thereof, and methods of identifying same
US11525000B2 (en) 2016-04-15 2022-12-13 Immunext, Inc. Anti-human VISTA antibodies and use thereof
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
WO2017184619A2 (en) 2016-04-18 2017-10-26 Celldex Therapeutics, Inc. Agonistic antibodies that bind human cd40 and uses 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
WO2017196663A1 (en) 2016-05-09 2017-11-16 Bristol-Myers Squibb Company Tl1a antibodies and uses thereof
US10968279B2 (en) 2016-05-09 2021-04-06 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
WO2017216724A1 (en) 2016-06-15 2017-12-21 Novartis Ag Methods for treating disease using inhibitors of bone morphogenetic protein 6 (bmp6)
US11111290B2 (en) 2016-07-12 2021-09-07 H. Lundbeck A/S Antibodies specific for hyperphosphorylated tau and methods of use thereof
WO2018011073A1 (en) 2016-07-12 2018-01-18 H. Lundbeck A/S Antibodies specific for hyperphosphorylated tau and methods of use thereof
US10647762B2 (en) 2016-07-12 2020-05-12 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
US10487142B2 (en) 2016-07-12 2019-11-26 H. Lundbeck A/S Antibodies specific for hyperphosphorylated tau and methods of use thereof
EP3878864A1 (en) 2016-07-12 2021-09-15 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
US11591392B2 (en) 2016-07-14 2023-02-28 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
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
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
WO2018049248A1 (en) 2016-09-09 2018-03-15 Icellhealth Consulting Llc Oncolytic virus equipped with bispecific engager molecules
WO2018064436A1 (en) 2016-09-30 2018-04-05 Janssen Biotech, Inc. Safe and effective method of treating psoriasis with anti-il23 specific antibody
WO2018071822A2 (en) 2016-10-13 2018-04-19 Massachusetts Institute Of Technology Antibodies that bind zika virus envelope protein and uses thereof
WO2018087720A1 (en) 2016-11-14 2018-05-17 Novartis Ag Compositions, methods, and therapeutic uses related to fusogenic protein minion
WO2018091444A1 (en) 2016-11-15 2018-05-24 H. Lundbeck A/S Agents, uses and methods for the treatment of synucleinopathy
US10889635B2 (en) 2016-11-15 2021-01-12 H. Lundbeck A/S Agents, uses and methods for the treatment of synucleinopathy
WO2018093841A1 (en) 2016-11-16 2018-05-24 Janssen Biotech, Inc. Method of treating psoriasis with anti-il-23 specific antibody
US11208474B2 (en) 2016-11-16 2021-12-28 Janssen Biotech, Inc. Method of treating psoriasis with anti-IL23 specific antibody
WO2018097308A1 (en) 2016-11-28 2018-05-31 中外製薬株式会社 Ligand-binding molecule having adjustable ligand binding activity
WO2018109058A1 (en) 2016-12-16 2018-06-21 H. Lundbeck A/S Agents, uses and methods
US11325968B2 (en) 2016-12-16 2022-05-10 H. Lundbeck A/S Alpha-synuclein antibodies
WO2018115225A1 (en) 2016-12-22 2018-06-28 H. Lundbeck A/S Monoclonal anti-alpha-synuclein antibodies for preventing tau aggregation
US10364286B2 (en) 2016-12-22 2019-07-30 H. Lundbeck A/S Monoclonal anti-alpha-synuclein antibodies for preventing tau aggregation
WO2018119380A2 (en) 2016-12-23 2018-06-28 Bristol-Myers Squibb Company Design of therapeutic immunoglobulin g4 for improved bioanalytical and bioprocessing properties
WO2018127519A1 (en) 2017-01-04 2018-07-12 H. Lundbeck A/S Antibodies specific for hyperphosphorylated tau for the treatment of ocular diseases
WO2018129451A2 (en) 2017-01-09 2018-07-12 Merrimack Pharmaceuticals, Inc. Anti-fgfr antibodies and methods of use
US11041020B2 (en) 2017-01-30 2021-06-22 Janssen Biotech, Inc. Methods for the treatment of active Psoriatic Arthritis
WO2018141910A1 (en) 2017-02-02 2018-08-09 Amgen Research (Munich) Gmbh Low ph pharmaceutical composition comprising t cell engaging antibody constructs
US11014982B2 (en) 2017-02-07 2021-05-25 Janssen Biotech, Inc. Anti-TNF antibodies, compositions, and methods for the treatment of active ankylosing spondylitis
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
US11142570B2 (en) 2017-02-17 2021-10-12 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
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
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
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
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
US10894833B2 (en) 2017-07-20 2021-01-19 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
WO2019058345A2 (en) 2017-09-25 2019-03-28 Janssen Biotech, Inc. Safe and effective method of treating lupus with anti-il12/il23 antibody
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
WO2019107384A1 (en) 2017-11-28 2019-06-06 中外製薬株式会社 Ligand-binding molecule having adjustable ligand-binding activity
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
US11572405B2 (en) 2018-01-12 2023-02-07 Bristol-Myers Squibb Company Combination therapy with anti-IL-8 antibodies and anti-PD-1 antibodies for treating cancer
WO2019151418A1 (en) 2018-01-31 2019-08-08 元一 加藤 Therapeutic agent for asthma containing il-6 inhibitor
WO2019171252A1 (en) 2018-03-05 2019-09-12 Janssen Biotech, Inc. Methods of treating crohn's disease with anti-il23 specific antibody
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
WO2019217455A1 (en) 2018-05-07 2019-11-14 Genmab A/S Methods of treating cancer with a combination of an anti-pd-1 antibody and an anti-tissue factor antibody-drug conjugate
WO2019217457A1 (en) 2018-05-07 2019-11-14 Genmab A/S Methods of treating cancer with a combination of an anti-pd-1 antibody and an anti-tissue factor antibody-drug conjugate
WO2019215701A1 (en) 2018-05-11 2019-11-14 Janssen Biotech, Inc. Methods of treating depression using il-23 antibodies
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
WO2020013238A1 (en) 2018-07-10 2020-01-16 田辺三菱製薬株式会社 Prevention or treatment method for peripheral neuropathy or pain accompanying disease in which peripheral neuropathy or astrocyte disorder is recognized
WO2020016838A2 (en) 2018-07-18 2020-01-23 Janssen Biotech, Inc. Sustained response predictors after treatment with anti-il23 specific antibody
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
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
WO2020065532A1 (en) 2018-09-24 2020-04-02 Janssen Biotech, Inc. Safe and effective method of treating ulcerative colitis with anti-il12/il23 antibody
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
WO2020092210A1 (en) 2018-10-30 2020-05-07 Genmab A/S Methods of treating cancer with a combination of an anti-vegf antibody and an anti-tissue factor antibody-drug conjugate
WO2020102501A1 (en) 2018-11-16 2020-05-22 Bristol-Myers Squibb Company Anti-nkg2a antibodies and uses thereof
US11548941B2 (en) 2018-11-20 2023-01-10 Janssen Biotech, Inc. Safe and effective method of treating psoriasis with anti-IL-23 specific antibody
WO2020104943A2 (en) 2018-11-20 2020-05-28 Janssen Biotech, Inc. Safe and effective method of treating psoriasis with anti-il-23 specific antibody
WO2020128864A1 (en) 2018-12-18 2020-06-25 Janssen Biotech, Inc. Safe and effective method of treating lupus with anti-il12/il23 antibody
WO2020148651A1 (en) 2019-01-15 2020-07-23 Janssen Biotech, Inc. Anti-tnf antibody compositions and methods for the treatment of juvenile idiopathic arthritis
WO2020154293A1 (en) 2019-01-22 2020-07-30 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
US11008395B2 (en) 2019-01-22 2021-05-18 Bristol Myers-Squibb Company Antibodies against IL-7R alpha subunit and uses thereof
WO2020152544A1 (en) 2019-01-23 2020-07-30 Janssen Biotech, Inc. Anti-tnf antibody compositions for use in methods for the treatment of psoriatic arthritis
WO2020153467A1 (en) 2019-01-24 2020-07-30 中外製薬株式会社 Novel cancer antigens and antibodies of said antigens
WO2020162452A1 (en) 2019-02-04 2020-08-13 国立大学法人愛媛大学 Car library and production method for scfv
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
WO2020183269A1 (en) 2019-03-14 2020-09-17 Janssen Biotech, Inc. Manufacturing methods for producing anti-tnf antibody compositions
WO2020183271A1 (en) 2019-03-14 2020-09-17 Janssen Biotech, Inc. Methods for producing anti-tnf antibody compositions
WO2020183270A1 (en) 2019-03-14 2020-09-17 Janssen Biotech, Inc. Methods for producing anti-tnf antibody compositions
WO2020183418A1 (en) 2019-03-14 2020-09-17 Janssen Biotech, Inc. Manufacturing methods for producing anti-il12/il23 antibody compositions
WO2020188466A1 (en) 2019-03-18 2020-09-24 Janssen Biotech, Inc. Method of treating psoriasis in pediatric subjects with anti-il12/il23 antibody
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
US11780911B2 (en) 2019-05-23 2023-10-10 Janssen Biotech, Inc. Method of treating inflammatory bowel disease with a combination therapy of antibodies to IL-23 and TNF alpha
WO2020245677A1 (en) 2019-06-03 2020-12-10 Janssen Biotech, Inc. Anti-tnf antibodies, compositions, and methods for the treatment of active ankylosing spondylitis
WO2020245676A1 (en) 2019-06-03 2020-12-10 Janssen Biotech, Inc. Anti-tnf antibody compositions, and methods for the treatment of psoriatic arthritis
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
WO2021028752A1 (en) 2019-08-15 2021-02-18 Janssen Biotech, Inc. Anti-tfn antibodies for treating type i diabetes
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
WO2021091815A1 (en) 2019-11-04 2021-05-14 Seagen Inc. Anti-cd30 antibody-drug conjugates and their use for the treatment of hiv infection
WO2021090272A1 (en) 2019-11-07 2021-05-14 Genmab A/S Methods of treating cancer with a combination of an anti-pd-1 antibody and an anti-tissue factor antibody-drug conjugate
WO2021089794A1 (en) 2019-11-07 2021-05-14 Genmab A/S Methods of treating cancer with a combination of a platinum-based agent and an anti-tissue factor antibody-drug conjugate
WO2021097344A1 (en) 2019-11-13 2021-05-20 Amgen Inc. Method for reduced aggregate formation in downstream processing of bispecific antigen-binding molecules
WO2021127528A1 (en) 2019-12-20 2021-06-24 Amgen Inc. Mesothelin-targeted cd40 agonistic multispecific antibody constructs for the treatment of solid tumors
WO2021126435A1 (en) 2019-12-20 2021-06-24 Novarock Biotherapeutics, Ltd. Anti-interleukin-23 p19 antibodies and methods of use thereof
WO2021132673A1 (en) 2019-12-26 2021-07-01 国立大学法人大阪大学 Agent for preventing or treating acute-phase neuromyelitis optica
WO2021130383A1 (en) 2019-12-27 2021-07-01 Affimed Gmbh Method for the production of bispecific fcyriii x cd30 antibody construct
WO2021145432A1 (en) 2020-01-15 2021-07-22 国立大学法人大阪大学 Agent for prevention or treatment of diabetic autonomic neuropathy
WO2021145435A1 (en) 2020-01-15 2021-07-22 国立大学法人大阪大学 Prophylactic or therapeutic agent for dementia
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
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
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
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
WO2021261546A1 (en) 2020-06-24 2021-12-30 国立大学法人 東京大学 Photosensitizing dye
WO2022002940A1 (en) 2020-06-29 2022-01-06 Genmab A/S Anti-tissue factor antibody-drug conjugates and their use in the treatment of cancer
WO2022014703A1 (en) 2020-07-17 2022-01-20 田辺三菱製薬株式会社 Agent for preventing or treating muscular disease
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
WO2022051591A2 (en) 2020-09-04 2022-03-10 Novarock Biotherapeutics, Ltd. Nectin-4 antibodies and uses thereof
WO2022074206A1 (en) 2020-10-08 2022-04-14 Affimed Gmbh Trispecific binders
WO2022096700A1 (en) 2020-11-06 2022-05-12 Amgen Research (Munich) Gmbh Polypeptide constructs selectively binding to cldn6 and cd3
WO2022097065A2 (en) 2020-11-06 2022-05-12 Novartis Ag ANTIBODY Fc VARIANTS
WO2022096704A1 (en) 2020-11-06 2022-05-12 Amgen Inc. Antigen binding domain with reduced clipping rate
WO2022096698A1 (en) 2020-11-06 2022-05-12 Amgen Inc. Polypeptide constructs binding to cd3
WO2022096716A2 (en) 2020-11-06 2022-05-12 Amgen Inc. Multitargeting bispecific antigen-binding molecules of increased selectivity
WO2022108931A2 (en) 2020-11-17 2022-05-27 Seagen Inc. Methods of treating cancer with a combination of tucatinib and an anti-pd-1/anti-pd-l1 antibody
WO2022130182A1 (en) 2020-12-14 2022-06-23 Novartis Ag Reversal binding agents for anti-natriuretic peptide receptor 1 (npr1) antibodies and uses thereof
WO2022176970A1 (en) 2021-02-18 2022-08-25 田辺三菱製薬株式会社 Novel anti-pad4 antibody
WO2022190034A1 (en) 2021-03-12 2022-09-15 Janssen Biotech, Inc. Method of treating psoriatic arthritis patients with inadequate response to tnf therapy with anti-il23 specific antibody
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
WO2022190033A1 (en) 2021-03-12 2022-09-15 Janssen Biotech, Inc. Safe and effective method of treating psoriatic arthritis with anti-il23 specific antibody
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
WO2023281463A1 (en) 2021-07-09 2023-01-12 Janssen Biotech, Inc. Manufacturing methods for producing anti-tnf antibody compositions
WO2023281462A1 (en) 2021-07-09 2023-01-12 Janssen Biotech, Inc. Manufacturing methods for producing anti-tnf antibody compositions
WO2023281466A1 (en) 2021-07-09 2023-01-12 Janssen Biotech, Inc. Manufacturing methods for producing anti-il12/il23 antibody compositions
WO2023007023A1 (en) 2021-07-30 2023-02-02 Affimed Gmbh Duplexbodies
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
WO2023076989A1 (en) 2021-10-29 2023-05-04 Seagen Inc. Methods of treating cancer with a combination of an anti-pd-1 antibody and an anti-cd30 antibody-drug conjugate
WO2023073615A1 (en) 2021-10-29 2023-05-04 Janssen Biotech, Inc. Methods of treating crohn's disease with anti-il23 specific antibody
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
WO2023084488A1 (en) 2021-11-15 2023-05-19 Janssen Biotech, Inc. Methods of treating crohn's disease with anti-il23 specific antibody
WO2023095000A1 (en) 2021-11-23 2023-06-01 Janssen Biotech, Inc. Method of treating ulcerative colitis with anti-il23 specific antibody
WO2023187707A1 (en) 2022-03-30 2023-10-05 Janssen Biotech, Inc. Method of treating mild to moderate psoriasis with il-23 specific antibody
WO2023209568A1 (en) 2022-04-26 2023-11-02 Novartis Ag Multispecific antibodies targeting il-13 and il-18
WO2023213960A1 (en) 2022-05-06 2023-11-09 Genmab A/S Methods of treating cancer with anti-tissue factor antibody-drug conjugates
WO2023218027A1 (en) 2022-05-12 2023-11-16 Amgen Research (Munich) Gmbh Multichain multitargeting bispecific antigen-binding molecules of increased selectivity
WO2023223265A1 (en) 2022-05-18 2023-11-23 Janssen Biotech, Inc. Method for evaluating and treating psoriatic arthritis with il23 antibody
WO2024013723A1 (en) 2022-07-15 2024-01-18 Pheon Therapeutics Ltd Antibody drug conjugates that bind cdcp1 and uses thereof
WO2024059675A2 (en) 2022-09-14 2024-03-21 Amgen Inc. Bispecific molecule stabilizing composition

Also Published As

Publication number Publication date
EP0754225A4 (en) 2001-01-31
JP2012143252A (en) 2012-08-02
JP2007054076A (en) 2007-03-08
CA2161351A1 (en) 1994-11-10
JPH08509612A (en) 1996-10-15
EP0754225A1 (en) 1997-01-22
JP5099405B2 (en) 2012-12-19
JP5550026B2 (en) 2014-07-16
CA2161351C (en) 2010-12-21
AU6819494A (en) 1994-11-21

Similar Documents

Publication Publication Date Title
US5770429A (en) Transgenic non-human animals capable of producing heterologous antibodies
US5625126A (en) Transgenic non-human animals for producing heterologous antibodies
CA2161351C (en) Transgenic non-human animals capable of producing heterologous antibodies
US5814318A (en) Transgenic non-human animals for producing heterologous antibodies
US7084260B1 (en) High affinity human antibodies and human antibodies against human antigens
US8293480B2 (en) Transgenic non-human animals for producing chimeric antibodies
AU747370B2 (en) Transgenic non-human animals capable of producing heterologous antibo dies
US5661016A (en) Transgenic non-human animals capable of producing heterologous antibodies of various isotypes
US5789650A (en) Transgenic non-human animals for producing heterologous antibodies
US5545806A (en) Ransgenic non-human animals for producing heterologous antibodies
WO1997013852A9 (en) Transgenic non-human animals capable of producing heterologous antibodies
WO1998024884A9 (en) Transgenic non-human animals capable of producing heterologous antibodies
AU3328493A (en) Transgenic non-human animals capable of producing heterologous antibodies
WO1999045962A1 (en) Transgenic non-human animals capable of producing heterologous antibodies
EP1288229A2 (en) Transgenic non human animals capable of producing heterologous antibodies
AU2003204055B2 (en) Transgenic non-human animals capable of producing heterologous antibodies
AU720612B2 (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 BY CA CH CN CZ DE DK ES FI GB HU JP KP KR KZ LK LU LV MG MN MW NL NO NZ PL PT RO RU SD SE SI SK TT UA US UZ VN

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 NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
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: 2161351

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 1994916581

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: 1994916581

Country of ref document: EP

WWW Wipo information: withdrawn in national office

Ref document number: 1994916581

Country of ref document: EP