US20120192300A1 - Common Light Chain Mouse - Google Patents

Common Light Chain Mouse Download PDF

Info

Publication number
US20120192300A1
US20120192300A1 US13/412,936 US201213412936A US2012192300A1 US 20120192300 A1 US20120192300 A1 US 20120192300A1 US 201213412936 A US201213412936 A US 201213412936A US 2012192300 A1 US2012192300 A1 US 2012192300A1
Authority
US
United States
Prior art keywords
human
mouse
light chain
region
sequence
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US13/412,936
Inventor
Robert Babb
John McWhirter
Lynn MacDonald
Sean Stevens
Samuel Davis
David R. Buckler
Karolina A. Hosiawa
Andrew J. Murphy
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Regeneron Pharmaceuticals Inc
Original Assignee
Regeneron Pharmaceuticals 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
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=43663643&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20120192300(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority claimed from US13/093,156 external-priority patent/US20120021409A1/en
Priority to US13/412,936 priority Critical patent/US20120192300A1/en
Application filed by Regeneron Pharmaceuticals Inc filed Critical Regeneron Pharmaceuticals Inc
Assigned to REGENERON PHARMACEUTICALS, INC. reassignment REGENERON PHARMACEUTICALS, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: BUCKLER, DAVID R., MURPHY, ANDREW J., BABB, ROBERT, DAVIS, SAMUEL, HOSIAWA, KAROLINA A., MACDONALD, LYNN, MCWHIRTER, JOHN, STEVENS, SEAN
Priority to US13/488,628 priority patent/US20130045492A1/en
Publication of US20120192300A1 publication Critical patent/US20120192300A1/en
Priority to EP13712618.1A priority patent/EP2822379A1/en
Priority to SG10201610294RA priority patent/SG10201610294RA/en
Priority to CN201380012671.3A priority patent/CN104244709B/en
Priority to PCT/US2013/029125 priority patent/WO2013134263A1/en
Priority to SG11201405088QA priority patent/SG11201405088QA/en
Priority to EP16203839.2A priority patent/EP3165086A1/en
Priority to RU2014140239A priority patent/RU2683514C2/en
Priority to NZ630637A priority patent/NZ630637A/en
Priority to CA2865029A priority patent/CA2865029A1/en
Priority to AU2013204140A priority patent/AU2013204140B2/en
Priority to JP2014561037A priority patent/JP2015509380A/en
Priority to KR20147026624A priority patent/KR20140136462A/en
Priority to MX2014010794A priority patent/MX353278B/en
Priority to IN8163DEN2014 priority patent/IN2014DN08163A/en
Priority to CN201611201918.8A priority patent/CN107090471A/en
Priority to US13/798,310 priority patent/US20130185821A1/en
Priority to US13/798,455 priority patent/US9796788B2/en
Priority to IL234207A priority patent/IL234207A0/en
Priority to US14/473,970 priority patent/US9969814B2/en
Priority to HK15106625.3A priority patent/HK1205873A1/en
Priority to US15/056,713 priority patent/US20160219847A1/en
Priority to AU2016202488A priority patent/AU2016202488C1/en
Priority to US15/700,973 priority patent/US10167344B2/en
Priority to US15/891,987 priority patent/US20190021295A1/en
Priority to US15/951,130 priority patent/US20190071519A1/en
Priority to JP2018088350A priority patent/JP2018138047A/en
Priority to US16/128,360 priority patent/US10412940B2/en
Priority to US16/159,496 priority patent/US20190090462A1/en
Priority to US16/530,030 priority patent/US11026407B2/en
Priority to US17/238,710 priority patent/US20210315189A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/46Hybrid immunoglobulins
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K67/00Rearing or breeding animals, not otherwise provided for; New breeds of animals
    • A01K67/027New breeds of vertebrates
    • A01K67/0275Genetically modified vertebrates, e.g. transgenic
    • A01K67/0276Knockout animals
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K67/00Rearing or breeding animals, not otherwise provided for; New breeds of animals
    • A01K67/027New breeds of vertebrates
    • A01K67/0275Genetically modified vertebrates, e.g. transgenic
    • A01K67/0278Humanized animals, e.g. knockin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/18Growth factors; Growth regulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/475Growth factors; Growth regulators
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/54Interleukins [IL]
    • 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
    • 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
    • C07K16/22Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against growth factors ; against growth regulators
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/24Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against cytokines, lymphokines or interferons
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/46Hybrid immunoglobulins
    • C07K16/461Igs containing Ig-regions, -domains or -residues form different species
    • C07K16/462Igs containing a variable region (Fv) from one specie and a constant region (Fc) from another
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/8509Vectors or expression systems specially adapted for eukaryotic hosts for animal cells for producing genetically modified animals, e.g. transgenic
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0603Embryonic cells ; Embryoid bodies
    • C12N5/0606Pluripotent embryonic cells, e.g. embryonic stem cells [ES]
    • 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
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/10Cells modified by introduction of foreign genetic material
    • 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
    • A01K2217/07Animals genetically altered by homologous recombination
    • A01K2217/072Animals genetically altered by homologous recombination maintaining or altering function, i.e. knock in
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • A01K2217/15Animals comprising multiple alterations of the genome, by transgenesis or homologous recombination, e.g. obtained by cross-breeding
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/21Immunoglobulins specific features characterized by taxonomic origin from primates, e.g. man
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/515Complete light chain, i.e. VL + CL
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/565Complementarity determining region [CDR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/567Framework region [FR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/30Non-immunoglobulin-derived peptide or protein having an immunoglobulin constant or Fc region, or a fragment thereof, attached thereto
    • 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
    • C12N2800/00Nucleic acids vectors
    • C12N2800/20Pseudochromosomes, minichrosomosomes
    • C12N2800/204Pseudochromosomes, minichrosomosomes of bacterial origin, e.g. BAC

Definitions

  • a genetically modified mouse that expresses antibodies having a common human variable/mouse constant light chain associated with diverse human variable/mouse constant heavy chains.
  • a method for making a human bispecific antibody from human variable region gene sequences of B cells of the mouse is provided.
  • Antibodies typically comprise a homodimeric heavy chain component, wherein each heavy chain monomer is associated with an identical light chain.
  • Antibodies having a heterodimeric heavy chain component e.g., bispecific antibodies
  • bispecific antibodies are desirable as therapeutic antibodies. But making bispecific antibodies having a suitable light chain component that can satisfactorily associate with each of the heavy chains of a bispecific antibody has proved problematic.
  • a light chain might be selected by surveying usage statistics for all light chain variable domains, identifying the most frequently employed light chain in human antibodies, and pairing that light chain in vitro with the two heavy chains of differing specificity.
  • a light chain might be selected by observing light chain sequences in a phage display library (e.g., a phage display library comprising human light chain variable region sequences, e.g., a human scFv library) and selecting the most commonly used light chain variable region from the library. The light chain can then be tested on the two different heavy chains of interest.
  • a phage display library e.g., a phage display library comprising human light chain variable region sequences, e.g., a human scFv library
  • a light chain might be selected by assaying a phage display library of light chain variable sequences using the heavy chain variable sequences of both heavy chains of interest as probes.
  • a light chain that associates with both heavy chain variable sequences might be selected as a light chain for the heavy chains.
  • a candidate light chain might be aligned with the heavy chains' cognate light chains, and modifications are made in the light chain to more closely match sequence characteristics common to the cognate light chains of both heavy chains. If the chances of immunogenicity need to be minimized, the modifications preferably result in sequences that are present in known human light chain sequences, such that proteolytic processing is unlikely to generate a T cell epitope based on parameters and methods known in the art for assessing the likelihood of immunogenicity (i.e., in silico as well as wet assays).
  • mice that express human immunoglobulin heavy and light chain variable domains, wherein the mice have a limited light chain variable repertoire.
  • a biological system for generating a human light chain variable domain that associates and expresses with a diverse repertoire of affinity-matured human heavy chain variable domains is provided.
  • Methods for making binding proteins comprising immunoglobulin variable domains are provided, comprising immunizing mice that have a limited immunoglobulin light chain repertoire with an antigen of interest, and employing an immunoglobulin variable region gene sequence of the mouse in a binding protein that specifically binds the antigen of interest.
  • Methods include methods for making human immunoglobulin heavy chain variable domains suitable for use in making multi-specific antigen-binding proteins.
  • mice that select suitable affinity-matured human immunoglobulin heavy chain variable domains derived from a repertoire of unrearranged human heavy chain variable region gene segments, wherein the affinity-matured human heavy chain variable domains associate and express with a single human light chain variable domain derived from one human light chain variable region gene segment. Genetically engineered mice that present a choice of two human light chain variable region gene segments are also provided.
  • mice that express a limited repertoire of human light chain variable domains, or a single human light chain variable domain, from a limited repertoire of human light chain variable region gene segments.
  • the mice are genetically engineered to include a single unrearranged human light chain variable region gene segment (or two human light chain variable region gene segments) that rearranges to form a rearranged human light chain variable region gene (or two rearranged light chain variable region genes) that express a single light chain (or that express either or both of two light chains).
  • the rearranged human light chain variable domains are capable of pairing with a plurality of affinity-matured human heavy chains selected by the mice, wherein the heavy chain variable regions specifically bind different epitopes.
  • mice that express a limited repertoire of human light chain variable domains, or a single human light chain variable domain, from a limited repertoire of human light chain variable region sequences.
  • the mice are genetically engineered to include a single V/J human light chain sequence (or two V/J sequences) that express a variable region of a single light chain (or that express either or both of two variable regions).
  • a light chain comprising the variable sequence is capable of pairing with a plurality of affinity-matured human heavy chains clonally selected by the mice, wherein the heavy chain variable regions specifically bind different epitopes.
  • a genetically modified mouse comprises a single human immunoglobulin light chain variable (V L ) region gene segment that is capable of rearranging with a human J gene segment (selected from one or a plurality of J L segments) and encoding a human V L domain of an immunoglobulin light chain.
  • the mouse comprises no more than two human V L gene segments, each of which is capable of rearranging with a human J gene segment (selected from one or a plurality of J L segments) and encoding a human V L domain of an immunoglobulin light chain.
  • the single human V L gene segment is operably linked to a human J L gene segment selected from J ⁇ 1, J ⁇ 2, J ⁇ 3, J ⁇ 4, and J ⁇ 5, wherein the single human V L gene segment is capable of rearranging to form a sequence encoding a light chain variable region gene with any of the one or more human J L gene segments.
  • the genetically modified mouse comprises an immunoglobulin light chain locus that does not comprise an endogenous mouse V L gene segment that is capable of rearranging to form an immunoglobulin light chain gene, wherein the V L locus comprises a single human V L gene segment that is capable of rearranging to encode a V L region of a light chain gene.
  • the human V L gene segment is a human V ⁇ 1-39J ⁇ 5 gene segment or a human V ⁇ 3-20J ⁇ 1 gene segment.
  • the genetically modified mouse comprises a V L locus that does not comprise an endogenous mouse V L gene segment that is capable of rearranging to form an immunoglobulin light chain gene, wherein the V L locus comprises no more than two human V L gene segments that are capable of rearranging to encode a V L region of a light chain gene.
  • the no more than 2 human V L gene segments are a human V ⁇ 1-39J ⁇ 5 gene segment and a human V ⁇ 3-20J ⁇ 1 gene segment.
  • a genetically modified mouse comprising a single rearranged (V/J) human immunoglobulin light chain variable (V L ) region (i.e., a V L /J L region) that encodes a human V L domain of an immunoglobulin light chain.
  • V L human immunoglobulin light chain variable
  • the mouse comprises no more than two rearranged human V L regions that are capable of encoding a human V L domain of an immunoglobulin light chain.
  • the V L region is a rearranged human V ⁇ 1-39/J sequence or a rearranged human V ⁇ 3-20/J sequence.
  • the human J L segment of the rearranged V L /J L sequence is selected from J ⁇ 1, J ⁇ 2, J ⁇ 3, J ⁇ 4, and J ⁇ 5.
  • the V L region is a human V ⁇ 1-39J ⁇ 5 sequence or a human V ⁇ 3-20J ⁇ K1 sequence.
  • the mouse has both a human V ⁇ 1-39J ⁇ 5 sequence and a human V ⁇ 3-20J ⁇ 1 sequence.
  • the human V L gene segment is operably linked to a human or mouse leader sequence.
  • the leader sequence is a mouse leader sequence.
  • the mouse leader sequence is a mouse V ⁇ 3-7 leader sequence.
  • the leader sequence is operably linked to an unrearranged human V L gene segment.
  • the leader sequence is operably linked to a rearranged human V L /J L sequence.
  • the V L gene segment is operably linked to an immunoglobulin promoter sequence.
  • the promoter sequence is a human promoter sequence.
  • the human immunoglobulin promoter is a human V ⁇ 3-15 promoter.
  • the promoter is operably linked to an unrearranged human V L gene segment.
  • the promoter is operably linked to a rearranged human V L /J L sequence.
  • the light chain locus comprises a leader sequence flanked 5′ (with respect to transcriptional direction of a V L gene segment) with a human immunoglobulin promoter and flanked 3′ with a human V L gene segment that rearranges with a human J segment and encodes a V L domain of a reverse chimeric light chain comprising an endogenous mouse light chain constant region (C L ).
  • the V L gene segment is at the mouse V ⁇ locus, and the mouse C L is a mouse C ⁇ .
  • the light chain locus comprises a leader sequence flanked 5′ (with respect to transcriptional direction of a V L gene segment) with a human immunoglobulin promoter and flanked 3′ with a rearranged human V L region (V L /J L sequence) and encodes a V L domain of a reverse chimeric light chain comprising an endogenous mouse light chain constant region (C L ).
  • V L /J L sequence is at the mouse kappa ( ⁇ ) locus
  • the mouse C L is a mouse C ⁇ .
  • the V L locus of the modified mouse is a ⁇ light chain locus
  • the ⁇ light chain locus comprises a mouse ⁇ intronic enhancer, a mouse ⁇ 3′ enhancer, or both an intronic enhancer and a 3′ enhancer.
  • the mouse comprises a nonfunctional immunoglobulin lambda ( ⁇ ) light chain locus.
  • the ⁇ light chain locus comprises a deletion of one or more sequences of the locus, wherein the one or more deletions renders the ⁇ light chain locus incapable of rearranging to form a light chain gene.
  • all or substantially all of the V L gene segments of the ⁇ light chain locus are deleted.
  • mouse makes a light chain that comprises a somatically mutated V L domain derived from a human V L gene segment.
  • the light chain comprises a somatically mutated V L domain derived from a human V L gene segment, and a mouse C ⁇ region.
  • the mouse does not express a ⁇ light chain.
  • the genetically modified mouse is capable of somatically hypermutating the human V L region sequence.
  • the mouse comprises a cell that comprises a rearranged immunoglobulin light chain gene derived from a human V L gene segment that is capable of rearranging and encoding a V L domain, and the rearranged immunoglobulin light chain gene comprises a somatically mutated V L domain.
  • the mouse comprises a cell that expresses a light chain comprising a somatically mutated human V L domain linked to a mouse CK, wherein the light chain associates with a heavy chain comprising a somatically mutated V H domain derived from a human V H gene segment and wherein the heavy chain comprises a mouse heavy chain constant region (C H ).
  • the heavy chain comprises a mouse C H 1, a mouse hinge, a mouse C H 2, and a mouse C H 3.
  • the heavy chain comprises a human C H 1, a hinge, a mouse C H 2, and a mouse C H 3.
  • the mouse comprises a replacement of endogenous mouse V H gene segments with one or more human V H gene segments, wherein the human V H gene segments are operably linked to a mouse C H region gene, such that the mouse rearranges the human V H gene segments and expresses a reverse chimeric immunoglobulin heavy chain that comprises a human V H domain and a mouse C H .
  • 90-100% of unrearranged mouse V H gene segments are replaced with at least one unrearranged human V H gene segment.
  • all or substantially all of the endogenous mouse V H gene segments are replaced with at least one unrearranged human V H gene segment.
  • the replacement is with at least 19, at least 39, or at least 80 or 81 unrearranged human V H gene segments.
  • the replacement is with at least 12 functional unrearranged human V H gene segments, at least 25 functional unrearranged human V H gene segments, or at least 43 functional unrearranged human V H gene segments.
  • the mouse comprises a replacement of all mouse D H and J H segments with at least one unrearranged human D H segment and at least one unrearranged human J H segment.
  • the at least one unrearranged human D H segment is selected from 1-1, D1-7, 1-26, 2-8, 2-15, 3-3, 3-10, 3-16, 3-22, 5-5, 5-12, 6-6, 6-13, 7-27, and a combination thereof.
  • the at least one unrearranged human J H segment is selected from 1, 2, 3, 4, 5, 6, and a combination thereof.
  • the one or more human V H gene segment is selected from a 1-2, 1-8, 1-24, 1-69, 2-5, 3-7, 3-9, 3-11, 3-13, 3-15, 3-20, 3-23, 3-30, 3-33, 3-48, 3-53, 4-31, 4-39, 4-59, 5-51, a 6-1 human V H gene segment, and a combination thereof.
  • the mouse comprises a B cell that expresses a binding protein that specifically binds an antigen of interest, wherein the binding protein comprises a light chain derived from a human V ⁇ 1-39/J ⁇ 5 rearrangement or a human V ⁇ 3-20/J ⁇ 1 rearrangement, and wherein the cell comprises a rearranged immunoglobulin heavy chain gene derived from a rearrangement of human V H gene segments selected from a 1-69, 2-5, 3-13, 3-23, 3-30, 3-33, 3-53, 4-39, 4-59, and 5-51 gene segment.
  • the one or more human V H gene segments are rearranged with a human heavy chain J H gene segment selected from 1, 2, 3, 4, 5, and 6.
  • the one or more human V H and J H gene segments are rearranged with a human D H gene segment selected from 1-1, 1-7, 1-26, 2-8, 2-15, 3-3, 3-10, 3-16, 3-22, 5-5, 5-12, 6-6, 6-13, and 7-27.
  • the light chain gene has 1, 2, 3, 4, or 5 or more somatic hypermutations.
  • the mouse comprises a B cell that comprises a rearranged immunoglobulin heavy chain variable region gene sequence comprising a V H /D H /J H region selected from 2-5/6-6/1, 2-5/3-22/1, 3-13/6-6/5, 3-23/2-8/4, 3-23/3-3/4, 3-23/3-10/4, 3-23/6-6/4, 3-23/7-27/4, 3-30/1-1/4, 3-30/1-7/4, 3-30/3-3/3, 3-30/3-3/4, 3-30/3-22/5, 3-30/5-5/2, 3-30/5-12/4, 3-30/6-6/1, 3-30/6-6/3, 3-30/6-6/4, 3-30/6-6/5, 3-30/6-13/4, 3-30/7-27/4, 3-30/7-27/5, 3-30/7-27/6, 3-33/1-7/4, 3-33/2-15/4, 4-39/1-26/3, 4-59/3-16/3, 4-59/3-16/4, 4-59/3-22/3, 5-51/3-16/6, 5-51/5-5/3, 5-51/6-13/5, 3-53/1-1/4, 1-69/6-6/5, and
  • the rearranged human V L region is a human V ⁇ 1-39J ⁇ 5 sequence
  • the mouse expresses a reverse chimeric light chain comprising (i) a V L domain derived from the human V L /J L sequence and (ii) a mouse C L ; wherein the light chain is associated with a reverse chimeric heavy chain comprising (i) a mouse C H and (ii) a somatically mutated human V H domain derived from a human V H gene segment selected from a 1-2, 1-8, 1-24, 1-69, 2-5, 3-7, 3-9, 3-11, 3-13, 3-15, 3-20, 3-23, 3-30, 3-33, 3-48, 3-53, 4-31, 4-39, 4-59, 5-51, a 6-1 human V H gene segment, and a combination thereof.
  • the mouse expresses a light chain that is somatically mutated.
  • the C L is a mouse C ⁇ .
  • the human V H gene segment is selected from a 2-5, 3-13, 3-23, 3-30, 4-59, 5-51, and 1-69 gene segment.
  • the somatically mutated human V H domain comprises a sequence derived from a D H segment selected from 1-1, 1-7, 2-8, 3-3, 3-10, 3-16, 3-22, 5-5, 5-12, 6-6, 6-13, and 7-27.
  • the somatically mutated human V H domain comprises a sequence derived from a J H segment selected from 1, 2, 3, 4, 5, and 6.
  • the somatically mutated human V H domain is encoded by a rearranged human V H /D H /J H sequence selected from 2-5/6-6/1, 2-5/3-22/1, 3-13/6-6/5, 3-23/2-8/4, 3-23/3-3/4, 3-23/3-10/4, 3-23/6-6/4, 3-23/7-27/4, 3-30/1-1/4, 3-30/1-7/4, 3-30/3-3/4, 3-30/3-22/5, 3-30/5-5/2, 3-30/5-12/4, 3-30/6-6/1, 3-30/6-6/3, 3-30/6-6/4, 3-30/6-6/5, 3-30/6-13/4, 3-30/7-27/4, 3-30/7-27/5, 3-30/7-27/6, 4-59/3-16/3, 4-59/3-16/4, 4-59/3-22/3, 5-51/5-5/3, 1-69/6-6/5, and 1-69/6-13/4.
  • the rearranged human V L region is a human V ⁇ 3-20J ⁇ 1 sequence
  • the mouse expresses a reverse chimeric light chain comprising (i) a V L domain derived from the rearranged human V L /J L sequence, and (ii) a mouse C L ; wherein the light chain is associated with a reverse chimeric heavy chain comprising (i) a mouse C H , and (ii) a somatically mutated human V H derived from a human V H gene segment selected from a 1-2, 1-8, 1-24, 1-69, 2-5, 3-7, 3-9, 3-11, 3-13, 3-15, 3-20, 3-23, 3-30, 3-33, 3-48, 3-53, 4-31, 4-39, 4-59, 5-51, a 6-1 human V H gene segment, and a combination thereof.
  • the mouse expresses a light chain that is somatically mutated.
  • the C L is a mouse C ⁇ .
  • the human V H gene segment is selected from a 3-30, 3-33, 3-53, 4-39, and 5-51 gene segment.
  • the somatically mutated human V H domain comprises a sequence derived from a D H segment selected from 1-1, 1-7, 1-26, 2-15, 3-3, 3-16, and 6-13.
  • the somatically mutated human V H domain comprises a sequence derived from a J H segment selected from 3, 4, 5, and 6.
  • the somatically mutated human V H domain is encoded by a rearranged human V H /D H /J H sequence selected from 3-30/1-1/4, 3-30/3-3/3, 3-33/1-7/4, 3-33/2-15/4, 4-39/1-26/3, 5-51/3-16/6, 5-51/6-13/5, and 3-53/1-1/4.
  • the mouse comprises both a rearranged human V ⁇ 1-39J ⁇ 5 sequence and a rearranged human V ⁇ 3-20J ⁇ 1 sequence, and the mouse expresses a reverse chimeric light chain comprising (i) a V L domain derived from the human V ⁇ 1-39J ⁇ 5 sequence or the human V ⁇ 3-20J ⁇ 1 sequence, and (ii) a mouse C L ; wherein the light chain is associated with a reverse chimeric heavy chain comprising (i) a mouse C H , and (ii) a somatically mutated human V H derived from a human V H gene segment selected from a 1-2, 1-8, 1-24, 1-69, 2-5, 3-7, 3-9, 3-11, 3-13, 3-15, 3-20, 3-23, 3-30, 3-33, 3-48, 3-53, 4-31, 4-39, 4-59, 5-51, a 6-1 human V H gene segment, and a combination thereof.
  • the mouse expresses a light chain that is somatically mutated.
  • 90-100% of the endogenous unrearranged mouse V H gene segments are replaced with at least one unrearranged human V H gene segment.
  • all or substantially all of the endogenous unrearranged mouse V H gene segments are replaced with at least one unrearranged human V H gene segment.
  • the replacement is with at least 18, at least 39, at least 80, or 81 unrearranged human V H gene segments.
  • the replacement is with at least 12 functional unrearranged human V H gene segments, at least 25 functional unrearranged human V H gene segments, or at least 43 unrearranged human V H gene segments.
  • the genetically modified mouse is a C57BL strain, in a specific embodiment selected from C57BL/A, C57BL/An, C57BL/GrFa, C57BL/KaLwN, C57BL/6, C57BL/6J, C57BL/6ByJ, C57BL/6NJ, C57BL/10, C57BL/10ScSn, C57BL/10Cr, and C57BL/Ola.
  • the genetically modified mouse is a mix of an aforementioned 129 strain and an aforementioned C57BL/6 strain.
  • the mouse is a mix of aforementioned 129 strains, or a mix of aforementioned BL/6 strains.
  • the 129 strain of the mix is a 129S6 (129/SvEvTac) strain.
  • the mouse expresses a reverse chimeric antibody comprising a light chain that comprises a mouse C ⁇ and a somatically mutated human V L domain derived from a rearranged human V ⁇ 1-39J ⁇ 5 sequence or a rearranged human V ⁇ 3-20J ⁇ 1 sequence, and a heavy chain that comprises a mouse C H and a somatically mutated human V H domain derived from a human V H gene segment selected from a 1-2, 1-8, 1-24, 1-69, 2-5, 3-7, 3-9, 3-11, 3-13, 3-15, 3-20, 3-23, 3-30, 3-33, 3-48, 3-53, 4-31, 4-39, 4-59, 5-51, and a 6-1 human V H gene segment, wherein the mouse does not express a fully mouse antibody and does not express a fully human antibody.
  • the mouse comprises a ⁇ light chain locus that comprises a replacement of endogenous mouse ⁇ light chain gene segments with the rearranged human V ⁇ 1-39J ⁇ 5 sequence or the rearranged human V ⁇ 3-20J ⁇ 1 sequence, and comprises a replacement of all or substantially all endogenous mouse V H gene segments with a complete or substantially complete repertoire of human V H gene segments.
  • a population of antigen-specific antibodies derived from a mouse as described herein wherein the antibodies comprise a light chain gene derived from a human V ⁇ 1-39/J ⁇ 5 rearrangement or a human V ⁇ 3-20/J ⁇ 1 rearrangement, and wherein the antibodies comprise a rearranged immunoglobulin heavy chain gene derived from a rearrangement of a human V H gene segment selected from a 1-2, 1-3, 1-8, 1-18, 1-24, 1-46, 1-58, 1-69, 2-5, 2-26, 2-70, 3-7, 3-9, 3-11, 3-13, 3-15, 3-16, 3-20, 3-21, 3-23, 3-30, 3-33, 3-43, 3-48, 3-53, 3-64, 3-72, 3-73, 4-31, 4-34, 4-39, 4-59, 5-51, and a 6-1 human V H gene segment.
  • the one or more human V H gene segments are rearranged with a human heavy chain J H gene segment selected from 1, 2, 3, 4, 5, and 6.
  • the light chain has 1, 2, 3, or 4 somatic hypermutations.
  • the light chain gene has 1 or 2 mutations.
  • the light chain gene is capable of incurring multiple mutations along its sequence.
  • the light chain is derived from a human V ⁇ 1-39/J ⁇ 5 rearrangement and the light chain has at least one or no more than four somatic hypermutations. In one embodiment, the light chain comprises at least two somatic hypermutations. In one embodiment, the light chain comprises at least three somatic hypermutations. In one embodiment, the light chain comprises at least four somatic hypermutations. In a specific embodiment, the mutations are present in one or more framework regions (FWs) of the light chain. In a specific embodiment, the mutations are present in one or more complementarity determining regions (CDRs) of the light chain. In a specific embodiment, the mutations are present in one or more FWs and/or one or more CDRs of the light chain.
  • FWs framework regions
  • CDRs complementarity determining regions
  • the framework regions are selected from framework 1 (FW1), framework 2 (FW2), framework 3 (FW3), and/or a combination thereof.
  • the CDRs are selected from CDR1, CDR2, CDR3, and/or a combination thereof.
  • the heavy chain comprises at least one mutation in one or more FWs or one or more CDRs. In one embodiment, the heavy chain comprises at least one mutation in one or more FWs and one or more CDRs. In one embodiment, the heavy chain comprises at least two mutations in one or more FWs and one or more CDRs. In one embodiment, the heavy chain comprises at least three mutations in one or more FWs and one or more CDRs. In one embodiment, the heavy chain comprises at least four mutations in one or more FWs and one or more CDRs.
  • the heavy chain comprises at least five or more than five mutations in one or more FWs and one or more CDRs; in a specific embodiment, the heavy chain comprises at least five or more than five mutations in two FWs; in a specific embodiment, the heavy chain comprises at least five or more than five mutations in one FW and one CDR.
  • the light chain is derived from a human V ⁇ 1-39/J ⁇ 5 rearrangement and about 9% of the V ⁇ 1-39/J ⁇ 5-derived light chains have at least one mutation present in FW1; in one embodiment, at least 9% of the light chains comprise one mutation present in FW1.
  • the light chain is derived from a human V ⁇ 1-39/J ⁇ 5 rearrangement and about 25% of the V ⁇ 1-39/J ⁇ 5-derived light chains have at least one or no more than two mutations present in CDR1; in one embodiment, at least 19% of the light chains have one mutation present in CDR1; in one embodiment, at least 5% of the light chains have two mutations present in CDR1.
  • the light chain is derived from a human V ⁇ 1-39/J ⁇ 5 rearrangement and about 20% of the V ⁇ 1-39/J ⁇ 5-derived light chains have at least one or no more than three mutations present in FW2; in one embodiment, at least 17% of the light chains have one mutation present in FW2; in one embodiment, at least 1% of the light chains have two mutations present in FW2; in one embodiment, at least 1% of the light chains have three mutations present in FW2.
  • the light chain is derived from a human V ⁇ 1-39/J ⁇ 5 rearrangement and about 10% of the V ⁇ 1-39/J ⁇ 5-derived light chains have at least one or no more than two mutations present in CDR2; in one embodiment, at least 10% of the light chains have one mutation present in CDR2; in one embodiment, at least 1% of the light chains have two mutations present in CDR2.
  • the light chain is derived from a human V ⁇ 1-39/J ⁇ 5 rearrangement and about 29% of the V ⁇ 1-39/J ⁇ 5-derived light chains have at least one or no more than four mutations present in FW3; in one embodiment, at least 21% of the light chains have one mutation present in FW3; in one embodiment, at least 5% of the light chains have two mutations present in FW3; in one embodiment, at least 2% of the light chains have three mutations present in FW3; in one embodiment, at least 2% of the light chains have four mutations present in FW3.
  • the light chain is derived from a human V ⁇ 1-39/J ⁇ 5 rearrangement and about 37% of the V ⁇ 1-39/J ⁇ 5-derived light chains have at least one or no more than four mutations present in CDR3; in one embodiment, at least 27% of the light chains have one mutation present in CDR3; in one embodiment, at least 8% of the light chains have two mutations present in CDR3; in one embodiment, at least 1% of the light chains have three mutations present in CDR3; in one embodiment, at least 1% of the light chains have four mutations present in CDR3.
  • a population of antigen-specific antibodies derived from a mouse as described herein wherein the antibodies comprise a light chain derived from a human V ⁇ 1-39/J ⁇ 5 rearrangement and about 9% of the V ⁇ 1-39/J ⁇ 5-derived light chains have one or more mutations present in FW1, about 25% of the V ⁇ 1-39/J ⁇ 5-derived light chains have one or more mutations present in CDR1, about 20% of the V ⁇ 1-39/J ⁇ 5-derived light chains have one or more mutations present in FW2, about 10% of the V ⁇ 1-39/J ⁇ 5-derived light chains have one or more mutations present in CDR2, about 29% of the V ⁇ 1-39/J ⁇ 5-derived light chains have one or more mutations present in FW3, and about 37% of the V ⁇ 1-39/J ⁇ 5-derived light chains have one or more mutations present in CDR3.
  • the light chain is derived from a human V ⁇ 1-39/J ⁇ 5 rearrangement and about 35% of the heavy chains have at least one mutation present in FW1; in one embodiment, at least 25% of the heavy chains have one mutation present in FW1; in one embodiment, at least 9% of the heavy chains have two mutations present in FW1; in one embodiment, at least 1% of the heavy chains have three mutations present in FW1; in one embodiment, at least 1% of the heavy chains have more than five mutations present in FW1.
  • the light chain is derived from a human V ⁇ 1-39J ⁇ 5 rearrangement and about 92% of the heavy chains have at least one or no more than four mutations present in CDR1; in one embodiment, at least 92% of the heavy chains have at least one, at least two, at least three, or at least four mutations present in CDR1; in one embodiment, at least 26% of the heavy chains have one mutation present in CDR1; in one embodiment, at least 44% of the heavy chains have two mutations present in CDR1; in one embodiment, at least 19% of the heavy chains have three mutations present in CDR1; in one embodiment, at least 3% of the heavy chains have four mutations present in CDR1.
  • the light chain is derived from a human V ⁇ 1-39/J ⁇ 5 rearrangement and about 66% of the heavy chains have at least one or no more than three mutations present in FW2; in one embodiment, at least 66% of the heavy chains have at least one, at least two, or at least three mutations present in FW2; in one embodiment, at least 35% of the heavy chains have one mutation present in FW2; in one embodiment, at least 23% of the heavy chains have two mutations present in FW2; in one embodiment, at least 8% of the heavy chains have three mutations present in FW2.
  • the light chain is derived from a human V ⁇ 1-39/J ⁇ 5 rearrangement and about 70% of the heavy chains have at least one or no more than four mutations present in CDR2; in one embodiment, at least 70% of the heavy chains have at least two, at least three, or at least four mutations present in CDR2; in one embodiment, at least 34% have one mutation present in CDR2; in one embodiment, at least 20% of the heavy chains have two mutations present in CDR2; in one embodiment, at least 12% of the heavy chains have three mutations present in CDR2; in one embodiment, at least 5% of the heavy chains have four mutations present in CDR2.
  • the light chain is derived from a human V ⁇ 1-39/J ⁇ 5 rearrangement and about 91% of the heavy chains have at least one or up to five or more mutations present in FW3; in one embodiment, at least 91% of the heavy chains have at least two, at least three, at least four, or at least five or more mutations present in FW3; in one embodiment, at least 19% of the heavy chains have one mutation present in FW3; in one embodiment, at least 33% of the heavy chains have two mutations present in FW3; in one embodiment, at least 22% of the heavy chains have three mutations present in FW3; in one embodiment, at least 11% of the heavy chains have four mutations present in FW3; in one embodiment, at least 7% of the heavy chains have five or more mutations present in FW3.
  • the light chain is derived from a human V ⁇ 1-39/J ⁇ 5 rearrangement and about 63% of the heavy chains have at least one or no more than two mutations present in CDR3; in one embodiment, at least 63% of the heavy chains have at one mutation present in CDR3; in one embodiment, at least 54% of the heavy chains have one mutation present in CDR3; in one embodiment, at least 9% of the heavy chains have two mutations present in CDR3.
  • a population of antigen-specific antibodies derived from a mouse as described herein wherein the antibodies comprise a light chain derived from a human V ⁇ 1-39/J ⁇ 5 rearrangement and at least 35% of the heavy chains have one or more mutations present in FW1, about 92% of the heavy chains have one or more mutations present in CDR1, about 66% of the heavy chains have one or more mutations present in FW2, about 70% of the heavy chains have one or more mutations present in CDR2, about 91% of the heavy chains have one or more mutations present in FW3, and about 63% of the heavy chains have one or more mutations present in CDR3.
  • the light chain is derived from a human V ⁇ 3-20/J ⁇ 1 rearrangement and the light chain gene has at least one or no more than two somatic hypermutations; in one embodiment, the light chain gene has at least two, at least three, at least four or more somatic hypermutations.
  • the mutations are present in one or more framework regions of the light chain. In a specific embodiment, the mutations are present in one or more CDR regions of the light chain. In a specific embodiment, the mutations are present in one or more framework regions and/or one or more CDR regions of the light chain.
  • the framework regions are selected from framework 1 (FW1), framework 2 (FW2), framework 3 (FW3), and/or a combination thereof.
  • the light chain is derived from a human V ⁇ 3-20/J ⁇ 1 rearrangement and about 10% of the V ⁇ 3-20/J ⁇ 1 -derived light chains have at least one mutation present in FW1; in one embodiment, at least 10% of the light chains have one mutation in FW1.
  • the light chain is derived from a human V ⁇ 3-20/J ⁇ 1 rearrangement and about 53% of the V ⁇ 3-20/J ⁇ 1 -derived light chains have at least one or no more than two mutations present in CDR1; in one embodiment, at least 27% of the light chains have one or more mutations in CDR1; in one embodiment, about 54% of the light chains have one or two mutations present in CDR1.
  • the light chain is derived from a human V ⁇ 3-20/J ⁇ 1 rearrangement and about 6% of the V ⁇ 3-20/J ⁇ 1-derived light chains have at least one or no more than two mutations present in FW2; in one embodiment, at least 6% of light chains have at least one mutation present in FW2; in one embodiment, at least 3% of the light chains have one mutation present in FW2; in one embodiment, at least 3% of the light chains have two mutations present in FW2.
  • the light chain is derived from a human V ⁇ 3-20/J ⁇ 1 rearrangement and at least about 3% of the V ⁇ 3-20/J ⁇ 1 -derived light chains have at least one mutation present in CDR2; in one embodiment, at least 3% of the light chains have one mutation in CDR2.
  • the light chain is derived from a human V ⁇ 3-20/J ⁇ 1 rearrangement and about 17% or more of the V ⁇ 3-20/J ⁇ 1-derived light chains have at least one or no more than two mutations present in FW3; in one embodiment, at least 20% of the light chain have one mutation present in FW3; in one embodiment, at least 17% of the light chains have two mutations present in FW3.
  • the light chain is derived from a human V ⁇ 3-20/J ⁇ 1 rearrangement and at least 43% of the V ⁇ 3-20/J ⁇ 1-derived light chains have at least one mutation present in CDR3; in one embodiment, at least 43% of the light chains have one mutation in CDR3.
  • a population of antigen-specific antibodies derived from a mouse as described herein wherein the antibodies comprise a light chain derived from a human V ⁇ 3-20/J ⁇ 1 rearrangement and about 10% of the V ⁇ 3-20/J ⁇ 1-derived light chains have one or more mutations present in at least, about 53% of the V ⁇ 3-20/J ⁇ 1-derived light chains have one or more mutations present in CDR1, about 6% of the V ⁇ 3-20/J ⁇ 1-derived light chains have one or more mutations present in FW2, about 3% of the V ⁇ 3-20/J ⁇ 1-derived light chains have one or more mutations present in CDR2, about 37% of the V ⁇ 3-20/J ⁇ 1 -derived light chains have one or more mutations present in FW3, and about 43% of the V ⁇ 3-20/J ⁇ 1-derived light chains have one or more mutations present in CDR3.
  • the light chain is derived from a human V ⁇ 3-20/J ⁇ 1 rearrangement and about 43% of the heavy chains have at least one or no more than two mutations present in FW1; in one embodiment, at least 41% of the heavy chains have at least one mutation present in FW1; in one embodiment, about 41% of the heavy chains have one mutation present in FW1; in one embodiment, about 2% of the heavy chains have two mutations present in FW1.
  • the light chain is derived from a human V ⁇ 3-20/ ⁇ 1 rearrangement and about 92% of the heavy chains have at least one or no more than four mutations present in CDR1; in one embodiment, at least 43% of heavy chains have at least one mutation present in CDR1; in one embodiment, at least 25% of heavy chains have at least two mutations present in CDR1; in one embodiment, at least 15% of heavy chains have at least 3 mutations present in CDR1; in one embodiment, at least 10% of heavy chains have 4 or more mutations present in CDR1.
  • the light chain is derived from a human V ⁇ 3-20/J ⁇ 1 rearrangement and about 46% of the heavy chains have at least one or no more than three mutations present in FW2; in one embodiment, at least 34% of heavy chains have at least one mutation present in FW2; in one embodiment, at least 10% of heavy chains have two or more mutations present in FW2; in one embodiment, at least 2% of heavy chains have three or more mutations present in FW2.
  • the light chain is derived from a human V ⁇ 3-20/J ⁇ 1 rearrangement and about 84% of the heavy chains have at least one or up to five or more than five mutations present in CDR2; in one embodiment, at least 39% of the heavy chains have one or more mutations present in CDR2; in one embodiment, at least 18% of the heavy chains have two or more mutations present in CDR2; in one embodiment, at least 21% of the heavy chains have three or more mutations present in CDR2; in one embodiment, at least 3% of the heavy chains have four or more mutations present in CDR2; in one embodiment, at least 2% of the heavy chains have five or more mutations present in CDR2.
  • the light chain is derived from a human V ⁇ 3-20/J ⁇ 1 rearrangement and about 92% of the heavy chains have at least one or up to five or more than five mutations present in FW3; in one embodiment, at least 21% of the light chains have at least one mutation present in FW3; in one embodiment, at least 20% of heavy chains have at least two mutations present in FW3; in one embodiment, at least 13% of the heavy chains have at least three mutations present in FW3; in one embodiment, at least 20% of the heavy chains have at least four mutations in FW3; in one embodiment, at least 18% of the heavy chains have at least 5 mutations in FW3.
  • the light chain is derived from a human V ⁇ 3-20/J ⁇ 1 rearrangement and about 7% of the heavy chains have at least one mutation present in CDR3; in one embodiment, about 7% of the heavy chains have one mutation in CDR3.
  • a population of antigen-specific antibodies derived from a mouse as described herein wherein the antibodies comprise a light chain derived from a human V ⁇ 3-20/J ⁇ 1 rearrangement and about 43% of the heavy chains have one or more mutations present in FW1, about 92% of the heavy chains have one or more mutations present in CDR1, about 46% of the heavy chains have one or more mutations present in FW2, about 84% of the heavy chains have one or more mutations present in CDR2, about 92% of the heavy chains have one or more mutations present in FW3, and about 7% of the heavy chains have one or more mutations present in CDR3.
  • a mouse that expresses an immunoglobulin light chain from a rearranged immunoglobulin light chain sequence is provided, wherein the rearranged immunoglobulin light chain sequence is present in the germline of the mouse, wherein the immunoglobulin light chain comprises a human variable sequence.
  • the germline of the mouse comprises a rearranged immunoglobulin light chain sequence that is derived from the same V segment and the same J segment as all non-surrogate light chain sequences present in every B cell of the mouse that comprises a rearranged light chain sequence.
  • the germline of the mouse lacks a functional unrearranged immunoglobulin light chain V gene segment. In one embodiment, the germline of the mouse lacks a functional unrearranged immunoglobulin light chain J gene segment.
  • the germline of the mouse comprises no more than one, no more than two, or no more than three rearranged (V/J) light chain sequences.
  • the rearranged V/J sequence comprises a ⁇ light chain sequence.
  • the ⁇ light chain sequence is a human ⁇ light chain sequence.
  • the ⁇ light chain sequence is selected from a human V ⁇ 1-39/J sequence, a human V ⁇ 3-20/J sequence, and a combination thereof.
  • the ⁇ light chain sequence is a human V ⁇ 1-39/J ⁇ 5 sequence.
  • the ⁇ light chain sequence is a human V ⁇ 3-20/J ⁇ 1 sequence.
  • the mouse further comprises in its germline a sequence selected from a mouse ⁇ intronic enhancer 5′ with respect to the rearranged immunoglobulin light chain sequence, a mouse ⁇ 3′ enhancer, and a combination thereof.
  • the mouse comprises an unrearranged human V H gene segment, an unrearranged human D H gene segment, and an unrearranged human J H gene segment, wherein said V H , D H , and J H gene segments are capable of rearranging to form an immunoglobulin heavy chain variable gene sequence operably linked to a heavy chain constant gene sequence.
  • the mouse comprises a plurality of human V H , D H , and J H gene segments.
  • the human V H , D H , and J H gene segments replace endogenous mouse V H , D H , and J H gene segments at the endogenous mouse immunoglobulin heavy chain locus.
  • the mouse comprises a replacement of all or substantially all functional mouse V H , D H , and J H gene segments with all or substantially all functional human V H , D H , and J H gene segments.
  • the mouse expresses an immunoglobulin light chain that comprises a mouse constant sequence. In one embodiment, the mouse expresses an immunoglobulin light chain that comprises a human constant sequence.
  • the mouse expresses an immunoglobulin heavy chain that comprises a mouse sequence selected from a C H 1 sequence, a hinge sequence, a C H 2 sequence, a C H 3 sequence, and a combination thereof.
  • the mouse expresses an immunoglobulin heavy chain that comprises a human sequence selected from a C H 1 sequence, a hinge sequence, a C H 2 sequence, a C H 3 sequence, and a combination thereof.
  • the rearranged immunoglobulin light chain sequence in the germline of the mouse is at an endogenous mouse immunoglobulin light chain locus.
  • the rearranged immunoglobulin light chain sequence in the germline of the mouse replaces all or substantially all mouse light chain V and J sequences at the endogenous mouse immunoglobulin light chain locus.
  • a mouse comprising a B cell population characterized by each B cell that comprises a non-surrogate light chain sequence comprises a rearranged light chain gene that is derived from a single human V segment and a single human J segment, wherein the only light chain variable sequence in the germline of the mouse is a rearranged sequence derived from the single human V segment and the single human J segment, and wherein each B ell that comprises the rearranged light chain gene further comprises a gene encoding a cognate human heavy chain variable domain, and wherein the rearranged light chain gene comprises at least one, at least two, at least three, or at least four somatic hypermutations.
  • a pluripotent, induced pluripotent, or totipotent cell derived from a mouse as described herein is provided.
  • the cell is a mouse embryonic stem (ES) cell.
  • tissue derived from a mouse as described herein is provided.
  • the tissue is derived from spleen, lymph node or bone marrow of a mouse as described herein.
  • nucleus derived from a mouse as described herein is provided.
  • the nucleus is from a diploid cell that is not a B cell.
  • a mouse cell that is isolated from a mouse as described herein.
  • the cell is an ES cell.
  • the cell is a lymphocyte.
  • the lymphocyte is a B cell.
  • the B cell expresses a chimeric heavy chain comprising a variable domain derived from a human gene segment; and a light chain derived from a rearranged human V ⁇ 1-39/J sequence, rearranged human V ⁇ 3-20/J sequence, or a combination thereof; wherein the heavy chain variable domain is fused to a mouse constant region and the light chain variable domain is fused to a mouse or a human constant region.
  • a hybridoma wherein the hybridoma is made with a B cell of a mouse as described herein.
  • the B cell is from a mouse as described herein that has been immunized with an immunogen comprising an epitope of interest, and the B cell expresses a binding protein that binds the epitope of interest, the binding protein has a somatically mutated human V H domain and a mouse C H , and has a human V L domain derived from a rearranged human V ⁇ 1-39J ⁇ 5 or a rearranged human V ⁇ 3-20J ⁇ 1 and a mouse C L .
  • a mouse embryo comprising a donor ES cell that is derived from a mouse as described herein.
  • a targeting vector comprising, from 5′ to 3′ in transcriptional direction with reference to the sequences of the 5′ and 3′ mouse homology arms of the vector, a 5′ mouse homology arm, a human or mouse immunoglobulin promoter, a human or mouse leader sequence, and a human V L region selected from a rearranged human V ⁇ 1-39J ⁇ 5 or a rearranged human V ⁇ 3-20J ⁇ 1, and a 3′ mouse homology arm.
  • the 5′ and 3′ homology arms target the vector to a sequence 5′ with respect to an enhancer sequence that is present 5′ and proximal to the mouse C ⁇ gene.
  • the promoter is a human immunoglobulin variable region gene segment promoter.
  • the promoter is a human V ⁇ 3-15 promoter.
  • the leader sequence is a mouse leader sequence.
  • the mouse leader sequence is a mouse V ⁇ 3-7 leader sequence.
  • a targeting vector is provided as described above, but in place of the 5′ mouse homology arm the human or mouse promoter is flanked 5′ with a site-specific recombinase recognition site (SRRS), and in place of the 3′ mouse homology arm the human V L region is flanked 3′ with an SRRS.
  • SRRS site-specific recombinase recognition site
  • a reverse chimeric antibody made by a mouse as described herein, wherein the reverse chimeric antibody comprises a light chain comprising a human V L and a mouse C L , and a heavy chain comprising a human V H and a mouse C H .
  • a method for making an antibody comprising expressing in a single cell (a) a first V H gene sequence of an immunized mouse as described herein fused with a human C H gene sequence; (b) a V L gene sequence of an immunized mouse as described herein fused with a human C L gene sequence; and, (c) maintaining the cell under conditions sufficient to express a fully human antibody, and isolating the antibody.
  • the cell comprises a second V H gene sequence of a second immunized mouse as described herein fused with a human C H gene sequence, the first V H gene sequence encodes a V H domain that recognizes a first epitope, and the second V H gene sequence encodes a V H domain that recognizes a second epitope, wherein the first epitope and the second epitope are not identical.
  • a method for making an epitope-binding protein comprising exposing a mouse as described herein with an immunogen that comprises an epitope of interest, maintaining the mouse under conditions sufficient for the mouse to generate an immunoglobulin molecule that specifically binds the epitope of interest, and isolating the immunoglobulin molecule that specifically binds the epitope of interest; wherein the epitope-binding protein comprises a heavy chain that comprises a somatically mutated human V H and a mouse C H , associated with a light chain comprising a mouse C L and a human V L derived from a rearranged human V ⁇ 1-39J ⁇ 5 or a rearranged human V ⁇ 3-20J ⁇ 1.
  • a cell that expresses an epitope-binding protein comprises: (a) a human nucleotide sequence encoding a human V L domain that is derived from a rearranged human V ⁇ 1-39J ⁇ 5 or a rearranged human V ⁇ 3-20J ⁇ 1, wherein the human nucleotide sequence is fused (directly or through a linker) to a human immunoglobulin light chain constant domain cDNA sequence (e.g., a human ⁇ constant domain DNA sequence); and, (b) a first human V H nucleotide sequence encoding a human V H domain derived from a first human V H nucleotide sequence, wherein the first human V H nucleotide sequence is fused (directly or through a linker) to a human immunoglobulin heavy chain constant domain cDNA sequence; wherein the epitope-binding protein recognizes a first epitope.
  • the epitope-binding protein binds the first epitope with a dissociation constant of lower than 10 ⁇ 6 M, lower than 10 ⁇ 8 M lower than 10 ⁇ 9 M, lower than 10 ⁇ 10 M, lower than 10 ⁇ 11 M, or lower than 10 ⁇ 12 M.
  • the cell comprises a second human nucleotide sequence encoding a second human V H domain, wherein the second human sequence is fused (directly or through a linker) to a human immunoglobulin heavy chain constant domain cDNA sequence, and wherein the second human V H domain does not specifically recognize the first epitope (e.g., displays a dissociation constant of, e.g., 10 ⁇ 6 M, 10 ⁇ 5 M, 10 ⁇ 4 M, or higher), and wherein the epitope-binding protein recognizes the first epitope and the second epitope, and wherein the first and the second immunoglobulin heavy chains each associate with an identical light chain of (a).
  • the second human sequence is fused (directly or through a linker) to a human immunoglobulin heavy chain constant domain cDNA sequence
  • the second human V H domain does not specifically recognize the first epitope (e.g., displays a dissociation constant of, e.g., 10 ⁇ 6 M, 10 ⁇ 5 M
  • the second V H domain binds the second epitope with a dissociation constant that is lower than 10 ⁇ 6 M, lower than 10 ⁇ 7 M, lower than 10 ⁇ 8 M, lower than 10 ⁇ 9 M, lower than 10 ⁇ 10 M, lower than 10 ⁇ 11 M, or lower than 10 ⁇ 12 M.
  • the epitope-binding protein comprises a first immunoglobulin heavy chain and a second immunoglobulin heavy chain, each associated with an identical light chain derived from a rearranged human V L region selected from a human V ⁇ 1-39J ⁇ 5 or a human V ⁇ 3-20J ⁇ 1, wherein the first immunoglobulin heavy chain binds a first epitope with a dissociation constant in the nanomolar to picomolar range, the second immunoglobulin heavy chain binds a second epitope with a dissociation constant in the nanomolar to picomolar range, the first epitope and the second epitope are not identical, the first immunoglobulin heavy chain does not bind the second epitope or binds the second epitope with a dissociation constant weaker than the micromolar range (e.g., the millimolar range), the second immunoglobulin heavy chain does not bind the first epitope or binds the first epitope with a dissociation constant weaker than the micromolar range (e
  • the first immunoglobulin heavy chain comprises a protein A-binding residue
  • the second immunoglobulin heavy chain lacks the protein A-binding residue
  • the cell is selected from CHO, COS, 293, HeLa, and a retinal cell expressing a viral nucleic acid sequence (e.g., a PERC.6TM cell).
  • a reverse chimeric antibody comprising a human V H and a mouse heavy chain constant domain, a human V L and a mouse light chain constant domain, wherein the antibody is made by a process that comprises immunizing a mouse as described herein with an immunogen comprising an epitope, and the antibody specifically binds the epitope of the immunogen with which the mouse was immunized.
  • the V L domain is somatically mutated.
  • the V H domain is somatically mutated.
  • both the V L domain and the V H domain are somatically mutated.
  • the V L is linked to a mouse C ⁇ domain.
  • a mouse comprising human V H gene segments replacing all or substantially all mouse V H gene segments at the endogenous mouse heavy chain locus; no more than one or two rearranged human light chain V L /J L sequences selected from a rearranged V ⁇ 1-39/J and a rearranged V ⁇ 3-20/J or a combination thereof, replacing all mouse light chain gene segments; wherein the human heavy chain variable gene segments are linked to a mouse constant gene, and the rearranged human light chain sequences are linked to a human or mouse constant gene.
  • a mouse ES cell comprising a replacement of all or substantially all mouse heavy chain variable gene segments with human heavy chain variable gene segments, and no more than one or two rearranged human light chain V L /J L sequences, wherein the human heavy chain variable gene segments are linked to a mouse immunoglobulin heavy chain constant gene, and the rearranged human light chain V L /J L sequences are linked to a mouse or human immunoglobulin light chain constant gene.
  • the light chain constant gene is a mouse constant gene.
  • an antigen-binding protein made by a mouse as described herein comprises a human immunoglobulin heavy chain variable region fused with a mouse constant region, and a human immunoglobulin light chain variable region derived from a V ⁇ 1-39 gene segment or a V ⁇ 3-20 gene segment, wherein the light chain constant region is a mouse constant region.
  • a fully human antigen-binding protein made from an immunoglobulin variable region gene sequence from a mouse as described herein, wherein the antigen-binding protein comprises a fully human heavy chain comprising a human variable region derived from a sequence of a mouse as described herein, and a fully human light chain comprising a V ⁇ 1-39 or a V ⁇ 3-20.
  • the light chain variable region comprises one to five somatic mutations.
  • the light chain variable region is a cognate light chain variable region that is paired in a B cell of the mouse with the heavy chain variable region.
  • the fully human antigen-binding protein comprises a first heavy chain and a second heavy chain, wherein the first heavy chain and the second heavy chain comprise non-identical variable regions independently derived from a mouse as described herein, and wherein each of the first and second heavy chains express from a host cell associated with a human light chain derived from a V ⁇ 1-39 gene segment or a V ⁇ 3-20 gene segment.
  • the first heavy chain comprises a first heavy chain variable region that specifically binds a first epitope of a first antigen
  • the second heavy chain comprises a second heavy chain variable region that specifically binds a second epitope of a second antigen.
  • the first antigen and the second antigen are different.
  • first antigen and the second antigen are the same, and the first epitope and the second epitope are not identical; in a specific embodiment, binding of the first epitope by a first molecule of the binding protein does not block binding of the second epitope by a second molecule of the binding protein.
  • a fully human binding protein derived from a human immunoglobulin sequence of a mouse as described herein comprises a first immunoglobulin heavy chain and a second immunoglobulin heavy chain, wherein the first immunoglobulin heavy chain comprises a first variable region that is not identical to a variable region of the second immunoglobulin heavy chain, and wherein the first immunoglobulin heavy chain comprises a wild type protein A binding determinant, and the second heavy chain lacks a wild type protein A binding determinant.
  • the first immunoglobulin heavy chain binds protein A under isolation conditions
  • the second immunoglobulin heavy chain does not bind protein A or binds protein A at least 10-fold, a hundred-fold, or a thousand-fold weaker than the first immunoglobulin heavy chain binds protein A under isolation conditions.
  • the first and the second heavy chains are IgG1 isotypes, wherein the second heavy chain comprises a modification selected from 95R (EU 435R), 96F (EU 436F), and a combination thereof, and wherein the first heavy chain lacks such modification.
  • a method for making a bispecific antigen-binding protein comprising exposing a first mouse as described herein to a first antigen of interest that comprises a first epitope, exposing a second mouse as described herein to a second antigen of interest that comprises a second epitope, allowing the first and the second mouse to each mount immune responses to the antigens of interest, identifying in the first mouse a first human heavy chain variable region that binds the first epitope of the first antigen of interest, identifying in the second mouse a second human heavy chain variable region that binds the second epitope of the second antigen of interest, making a first fully human heavy chain gene that encodes a first heavy chain that binds the first epitope of the first antigen of interest, making a second fully human heavy chain gene that encodes a second heavy chain that binds the second epitope of the second antigen of interest, expressing the first heavy chain and the second heavy chain in a cell that expresses a single fully human light chain
  • the first antigen and the second antigen are not identical.
  • first antigen and the second antigen are identical, and the first epitope and the second epitope are not identical. In one embodiment, binding of the first heavy chain variable region to the first epitope does not block binding of the second heavy chain variable region to the second epitope.
  • the first antigen is selected from a soluble antigen and a cell surface antigen (e.g., a tumor antigen), and the second antigen comprises a cell surface receptor.
  • the cell surface receptor is an immunoglobulin receptor.
  • the immunoglobulin receptor is an Fc receptor.
  • the first antigen and the second antigen are the same cell surface receptor, and binding of the first heavy chain to the first epitope does not block binding of the second heavy chain to the second epitope.
  • the light chain variable domain of the light chain comprises 2 to 5 somatic mutations. In one embodiment, the light chain variable domain is a somatically mutated cognate light chain expressed in a B cell of the first or the second immunized mouse with either the first or the second heavy chain variable domain.
  • the first fully human heavy chain bears an amino acid modification that reduces its affinity to protein A, and he second fully human heavy chain does not comprise a modification that reduces its affinity to protein A.
  • an antibody or a bispecific antibody comprising a human heavy chain variable domain made in accordance with the invention is provided.
  • use of a mouse as described herein to make a fully human antibody or a fully human bispecific antibody is provided.
  • a genetically modified mouse, embryo, or cell described herein comprises a ⁇ light chain locus that retains endogenous regulatory or control elements, e.g., a mouse ⁇ intronic enhancer, a mouse ⁇ 3′ enhancer, or both an intronic enhancer and a 3′ enhancer, wherein the regulatory or control elements facilitate somatic mutation and affinity maturation of an expressed sequence of the ⁇ light chain locus.
  • endogenous regulatory or control elements e.g., a mouse ⁇ intronic enhancer, a mouse ⁇ 3′ enhancer, or both an intronic enhancer and a 3′ enhancer
  • a mouse in one aspect, comprises a B cell population characterized by having immunoglobulin light chains derived from no more than one, or no more than two, rearranged or unrearranged immunoglobulin light chain V and J gene segments, wherein the mouse exhibits a ⁇ : ⁇ light chain ratio that is about the same as a mouse that comprises a wild type complement of immunoglobulin light chain V and J gene segments.
  • the immunoglobulin light chains are derived from no more than one, or no more than two, rearranged immunoglobulin light chain V and J gene segments. In a specific embodiment, the light chains are derived from no more than one rearranged immunoglobulin light chain V and J gene segments.
  • a mouse as described herein expresses an immunoglobulin light chain derived from no more than one, or no more than two, human V ⁇ /J ⁇ sequences, wherein the mouse comprises a replacement of all or substantially all endogenous mouse heavy chain variable region gene segments with one or more human heavy chain variable region gene segments, and the mouse exhibits a ratio of (a) CD19 + B cells that express an immunoglobulin having a ⁇ light chain, to (b) CD19 + B cells that express an immunoglobulin having a ⁇ light chain, of about 1 to about 20.
  • the mouse expresses a single ⁇ light chain derived from a human V ⁇ 1-39J ⁇ 5 sequence, and the ratio of CD19 + B cells that express an immunoglobulin having a 2 light chain to CD19 + B cells that express an immunoglobulin having a ⁇ light chain is about 1 to about 20; in one embodiment, the ratio is about 1 to at least about 66; in a specific embodiment, the ratio is about 1 to 66.
  • the mouse expresses a single ⁇ light chain derived from a human V ⁇ 3-20J ⁇ 5 sequence, and the ratio of CD19 + B cells that express an immunoglobulin having a ⁇ light chain to CD19 + B cells that express an immunoglobulin having a ⁇ light chain is about 1 to about 20; in one embodiment, the ratio is about 1 to about 21. In specific embodiments, the ratio is 1 to 20, or 1 to 21.
  • a genetically modified mouse that expresses a single rearranged ⁇ light chain, wherein the mouse comprises a functional ⁇ light chain locus, and wherein the mouse expresses a B cell population that comprises Ig ⁇ + cells that express a ⁇ light chain derived from the same single rearranged ⁇ light chain.
  • the percent of Ig ⁇ + Ig ⁇ + B cells in the mouse is about the same as in a wild type mouse. In a specific embodiment, the percent of Ig ⁇ + Ig ⁇ + B cells in the mouse is about 2 to about 6 percent.
  • the percent of Ig ⁇ + Ig ⁇ + B cells in a mouse wherein the single rearranged ⁇ light chain is derived from a V ⁇ 1-39J ⁇ 5 sequence is about 2 to about 3; in a specific embodiment, the percent is about 2.6. In a specific embodiment, the percent of Ig ⁇ + Ig ⁇ + B cells in a mouse wherein the single rearranged ⁇ light chain is derived from a V ⁇ 3-20J ⁇ 1 sequence is about 4 to about 8; in a specific embodiment, about 6.
  • a genetically modified mouse wherein the mouse expresses a single rearranged ⁇ light chain derived from a human V ⁇ and J ⁇ gene segment, wherein the mouse expresses a B cell population that comprises a single ⁇ light chain derived from the single rearranged ⁇ light chain sequence, wherein the genetically modified mouse has not been rendered resistant to somatic hypermutations.
  • at least 90% of the ⁇ light chains expressed on a B cell of the mouse exhibit from at least one to about five somatic hypermutations.
  • a genetically modified mouse is provided that is modified to express a single ⁇ light chain derived from no more than one, or no more than two, rearranged ⁇ light chain sequences, wherein the mouse exhibits a ⁇ light chain usage that is about two-fold or more, at least about three-fold or more, or at least about four-fold or more greater than the ⁇ light chain usage exhibited by a wild type mouse, or greater than the ⁇ light chain usage exhibited by a mouse of the same strain that comprises a wild type repertoire of ⁇ light chain gene segments.
  • the mouse expresses the single ⁇ light chain from no more than one rearranged ⁇ light chain sequence.
  • the rearranged ⁇ light chain sequence is selected from a V ⁇ 1-39J ⁇ 5 and V ⁇ 3-20J ⁇ 1 sequence. In one embodiment, the rearranged ⁇ light chain sequence is a V ⁇ 1-39J ⁇ 5 sequence. In one embodiment, the rearranged ⁇ light chain sequence is a V ⁇ 3-20J ⁇ 1 sequence.
  • a genetically modified mouse that expresses a single ⁇ light chain derived from no more than one, or no more than two, rearranged ⁇ light chain sequences, wherein the mouse exhibits a ⁇ light chain usage that is about 100-fold or more, at least about 200-fold or more, at least about 300-fold or more, at least about 400-fold or more, at least about 500-fold or more, at least about 600-fold or more, at least about 700-fold or more, at least about 800-fold or more, at least about 900-fold or more, at least about 1000-fold or more greater than the same ⁇ light chain usage exhibited by a mouse bearing a complete or substantially complete human ⁇ light chain locus.
  • the mouse bearing a complete or substantially complete human ⁇ light chain locus lacks a functional unrearranged mouse ⁇ light chain sequence.
  • the mouse expresses the single ⁇ light chain from no more than one rearranged ⁇ light chain sequence.
  • the mouse comprises one copy of a rearranged ⁇ light chain sequence (e.g., a heterozygote).
  • the mouse comprises two copies of a rearranged ⁇ light chain sequence (e.g., a homozygote).
  • the rearranged ⁇ light chain sequence is selected from a V ⁇ 1-39J ⁇ 5 and V ⁇ 3-20J ⁇ 1 sequence.
  • the rearranged K light chain sequence is a V ⁇ 1-39J ⁇ 5 sequence.
  • the rearranged ⁇ light chain sequence is a V ⁇ 3-20J ⁇ 1 sequence.
  • a genetically modified mouse that expresses a single light chain derived from no more than one, or no more than two, rearranged light chain sequences, wherein the light chain in the genetically modified mouse exhibits a level of expression that is at least 10-fold to about 1,000-fold, 100-fold to about 1,000-fold, 200-fold to about 1,000-fold, 300-fold to about 1,000-fold, 400-fold to about 1,000-fold, 500-fold to about 1,000-fold, 600-fold to about 1,000-fold, 700-fold to about 1,000-fold, 800-fold to about 1,000-fold, or 900-fold to about 1,000-fold higher than expression of the same rearranged light chain exhibited by a mouse bearing a complete or substantially complete light chain locus.
  • the light chain comprises a human sequence.
  • the human sequence is a ⁇ sequence.
  • the human sequence is a ⁇ sequence.
  • the light chain is a fully human light chain.
  • the level of expression is characterized by quantitating mRNA of transcribed light chain sequence, and comparing it to transcribed light chain sequence of a mouse bearing a complete or substantially complete light chain locus.
  • a genetically modified mouse that expresses a single ⁇ light chain derived from no more than one, or no more than two, rearranged ⁇ light chain sequences, wherein the mouse, upon immunization with antigen, exhibits a serum titer that is comparable to a wild type mouse immunized with the same antigen.
  • the mouse expresses a single ⁇ light chain from no more than one rearranged ⁇ light chain sequence.
  • the serum titer is characterized as total immunoglobulin.
  • the serum titer is characterized as IgM specific titer.
  • the serum titer is characterized as IgG specific titer.
  • the rearranged ⁇ light chain sequence is selected from a V ⁇ 1-39J ⁇ 5 and V ⁇ 3-20J ⁇ 1 sequence. In one embodiment, the rearranged ⁇ light chain sequence is a V ⁇ 1-39J ⁇ 5 sequence. In one embodiment, the rearranged ⁇ light chain sequence is a V ⁇ 3-20J ⁇ 1 sequence.
  • a genetically modified mouse that expresses a population of antigen-specific antibodies, wherein all of the immunoglobulin light chains of the population of antigen-specific antibodies comprise a human light chain variable (V L ) region derived from the same single human V L gene segment and the immunoglobulin heavy chains comprise a human heavy chain variable (V H ) region derived from one of a plurality of human V H gene segments.
  • V L human light chain variable
  • V H human heavy chain variable
  • the human V H gene segments are selected from V H 1-2, V H 1-3, V H 1-8, V H 1-18, V H 1-24, V H 1-46, V H 1-58, V H 1-69, V H 2-5, V H 2-26, V H 2-70, V H 3-7, V H 3-9, V H 3-11, V H 3-13, V H 3-15, V H 3-20, V H 3-21, V H 3-23, V H 3-30, V H 3-33, V H 3-43, V H 3-48, V H 3-53, V H 3-64, V H 3-72, V H 3-73, V H 4-31, V H 4-34, V H 4-39, V H 4-59, V H 5-51, and V H 6-1.
  • same single human V L gene segment is selected from a human V ⁇ 1-39 gene segment and a human V ⁇ 3-20 gene segment.
  • all of the immunoglobulin light chains comprise a human light chain J (J L ) gene segment selected from a J ⁇ and a J ⁇ gene segment.
  • the human J L gene segment is selected from a human J ⁇ 1 and a J ⁇ 5 gene segment.
  • the mouse lacks a sequence selected from a mouse immunoglobulin V L gene segment, a mouse immunoglobulin J L gene segment, and a combination thereof.
  • the human V L region is operably linked to a human, mouse, or rat immunoglobulin light chain constant (C L ) region.
  • the human V L region is operably linked to a mouse C ⁇ region.
  • the human V L region is operably linked to a rat C ⁇ region.
  • the human V L region is expressed from an endogenous immunoglobulin light chain locus.
  • the human V H region is operably linked to a human, mouse, or rat immunoglobulin heavy chain constant (C H ) region.
  • the (C H ) region comprises a human sequence selected from a C H 1, a hinge, a C H 2, a C H 3, a C H 4, and/or a combination thereof.
  • the human V H region is expressed from an endogenous immunoglobulin heavy chain locus.
  • a genetically modified mouse that expresses a plurality of immunoglobulin heavy chains associated with a single light chain.
  • the heavy chain comprises a human sequence.
  • the human sequence is selected from a variable sequence, a C H 1, a hinge, a C H 2, a C H 3, and a combination thereof.
  • the single light chain comprises a human sequence.
  • the human sequence is selected from a variable sequence, a constant sequence, and a combination thereof.
  • the mouse comprises a disabled endogenous immunoglobulin locus and expresses the heavy chain and/or the light chain from a transgene or extrachromosomal episome.
  • the mouse comprises a replacement at an endogenous mouse locus of some or all endogenous mouse heavy chain gene segments (i.e., V, D, J), and/or some or all endogenous mouse heavy chain constant sequences (e.g., C H 1, hinge, C H 2, C H 3, or a combination thereof), and/or some or all endogenous mouse light chain sequences (e.g., V, J, constant, or a combination thereof), with one or more human immunoglobulin sequences.
  • endogenous mouse heavy chain gene segments i.e., V, D, J
  • endogenous mouse heavy chain constant sequences e.g., C H 1, hinge, C H 2, C H 3, or a combination thereof
  • endogenous mouse light chain sequences e.g., V, J, constant, or a combination thereof
  • a mouse suitable for making antibodies that have the same light chain wherein all or substantially all antibodies made in the mouse are expressed with the same light chain.
  • the light chain is expressed from an endogenous light chain locus.
  • a method for making a light chain for a human antibody comprising obtaining from a mouse as described herein a light chain sequence and a heavy chain sequence, and employing the light chain sequence and the heavy chain sequence in making a human antibody.
  • the human antibody is a bispecific antibody.
  • a method for identifying a human heavy chain variable domain that is capable of binding an antigen of interest with an engineered light chain as described herein comprises providing a heavy chain variable domain derived from a first antibody that is capable of binding the antigen, repairing the heavy chain variable domain with a germline light chain sequence and transfecting a cell so that each are expressed to form a second antibody, exposing the second antibody to the antigen, and measuring binding of the second antibody to the antigen.
  • the light chain of the first antibody comprises a human V ⁇ 1-39 sequence. In one embodiment, the light chain of the first antibody comprises a human V ⁇ 3-20 sequence. In one embodiment, the germline light chain sequence comprises a human V ⁇ 1-39 or V ⁇ 3-20 sequence. In various embodiments, binding of the second antibody to the antigen is determined by comparison of binding of the first antibody to the antigen.
  • FIG. 1 illustrates a targeting strategy for replacing endogenous mouse immunoglobulin light chain variable region gene segments with a human V ⁇ 1-39J ⁇ 5 gene region.
  • FIG. 2 illustrates a targeting strategy for replacing endogenous mouse immunoglobulin light chain variable region gene segments with a human V ⁇ 3-20J ⁇ 1 gene region.
  • FIG. 3 illustrates a targeting strategy for replacing endogenous mouse immunoglobulin light chain variable region gene segments with a human VpreB/J ⁇ 5 gene region.
  • FIG. 4 shows the percent of CD19 + B cells (y-axis) from peripheral blood for wild type mice (WT), mice homozygous for an engineered human rearranged V ⁇ 1-39J ⁇ 5 light chain region (V ⁇ 1-39J ⁇ 5 HO) and mice homozygous for an engineered human rearranged V ⁇ 3-20J ⁇ 1 light chain region (V ⁇ 3-20J ⁇ 1 HO).
  • FIG. 5A shows the relative mRNA expression (y-axis) of a V ⁇ 1-39-derived light chain in a quantitative PCR assay using probes specific for the junction of an engineered human rearranged V ⁇ 1-39J ⁇ 5 light chain region (V ⁇ 1-39J ⁇ 5 Junction Probe) and the human V ⁇ 1-39 gene segment (V ⁇ 1-39 Probe) in a mouse homozygous for a replacement of the endogenous V ⁇ and J ⁇ gene segments with human V ⁇ and J ⁇ gene segments (H ⁇ ), a wild type mouse (WT), and a mouse heterozygous for an engineered human rearranged V ⁇ 1-39J ⁇ 5 light chain region (V ⁇ 1-39J ⁇ 5 HET). Signals are normalized to expression of mouse CK. N.D.: not detected.
  • FIG. 5B shows the relative mRNA expression (y-axis) of a V ⁇ 1-39-derived light chain in a quantitative PCR assay using probes specific for the junction of an engineered human rearranged V ⁇ 1-39J ⁇ 5 light chain region (V ⁇ 1-39J ⁇ 5 Junction Probe) and the human V ⁇ 1-39 gene segment (V ⁇ 1-39 Probe) in a mouse homozygous for a replacement of the endogenous V ⁇ and J ⁇ gene segments with human V ⁇ and J ⁇ gene segments (H ⁇ ), a wild type mouse (WT), and a mouse homozygous for an engineered human rearranged V ⁇ 1-39J ⁇ 5 light chain region (V ⁇ 1-39J ⁇ 5 HO). Signals are normalized to expression of mouse C ⁇ .
  • FIG. 5C shows the relative mRNA expression (y-axis) of a V ⁇ 3-20-derived light chain in a quantitative PCR assay using probes specific for the junction of an engineered human rearranged V ⁇ 3-20J ⁇ 1 light chain region (V ⁇ 3-20J ⁇ 1 Junction Probe) and the human V ⁇ 3-20 gene segment (V ⁇ 3-20 Probe) in a mouse homozygous for a replacement of the endogenous V ⁇ and J ⁇ gene segments with human V ⁇ and J ⁇ gene segments (H ⁇ ), a wild type mouse (WT), and a mouse heterozygous (HET) and homozygous (HO) for an engineered human rearranged V ⁇ 3-20J ⁇ 1 light chain region.
  • Signals are normalized to expression of mouse CK.
  • antibody includes immunoglobulin molecules comprising four polypeptide chains, two heavy (H) chains and two light (L) chains inter-connected by disulfide bonds.
  • Each heavy chain comprises a heavy chain variable (V H ) region and a heavy chain constant region (C H ).
  • the heavy chain constant region comprises three domains, C H 1, C H 2 and C H 3.
  • Each light chain comprises a light chain variable (V L ) region and a light chain constant region (C L ).
  • the V H and V L regions can be further subdivided into regions of hypervariability, termed complementarity determining regions (CDR), interspersed with regions that are more conserved, termed framework regions (FR).
  • CDR complementarity determining regions
  • Each V H and V L comprises three CDRs and four FRs, arranged from amino-terminus to carboxy-terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4 (heavy chain CDRs may be abbreviated as HCDR1, HCDR2 and HCDR3; light chain CDRs may be abbreviated as LCDR1, LCDR2 and LCDR3.
  • the term “high affinity” antibody refers to an antibody that has a K D with respect to its target epitope about of 10 ⁇ 9 M or lower (e.g., about 1 ⁇ 10 ⁇ 9 M, 1 ⁇ 10 ⁇ 10 M, 1 ⁇ 10 ⁇ 11 M, or about 1 ⁇ 10 ⁇ 12 M).
  • K D is measured by surface plasmon resonance, e.g., BIACORETM; in another embodiment, K D is measured by ELISA.
  • bispecific antibody includes an antibody capable of selectively binding two or more epitopes.
  • Bispecific antibodies generally comprise two nonidentical heavy chains, with each heavy chain specifically binding a different epitope—either on two different molecules (e.g., different epitopes on two different immunogens) or on the same molecule (e.g., different epitopes on the same immunogen). If a bispecific antibody is capable of selectively binding two different epitopes (a first epitope and a second epitope), the affinity of the first heavy chain for the first epitope will generally be at least one to two or three or four or more orders of magnitude lower than the affinity of the first heavy chain for the second epitope, and vice versa.
  • Epitopes specifically bound by the bispecific antibody can be on the same or a different target (e.g., on the same or a different protein).
  • Bispecific antibodies can be made, for example, by combining heavy chains that recognize different epitopes of the same immunogen.
  • nucleic acid sequences encoding heavy chain variable sequences that recognize different epitopes of the same immunogen can be fused to nucleic acid sequences encoding the same or different heavy chain constant regions, and such sequences can be expressed in a cell that expresses an immunoglobulin light chain.
  • a typical bispecific antibody has two heavy chains each having three heavy chain CDRs, followed by (N-terminal to C-terminal) a C H 1 domain, a hinge, a C H 2 domain, and a C H 3 domain, and an immunoglobulin light chain that either does not confer epitope-binding specificity but that can associate with each heavy chain, or that can associate with each heavy chain and that can bind one or more of the epitopes bound by the heavy chain epitope-binding regions, or that can associate with each heavy chain and enable binding or one or both of the heavy chains to one or both epitopes.
  • cell includes any cell that is suitable for expressing a recombinant nucleic acid sequence.
  • Cells include those of prokaryotes and eukaryotes (single-cell or multiple-cell), bacterial cells (e.g., strains of E. coli, Bacillus spp., Streptomyces spp., etc.), mycobacteria cells, fungal cells, yeast cells (e.g., S. cerevisiae, S. pombe, P. pastoris, P.
  • the cell is a human, monkey, ape, hamster, rat, or mouse cell.
  • the cell is eukaryotic and is selected from the following cells: CHO (e.g., CHO K1, DXB-11 CHO, Veggie-CHO), COS (e.g., COS-7), retinal cell, Vero, CV1, kidney (e.g., HEK293, 293 EBNA, MSR 293, MDCK, HaK, BHK), HeLa, HepG2, WI38, MRC 5, Colo205, HB 8065, HL-60, (e.g., BHK21), Jurkat, Daudi, A431 (epidermal), CV-1, U937, 3T3, L cell, C127 cell, SP2/0, NS-0, MMT 060562, Sertoli cell, BRL 3A cell, HT1080 cell, myeloma cell, tumor cell, and a cell line derived from an aforementioned cell.
  • the cell comprises one or more viral genes, e.g., a retinal cell that expresses a viral
  • CDR complementarity determining region
  • a CDR includes an amino acid sequence encoded by a nucleic acid sequence of an organism's immunoglobulin genes that normally (i.e., in a wild type animal) appears between two framework regions in a variable region of a light or a heavy chain of an immunoglobulin molecule (e.g., an antibody or a T cell receptor).
  • a CDR can be encoded by, for example, a germline sequence or a rearranged or unrearranged sequence, and, for example, by a naive or a mature B cell or a T cell.
  • a CDR can be somatically mutated (e.g., vary from a sequence encoded in an animal's germline), humanized, and/or modified with amino acid substitutions, additions, or deletions.
  • CDRs can be encoded by two or more sequences (e.g., germline sequences) that are not contiguous (e.g., in an unrearranged nucleic acid sequence) but are contiguous in a B cell nucleic acid sequence, e.g., as the result of splicing or connecting the sequences (e.g., V-D-J recombination to form a heavy chain CDR3).
  • conservative amino acid substitution when used to describe a conservative amino acid substitution, includes substitution of an amino acid residue by another amino acid residue having a side chain R group with similar chemical properties (e.g., charge or hydrophobicity). In general, a conservative amino acid substitution will not substantially change the functional properties of interest of a protein, for example, the ability of a variable region to specifically bind a target epitope with a desired affinity.
  • groups of amino acids that have side chains with similar chemical properties include aliphatic side chains such as glycine, alanine, valine, leucine, and isoleucine; aliphatic-hydroxyl side chains such as serine and threonine; amide-containing side chains such as asparagine and glutamine; aromatic side chains such as phenylalanine, tyrosine, and tryptophan; basic side chains such as lysine, arginine, and histidine; acidic side chains such as aspartic acid and glutamic acid; and, sulfur-containing side chains such as cysteine and methionine.
  • aliphatic side chains such as glycine, alanine, valine, leucine, and isoleucine
  • aliphatic-hydroxyl side chains such as serine and threonine
  • amide-containing side chains such as asparagine and glutamine
  • aromatic side chains such as phenylalanine, tyrosine, and trypto
  • Conservative amino acids substitution groups include, for example, valine/leucine/isoleucine, phenylalanine/tyrosine, lysine/arginine, alanine/valine, glutamate/aspartate, and asparagine/glutamine.
  • a conservative amino acid substitution can be substitution of any native residue in a protein with alanine, as used in, for example, alanine scanning mutagenesis.
  • a conservative substitution is made that has a positive value in the PAM250 log-likelihood matrix disclosed in Gonnet et al. (1992) Exhaustive Matching of the Entire Protein Sequence Database, Science 256:1443-45, hereby incorporated by reference.
  • the substitution is a moderately conservative substitution wherein the substitution has a nonnegative value in the PAM250 log-likelihood matrix.
  • residue positions in an immunoglobulin light chain or heavy chain differ by one or more conservative amino acid substitutions.
  • residue positions in an immunoglobulin light chain or functional fragment thereof e.g., a fragment that allows expression and secretion from, e.g., a B cell
  • residue positions in an immunoglobulin light chain or functional fragment thereof are not identical to a light chain whose amino acid sequence is listed herein, but differs by one or more conservative amino acid substitutions.
  • epitope-binding protein includes a protein having at least one CDR and that is capable of selectively recognizing an epitope, e.g., is capable of binding an epitope with a K D that is at about one micromolar or lower (e.g., a K D that is about 1 ⁇ 10 ⁇ 6 M, 1 ⁇ 10 ⁇ 7 M, 1 ⁇ 10 ⁇ 9 M, 1 ⁇ 10 ⁇ 9 M, 1 ⁇ 10 ⁇ 10 M, 1 ⁇ 10 ⁇ 11 M, or about 1 ⁇ 10 ⁇ 12 M).
  • Therapeutic epitope-binding proteins e.g., therapeutic antibodies
  • the phrase “functional fragment” includes fragments of epitope-binding proteins that can be expressed, secreted, and specifically bind to an epitope with a K D in the micromolar, nanomolar, or picomolar range. Specific recognition includes having a K D that is at least in the micromolar range, the nanomolar range, or the picomolar range.
  • germline includes reference to an immunoglobulin nucleic acid sequence in a non-somatically mutated cell, e.g., a non-somatically mutated B cell or pre-B cell or hematopoietic cell.
  • heavy chain or “immunoglobulin heavy chain” includes an immunoglobulin heavy chain constant region sequence from any organism.
  • Heavy chain variable domains include three heavy chain CDRs and four FR regions, unless otherwise specified. Fragments of heavy chains include CDRs, CDRs and FRs, and combinations thereof.
  • a typical heavy chain has, following the variable domain (from N-terminal to C-terminal), a C H 1 domain, a hinge, a C H 2 domain, and a C H 3 domain.
  • a functional fragment of a heavy chain includes a fragment that is capable of specifically recognizing an epitope (e.g., recognizing the epitope with a K D in the micromolar, nanomolar, or picomolar range), that is capable of expressing and secreting from a cell, and that comprises at least one CDR.
  • an epitope e.g., recognizing the epitope with a K D in the micromolar, nanomolar, or picomolar range
  • identity when used in connection with sequence includes identity as determined by a number of different algorithms known in the art that can be used to measure nucleotide and/or amino acid sequence identity. In some embodiments described herein, identities are determined using a ClustalW v. 1.83 (slow) alignment employing an open gap penalty of 10.0, an extend gap penalty of 0.1, and using a Gonnet similarity matrix (MACVECTORTM 10.0.2, MacVector Inc., 2008).
  • ClustalW v. 1.83 (slow) alignment employing an open gap penalty of 10.0, an extend gap penalty of 0.1, and using a Gonnet similarity matrix (MACVECTORTM 10.0.2, MacVector Inc., 2008).
  • the length of the sequences compared with respect to identity of sequences will depend upon the particular sequences, but in the case of a light chain constant domain, the length should contain sequence of sufficient length to fold into a light chain constant domain that is capable of self-association to form a canonical light chain constant domain, e.g., capable of forming two beta sheets comprising beta strands and capable of interacting with at least one C H 1 domain of a human or a mouse. In the case of a C H 1 domain, the length of sequence should contain sequence of sufficient length to fold into a C H 1 domain that is capable of forming two beta sheets comprising beta strands and capable of interacting with at least one light chain constant domain of a mouse or a human.
  • immunoglobulin molecule includes two immunoglobulin heavy chains and two immunoglobulin light chains.
  • the heavy chains may be identical or different, and the light chains may be identical or different.
  • light chain includes an immunoglobulin light chain sequence from any organism, and unless otherwise specified includes human ⁇ and ⁇ . light chains and a VpreB, as well as surrogate light chains.
  • Light chain variable (V L ) domains typically include three light chain CDRs and four framework (FR) regions, unless otherwise specified.
  • FR framework
  • a full-length light chain includes, from amino terminus to carboxyl terminus, a V L domain that includes FR1-CDR1-FR2-CDR2-FR3-CDR3-FR4, and a light chain constant domain.
  • Light chains include those, e.g., that do not selectively bind either a first or a second epitope selectively bound by the epitope-binding protein in which they appear.
  • Light chains also include those that bind and recognize, or assist the heavy chain with binding and recognizing, one or more epitopes selectively bound by the epitope-binding protein in which they appear.
  • Common light chains are those derived from a rearranged human V ⁇ 1-39J ⁇ 5 sequence or a rearranged human V ⁇ 3-20J ⁇ 1 sequence, and include somatically mutated (e.g., affinity matured) versions.
  • micromolar range is intended to mean 1-999 micromolar; the phrase “nanomolar range” is intended to mean 1-999 nanomolar; the phrase “picomolar range” is intended to mean 1-999 picomolar.
  • the phrase “somatically mutated” includes reference to a nucleic acid sequence from a B cell that has undergone class-switching, wherein the nucleic acid sequence of an immunoglobulin variable region (e.g., a heavy chain variable domain or including a heavy chain CDR or FR sequence) in the class-switched B cell is not identical to the nucleic acid sequence in the B cell prior to class-switching, such as, for example, a difference in a CDR or framework nucleic acid sequence between a B cell that has not undergone class-switching and a B cell that has undergone class-switching.
  • an immunoglobulin variable region e.g., a heavy chain variable domain or including a heavy chain CDR or FR sequence
  • “Somatically mutated” includes reference to nucleic acid sequences from affinity-matured B cells that are not identical to corresponding immunoglobulin variable region sequences in B cells that are not affinity-matured (i.e., sequences in the genome of germline cells).
  • the phrase “somatically mutated” also includes reference to an immunoglobulin variable region nucleic acid sequence from a B cell after exposure of the B cell to an epitope of interest, wherein the nucleic acid sequence differs from the corresponding nucleic acid sequence prior to exposure of the B cell to the epitope of interest.
  • mutated refers to sequences from antibodies that have been generated in an animal, e.g., a mouse having human immunoglobulin variable region nucleic acid sequences, in response to an immunogen challenge, and that result from the selection processes inherently operative in such an animal.
  • nucleic acid sequences that exist in the germline of an animal cell.
  • variable domain includes an amino acid sequence of an immunoglobulin light or heavy chain (modified as desired) that comprises the following amino acid regions, in sequence from N-terminal to C-terminal (unless otherwise indicated): FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4.
  • mouse heavy chain immunoglobulin variable regions that pair with a common human light chain is of limited practical utility. More in vitro engineering efforts would be expended in a trial-and-error process to try to humanize the mouse heavy chain variable sequences while hoping to retain epitope specificity and affinity while maintaining the ability to couple with the common human light chain, with uncertain outcome. At the end of such a process, the final product may maintain some of the specificity and affinity, and associate with the common light chain, but ultimately immunogenicity in a human would likely remain a profound risk.
  • a suitable mouse for making human therapeutics would include a suitably large repertoire of human heavy chain variable region gene segments in place of endogenous mouse heavy chain variable region gene segments.
  • the human heavy chain variable region gene segments should be able to rearrange and recombine with an endogenous mouse heavy chain constant domain to form a reverse chimeric heavy chain (i.e., a heavy chain comprising a human variable domain and a mouse constant region).
  • the heavy chain should be capable of class switching and somatic hypermutation so that a suitably large repertoire of heavy chain variable domains are available for the mouse to select one that can associate with the limited repertoire of human light chain variable regions.
  • a mouse that selects a common light chain for a plurality of heavy chains has a practical utility.
  • antibodies that express in a mouse that can only express a common light chain will have heavy chains that can associate and express with an identical or substantially identical light chain. This is particularly useful in making bispecific antibodies.
  • a mouse can be immunized with a first immunogen to generate a B cell that expresses an antibody that specifically binds a first epitope.
  • the mouse (or a mouse genetically the same) can be immunized with a second immunogen to generate a B cell that expresses an antibody that specifically binds the second epitope.
  • Variable heavy regions can be cloned from the B cells and expresses with the same heavy chain constant region, and the same light chain, and expressed in a cell to make a bispecific antibody, wherein the light chain component of the bispecific antibody has been selected by a mouse to associate and express with the light chain component.
  • the inventors have engineered a mouse for generating immunoglobulin light chains that will suitably pair with a rather diverse family of heavy chains, including heavy chains whose variable regions depart from germline sequences, e.g., affinity matured or somatically mutated variable regions.
  • the mouse is devised to pair human light chain variable domains with human heavy chain variable domains that comprise somatic mutations, thus enabling a route to high affinity binding proteins suitable for use as human therapeutics.
  • the genetically engineered mouse through the long and complex process of antibody selection within an organism, makes biologically appropriate choices in pairing a diverse collection of human heavy chain variable domains with a limited number of human light chain options.
  • the mouse is engineered to present a limited number of human light chain variable domain options in conjunction with a wide diversity of human heavy chain variable domain options.
  • the mouse Upon challenge with an immunogen, the mouse maximizes the number of solutions in its repertoire to develop an antibody to the immunogen, limited largely or solely by the number or light chain options in its repertoire. In various embodiments, this includes allowing the mouse to achieve suitable and compatible somatic mutations of the light chain variable domain that will nonetheless be compatible with a relatively large variety of human heavy chain variable domains, including in particular somatically mutated human heavy chain variable domains.
  • the mouse is engineered to render nonfunctional or substantially nonfunctional its ability to make, or rearrange, a native mouse light chain variable domain. This can be achieved, e.g., by deleting the mouse's light chain variable region gene segments.
  • the endogenous mouse locus can then be modified by an exogenous suitable human light chain variable region gene segment of choice, operably linked to the endogenous mouse light chain constant domain, in a manner such that the exogenous human variable region gene segments can combine with the endogenous mouse light chain constant region gene and form a rearranged reverse chimeric light chain gene (human variable, mouse constant).
  • the light chain variable region is capable of being somatically mutated.
  • the appropriate enhancer(s) is retained in the mouse.
  • the mouse ⁇ intronic enhancer and mouse ⁇ 3′ enhancer are functionally maintained, or undisrupted.
  • a genetically engineered mouse that expresses a limited repertoire of reverse chimeric (human variable, mouse constant) light chains associated with a diversity of reverse chimeric (human variable, mouse constant) heavy chains.
  • the endogenous mouse ⁇ light chain gene segments are deleted and replaced with a single (or two) rearranged human light chain region, operably linked to the endogenous mouse C ⁇ gene.
  • the mouse ⁇ intronic enhancer and the mouse ⁇ 3′ enhancer are maintained.
  • the mouse also comprises a nonfunctional ⁇ light chain locus, or a deletion thereof or a deletion that renders the locus unable to make a ⁇ light chain.
  • a genetically engineered mouse comprises a light chain variable region locus lacking endogenous mouse light chain V L and J L gene segments and comprising a rearranged human light chain variable region, in one embodiment a rearranged human V L /J L sequence, operably linked to a mouse constant region, wherein the locus is capable of undergoing somatic hypermutation, and wherein the locus expresses a light chain comprising the human V L /J L sequence linked to a mouse constant region.
  • the locus comprises a mouse ⁇ 3′ enhancer, which is correlated with a normal, or wild type, level of somatic hypermutation.
  • the genetically engineered mouse in various embodiments when immunized with an antigen of interest generates B cells that exhibit a diversity of rearrangements of human immunoglobulin heavy chain variable regions that express and function with one or with two rearranged light chains, including embodiments where the one or two light chains comprise human light chain variable regions that comprise, e.g., 1 to 5 somatic mutations.
  • the human light chains so expressed are capable of associating and expressing with any human immunoglobulin heavy chain variable region expressed in the mouse.
  • compositions and methods of described herein can be used to make binding proteins that bind more than one epitope with high affinity, e.g., bispecific antibodies.
  • Advantages of the invention include the ability to select suitably high binding (e.g., affinity matured) heavy chain immunoglobulin chains each of which will associate with a single light chain.
  • each construct encodes a human heavy chain variable domain that binds a different epitope.
  • One of the human V L s e.g., human V ⁇ 1-39J ⁇ 5 or human V ⁇ 3-20J ⁇ 1
  • a suitable human constant region gene e.g., a human ⁇ constant gene.
  • one of the heavy chains is modified to omit a Protein A-binding determinant, resulting in a differential affinity of a homodimeric binding protein from a heterodimeric binding protein.
  • Compositions and methods that address this issue are described in U.S. Ser. No. 12/832,838, filed 25 Jun. 2010, entitled “Readily Isolated Bispecific Antibodies with Native Immunoglobulin Format,” published as US 2010/0331527A1, hereby incorporated by reference.
  • an epitope-binding protein as described herein wherein human V L and V H sequences are derived from mice described herein that have been immunized with an antigen comprising an epitope of interest.
  • an epitope-binding protein comprises a first and a second polypeptide, the first polypeptide comprising, from N-terminal to C-terminal, a first epitope-binding region that selectively binds a first epitope, followed by a constant region that comprises a first C H 3 region of a human IgG selected from IgG1, IgG2, IgG4, and a combination thereof; and, a second polypeptide comprising, from N-terminal to C-terminal, a second epitope-binding region that selectively binds a second epitope, followed by a constant region that comprises a second C H 3 region of a human IgG selected from IgG1, IgG2, IgG4, and a combination thereof, wherein the second C H 3 region comprises a modification that reduces or eliminates binding of the second C H 3 domain to protein A.
  • the second C H 3 region comprises an H95R modification (by IMGT exon numbering; H435R by EU numbering). In another embodiment, the second C H 3 region further comprises a Y96F modification (IMGT; Y436F by EU).
  • the second C H 3 region is from a modified human IgG1, and further comprises a modification selected from the group consisting of D16E, L18M, N44S, K52N, V57M, and V82I (IMGT; D356E, L358M, N384S, K392N, V397M, and V422I by EU).
  • the second C H 3 region is from a modified human IgG2, and further comprises a modification selected from the group consisting of N44S, K52N, and V82I (IMGT; N384S, K392N, and V422I by EU).
  • IMGT N44S, K52N, and V82I
  • the second C H 3 region is from a modified human IgG4, and further comprises a modification selected from the group consisting of Q15R, N44S, K52N, V57M, R69K, E79Q, and V82I (IMGT; Q355R, N384S, K392N, V397M, R409K, E419Q, and V422I by EU).
  • IMGT Q15R, N44S, K52N, V57M, R69K, E79Q, and V82I
  • One method for making an epitope-binding protein that binds more than one epitope is to immunize a first mouse in accordance with the invention with an antigen that comprises a first epitope of interest, wherein the mouse comprises an endogenous immunoglobulin light chain variable region locus that does not contain an endogenous mouse V L that is capable of rearranging and forming a light chain, wherein at the endogenous mouse immunoglobulin light chain variable region locus is a single rearranged human V L region operably linked to the mouse endogenous light chain constant region gene, and the rearranged human V L region is selected from a human V ⁇ 1-39J ⁇ 5 and a human V ⁇ 3-20J ⁇ 1, and the endogenous mouse V H gene segments have been replaced in whole or in part with human V H gene segments, such that immunoglobulin heavy chains made by the mouse are solely or substantially heavy chains that comprise human variable domains and mouse constant domains.
  • a reverse chimeric antibody comprising only one of two human light chain variable domains (e.g., one of human V ⁇ 1-39J ⁇ 5 or human V ⁇ 3-20J ⁇ 1).
  • the nucleotide sequence of the V H (and, optionally, the V L ) can be retrieved (e.g., by PCR) and cloned into an expression construct in frame with a suitable human immunoglobulin constant domain.
  • This process can be repeated to identify a second V H domain that binds a second epitope, and a second V H gene sequence can be retrieved and cloned into an expression vector in frame to a second suitable immunoglobulin constant domain.
  • the first and the second immunoglobulin constant domains can the same or different isotype, and one of the immunoglobulin constant domains (but not the other) can be modified as described herein or in US 2010/0331527A1, and epitope-binding protein can be expressed in a suitable cell and isolated based on its differential affinity for Protein A as compared to a homodimeric epitope-binding protein, e.g., as described in US 2010/0331527A1.
  • a method for making a bispecific epitope-binding protein comprising identifying a first affinity-matured (e.g., comprising one or more somatic hypermutations) human V H nucleotide sequence (V H 1) from a mouse as described herein, identifying a second affinity-matured (e.g., comprising one or more somatic hypermutations) human V H nucleotide sequence (V H 2) from a mouse as described herein, cloning V H 1 in frame with a human heavy chain lacking a Protein A-determinant modification as described in US 2010/0331527A1 for form heavy chain 1 (HC1), cloning V H 2 in frame with a human heavy chain comprising a Protein A-determinant as described in US 2010/0331527A1 to form heavy chain 2 (HC2), introducing an expression vector comprising HC1 and the same or a different expression vector comprising HC2 into a cell, wherein the cell also expresses a human immunoglobin
  • HC1 is an IgG1
  • HC2 is an IgG1 that comprises the modification H95R (IMGT; H435R by EU) and further comprises the modification Y96F (IMGT; Y436F by EU).
  • the VH domain encoded by V H 1, the V H domain encoded by V H 2, or both, are somatically mutated.
  • a variety of human variable regions from affinity-matured antibodies raised against four different antigens were expressed with either their cognate light chain, or at least one of a human light chain selected from human V ⁇ 1-39J ⁇ 5, human V ⁇ 3-20J ⁇ 1, or human VpreBJ ⁇ 5 (see Example 1).
  • somatically mutated high affinity heavy chains from different gene families paired successfully with rearranged human germline V ⁇ 1-39J ⁇ 5 and V ⁇ 3-20J ⁇ 1 regions and were secreted from cells expressing the heavy and light chains.
  • V H domains derived from the following human V H gene families expressed favorably: 1-2, 1-8, 1-24, 2-5, 3-7, 3-9, 3-11, 3-13, 3-15, 3-20, 3-23, 3-30, 3-33, 3-48, 4-31, 4-39, 4-59, 5-51, and 6-1.
  • a mouse that is engineered to express a limited repertoire of human V L domains from one or both of V ⁇ 1-39J ⁇ 5 and V ⁇ 3-20J ⁇ 1 will generate a diverse population of somatically mutated human V H domains from a V H locus modified to replace mouse V H gene segments with human V H gene segments.
  • Mice genetically engineered to express reverse chimeric (human variable, mouse constant) immunoglobulin heavy chains associated with a single rearranged light chain e.g., a V ⁇ 1-39/J or a V ⁇ 3-20/J
  • a single rearranged light chain e.g., a V ⁇ 1-39/J or a V ⁇ 3-20/J
  • B cells that comprised a diversity of human V H rearrangements and expressed a diversity of high-affinity antigen-specific antibodies with diverse properties with respect to their ability to block binding of the antigen to its ligand, and with respect to their ability to bind variants of the antigen (see Examples 5 through 10).
  • mice and methods described herein are useful in making and selecting human immunoglobulin heavy chain variable domains, including somatically mutated human heavy chain variable domains, that result from a diversity of rearrangements, that exhibit a wide variety of affinities (including exhibiting a K D of about a nanomolar or less), a wide variety of specificities (including binding to different epitopes of the same antigen), and that associate and express with the same or substantially the same human immunoglobulin light chain variable region.
  • An in vitro expression system was constructed to determine if a single rearranged human germline light chain could be co-expressed with human heavy chains from antigen specific human antibodies.
  • VELOCIMMUNE® Methods for generating human antibodies in genetically modified mice are known (see e.g., U.S. Pat. No. 6,596,541, Regeneron Pharmaceuticals, VELOCIMMUNE®).
  • the VELOCIMMUNE® technology involves generation of a genetically modified mouse having a genome comprising human heavy and light chain variable regions operably linked to endogenous mouse constant region loci such that the mouse produces an antibody comprising a human variable region and a mouse constant region in response to antigenic stimulation.
  • the DNA encoding the variable regions of the heavy and light chains of the antibodies produced from a VELOCIMMUNE® mouse are fully human. Initially, high affinity chimeric antibodies are isolated having a human variable region and a mouse constant region.
  • the antibodies are characterized and selected for desirable characteristics, including affinity, selectivity, epitope, etc.
  • the mouse constant regions are replaced with a desired human constant region to generate a fully human antibody containing a non-IgM isotype, for example, wild type or modified IgG1, IgG2, IgG3 or IgG4. While the constant region selected may vary according to specific use, high affinity antigen-binding and target specificity characteristics reside in the variable region.
  • a VELOCIMMUNE® mouse was immunized with a growth factor that promotes angiogenesis (Antigen C) and antigen-specific human antibodies were isolated and sequenced for V gene usage using standard techniques recognized in the art. Selected antibodies were cloned onto human heavy and light chain constant regions and 69 heavy chains were selected for pairing with one of three human light chains: (1) the cognate ⁇ light chain linked to a human ⁇ constant region, (2) a rearranged human germline V ⁇ 1-39J ⁇ 5 linked to a human ⁇ constant region, or (3) a rearranged human germline V ⁇ 3-20J ⁇ 1 linked to a human ⁇ constant region. Each heavy chain and light chain pair were co-transfected in CHO-K1 cells using standard techniques.
  • Presence of antibody in the supernatant was detected by anti-human IgG in an ELISA assay.
  • Antibody titer (ng/ml) was determined for each heavy chain/light chain pair and titers with the different rearranged germline light chains were compared to the titers obtained with the parental antibody molecule (i.e., heavy chain paired with cognate light chain) and percent of native titer was calculated (Table 1).
  • V H Heavy chain variable gene.
  • ND no expression detected under current experimental conditions.
  • VELOCIMMUNE® mice were immunized with several different antigens and selected heavy chains of antigen specific human antibodies were tested for their ability to pair with different rearranged human germline light chains (as described above).
  • the antigens used in this experiment included an enzyme involved in cholesterol homeostasis (Antigen A), a serum hormone involved in regulating glucose homeostasis (Antigen B), a growth factor that promotes angiogenesis (Antigen C) and a cell-surface receptor (Antigen D).
  • Antigen specific antibodies were isolated from mice of each immunization group and the heavy chain and light chain variable regions were cloned and sequenced.
  • V gene usage was determined and selected heavy chains were paired with either their cognate light chain or a rearranged human germline V ⁇ 1-39J ⁇ 5 region. Each heavy/light chain pair was co-transfected in CHO-K1 cells and the presence of antibody in the supernatant was detected by anti-human IgG in an ELISA assay. Antibody titer ( ⁇ g/ml) was determined for each heavy chain/light chain pairing and titers with the different rearranged human germline light chains were compared to the titers obtained with the parental antibody molecule (i.e., heavy chain paired with cognate light chain) and percent of native titer was calculated (Table 2).
  • V H Heavy chain variable gene.
  • V ⁇ ⁇ light chain variable gene.
  • ND no expression detected under current experimental conditions.
  • both rearranged human germline light chains conferred an increase in expression as compared to the cognate light chain of the parental antibody.
  • the rearranged human germline V ⁇ 1-39J ⁇ 5 region conferred an increase in expression of several heavy chains specific for a range of different classes of antigens as compared to the cognate light chain for the parental antibodies.
  • Antibody titer was increased by more than two-fold for about 35% (15/43) of the heavy chains as compared to the cognate light chain of the parental antibodies. For two heavy chains (315 and 316), the increase was greater than ten-fold as compared to the parental antibody.
  • V H 3 family three heavy chains are over represented in comparison to other heavy chain variable region gene families. This demonstrates a favorable relationship of human V H 3 heavy chains to pair with rearranged human germline V ⁇ 1-39J ⁇ 5 and V ⁇ 3-20J ⁇ 1 light chains.
  • a DNA segment containing exon 1 (encoding the leader peptide) and intron 1 of the mouse V ⁇ 3-7 gene was made by de novo DNA synthesis (Integrated DNA Technologies). Part of the 5′ untranslated region up to a naturally occurring Blpl restriction enzyme site was included. Exons of human V ⁇ 1-39 and V ⁇ 3-20 genes were PCR amplified from human genomic BAC libraries. The forward primers had a 5′ extension containing the splice acceptor site of intron 1 of the mouse V ⁇ 3-7 gene.
  • the reverse primer used for PCR of the human V ⁇ 1-39 sequence included an extension encoding human J ⁇ 5, whereas the reverse primer used for PCR of the human V ⁇ 3-20 sequence included an extension encoding human J ⁇ 1.
  • the human VpreBJ ⁇ 5 sequence was made by de novo DNA synthesis (Integrated DNA Technologies). A portion of the human J ⁇ -C ⁇ intron including the splice donor site was PCR amplified from plasmid pBS-296-HA18-PIScel. The forward PCR primer included an extension encoding part of either a human J ⁇ 5, J ⁇ 1, or J ⁇ 5 sequence. The reverse primer included a PI-Scel site, which was previously engineered into the intron.
  • mice V ⁇ 3-7 exon1/intron 1, human variable light chain exons, and human J ⁇ -C ⁇ intron fragments were sewn together by overlap extension PCR, digested with Blpl and PI-Scel, and ligated into plasmid pBS-296-HA18-PIScel, which contained the promoter from the human V ⁇ 3-15 variable gene segment.
  • a loxed hygromycin cassette within plasmid pBS-296-HA18-PIScel was replaced with a FRTed hygromycin cassette flanked by NotI and AscI sites.
  • the NotI/PI-Scel fragment of this plasmid was ligated into modified mouse BAC 254m04, which contained part of the mouse J ⁇ -C ⁇ intron, the mouse CK exon, and about 75 kb of genomic sequence downstream of the mouse ⁇ locus, which provided a 3′ homology arm for homologous recombination in mouse ES cells.
  • the Notl/Ascl fragment of this BAC was then ligated into modified mouse BAC 302g12, which contained a FRTed neomycin cassette and about 23 kb of genomic sequence upstream of the endogenous ⁇ locus for homologous recombination in mouse ES cells.
  • FIG. 1 Rearranged Human Germline V ⁇ 1-39J ⁇ 5 Targeting Vector ( FIG. 1 ). Restriction enzyme sites were introduced at the 5′ and 3′ ends of an engineered light chain insert for cloning into a targeting vector: an Ascl site at the 5′ end and a PI-Scel site at the 3′ end.
  • the targeting construct from 5′ to 3′ included a 5′ homology arm containing sequence 5′ to the endogenous mouse ⁇ light chain locus obtained from mouse BAC clone 302g12, a FRTed neomycin resistance gene, an genomic sequence including the human V ⁇ 3-15 promoter, a leader sequence of the mouse V ⁇ 3-7 variable gene segment, a intron sequence of the mouse V ⁇ 3-7 variable gene segment, an open reading frame of a rearranged human germline V ⁇ 1-39J ⁇ 5 region, a genomic sequence containing a portion of the human J ⁇ -C ⁇ intron, and a 3′ homology arm containing sequence 3′ of the endogenous mouse J ⁇ 5 gene segment obtained from mouse BAC clone 254m04 ( FIG.
  • Targeted insertion of the rearranged human germline V ⁇ 1-39J ⁇ 5 region into BAC DNA was confirmed by polymerase chain reaction (PCR) using primers located at sequences within the rearranged human germline light chain region. Briefly, the intron sequence 3′ to the mouse V ⁇ 3-7 leader sequence was confirmed with primers ULC-m1F (AGGTGAGGGT ACAGATAAGT GTTATGAG; SEQ ID NO:2) and ULC-m1R (TGACAAATGC CCTAATTATA GTGATCA; SEQ ID NO:3).
  • the open reading frame of the rearranged human germline V ⁇ 1-39J ⁇ 5 region was confirmed with primers 1633-h2F (GGGCAAGTCA GAGCATTAGC A; SEQ ID NO:4) and 1633-h2R (TGCAAACTGG ATGCAGCATA G; SEQ ID NO:5).
  • the neomycin cassette was confirmed with primers neoF (GGTGGAGAGG CTATTCGGC; SEQ ID NO:6) and neoR (GAACACGGCG GCATCAG; SEQ ID NO:7).
  • Targeted BAC DNA was then used to electroporate mouse ES cells to created modified ES cells for generating chimeric mice that express a rearranged human germline V ⁇ 1-39J ⁇ 5 region.
  • Positive ES cell clones were confirmed by TAQMANTM screening and karyotyping using probes specific for the engineered V ⁇ 1-39J ⁇ 5 light chain region inserted into the endogenous locus. Briefly, probe neoP (TGGGCACAAC AGACAATCGG CTG; SEQ ID NO:8) which binds within the neomycin marker gene, probe ULC-m1P (CCATTATGAT GCTCCATGCC TCTCTGTTC; SEQ ID NO:9) which binds within the intron sequence 3′ to the mouse V ⁇ 3-7 leader sequence, and probe 1633h2P (ATCAGCAGAA ACCAGGGAAA GCCCCT; SEQ ID NO:10) which binds within the rearranged human germline V ⁇ 1-39J ⁇ 5 open reading frame. Positive ES cell clones were then used to implant female mice to give rise to a litter of pups expressing the germline V ⁇ 1-39J ⁇ 5 light chain region.
  • ES cells bearing the rearranged human germline V ⁇ 1-39J ⁇ 5 light chain region are transfected with a construct that expresses FLP in order to remove the FRTed neomycin cassette introduced by the targeting construct.
  • the neomycin cassette is removed by breeding to mice that express FLP recombinase (e.g., U.S. Pat. No. 6,774,279).
  • the neomycin cassette is retained in the mice.
  • FIG. 2 Rearranged Human Germline V ⁇ 3-20J ⁇ 1 Targeting Vector
  • a targeting construct including, from 5′ to 3′, a 5′ homology arm containing sequence 5′ to the endogenous mouse ⁇ light chain locus obtained from mouse BAC clone 302g12, a FRTed neomycin resistance gene, a genomic sequence including the human V ⁇ 3-15 promoter, a leader sequence of the mouse V ⁇ 3-7 variable gene segment, an intron sequence of the mouse V ⁇ 3-7 variable gene segment, an open reading frame of a rearranged human germline V ⁇ 3-20J ⁇ 1 region, a genomic sequence containing a portion of the human J ⁇ -C ⁇ intron, and a 3′ homology arm containing sequence 3′ of the endogenous mouse J ⁇ 5 gene segment obtained from mouse BAC clone 254m04 ( FIG. 2 , middle).
  • Targeted insertion of the rearranged human germline V ⁇ 3-20J ⁇ 1 region into BAC DNA was confirmed by polymerase chain reaction (PCR) using primers located at sequences within the rearranged human germline V ⁇ 3-20J ⁇ 1 light chain region.
  • PCR polymerase chain reaction
  • the intron sequence 3′ to the mouse V ⁇ 3-7 leader sequence was confirmed with primers ULC-m1F (SEQ ID NO:2) and ULC-m1R (SEQ ID NO:3).
  • the open reading frame of the rearranged human germline V ⁇ 3-20J ⁇ 1 region was confirmed with primers 1635-h2F (TCCAGGCACC CTGTCTTTG; SEQ ID NO:12) and 1635-h2R (AAGTAGCTGC TGCTAACACT CTGACT; SEQ ID NO:13).
  • neomycin cassette was confirmed with primers neoF (SEQ ID NO:6) and neoR (SEQ ID NO:7).
  • Targeted BAC DNA was then used to electroporate mouse ES cells to created modified ES cells for generating chimeric mice that express the rearranged human germline V ⁇ 3-20J ⁇ 1 light chain.
  • Positive ES cell clones were confirmed by TAQMANTM screening and karyotyping using probes specific for the engineered V ⁇ 3-20J ⁇ 1 light chain region inserted into the endogenous ⁇ light chain locus. Briefly, probe neoP (SEQ ID NO:8) which binds within the neomycin marker gene, probe ULC-m1P (SEQ ID NO:9) which binds within the mouse V ⁇ 3-7 leader sequence, and probe 1635h2P (AAAGAGCCAC CCTCTCCTGC AGGG; SEQ ID NO:14) which binds within the human V ⁇ 3-20J ⁇ 1 open reading frame. Positive ES cell clones were then used to implant female mice. A litter of pups expressing the human germline V ⁇ 3-20J ⁇ 1 light chain region.
  • ES cells bearing human germline V ⁇ 3-20J ⁇ 1 light chain region can be transfected with a construct that expresses FLP in order to remove the FRTed neomycin cassette introduced by the targeting construct.
  • the neomycin cassette may be removed by breeding to mice that express FLP recombinase (e.g., U.S. Pat. No. 6,774,279).
  • the neomycin cassette is retained in the mice.
  • an engineered light chain locus expressing a rearranged human germline VpreBJ ⁇ 5 region was made using a targeting construct including, from 5′ to 3′, a 5′ homology arm containing sequence 5′ to the endogenous mouse ⁇ light chain locus obtained from mouse BAC clone 302g12, a FRTed neomycin resistance gene, an genomic sequence including the human V ⁇ 3-15 promoter, a leader sequence of the mouse V ⁇ 3-7 variable gene segment, an intron sequence of the mouse V ⁇ 3-7 variable gene segment, an open reading frame of a rearranged human germline VpreBJ ⁇ 5 region, a genomic sequence containing a portion of the human J ⁇ -C ⁇ intron, and a 3′ homology arm containing sequence 3′ of the endogenous mouse J ⁇ 5 gene segment obtained from mouse BAC clone 254m04 ( FIG. 3 , middle).
  • Targeted insertion of the rearranged human germline VpreBJ ⁇ 5 region into BAC DNA was confirmed by polymerase chain reaction (PCR) using primers located at sequences within the rearranged human germline VpreBJ ⁇ 5 region light chain region.
  • PCR polymerase chain reaction
  • the intron sequence 3′ to the mouse V ⁇ 3-7 leader sequence was confirmed with primers ULC-m1F (SEQ ID NO:2) and ULC-m1R (SEQ ID NO:3).
  • the open reading frame of the rearranged human germline VpreBJ ⁇ 5 region was confirmed with primers 1616-h1F (TGTCCTCGGC CCTTGGA; SEQ ID NO:16) and 1616-h1R (CCGATGTCAT GGTCGTTCCT; SEQ ID NO:17).
  • neomycin cassette was confirmed with primers neoF (SEQ ID NO:6) and neoR (SEQ ID NO:7).
  • Targeted BAC DNA was then used to electroporate mouse ES cells to created modified ES cells for generating chimeric mice that express the rearranged human germline VpreBJ ⁇ 5 light chain.
  • Positive ES cell clones are confirmed by TAQMANTM screening and karyotyping using probes specific for the engineered VpreBJ ⁇ 5 light chain region inserted into the endogenous ⁇ light chain locus. Briefly, probe neoP (SEQ ID NO:8) which binds within the neomycin marker gene, probe ULC-m1P (SEQ ID NO:9) which binds within the mouse IgV ⁇ 3-7 leader sequence, and probe 1616h1P (ACAATCCGCC TCACCTGCAC CCT; SEQ ID NO:18) which binds within the human VpreBJ ⁇ 5 open reading frame. Positive ES cell clones are then used to implant female mice to give rise to a litter of pups expressing a germline light chain region.
  • ES cells bearing the rearranged human germline VpreBJ ⁇ 5 light chain region are transfected with a construct that expresses FLP in order to remove the FRTed neomycin cassette introduced by the targeting construct.
  • the neomycin cassette is removed by breeding to mice that express FLP recombinase (e.g., U.S. Pat. No. 6,774,279).
  • the neomycin cassette is retained in the mice.
  • Targeted ES cells described above were used as donor ES cells and introduced into an 8-cell stage mouse embryo by the VELOCIMOUSE® method (see, e.g., U.S. Pat. No. 7,294,754 and Poueymirou et al. (2007) F0 generation mice that are essentially fully derived from the donor gene-targeted ES cells allowing immediate phenotypic analyses Nature Biotech. 25(1):91-99.
  • VELOCIMICE® independently bearing an engineered human germline V ⁇ 1-39J ⁇ 5 light chain region, a V ⁇ 3-20J ⁇ 1 light chain region or a VpreBJ ⁇ 5 light chain region are identified by genotyping using a modification of allele assay (Valenzuela et al., supra) that detects the presence of the unique rearranged human germline light chain region.
  • Pups are genotyped and a pup heterozygous or homozygous for the unique rearranged human germline light chain region are selected for characterizing expression of the rearranged human germline light chain region.
  • CD19 + B cells were purified from the spleens of wild type, mice homozygous for a replacement of the mouse heavy chain and ⁇ light chain variable region loci with corresponding human heavy chain and ⁇ light chain variable region loci (H ⁇ ), as well as mice homozygous and heterozygous for each rearranged human light chain region (V ⁇ 1-39J ⁇ 5 or V ⁇ 3-20J ⁇ 1) using mouse CD19 Microbeads (Miltenyi Biotec) according to manufacturer's specifications.
  • Total RNA was purified from CD19 + B cells using RNeasy Mini kit (Qiagen) according to manufacturer's specifications and genomic RNA was removed using an RNase-free DNase on-column treatment (Qiagen).
  • Common light chain mice bearing either a V ⁇ 1-39J ⁇ 5 or V ⁇ 3-20J ⁇ 1 common light chain at the endogenous mouse ⁇ light chain locus were immunized with (3-galactosidase and antibody titer was measured.
  • ELISA plates (Nunc) were coated with 1 ⁇ g/mL ⁇ -galactosidase overnight at 4° C. Excess antigen was washed off before blocking with PBS with 1% BSA for one hour at room temperature. Serial dilutions of serum were added to the plates and incubated for one hour at room temperature before washing.
  • V ⁇ 1-39J ⁇ 5 and V ⁇ 3-20J ⁇ 1 common light chain mice demonstrated a near wild type pattern (Table 3 and FIG. 4 ).
  • VpreBJ ⁇ 5 common light chain mice demonstrated fewer peripheral B cells, of which about 1-2% express the engineered human light chain region (data not shown).
  • the expression levels of the V ⁇ 1-39J ⁇ 5 and V ⁇ 3-20J ⁇ 1 rearranged human light chain regions from the endogenous ⁇ light chain locus were elevated in comparison to an endogenous ⁇ light chain locus containing a complete replacement of mouse V ⁇ and J ⁇ gene segments with human V ⁇ and J ⁇ gene segments ( FIGS. 5A , 5 B and 5 C).
  • VpreBJ ⁇ 5 rearranged human light chain region demonstrated similar high expression from the endogenous ⁇ light chain locus in both heterozygous and homozygous mice (data not shown). This demonstrates that in direct competition with the mouse ⁇ , ⁇ , or both endogenous light chain loci, a single rearranged human V L /J L sequence can yield better than wild type level expression from the endogenous ⁇ light chain locus and give rise to normal splenic and blood B cell frequency.
  • This Example describes several other genetically modified mouse strains that can be bred to any one of the common light chain mice described herein to create multiple genetically modified mouse strains harboring multiple genetically modified immunoglobulin loci.
  • mice bearing one of the rearranged human germline light chain regions are bred to another mouse containing a deletion in the endogenous ⁇ light chain locus.
  • the progeny obtained will express, as their only light chain, the rearranged human germline light chain region as described in Example 2. Breeding is performed by standard techniques recognized in the art and, alternatively, by a commercial breeder (e.g., The Jackson Laboratory).
  • Mouse strains bearing an engineered light chain locus and a deletion of the endogenous ⁇ light chain locus are screened for presence of the unique light chain region and absence of endogenous mouse ⁇ light chains.
  • mice bearing an engineered human germline light chain locus are bred with mice that contain a replacement of the endogenous mouse heavy chain variable gene locus with the human heavy chain variable gene locus (see U.S. Pat. No. 6,596,541; the VELOCIMMUNE® mouse, Regeneron Pharmaceuticals, Inc.).
  • the VELOCIMMUNE® mouse comprises a genome comprising human heavy chain variable regions operably linked to endogenous mouse constant region loci such that the mouse produces antibodies comprising a human heavy chain variable region and a mouse heavy chain constant region in response to antigenic stimulation.
  • the DNA encoding the variable regions of the heavy chains of the antibodies is isolated and operably linked to DNA encoding the human heavy chain constant regions.
  • the DNA is then expressed in a cell capable of expressing the fully human heavy chain of the antibody.
  • mice bearing a replacement of the endogenous mouse V H locus with the human VH locus and a single rearranged human germline V L region at the endogenous ⁇ light chain locus are obtained.
  • Reverse chimeric antibodies containing somatically mutated heavy chains (human V H and mouse C H ) with a single human light chain (human V L and mouse C L ) are obtained upon immunization with an antigen of interest.
  • V H and V L nucleotide sequences of B cells expressing the antibodies are identified and fully human antibodies are made by fusion the V H and V L nucleotide sequences to human C H and C L nucleotide sequences in a suitable expression system.
  • mice After breeding mice that contain the engineered human light chain region to various desired strains containing modifications and deletions of other endogenous Ig loci (as described in Example 4), selected mice can be immunized with an antigen of interest.
  • a VELOCIMMUNE® mouse containing one of the single rearranged human germline light chain regions is challenged with an antigen, and lymphatic cells (such as B-cells) are recovered from serum of the animals.
  • lymphatic cells such as B-cells
  • the lymphatic cells are fused with a myeloma cell line to prepare immortal hybridoma cell lines, and such hybridoma cell lines are screened and selected to identify hybridoma cell lines that produce antibodies containing human heavy chain variables and a rearranged human germline light chains which are specific to the antigen used for immunization.
  • DNA encoding the variable regions of the heavy chains and the light chain are isolated and linked to desirable isotypic constant regions of the heavy chain and light chain.
  • the single light chain of each antibody may be somatically mutated. This adds additional diversity to the antigen-specific repertoire comprising a single light chain and diverse heavy chain sequences.
  • the resulting cloned antibody sequences are subsequently expressed in a cell, such as a CHO cell.
  • DNA encoding the antigen-specific chimeric antibodies or the variable domains of the light and heavy chains are identified directly from antigen-specific lymphocytes.
  • high affinity chimeric antibodies are isolated having a human variable region and a mouse constant region.
  • the antibodies are characterized and selected for desirable characteristics, including affinity, selectivity, epitope, etc.
  • the mouse constant regions are replaced with a desired human constant region to generate the fully human antibody containing a somatically mutated human heavy chain and a single light chain derived from a rearranged human germline light chain region of the invention.
  • Suitable human constant regions include, for example wild type or modified IgG1 or IgG4.
  • Antigen E Two to three micrograms of Antigen E are mixed with 10 ⁇ g of CpG oligonucleotide (Cat # tlrl-modn—ODN1826 oligonucleotide; InVivogen, San Diego, Calif.) and 25 ⁇ g of Adju-Phos (Aluminum phosphate gel adjuvant, Cat# H-71639-250; Brenntag Biosector, Frederikssund, Denmark) prior to injection. A total of six injections are given prior to the final antigen recall, which is given 3-5 days prior to sacrifice. Bleeds after the 4th and 6th injection are collected and the antibody immune response is monitored by a standard antigen-specific immunoassay.
  • CpG oligonucleotide Cat # tlrl-modn—ODN1826 oligonucleotide; InVivogen, San Diego, Calif.
  • Adju-Phos Alluminum phosphate gel adjuvant, Cat# H-71639-250;
  • splenocytes are harvested and fused with mouse myeloma cells to preserve their viability and form hybridoma cell lines.
  • the hybridoma cell lines are screened and selected to identify cell lines that produce Antigen E-specific common light chain antibodies.
  • anti-Antigen E-specific common light chain antibodies i.e., antibodies possessing human heavy chain variable domains, the same human light chain variable domain, and mouse constant domains.
  • anti-Antigen E common light chain antibodies are isolated directly from antigen-positive B cells without fusion to myeloma cells, as described in U.S. 2007/0280945A1, herein specifically incorporated by reference in its entirety.
  • several fully human anti-Antigen E common light chain antibodies i.e., antibodies possessing human heavy chain variable domains, either an engineered human V ⁇ 1-39J ⁇ 5 light chain or an engineered human V ⁇ 3-20J ⁇ 1 light chain region, and human constant domains
  • nucleic acids encoding heavy chain antibody variable regions were cloned and sequenced. From the nucleic acid sequences and predicted amino acid sequences of the antibodies, gene usage was identified for the heavy chain variable region (HCVR) of selected common light chain antibodies obtained from immunized VELOCIMMUNE® mice containing either the engineered human V ⁇ 1-39J ⁇ 5 light chain or engineered human V ⁇ 3-20J ⁇ 1 light chain region.
  • HCVR heavy chain variable region
  • mice according to the invention generate antigen-specific common light chain antibodies from a variety of human heavy chain gene segments, due to a variety of rearrangements, when employing either a mouse that expresses a light chain from only a human V ⁇ 1-39- or a human V ⁇ 3-20-derived light chain.
  • Human V H gene segments of the 2, 3, 4, and 5 families rearranged with a variety of human D H segments and human J H segments to yield antigen-specific antibodies.
  • the extracellular domain (ECD) of Antigen E was conjugated to two myc epitope tags and a 6 ⁇ histidine tag (Antigen E-mmH) and amine-coupled to carboxylated microspheres at a concentration of 20 ⁇ g/mL in MES buffer. The mixture was incubated for two hours at room temperature followed by bead deactivation with 1M Tris pH 8.0 followed by washing in PBS with 0.05% (v/v) Tween-20. The beads were then blocked with PBS (Irvine Scientific, Santa Ana, Calif.) containing 2% (w/v) BSA (Sigma-Aldrich Corp., St. Louis, Mo.).
  • Detection of biotinylated-Ligand Y bound to Antigen E-myc-myc-6His labeled beads was determined with R-Phycoerythrin conjugated to Streptavidin (Moss Inc, Pasadena, Md.) followed by measurement in a LUMINEXTM flow cytometry-based analyzer. Background Mean Fluorescence Intensity (MFI) of a sample without Ligand Y was subtracted from all samples. Percent blocking was calculated by division of the background-subtracted MFI of each sample by the adjusted negative control value, multiplying by 100 and subtracting the resulting value from 100.
  • MFI Green Fluorescence Intensity
  • Ligand Y was amine-coupled to carboxylated microspheres at a concentration of 20 ⁇ g/mL diluted in MES buffer. The mixture and incubated two hours at room temperature followed by deactivation of beads with 1M Tris pH 8 then washing in PBS with 0.05% (v/v) Tween-20. The beads were then blocked with PBS (Irvine Scientific, Santa Ana, Calif.) containing 2% (w/v) BSA (Sigma-Aldrich Corp., St. Louis, Mo.). In a 96-well filter plate, supernatants containing Antigen E-specific common light chain antibodies were diluted 1:15 in buffer.
  • a negative control containing a mock supernatant with the same media components as for the antibody supernatant was prepared.
  • a biotinylated-Antigen E-mmH was added to a final concentration of 0.42 nM and incubated overnight at 4° C.
  • Ligand Y-labeled beads were then added to the antibody/Antigen E mixture and incubated for two hours at room temperature. Detection of biotinylated-Antigen E-mmH bound to Ligand Y-beads was determined with R-Phycoerythrin conjugated to Streptavidin (Moss Inc, Pasadena, Md.) followed by measurement in a LUMINEXTM flow cytometry-based analyzer.
  • MFI Background Mean Fluorescence Intensity
  • Tables 7 and 8 show the percent blocking for all 98 anti-Antigen E common light chain antibodies tested in both LUMINEXTM assays. ND: not determined under current experimental conditions.
  • Tables 7 and 8 establish that the rearrangements described in Tables 5 and 6 generated anti-Antigen E-specific common light chain antibodies that blocked binding of Ligand Y to its cognate receptor Antigen E with varying degrees of efficacy, which is consistent with the anti-Antigen E common light chain antibodies of Tables 5 and 6 comprising antibodies with overlapping and non-overlapping epitope specificity with respect to Antigen E.
  • Human common light chain antibodies raised against Antigen E were tested for their ability to block Antigen E binding to a Ligand Y-coated surface in an ELISA assay.
  • Ligand Y was coated onto 96-well plates at a concentration of 2 pg/mL diluted in PBS and incubated overnight followed by washing four times in PBS with 0.05% Tween-20. The plate was then blocked with PBS (Irvine Scientific, Santa Ana, Calif.) containing 0.5% (w/v) BSA (Sigma-Aldrich Corp., St. Louis, Mo.) for one hour at room temperature. In a separate plate, supernatants containing anti-Antigen E common light chain antibodies were diluted 1:10 in buffer. A mock supernatant with the same components of the antibodies was used as a negative control. Antigen E-mmH (described above) was added to a final concentration of 0.150 nM and incubated for one hour at room temperature.
  • PBS Irvine Scientific, Santa Ana, Calif.
  • BSA Sigma-Aldrich Corp., St. Louis, Mo.
  • the antibody/Antigen E-mmH mixture was then added to the plate containing Ligand Y and incubated for one hour at room temperature. Detection of Antigen E-mmH bound to Ligand Y was determined with Horse-Radish Peroxidase (HRP) conjugated to anti-Penta-His antibody (Qiagen, Valencia, Calif.) and developed by standard colorimetric response using tetramethylbenzidine (TMB) substrate (BD Biosciences, San Jose, Calif.) neutralized by sulfuric acid. Absorbance was read at OD450 for 0.1 sec. Background absorbance of a sample without Antigen E was subtracted from all samples. Percent blocking was calculated by division of the background-subtracted MFI of each sample by the adjusted negative control value, multiplying by 100 and subtracting the resulting value from 100.
  • HRP Horse-Radish Peroxidase
  • TMB tetramethylbenzidine
  • Tables 9 and 10 show the percent blocking for all 98 anti-Antigen E common light chain antibodies tested in the ELISA assay. ND: not determined under current experimental conditions.
  • K D Equilibrium dissociation constants for selected antibody supernatants were determined by SPR (Surface Plasmon Resonance) using a BIACORETM T100 instrument (GE Healthcare). All data was obtained using HBS-EP (10mM Hepes, 150 mM NaCl, 0.3 mM EDTA, 0.05% Surfactant P20, pH 7.4) as both the running and sample buffers, at 25° C. Antibodies were captured from crude supernatant samples on a CM5 sensor chip surface previously derivatized with a high density of anti-human Fc antibodies using standard amine coupling chemistry.
  • the binding affinities of common light chain antibodies comprising the rearrangements shown in Tables 5 and 6 vary, with nearly all exhibiting a K D in the nanomolar range.
  • the affinity data is consistent with the common light chain antibodies resulting from the combinatorial association of rearranged variable domains described in Tables 5 and 6 being high-affinity, clonally selected, and somatically mutated.
  • the common light chain antibodies described in Tables 5 and 6 comprise a collection of diverse, high-affinity antibodies that exhibit specificity for one or more epitopes on Antigen E.
  • Selected anti-Antigen E common light chain antibodies were tested for their ability to bind to the ECD of Antigen E and Antigen E ECD variants, including the cynomolgous monkey ortholog (Mf Antigen E), which differs from the human protein in approximately 10% of its amino acid residues; a deletion mutant of Antigen E lacking the last 10 amino acids from the C-terminal end of the ECD (Antigen E-ACT); and two mutants containing an alanine substitution at suspected locations of interaction with Ligand Y (Antigen E-Ala1 and AntigenE-Ala2).
  • the Antigen E proteins were produced in CHO cells and each contained a myc-myc-His C-terminal tag.
  • Antigen E ECD protein or variant protein (described above) from 1 mL of culture medium was captured by incubation for 2 hr at room temperature with 1 ⁇ 10 6 microsphere (LUMINEXTM) beads covalently coated with an anti-myc monoclonal antibody (MAb 9E10, hybridoma cell line CRL-1729TM; ATCC, Manassas, Va.). The beads were then washed with PBS before use. Supernatants containing anti-Antigen E common light chain antibodies were diluted 1:4 in buffer and added to 96-well filter plates. A mock supernatant with no antibody was used as negative control.
  • the beads containing the captured Antigen E proteins were then added to the antibody samples (3000 beads per well) and incubated overnight at 4° C. The following day, the sample beads were washed and the bound common light chain antibody was detected with a R-phycoerythrin-conjugated anti-human IgG antibody.
  • the fluorescence intensity of the beads (approximately 100 beads counted for each antibody sample binding to each Antigen E protein) was measured with a LUMINEXTM flow cytometry-based analyzer, and the median fluorescence intensity (MFI) for at least 100 counted beads per bead/antibody interaction was recorded. Results are shown in Tables 13 and 14.
  • the anti-Antigen E common light chain antibody supernatants exhibited high specific binding to the beads linked to Antigen E-ECD.
  • the negative control mock supernatant resulted in negligible signal ( ⁇ 10 MFI) when combined with the Antigen E-ECD bead sample, whereas the supernatants containing anti-Antigen E common light chain antibodies exhibited strong binding signal (average MFI of 2627 for 98 antibody supernatants; MFI>500 for 91/98 antibody samples).
  • the relative binding of the antibodies to the variants were determined. All four Antigen E variants were captured to the anti-myc LUMINEXTM beads as described above for the native Antigen E-ECD binding studies, and the relative binding ratios (MFI variant /MFI Antigen E-ECD ) were determined.
  • Heavy chains of selected antigen-specific common light chain antibodies were tested for binding to Antigen E after repairing the heavy chains with either a germline V ⁇ 1-39J ⁇ 5 or a germline V ⁇ 3-20J ⁇ 1 engineered light chain (as described in Example 1).
  • V ⁇ 1-39J ⁇ 5 and V ⁇ 3-20J ⁇ 1 247 heavy chains of Antigen E-specific common light chain antibodies (V ⁇ 1-39J ⁇ 5 and V ⁇ 3-20J ⁇ 1) were transfected with either a germline V ⁇ 1-39 or a germline V ⁇ 3-20 engineered light chain and rescreened for binding to Antigen E by a LUMINEXTM assay (as described in Example 7 and Example 10). Binding to Antigen E was confirmed by BIACORETM (as described in Example 9). The results are shown in Table 15.
  • Heavy and light chain sequences (>6000) of antibodies raised in VELCOIMMUNE® mice (e.g., U.S. Pat. No. 6,596,541 and U.S. Pat. No. 7,105,348) were compiled with heavy and light chain sequences (>600) of common light chain antibodies obtained by a multi-antigen immunization scheme employing the engineered light chain mice (described above) to compare heavy chain gene segment usage and somatic hypermutation frequencies of the antibody chains.
  • Heavy Chain Gene Usage Heavy and light chain sequences obtained from VELOCIMMUNE® mice containing a replacement of the endogenous mouse heavy chain locus with human V H , D H , and J H gene segments and a replacement of the endogenous mouse ⁇ light chain locus with either the engineered germline V ⁇ 1-39J ⁇ 5 human light chain region or the engineered germline V ⁇ 3-20J ⁇ 1 human light chain region (as described in Example 2) immunized with a human cell-surface receptor (Antigen E), a heterodimer of two human cell-surface glycoproteins (Antigen F), a human cytokine receptor (Antigen G) and a human tumor differentiation antigen (Antigen H) were analyzed for heavy chain gene segment usage and V H and J H gene segments were recorded. Results are shown in Tables 16-18. Percentages in Tables 16-18 represent rounded values and in some cases may not equal 100% when added together.
  • Table 16 sets forth the percent heavy chain family usage for antibodies from VELCOIMMUNE® mice (VI), antibodies from VELCOIMMUNE® mice having a cognate W1-39 light chain (VI-V ⁇ 1-39), antibodies from V ⁇ 1-39 engineered light chain mice (V ⁇ 1-39), antibodies from VELCOIMMUNE® mice having a cognate V ⁇ 3-20 light chain (VI-V ⁇ 3-20), and antibodies from V ⁇ 3-20 engineered light chain mice (V ⁇ 3-20).
  • Table 17 sets forth the percent V H and J H gene usage for antibodies from VELCOIMMUNE® mice (VI), antibodies from VELCOIMMUNE® mice having a cognate V ⁇ 1-39 light chain (VI-V ⁇ 1-39), antibodies from V ⁇ 1-39 engineered light chain mice (V ⁇ 1-39), antibodies from VELCOIMMUNE® mice having a cognate V ⁇ 3-20 light chain (VI-V ⁇ 3-20), and antibodies from V ⁇ 3-20 engineered light chain mice (V ⁇ 3-20).
  • Table 18 sets forth the percent V H gene usage for antibodies from V ⁇ 1-39 engineered light chain mice (V ⁇ 1-39 Mice) from each immunization group (Antigens E, F, G and H) and the percent V H gene usage for antibodies from V ⁇ 3-20 engineered light chain mice (V ⁇ 3-20 Mice) from selected immunization groups (Antigens E and G).
  • V H family III subgroups V H 3-7, V H 3-9, V H 3-11, V H 3-13, V H 3-20, V H 3-23, V H 3-30, V H 3-33 and V H 3-48.
  • Notable usage of other V H family subgroups was characterized by usage of V H 1-18, V H 1-69, V H 2-5, V H 4-59 and V H 6-1.
  • V H family III For antigens tested in V ⁇ 3-20J ⁇ 1 engineered light chain mice, heavy chain gene usage was characterized by a preponderance of V H family III, V H family IV and V H family V subgroups (V H 3-11, V H 3-30, V H 3-33, V H 4-39, V H 4-59 and V H 5-51). Notable usage of other V H family subgroups was characterized by usage of V H 1-18, V H 1-69, V H 2-70 and V H 6-1.
  • Somatic Hypermutation Frequency Heavy and light chains from antibodies raised in VELCOIMMUNE® mice and the engineered light chain mice (described above) were aligned to germline sequences according to the heavy and light chain gene usage demonstrated for each heavy and/or light chain. Amino acid changes for each framework region (FW) and complementarity determining region (CDR) for both heavy and light chain of each sequence were calculated. Results are shown in Tables 19-22. Percentages in Tables 21-24 represent rounded values and in some cases may not equal 100% when added together.
  • Table 19 sets forth the number of amino acid (AA) changes observed in each FW and CDR region of heavy chains of antibodies from VELCOIMMUNE® mice, heavy chains of antibodies from V ⁇ 1-39 engineered light chain mice (V ⁇ 1-39 Mice) and heavy chains of antibodies from V ⁇ 3-20 engineered light chain mice (V ⁇ 3-20 Mice).
  • Table 20 sets forth the number of amino acid (AA) changes observed in each FW and CDR region of light chains of antibodies from VELCOIMMUNE® mice, the light chain of antibodies from V ⁇ 1-39 engineered mice (V ⁇ 1-39 Mice) and the light chain of antibodies from V ⁇ 3-20 engineered mice (V ⁇ 3-20 Mice).
  • Table 21 sets forth the number of amino acid (AA) changes observed in each FW and CDR region of heavy chains of antibodies from V ⁇ 1-39 engineered light chain mice (V ⁇ 1-39 Mice) for selected immunization groups (Antigens E, F and H).
  • Table 22 sets forth the number of amino acid (AA) changes observed in each FW and CDR region of heavy chains of antibodies from V ⁇ 3-20 engineered light chain mice (V ⁇ 3-20 Mice) for selected immunization groups (Antigens E and G).
  • VI-V ⁇ 1-39 V ⁇ 1-39 VI-V ⁇ 3-20 V ⁇ 3-20 1 9.0 14.8 3.3 7.1 4.9 2 2.2 1.8 4.6 0 1.6 3 77.8 69.8 77.3 61.4 29.5 4 8.4 8.3 11.2 27.1 39.3 5 0.9 0 0.7 4.3 23.0 6 1.7 5.3 3.0 0 1.6

Abstract

A genetically modified mouse is provided, wherein the mouse expresses an immunoglobulin light chain repertoire characterized by a limited number of light chain variable domains. Mice are provided that express just one or a few immunoglobulin light chain variable domains from a limited repertoire in their germline. Methods for making light chain variable regions in mice, including human light chain variable regions, are provided. Methods for making human variable regions suitable for use in multispecific binding proteins, e.g., bispecific antibodies, are provided.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application is a continuation-in-part of U.S. Ser. No. 13/093,156 filed Apr. 25, 2011, which is a continuation-in-part of U.S. Ser. No. 13/022,759 filed Feb. 8, 2011, which is a nonprovisional application of U.S. Provisional Application Ser. No. 61/302,282, filed Feb. 8, 2010; which applications are hereby incorporated by reference in their entirety.
  • FIELD
  • A genetically modified mouse is provided that expresses antibodies having a common human variable/mouse constant light chain associated with diverse human variable/mouse constant heavy chains. A method for making a human bispecific antibody from human variable region gene sequences of B cells of the mouse is provided.
  • BACKGROUND
  • Antibodies typically comprise a homodimeric heavy chain component, wherein each heavy chain monomer is associated with an identical light chain. Antibodies having a heterodimeric heavy chain component (e.g., bispecific antibodies) are desirable as therapeutic antibodies. But making bispecific antibodies having a suitable light chain component that can satisfactorily associate with each of the heavy chains of a bispecific antibody has proved problematic.
  • In one approach, a light chain might be selected by surveying usage statistics for all light chain variable domains, identifying the most frequently employed light chain in human antibodies, and pairing that light chain in vitro with the two heavy chains of differing specificity.
  • In another approach, a light chain might be selected by observing light chain sequences in a phage display library (e.g., a phage display library comprising human light chain variable region sequences, e.g., a human scFv library) and selecting the most commonly used light chain variable region from the library. The light chain can then be tested on the two different heavy chains of interest.
  • In another approach, a light chain might be selected by assaying a phage display library of light chain variable sequences using the heavy chain variable sequences of both heavy chains of interest as probes. A light chain that associates with both heavy chain variable sequences might be selected as a light chain for the heavy chains.
  • In another approach, a candidate light chain might be aligned with the heavy chains' cognate light chains, and modifications are made in the light chain to more closely match sequence characteristics common to the cognate light chains of both heavy chains. If the chances of immunogenicity need to be minimized, the modifications preferably result in sequences that are present in known human light chain sequences, such that proteolytic processing is unlikely to generate a T cell epitope based on parameters and methods known in the art for assessing the likelihood of immunogenicity (i.e., in silico as well as wet assays).
  • All of the above approaches rely on in vitro methods that subsume a number of a priori restraints, e.g., sequence identity, ability to associate with specific pre-selected heavy chains, etc. There is a need in the art for compositions and methods that do not rely on manipulating in vitro conditions, but that instead employ more biologically sensible approaches to making human epitope-binding proteins that include a common light chain.
  • SUMMARY
  • Genetically modified mice that express human immunoglobulin heavy and light chain variable domains, wherein the mice have a limited light chain variable repertoire, are provided. A biological system for generating a human light chain variable domain that associates and expresses with a diverse repertoire of affinity-matured human heavy chain variable domains is provided. Methods for making binding proteins comprising immunoglobulin variable domains are provided, comprising immunizing mice that have a limited immunoglobulin light chain repertoire with an antigen of interest, and employing an immunoglobulin variable region gene sequence of the mouse in a binding protein that specifically binds the antigen of interest. Methods include methods for making human immunoglobulin heavy chain variable domains suitable for use in making multi-specific antigen-binding proteins.
  • Genetically engineered mice are provided that select suitable affinity-matured human immunoglobulin heavy chain variable domains derived from a repertoire of unrearranged human heavy chain variable region gene segments, wherein the affinity-matured human heavy chain variable domains associate and express with a single human light chain variable domain derived from one human light chain variable region gene segment. Genetically engineered mice that present a choice of two human light chain variable region gene segments are also provided.
  • Genetically engineered mice are provided that express a limited repertoire of human light chain variable domains, or a single human light chain variable domain, from a limited repertoire of human light chain variable region gene segments. The mice are genetically engineered to include a single unrearranged human light chain variable region gene segment (or two human light chain variable region gene segments) that rearranges to form a rearranged human light chain variable region gene (or two rearranged light chain variable region genes) that express a single light chain (or that express either or both of two light chains). The rearranged human light chain variable domains are capable of pairing with a plurality of affinity-matured human heavy chains selected by the mice, wherein the heavy chain variable regions specifically bind different epitopes.
  • Genetically engineered mice are provided that express a limited repertoire of human light chain variable domains, or a single human light chain variable domain, from a limited repertoire of human light chain variable region sequences. The mice are genetically engineered to include a single V/J human light chain sequence (or two V/J sequences) that express a variable region of a single light chain (or that express either or both of two variable regions). A light chain comprising the variable sequence is capable of pairing with a plurality of affinity-matured human heavy chains clonally selected by the mice, wherein the heavy chain variable regions specifically bind different epitopes.
  • In one aspect, a genetically modified mouse is provided that comprises a single human immunoglobulin light chain variable (VL) region gene segment that is capable of rearranging with a human J gene segment (selected from one or a plurality of JL segments) and encoding a human VL domain of an immunoglobulin light chain. In another aspect, the mouse comprises no more than two human VL gene segments, each of which is capable of rearranging with a human J gene segment (selected from one or a plurality of JL segments) and encoding a human VL domain of an immunoglobulin light chain.
  • In one embodiment, the single human VL gene segment is operably linked to a human JL gene segment selected from Jκ1, Jκ2, Jκ3, Jκ4, and Jκ5, wherein the single human VL gene segment is capable of rearranging to form a sequence encoding a light chain variable region gene with any of the one or more human JL gene segments.
  • In one embodiment, the genetically modified mouse comprises an immunoglobulin light chain locus that does not comprise an endogenous mouse VL gene segment that is capable of rearranging to form an immunoglobulin light chain gene, wherein the VL locus comprises a single human VL gene segment that is capable of rearranging to encode a VL region of a light chain gene. In a specific embodiment, the human VL gene segment is a human Vκ1-39Jκ5 gene segment or a human Vκ3-20Jκ1 gene segment. In one embodiment, the genetically modified mouse comprises a VL locus that does not comprise an endogenous mouse VL gene segment that is capable of rearranging to form an immunoglobulin light chain gene, wherein the VL locus comprises no more than two human VL gene segments that are capable of rearranging to encode a VL region of a light chain gene. In a specific embodiment, the no more than 2 human VL gene segments are a human Vκ1-39Jκ5 gene segment and a human Vκ3-20Jκ1 gene segment.
  • In one aspect, a genetically modified mouse is provided that comprises a single rearranged (V/J) human immunoglobulin light chain variable (VL) region (i.e., a VL/JL region) that encodes a human VL domain of an immunoglobulin light chain. In another aspect, the mouse comprises no more than two rearranged human VL regions that are capable of encoding a human VL domain of an immunoglobulin light chain.
  • In one embodiment, the VL region is a rearranged human Vκ1-39/J sequence or a rearranged human Vκ3-20/J sequence. In one embodiment, the human JL segment of the rearranged VL/JL sequence is selected from Jκ1, Jκ2, Jκ3, Jκ4, and Jκ5. In a specific embodiment, the VL region is a human Vκ1-39Jκ5 sequence or a human Vκ3-20JκK1 sequence. In a specific embodiment, the mouse has both a human Vκ1-39Jκ5 sequence and a human Vκ3-20Jκ1 sequence.
  • In one embodiment, the human VL gene segment is operably linked to a human or mouse leader sequence. In one embodiment, the leader sequence is a mouse leader sequence. In a specific embodiment, the mouse leader sequence is a mouse Vκ3-7 leader sequence. In a specific embodiment, the leader sequence is operably linked to an unrearranged human VL gene segment. In a specific embodiment, the leader sequence is operably linked to a rearranged human VL/JL sequence.
  • In one embodiment, the VL gene segment is operably linked to an immunoglobulin promoter sequence. In one embodiment, the promoter sequence is a human promoter sequence. In a specific embodiment, the human immunoglobulin promoter is a human Vκ3-15 promoter. In a specific embodiment, the promoter is operably linked to an unrearranged human VL gene segment. In a specific embodiment, the promoter is operably linked to a rearranged human VL/JL sequence.
  • In one embodiment, the light chain locus comprises a leader sequence flanked 5′ (with respect to transcriptional direction of a VL gene segment) with a human immunoglobulin promoter and flanked 3′ with a human VL gene segment that rearranges with a human J segment and encodes a VL domain of a reverse chimeric light chain comprising an endogenous mouse light chain constant region (CL). In a specific embodiment, the VL gene segment is at the mouse Vκ locus, and the mouse CL is a mouse Cκ.
  • In one embodiment, the light chain locus comprises a leader sequence flanked 5′ (with respect to transcriptional direction of a VL gene segment) with a human immunoglobulin promoter and flanked 3′ with a rearranged human VL region (VL/JL sequence) and encodes a VL domain of a reverse chimeric light chain comprising an endogenous mouse light chain constant region (CL). In a specific embodiment, the rearranged human VL/JL sequence is at the mouse kappa (κ) locus, and the mouse CL is a mouse Cκ.
  • In one embodiment, the VL locus of the modified mouse is a κ light chain locus, and the κ light chain locus comprises a mouse κ intronic enhancer, a mouse κ3′ enhancer, or both an intronic enhancer and a 3′ enhancer.
  • In one embodiment, the mouse comprises a nonfunctional immunoglobulin lambda (λ) light chain locus. In a specific embodiment, the λ light chain locus comprises a deletion of one or more sequences of the locus, wherein the one or more deletions renders the λ light chain locus incapable of rearranging to form a light chain gene. In another embodiment, all or substantially all of the VL gene segments of the λ light chain locus are deleted.
  • In one embodiment, mouse makes a light chain that comprises a somatically mutated VL domain derived from a human VL gene segment. In one embodiment, the light chain comprises a somatically mutated VL domain derived from a human VL gene segment, and a mouse Cκ region. In one embodiment, the mouse does not express a λ light chain.
  • In one embodiment, the genetically modified mouse is capable of somatically hypermutating the human VL region sequence. In a specific embodiment, the mouse comprises a cell that comprises a rearranged immunoglobulin light chain gene derived from a human VL gene segment that is capable of rearranging and encoding a VL domain, and the rearranged immunoglobulin light chain gene comprises a somatically mutated VL domain.
  • In one embodiment, the mouse comprises a cell that expresses a light chain comprising a somatically mutated human VL domain linked to a mouse CK, wherein the light chain associates with a heavy chain comprising a somatically mutated VH domain derived from a human VH gene segment and wherein the heavy chain comprises a mouse heavy chain constant region (CH). In a specific embodiment, the heavy chain comprises a mouse C H1, a mouse hinge, a mouse CH2, and a mouse CH3. In a specific embodiment, the heavy chain comprises a human C H1, a hinge, a mouse CH2, and a mouse CH3.
  • In one embodiment, the mouse comprises a replacement of endogenous mouse VH gene segments with one or more human VH gene segments, wherein the human VH gene segments are operably linked to a mouse CH region gene, such that the mouse rearranges the human VH gene segments and expresses a reverse chimeric immunoglobulin heavy chain that comprises a human VH domain and a mouse CH. In one embodiment, 90-100% of unrearranged mouse VH gene segments are replaced with at least one unrearranged human VH gene segment. In a specific embodiment, all or substantially all of the endogenous mouse VH gene segments are replaced with at least one unrearranged human VH gene segment. In one embodiment, the replacement is with at least 19, at least 39, or at least 80 or 81 unrearranged human VH gene segments. In one embodiment, the replacement is with at least 12 functional unrearranged human VH gene segments, at least 25 functional unrearranged human VH gene segments, or at least 43 functional unrearranged human VH gene segments. In one embodiment, the mouse comprises a replacement of all mouse DH and JH segments with at least one unrearranged human DH segment and at least one unrearranged human JH segment. In one embodiment, the at least one unrearranged human DH segment is selected from 1-1, D1-7, 1-26, 2-8, 2-15, 3-3, 3-10, 3-16, 3-22, 5-5, 5-12, 6-6, 6-13, 7-27, and a combination thereof. In one embodiment, the at least one unrearranged human JH segment is selected from 1, 2, 3, 4, 5, 6, and a combination thereof. In a specific embodiment, the one or more human VH gene segment is selected from a 1-2, 1-8, 1-24, 1-69, 2-5, 3-7, 3-9, 3-11, 3-13, 3-15, 3-20, 3-23, 3-30, 3-33, 3-48, 3-53, 4-31, 4-39, 4-59, 5-51, a 6-1 human VH gene segment, and a combination thereof.
  • In one embodiment, the mouse comprises a B cell that expresses a binding protein that specifically binds an antigen of interest, wherein the binding protein comprises a light chain derived from a human Vκ1-39/Jκ5 rearrangement or a human Vκ3-20/Jκ1 rearrangement, and wherein the cell comprises a rearranged immunoglobulin heavy chain gene derived from a rearrangement of human VH gene segments selected from a 1-69, 2-5, 3-13, 3-23, 3-30, 3-33, 3-53, 4-39, 4-59, and 5-51 gene segment. In one embodiment, the one or more human VH gene segments are rearranged with a human heavy chain JH gene segment selected from 1, 2, 3, 4, 5, and 6. In one embodiment, the one or more human VH and JH gene segments are rearranged with a human DH gene segment selected from 1-1, 1-7, 1-26, 2-8, 2-15, 3-3, 3-10, 3-16, 3-22, 5-5, 5-12, 6-6, 6-13, and 7-27. In a specific embodiment, the light chain gene has 1, 2, 3, 4, or 5 or more somatic hypermutations.
  • In one embodiment, the mouse comprises a B cell that comprises a rearranged immunoglobulin heavy chain variable region gene sequence comprising a VH/DH/JH region selected from 2-5/6-6/1, 2-5/3-22/1, 3-13/6-6/5, 3-23/2-8/4, 3-23/3-3/4, 3-23/3-10/4, 3-23/6-6/4, 3-23/7-27/4, 3-30/1-1/4, 3-30/1-7/4, 3-30/3-3/3, 3-30/3-3/4, 3-30/3-22/5, 3-30/5-5/2, 3-30/5-12/4, 3-30/6-6/1, 3-30/6-6/3, 3-30/6-6/4, 3-30/6-6/5, 3-30/6-13/4, 3-30/7-27/4, 3-30/7-27/5, 3-30/7-27/6, 3-33/1-7/4, 3-33/2-15/4, 4-39/1-26/3, 4-59/3-16/3, 4-59/3-16/4, 4-59/3-22/3, 5-51/3-16/6, 5-51/5-5/3, 5-51/6-13/5, 3-53/1-1/4, 1-69/6-6/5, and 1-69/6-13/4. In a specific embodiment, the B cell expresses a binding protein comprising a human immunoglobulin heavy chain variable region fused with a mouse heavy chain constant region, and a human immunoglobulin light chain variable region fused with a mouse light chain constant region.
  • In one embodiment, the rearranged human VL region is a human Vκ1-39Jκ5 sequence, and the mouse expresses a reverse chimeric light chain comprising (i) a VL domain derived from the human VL/JL sequence and (ii) a mouse CL; wherein the light chain is associated with a reverse chimeric heavy chain comprising (i) a mouse CH and (ii) a somatically mutated human VH domain derived from a human VH gene segment selected from a 1-2, 1-8, 1-24, 1-69, 2-5, 3-7, 3-9, 3-11, 3-13, 3-15, 3-20, 3-23, 3-30, 3-33, 3-48, 3-53, 4-31, 4-39, 4-59, 5-51, a 6-1 human VH gene segment, and a combination thereof. In one embodiment, the mouse expresses a light chain that is somatically mutated. In one embodiment the CL is a mouse Cκ. In a specific embodiment, the human VH gene segment is selected from a 2-5, 3-13, 3-23, 3-30, 4-59, 5-51, and 1-69 gene segment. In a specific embodiment, the somatically mutated human VH domain comprises a sequence derived from a DH segment selected from 1-1, 1-7, 2-8, 3-3, 3-10, 3-16, 3-22, 5-5, 5-12, 6-6, 6-13, and 7-27. In a specific embodiment, the somatically mutated human VH domain comprises a sequence derived from a JH segment selected from 1, 2, 3, 4, 5, and 6. In a specific embodiment, the somatically mutated human VH domain is encoded by a rearranged human VH/DH/JH sequence selected from 2-5/6-6/1, 2-5/3-22/1, 3-13/6-6/5, 3-23/2-8/4, 3-23/3-3/4, 3-23/3-10/4, 3-23/6-6/4, 3-23/7-27/4, 3-30/1-1/4, 3-30/1-7/4, 3-30/3-3/4, 3-30/3-22/5, 3-30/5-5/2, 3-30/5-12/4, 3-30/6-6/1, 3-30/6-6/3, 3-30/6-6/4, 3-30/6-6/5, 3-30/6-13/4, 3-30/7-27/4, 3-30/7-27/5, 3-30/7-27/6, 4-59/3-16/3, 4-59/3-16/4, 4-59/3-22/3, 5-51/5-5/3, 1-69/6-6/5, and 1-69/6-13/4.
  • In one embodiment, the rearranged human VL region is a human Vκ3-20Jκ1 sequence, and the mouse expresses a reverse chimeric light chain comprising (i) a VL domain derived from the rearranged human VL/JL sequence, and (ii) a mouse CL; wherein the light chain is associated with a reverse chimeric heavy chain comprising (i) a mouse CH, and (ii) a somatically mutated human VH derived from a human VH gene segment selected from a 1-2, 1-8, 1-24, 1-69, 2-5, 3-7, 3-9, 3-11, 3-13, 3-15, 3-20, 3-23, 3-30, 3-33, 3-48, 3-53, 4-31, 4-39, 4-59, 5-51, a 6-1 human VH gene segment, and a combination thereof. In one embodiment, the mouse expresses a light chain that is somatically mutated. In one embodiment the CL is a mouse Cκ. In a specific embodiment, the human VH gene segment is selected from a 3-30, 3-33, 3-53, 4-39, and 5-51 gene segment. In a specific embodiment, the somatically mutated human VH domain comprises a sequence derived from a DH segment selected from 1-1, 1-7, 1-26, 2-15, 3-3, 3-16, and 6-13. In a specific embodiment, the somatically mutated human VH domain comprises a sequence derived from a JH segment selected from 3, 4, 5, and 6. In a specific embodiment, the somatically mutated human VH domain is encoded by a rearranged human VH/DH/JH sequence selected from 3-30/1-1/4, 3-30/3-3/3, 3-33/1-7/4, 3-33/2-15/4, 4-39/1-26/3, 5-51/3-16/6, 5-51/6-13/5, and 3-53/1-1/4.
  • In one embodiment, the mouse comprises both a rearranged human Vκ1-39Jκ5 sequence and a rearranged human Vκ3-20Jκ1 sequence, and the mouse expresses a reverse chimeric light chain comprising (i) a VL domain derived from the human Vκ1-39Jκ5 sequence or the human Vκ3-20Jκ1 sequence, and (ii) a mouse CL; wherein the light chain is associated with a reverse chimeric heavy chain comprising (i) a mouse CH, and (ii) a somatically mutated human VH derived from a human VH gene segment selected from a 1-2, 1-8, 1-24, 1-69, 2-5, 3-7, 3-9, 3-11, 3-13, 3-15, 3-20, 3-23, 3-30, 3-33, 3-48, 3-53, 4-31, 4-39, 4-59, 5-51, a 6-1 human VH gene segment, and a combination thereof. In one embodiment, the mouse expresses a light chain that is somatically mutated. In one embodiment the CL is a mouse Cκ.
  • In one embodiment, 90-100% of the endogenous unrearranged mouse VH gene segments are replaced with at least one unrearranged human VH gene segment. In a specific embodiment, all or substantially all of the endogenous unrearranged mouse VH gene segments are replaced with at least one unrearranged human VH gene segment. In one embodiment, the replacement is with at least 18, at least 39, at least 80, or 81 unrearranged human VH gene segments. In one embodiment, the replacement is with at least 12 functional unrearranged human VH gene segments, at least 25 functional unrearranged human VH gene segments, or at least 43 unrearranged human VH gene segments.
  • In one embodiment, the genetically modified mouse is a C57BL strain, in a specific embodiment selected from C57BL/A, C57BL/An, C57BL/GrFa, C57BL/KaLwN, C57BL/6, C57BL/6J, C57BL/6ByJ, C57BL/6NJ, C57BL/10, C57BL/10ScSn, C57BL/10Cr, and C57BL/Ola. In a specific embodiment, the genetically modified mouse is a mix of an aforementioned 129 strain and an aforementioned C57BL/6 strain. In another specific embodiment, the mouse is a mix of aforementioned 129 strains, or a mix of aforementioned BL/6 strains. In a specific embodiment, the 129 strain of the mix is a 129S6 (129/SvEvTac) strain.
  • In one embodiment, the mouse expresses a reverse chimeric antibody comprising a light chain that comprises a mouse Cκ and a somatically mutated human VL domain derived from a rearranged human Vκ1-39Jκ5 sequence or a rearranged human Vκ3-20Jκ1 sequence, and a heavy chain that comprises a mouse CH and a somatically mutated human VH domain derived from a human VH gene segment selected from a 1-2, 1-8, 1-24, 1-69, 2-5, 3-7, 3-9, 3-11, 3-13, 3-15, 3-20, 3-23, 3-30, 3-33, 3-48, 3-53, 4-31, 4-39, 4-59, 5-51, and a 6-1 human VH gene segment, wherein the mouse does not express a fully mouse antibody and does not express a fully human antibody. In one embodiment the mouse comprises a κ light chain locus that comprises a replacement of endogenous mouse κ light chain gene segments with the rearranged human Vκ1-39Jκ5 sequence or the rearranged human Vκ3-20Jκ1 sequence, and comprises a replacement of all or substantially all endogenous mouse VH gene segments with a complete or substantially complete repertoire of human VH gene segments.
  • In one aspect, a population of antigen-specific antibodies derived from a mouse as described herein is provided, wherein the antibodies comprise a light chain gene derived from a human Vκ1-39/Jκ5 rearrangement or a human Vκ3-20/Jκ1 rearrangement, and wherein the antibodies comprise a rearranged immunoglobulin heavy chain gene derived from a rearrangement of a human VH gene segment selected from a 1-2, 1-3, 1-8, 1-18, 1-24, 1-46, 1-58, 1-69, 2-5, 2-26, 2-70, 3-7, 3-9, 3-11, 3-13, 3-15, 3-16, 3-20, 3-21, 3-23, 3-30, 3-33, 3-43, 3-48, 3-53, 3-64, 3-72, 3-73, 4-31, 4-34, 4-39, 4-59, 5-51, and a 6-1 human VH gene segment. In one embodiment, the one or more human VH gene segments are rearranged with a human heavy chain JH gene segment selected from 1, 2, 3, 4, 5, and 6. In a specific embodiment, the light chain has 1, 2, 3, 4, or 5 or more somatic hypermutations.
  • In one embodiment, the light chain has 1, 2, 3, or 4 somatic hypermutations. In one embodiment, the light chain gene has 1 or 2 mutations. In various embodiments, the light chain gene is capable of incurring multiple mutations along its sequence.
  • In one embodiment, the light chain is derived from a human Vκ1-39/Jκ5 rearrangement and the light chain has at least one or no more than four somatic hypermutations. In one embodiment, the light chain comprises at least two somatic hypermutations. In one embodiment, the light chain comprises at least three somatic hypermutations. In one embodiment, the light chain comprises at least four somatic hypermutations. In a specific embodiment, the mutations are present in one or more framework regions (FWs) of the light chain. In a specific embodiment, the mutations are present in one or more complementarity determining regions (CDRs) of the light chain. In a specific embodiment, the mutations are present in one or more FWs and/or one or more CDRs of the light chain. In various embodiments, the framework regions are selected from framework 1 (FW1), framework 2 (FW2), framework 3 (FW3), and/or a combination thereof. In various embodiments, the CDRs are selected from CDR1, CDR2, CDR3, and/or a combination thereof.
  • In one embodiment, the heavy chain comprises at least one mutation in one or more FWs or one or more CDRs. In one embodiment, the heavy chain comprises at least one mutation in one or more FWs and one or more CDRs. In one embodiment, the heavy chain comprises at least two mutations in one or more FWs and one or more CDRs. In one embodiment, the heavy chain comprises at least three mutations in one or more FWs and one or more CDRs. In one embodiment, the heavy chain comprises at least four mutations in one or more FWs and one or more CDRs. In one embodiment, the heavy chain comprises at least five or more than five mutations in one or more FWs and one or more CDRs; in a specific embodiment, the heavy chain comprises at least five or more than five mutations in two FWs; in a specific embodiment, the heavy chain comprises at least five or more than five mutations in one FW and one CDR.
  • In one embodiment, the light chain is derived from a human Vκ1-39/Jκ5 rearrangement and about 9% of the Vκ1-39/Jκ5-derived light chains have at least one mutation present in FW1; in one embodiment, at least 9% of the light chains comprise one mutation present in FW1. In one embodiment, the light chain is derived from a human Vκ1-39/Jκ5 rearrangement and about 25% of the Vκ1-39/Jκ5-derived light chains have at least one or no more than two mutations present in CDR1; in one embodiment, at least 19% of the light chains have one mutation present in CDR1; in one embodiment, at least 5% of the light chains have two mutations present in CDR1.
  • In one embodiment, the light chain is derived from a human Vκ1-39/Jκ5 rearrangement and about 20% of the Vκ1-39/Jκ5-derived light chains have at least one or no more than three mutations present in FW2; in one embodiment, at least 17% of the light chains have one mutation present in FW2; in one embodiment, at least 1% of the light chains have two mutations present in FW2; in one embodiment, at least 1% of the light chains have three mutations present in FW2.
  • In one embodiment, the light chain is derived from a human Vκ1-39/Jκ5 rearrangement and about 10% of the Vκ1-39/Jκ5-derived light chains have at least one or no more than two mutations present in CDR2; in one embodiment, at least 10% of the light chains have one mutation present in CDR2; in one embodiment, at least 1% of the light chains have two mutations present in CDR2.
  • In one embodiment, the light chain is derived from a human Vκ1-39/Jκ5 rearrangement and about 29% of the Vκ1-39/Jκ5-derived light chains have at least one or no more than four mutations present in FW3; in one embodiment, at least 21% of the light chains have one mutation present in FW3; in one embodiment, at least 5% of the light chains have two mutations present in FW3; in one embodiment, at least 2% of the light chains have three mutations present in FW3; in one embodiment, at least 2% of the light chains have four mutations present in FW3.
  • In one embodiment, the light chain is derived from a human Vκ1-39/Jκ5 rearrangement and about 37% of the Vκ1-39/Jκ5-derived light chains have at least one or no more than four mutations present in CDR3; in one embodiment, at least 27% of the light chains have one mutation present in CDR3; in one embodiment, at least 8% of the light chains have two mutations present in CDR3; in one embodiment, at least 1% of the light chains have three mutations present in CDR3; in one embodiment, at least 1% of the light chains have four mutations present in CDR3.
  • In one embodiment, a population of antigen-specific antibodies derived from a mouse as described herein is provided, wherein the antibodies comprise a light chain derived from a human Vκ1-39/Jκ5 rearrangement and about 9% of the Vκ1-39/Jκ5-derived light chains have one or more mutations present in FW1, about 25% of the Vκ1-39/Jκ5-derived light chains have one or more mutations present in CDR1, about 20% of the Vκ1-39/Jκ5-derived light chains have one or more mutations present in FW2, about 10% of the Vκ1-39/Jκ5-derived light chains have one or more mutations present in CDR2, about 29% of the Vκ1-39/Jκ5-derived light chains have one or more mutations present in FW3, and about 37% of the Vκ1-39/Jκ5-derived light chains have one or more mutations present in CDR3.
  • In one embodiment, the light chain is derived from a human Vκ1-39/Jκ5 rearrangement and about 35% of the heavy chains have at least one mutation present in FW1; in one embodiment, at least 25% of the heavy chains have one mutation present in FW1; in one embodiment, at least 9% of the heavy chains have two mutations present in FW1; in one embodiment, at least 1% of the heavy chains have three mutations present in FW1; in one embodiment, at least 1% of the heavy chains have more than five mutations present in FW1.
  • In one embodiment, the light chain is derived from a human Vκ1-39Jκ5 rearrangement and about 92% of the heavy chains have at least one or no more than four mutations present in CDR1; in one embodiment, at least 92% of the heavy chains have at least one, at least two, at least three, or at least four mutations present in CDR1; in one embodiment, at least 26% of the heavy chains have one mutation present in CDR1; in one embodiment, at least 44% of the heavy chains have two mutations present in CDR1; in one embodiment, at least 19% of the heavy chains have three mutations present in CDR1; in one embodiment, at least 3% of the heavy chains have four mutations present in CDR1.
  • In one embodiment, the light chain is derived from a human Vκ1-39/Jκ5 rearrangement and about 66% of the heavy chains have at least one or no more than three mutations present in FW2; in one embodiment, at least 66% of the heavy chains have at least one, at least two, or at least three mutations present in FW2; in one embodiment, at least 35% of the heavy chains have one mutation present in FW2; in one embodiment, at least 23% of the heavy chains have two mutations present in FW2; in one embodiment, at least 8% of the heavy chains have three mutations present in FW2.
  • In one embodiment, the light chain is derived from a human Vκ1-39/Jκ5 rearrangement and about 70% of the heavy chains have at least one or no more than four mutations present in CDR2; in one embodiment, at least 70% of the heavy chains have at least two, at least three, or at least four mutations present in CDR2; in one embodiment, at least 34% have one mutation present in CDR2; in one embodiment, at least 20% of the heavy chains have two mutations present in CDR2; in one embodiment, at least 12% of the heavy chains have three mutations present in CDR2; in one embodiment, at least 5% of the heavy chains have four mutations present in CDR2.
  • In one embodiment, the light chain is derived from a human Vκ1-39/Jκ5 rearrangement and about 91% of the heavy chains have at least one or up to five or more mutations present in FW3; in one embodiment, at least 91% of the heavy chains have at least two, at least three, at least four, or at least five or more mutations present in FW3; in one embodiment, at least 19% of the heavy chains have one mutation present in FW3; in one embodiment, at least 33% of the heavy chains have two mutations present in FW3; in one embodiment, at least 22% of the heavy chains have three mutations present in FW3; in one embodiment, at least 11% of the heavy chains have four mutations present in FW3; in one embodiment, at least 7% of the heavy chains have five or more mutations present in FW3.
  • In one embodiment, the light chain is derived from a human Vκ1-39/Jκ5 rearrangement and about 63% of the heavy chains have at least one or no more than two mutations present in CDR3; in one embodiment, at least 63% of the heavy chains have at one mutation present in CDR3; in one embodiment, at least 54% of the heavy chains have one mutation present in CDR3; in one embodiment, at least 9% of the heavy chains have two mutations present in CDR3.
  • In one embodiment, a population of antigen-specific antibodies derived from a mouse as described herein is provided, wherein the antibodies comprise a light chain derived from a human Vκ1-39/Jκ5 rearrangement and at least 35% of the heavy chains have one or more mutations present in FW1, about 92% of the heavy chains have one or more mutations present in CDR1, about 66% of the heavy chains have one or more mutations present in FW2, about 70% of the heavy chains have one or more mutations present in CDR2, about 91% of the heavy chains have one or more mutations present in FW3, and about 63% of the heavy chains have one or more mutations present in CDR3.
  • In one embodiment, the light chain is derived from a human Vκ3-20/Jκ1 rearrangement and the light chain gene has at least one or no more than two somatic hypermutations; in one embodiment, the light chain gene has at least two, at least three, at least four or more somatic hypermutations. In a specific embodiment, the mutations are present in one or more framework regions of the light chain. In a specific embodiment, the mutations are present in one or more CDR regions of the light chain. In a specific embodiment, the mutations are present in one or more framework regions and/or one or more CDR regions of the light chain. In various embodiments, the framework regions are selected from framework 1 (FW1), framework 2 (FW2), framework 3 (FW3), and/or a combination thereof.
  • In one embodiment, the light chain is derived from a human Vκ3-20/Jκ1 rearrangement and about 10% of the Vκ3-20/Jκ1 -derived light chains have at least one mutation present in FW1; in one embodiment, at least 10% of the light chains have one mutation in FW1.
  • In one embodiment, the light chain is derived from a human Vκ3-20/Jκ1 rearrangement and about 53% of the Vκ3-20/Jκ1 -derived light chains have at least one or no more than two mutations present in CDR1; in one embodiment, at least 27% of the light chains have one or more mutations in CDR1; in one embodiment, about 54% of the light chains have one or two mutations present in CDR1.
  • In one embodiment, the light chain is derived from a human Vκ3-20/Jκ1 rearrangement and about 6% of the Vκ3-20/Jκ1-derived light chains have at least one or no more than two mutations present in FW2; in one embodiment, at least 6% of light chains have at least one mutation present in FW2; in one embodiment, at least 3% of the light chains have one mutation present in FW2; in one embodiment, at least 3% of the light chains have two mutations present in FW2.
  • In one embodiment, the light chain is derived from a human Vκ3-20/Jκ1 rearrangement and at least about 3% of the Vκ3-20/Jκ1 -derived light chains have at least one mutation present in CDR2; in one embodiment, at least 3% of the light chains have one mutation in CDR2.
  • In one embodiment, the light chain is derived from a human Vκ3-20/Jκ1 rearrangement and about 17% or more of the Vκ3-20/Jκ1-derived light chains have at least one or no more than two mutations present in FW3; in one embodiment, at least 20% of the light chain have one mutation present in FW3; in one embodiment, at least 17% of the light chains have two mutations present in FW3.
  • In one embodiment, the light chain is derived from a human Vκ3-20/Jκ1 rearrangement and at least 43% of the Vκ3-20/Jκ1-derived light chains have at least one mutation present in CDR3; in one embodiment, at least 43% of the light chains have one mutation in CDR3.
  • In one embodiment, a population of antigen-specific antibodies derived from a mouse as described herein is provided, wherein the antibodies comprise a light chain derived from a human Vκ3-20/Jκ1 rearrangement and about 10% of the Vκ3-20/Jκ1-derived light chains have one or more mutations present in at least, about 53% of the Vκ3-20/Jκ1-derived light chains have one or more mutations present in CDR1, about 6% of the Vκ3-20/Jκ1-derived light chains have one or more mutations present in FW2, about 3% of the Vκ3-20/Jκ1-derived light chains have one or more mutations present in CDR2, about 37% of the Vκ3-20/Jκ1 -derived light chains have one or more mutations present in FW3, and about 43% of the Vκ3-20/Jκ1-derived light chains have one or more mutations present in CDR3.
  • In one embodiment, the light chain is derived from a human Vκ3-20/Jκ1 rearrangement and about 43% of the heavy chains have at least one or no more than two mutations present in FW1; in one embodiment, at least 41% of the heavy chains have at least one mutation present in FW1; in one embodiment, about 41% of the heavy chains have one mutation present in FW1; in one embodiment, about 2% of the heavy chains have two mutations present in FW1.
  • In one embodiment, the light chain is derived from a human Vκ3-20/κ1 rearrangement and about 92% of the heavy chains have at least one or no more than four mutations present in CDR1; in one embodiment, at least 43% of heavy chains have at least one mutation present in CDR1; in one embodiment, at least 25% of heavy chains have at least two mutations present in CDR1; in one embodiment, at least 15% of heavy chains have at least 3 mutations present in CDR1; in one embodiment, at least 10% of heavy chains have 4 or more mutations present in CDR1.
  • In one embodiment, the light chain is derived from a human Vκ3-20/Jκ1 rearrangement and about 46% of the heavy chains have at least one or no more than three mutations present in FW2; in one embodiment, at least 34% of heavy chains have at least one mutation present in FW2; in one embodiment, at least 10% of heavy chains have two or more mutations present in FW2; in one embodiment, at least 2% of heavy chains have three or more mutations present in FW2.
  • In one embodiment, the light chain is derived from a human Vκ3-20/Jκ1 rearrangement and about 84% of the heavy chains have at least one or up to five or more than five mutations present in CDR2; in one embodiment, at least 39% of the heavy chains have one or more mutations present in CDR2; in one embodiment, at least 18% of the heavy chains have two or more mutations present in CDR2; in one embodiment, at least 21% of the heavy chains have three or more mutations present in CDR2; in one embodiment, at least 3% of the heavy chains have four or more mutations present in CDR2; in one embodiment, at least 2% of the heavy chains have five or more mutations present in CDR2.
  • In one embodiment, the light chain is derived from a human Vκ3-20/Jκ1 rearrangement and about 92% of the heavy chains have at least one or up to five or more than five mutations present in FW3; in one embodiment, at least 21% of the light chains have at least one mutation present in FW3; in one embodiment, at least 20% of heavy chains have at least two mutations present in FW3; in one embodiment, at least 13% of the heavy chains have at least three mutations present in FW3; in one embodiment, at least 20% of the heavy chains have at least four mutations in FW3; in one embodiment, at least 18% of the heavy chains have at least 5 mutations in FW3.
  • In one embodiment, the light chain is derived from a human Vκ3-20/Jκ1 rearrangement and about 7% of the heavy chains have at least one mutation present in CDR3; in one embodiment, about 7% of the heavy chains have one mutation in CDR3.
  • In one embodiment, a population of antigen-specific antibodies derived from a mouse as described herein is provided, wherein the antibodies comprise a light chain derived from a human Vκ3-20/Jκ1 rearrangement and about 43% of the heavy chains have one or more mutations present in FW1, about 92% of the heavy chains have one or more mutations present in CDR1, about 46% of the heavy chains have one or more mutations present in FW2, about 84% of the heavy chains have one or more mutations present in CDR2, about 92% of the heavy chains have one or more mutations present in FW3, and about 7% of the heavy chains have one or more mutations present in CDR3.
  • In one aspect, a mouse that expresses an immunoglobulin light chain from a rearranged immunoglobulin light chain sequence is provided, wherein the rearranged immunoglobulin light chain sequence is present in the germline of the mouse, wherein the immunoglobulin light chain comprises a human variable sequence. In one embodiment, the germline of the mouse comprises a rearranged immunoglobulin light chain sequence that is derived from the same V segment and the same J segment as all non-surrogate light chain sequences present in every B cell of the mouse that comprises a rearranged light chain sequence.
  • In one embodiment, the germline of the mouse lacks a functional unrearranged immunoglobulin light chain V gene segment. In one embodiment, the germline of the mouse lacks a functional unrearranged immunoglobulin light chain J gene segment.
  • In one embodiment, the germline of the mouse comprises no more than one, no more than two, or no more than three rearranged (V/J) light chain sequences.
  • In one embodiment, the rearranged V/J sequence comprises a κ light chain sequence. In a specific embodiment, the κ light chain sequence is a human κ light chain sequence. In a specific embodiment, the κ light chain sequence is selected from a human Vκ1-39/J sequence, a human Vκ3-20/J sequence, and a combination thereof. In a specific embodiment, the κ light chain sequence is a human Vκ1-39/Jκ5 sequence. In a specific embodiment, the κ light chain sequence is a human Vκ3-20/Jκ1 sequence.
  • In one embodiment, the mouse further comprises in its germline a sequence selected from a mouse κ intronic enhancer 5′ with respect to the rearranged immunoglobulin light chain sequence, a mouse κ 3′ enhancer, and a combination thereof.
  • In one embodiment, the mouse comprises an unrearranged human VH gene segment, an unrearranged human DH gene segment, and an unrearranged human JH gene segment, wherein said VH, DH, and JH gene segments are capable of rearranging to form an immunoglobulin heavy chain variable gene sequence operably linked to a heavy chain constant gene sequence. In one embodiment, the mouse comprises a plurality of human VH, DH, and JH gene segments. In a specific embodiment, the human VH, DH, and JH gene segments replace endogenous mouse VH, DH, and JH gene segments at the endogenous mouse immunoglobulin heavy chain locus. In a specific embodiment, the mouse comprises a replacement of all or substantially all functional mouse VH, DH, and JH gene segments with all or substantially all functional human VH, DH, and JH gene segments.
  • In one embodiment, the mouse expresses an immunoglobulin light chain that comprises a mouse constant sequence. In one embodiment, the mouse expresses an immunoglobulin light chain that comprises a human constant sequence.
  • In one embodiment, the mouse expresses an immunoglobulin heavy chain that comprises a mouse sequence selected from a C H1 sequence, a hinge sequence, a CH2 sequence, a CH3 sequence, and a combination thereof.
  • In one embodiment, the mouse expresses an immunoglobulin heavy chain that comprises a human sequence selected from a C H1 sequence, a hinge sequence, a CH2 sequence, a CH3 sequence, and a combination thereof.
  • In one embodiment, the rearranged immunoglobulin light chain sequence in the germline of the mouse is at an endogenous mouse immunoglobulin light chain locus. In a specific embodiment, the rearranged immunoglobulin light chain sequence in the germline of the mouse replaces all or substantially all mouse light chain V and J sequences at the endogenous mouse immunoglobulin light chain locus.
  • In one aspect, a mouse is provided that comprises a B cell population characterized by each B cell that comprises a non-surrogate light chain sequence comprises a rearranged light chain gene that is derived from a single human V segment and a single human J segment, wherein the only light chain variable sequence in the germline of the mouse is a rearranged sequence derived from the single human V segment and the single human J segment, and wherein each B ell that comprises the rearranged light chain gene further comprises a gene encoding a cognate human heavy chain variable domain, and wherein the rearranged light chain gene comprises at least one, at least two, at least three, or at least four somatic hypermutations.
  • In one aspect, a pluripotent, induced pluripotent, or totipotent cell derived from a mouse as described herein is provided. In a specific embodiment, the cell is a mouse embryonic stem (ES) cell.
  • In one aspect, a tissue derived from a mouse as described herein is provided. In one embodiment, the tissue is derived from spleen, lymph node or bone marrow of a mouse as described herein.
  • In one aspect, a nucleus derived from a mouse as described herein is provided. In one embodiment, the nucleus is from a diploid cell that is not a B cell.
  • In one aspect, a mouse cell is provided that is isolated from a mouse as described herein. In one embodiment, the cell is an ES cell. In one embodiment, the cell is a lymphocyte. In one embodiment, the lymphocyte is a B cell. In one embodiment, the B cell expresses a chimeric heavy chain comprising a variable domain derived from a human gene segment; and a light chain derived from a rearranged human Vκ1-39/J sequence, rearranged human Vκ3-20/J sequence, or a combination thereof; wherein the heavy chain variable domain is fused to a mouse constant region and the light chain variable domain is fused to a mouse or a human constant region.
  • In one aspect, a hybridoma is provided, wherein the hybridoma is made with a B cell of a mouse as described herein. In a specific embodiment, the B cell is from a mouse as described herein that has been immunized with an immunogen comprising an epitope of interest, and the B cell expresses a binding protein that binds the epitope of interest, the binding protein has a somatically mutated human VH domain and a mouse CH, and has a human VL domain derived from a rearranged human Vκ1-39Jκ5 or a rearranged human Vκ3-20Jκ1 and a mouse CL.
  • In one aspect, a mouse embryo is provided, wherein the embryo comprises a donor ES cell that is derived from a mouse as described herein.
  • In one aspect, a targeting vector is provided, comprising, from 5′ to 3′ in transcriptional direction with reference to the sequences of the 5′ and 3′ mouse homology arms of the vector, a 5′ mouse homology arm, a human or mouse immunoglobulin promoter, a human or mouse leader sequence, and a human VL region selected from a rearranged human Vκ1-39Jκ5 or a rearranged human Vκ3-20Jκ1, and a 3′ mouse homology arm. In one embodiment, the 5′ and 3′ homology arms target the vector to a sequence 5′ with respect to an enhancer sequence that is present 5′ and proximal to the mouse Cκ gene. In one embodiment, the promoter is a human immunoglobulin variable region gene segment promoter. In a specific embodiment, the promoter is a human Vκ3-15 promoter. In one embodiment, the leader sequence is a mouse leader sequence. In a specific embodiment, the mouse leader sequence is a mouse Vκ3-7 leader sequence.
  • In one aspect, a targeting vector is provided as described above, but in place of the 5′ mouse homology arm the human or mouse promoter is flanked 5′ with a site-specific recombinase recognition site (SRRS), and in place of the 3′ mouse homology arm the human VL region is flanked 3′ with an SRRS.
  • In one aspect, a reverse chimeric antibody made by a mouse as described herein, wherein the reverse chimeric antibody comprises a light chain comprising a human VL and a mouse CL, and a heavy chain comprising a human VH and a mouse CH.
  • In one aspect, a method for making an antibody is provided, comprising expressing in a single cell (a) a first VH gene sequence of an immunized mouse as described herein fused with a human CH gene sequence; (b) a VL gene sequence of an immunized mouse as described herein fused with a human CL gene sequence; and, (c) maintaining the cell under conditions sufficient to express a fully human antibody, and isolating the antibody. In one embodiment, the cell comprises a second VH gene sequence of a second immunized mouse as described herein fused with a human CH gene sequence, the first VH gene sequence encodes a VH domain that recognizes a first epitope, and the second VH gene sequence encodes a VH domain that recognizes a second epitope, wherein the first epitope and the second epitope are not identical.
  • In one aspect, a method for making an epitope-binding protein is provided, comprising exposing a mouse as described herein with an immunogen that comprises an epitope of interest, maintaining the mouse under conditions sufficient for the mouse to generate an immunoglobulin molecule that specifically binds the epitope of interest, and isolating the immunoglobulin molecule that specifically binds the epitope of interest; wherein the epitope-binding protein comprises a heavy chain that comprises a somatically mutated human VH and a mouse CH, associated with a light chain comprising a mouse CL and a human VL derived from a rearranged human Vκ1-39Jκ5 or a rearranged human Vκ3-20Jκ1.
  • In one aspect, a cell that expresses an epitope-binding protein is provided, wherein the cell comprises: (a) a human nucleotide sequence encoding a human VL domain that is derived from a rearranged human Vκ1-39Jκ5 or a rearranged human Vκ3-20Jκ1, wherein the human nucleotide sequence is fused (directly or through a linker) to a human immunoglobulin light chain constant domain cDNA sequence (e.g., a human κ constant domain DNA sequence); and, (b) a first human VH nucleotide sequence encoding a human VH domain derived from a first human VH nucleotide sequence, wherein the first human VH nucleotide sequence is fused (directly or through a linker) to a human immunoglobulin heavy chain constant domain cDNA sequence; wherein the epitope-binding protein recognizes a first epitope. In one embodiment, the epitope-binding protein binds the first epitope with a dissociation constant of lower than 10−6 M, lower than 10−8 M lower than 10−9 M, lower than 10−10 M, lower than 10−11 M, or lower than 10−12 M.
  • In one embodiment, the cell comprises a second human nucleotide sequence encoding a second human VH domain, wherein the second human sequence is fused (directly or through a linker) to a human immunoglobulin heavy chain constant domain cDNA sequence, and wherein the second human VH domain does not specifically recognize the first epitope (e.g., displays a dissociation constant of, e.g., 10−6 M, 10−5 M, 10−4 M, or higher), and wherein the epitope-binding protein recognizes the first epitope and the second epitope, and wherein the first and the second immunoglobulin heavy chains each associate with an identical light chain of (a).
  • In one embodiment, the second VH domain binds the second epitope with a dissociation constant that is lower than 10−6 M, lower than 10−7M, lower than 10−8 M, lower than 10−9 M, lower than 10−10 M, lower than 10−11 M, or lower than 10−12 M.
  • In one embodiment, the epitope-binding protein comprises a first immunoglobulin heavy chain and a second immunoglobulin heavy chain, each associated with an identical light chain derived from a rearranged human VL region selected from a human Vκ1-39Jκ5 or a human Vκ3-20Jκ1, wherein the first immunoglobulin heavy chain binds a first epitope with a dissociation constant in the nanomolar to picomolar range, the second immunoglobulin heavy chain binds a second epitope with a dissociation constant in the nanomolar to picomolar range, the first epitope and the second epitope are not identical, the first immunoglobulin heavy chain does not bind the second epitope or binds the second epitope with a dissociation constant weaker than the micromolar range (e.g., the millimolar range), the second immunoglobulin heavy chain does not bind the first epitope or binds the first epitope with a dissociation constant weaker than the micromolar range (e.g., the millimolar range), and one or more of the VL, the VH of the first immunoglobulin heavy chain, and the VH of the second immunoglobulin heavy chain, are somatically mutated.
  • In one embodiment, the first immunoglobulin heavy chain comprises a protein A-binding residue, and the second immunoglobulin heavy chain lacks the protein A-binding residue.
  • In one embodiment, the cell is selected from CHO, COS, 293, HeLa, and a retinal cell expressing a viral nucleic acid sequence (e.g., a PERC.6™ cell).
  • In one aspect, a reverse chimeric antibody is provided, comprising a human VH and a mouse heavy chain constant domain, a human VL and a mouse light chain constant domain, wherein the antibody is made by a process that comprises immunizing a mouse as described herein with an immunogen comprising an epitope, and the antibody specifically binds the epitope of the immunogen with which the mouse was immunized. In one embodiment, the VL domain is somatically mutated. In one embodiment the VH domain is somatically mutated. In one embodiment, both the VL domain and the VH domain are somatically mutated. In one embodiment, the VL is linked to a mouse Cκ domain.
  • In one aspect, a mouse is provided, comprising human VH gene segments replacing all or substantially all mouse VH gene segments at the endogenous mouse heavy chain locus; no more than one or two rearranged human light chain VL/JL sequences selected from a rearranged Vκ1-39/J and a rearranged Vκ3-20/J or a combination thereof, replacing all mouse light chain gene segments; wherein the human heavy chain variable gene segments are linked to a mouse constant gene, and the rearranged human light chain sequences are linked to a human or mouse constant gene.
  • In one aspect, a mouse ES cell comprising a replacement of all or substantially all mouse heavy chain variable gene segments with human heavy chain variable gene segments, and no more than one or two rearranged human light chain VL/JL sequences, wherein the human heavy chain variable gene segments are linked to a mouse immunoglobulin heavy chain constant gene, and the rearranged human light chain VL/JL sequences are linked to a mouse or human immunoglobulin light chain constant gene. In a specific embodiment, the light chain constant gene is a mouse constant gene.
  • In one aspect, an antigen-binding protein made by a mouse as described herein is provided. In a specific embodiment, the antigen-binding protein comprises a human immunoglobulin heavy chain variable region fused with a mouse constant region, and a human immunoglobulin light chain variable region derived from a Vκ1-39 gene segment or a Vκ3-20 gene segment, wherein the light chain constant region is a mouse constant region.
  • In one aspect, a fully human antigen-binding protein made from an immunoglobulin variable region gene sequence from a mouse as described herein is provided, wherein the antigen-binding protein comprises a fully human heavy chain comprising a human variable region derived from a sequence of a mouse as described herein, and a fully human light chain comprising a Vκ1-39 or a Vκ3-20. In one embodiment, the light chain variable region comprises one to five somatic mutations. In one embodiment, the light chain variable region is a cognate light chain variable region that is paired in a B cell of the mouse with the heavy chain variable region.
  • In one embodiment, the fully human antigen-binding protein comprises a first heavy chain and a second heavy chain, wherein the first heavy chain and the second heavy chain comprise non-identical variable regions independently derived from a mouse as described herein, and wherein each of the first and second heavy chains express from a host cell associated with a human light chain derived from a Vκ1-39 gene segment or a Vκ3-20 gene segment. In one embodiment, the first heavy chain comprises a first heavy chain variable region that specifically binds a first epitope of a first antigen, and the second heavy chain comprises a second heavy chain variable region that specifically binds a second epitope of a second antigen. In a specific embodiment, the first antigen and the second antigen are different. In a specific embodiment, the first antigen and the second antigen are the same, and the first epitope and the second epitope are not identical; in a specific embodiment, binding of the first epitope by a first molecule of the binding protein does not block binding of the second epitope by a second molecule of the binding protein.
  • In one aspect, a fully human binding protein derived from a human immunoglobulin sequence of a mouse as described herein comprises a first immunoglobulin heavy chain and a second immunoglobulin heavy chain, wherein the first immunoglobulin heavy chain comprises a first variable region that is not identical to a variable region of the second immunoglobulin heavy chain, and wherein the first immunoglobulin heavy chain comprises a wild type protein A binding determinant, and the second heavy chain lacks a wild type protein A binding determinant. In one embodiment, the first immunoglobulin heavy chain binds protein A under isolation conditions, and the second immunoglobulin heavy chain does not bind protein A or binds protein A at least 10-fold, a hundred-fold, or a thousand-fold weaker than the first immunoglobulin heavy chain binds protein A under isolation conditions. In a specific embodiment, the first and the second heavy chains are IgG1 isotypes, wherein the second heavy chain comprises a modification selected from 95R (EU 435R), 96F (EU 436F), and a combination thereof, and wherein the first heavy chain lacks such modification.
  • In one aspect, a method for making a bispecific antigen-binding protein is provided, comprising exposing a first mouse as described herein to a first antigen of interest that comprises a first epitope, exposing a second mouse as described herein to a second antigen of interest that comprises a second epitope, allowing the first and the second mouse to each mount immune responses to the antigens of interest, identifying in the first mouse a first human heavy chain variable region that binds the first epitope of the first antigen of interest, identifying in the second mouse a second human heavy chain variable region that binds the second epitope of the second antigen of interest, making a first fully human heavy chain gene that encodes a first heavy chain that binds the first epitope of the first antigen of interest, making a second fully human heavy chain gene that encodes a second heavy chain that binds the second epitope of the second antigen of interest, expressing the first heavy chain and the second heavy chain in a cell that expresses a single fully human light chain derived from a human Vκ1-39 or a human Vκ3-20 gene segment to form a bispecific antigen-binding protein, and isolating the bispecific antigen-binding protein.
  • In one embodiment, the first antigen and the second antigen are not identical.
  • In one embodiment, the first antigen and the second antigen are identical, and the first epitope and the second epitope are not identical. In one embodiment, binding of the first heavy chain variable region to the first epitope does not block binding of the second heavy chain variable region to the second epitope.
  • In one embodiment, the first antigen is selected from a soluble antigen and a cell surface antigen (e.g., a tumor antigen), and the second antigen comprises a cell surface receptor. In a specific embodiment, the cell surface receptor is an immunoglobulin receptor. In a specific embodiment, the immunoglobulin receptor is an Fc receptor. In one embodiment, the first antigen and the second antigen are the same cell surface receptor, and binding of the first heavy chain to the first epitope does not block binding of the second heavy chain to the second epitope.
  • In one embodiment, the light chain variable domain of the light chain comprises 2 to 5 somatic mutations. In one embodiment, the light chain variable domain is a somatically mutated cognate light chain expressed in a B cell of the first or the second immunized mouse with either the first or the second heavy chain variable domain.
  • In one embodiment, the first fully human heavy chain bears an amino acid modification that reduces its affinity to protein A, and he second fully human heavy chain does not comprise a modification that reduces its affinity to protein A.
  • In one aspect, an antibody or a bispecific antibody comprising a human heavy chain variable domain made in accordance with the invention is provided. In another aspect, use of a mouse as described herein to make a fully human antibody or a fully human bispecific antibody is provided.
  • In one aspect, a genetically modified mouse, embryo, or cell described herein comprises a κ light chain locus that retains endogenous regulatory or control elements, e.g., a mouse κ intronic enhancer, a mouse κ 3′ enhancer, or both an intronic enhancer and a 3′ enhancer, wherein the regulatory or control elements facilitate somatic mutation and affinity maturation of an expressed sequence of the κ light chain locus.
  • In one aspect, a mouse is provided that comprises a B cell population characterized by having immunoglobulin light chains derived from no more than one, or no more than two, rearranged or unrearranged immunoglobulin light chain V and J gene segments, wherein the mouse exhibits a κ:λ light chain ratio that is about the same as a mouse that comprises a wild type complement of immunoglobulin light chain V and J gene segments.
  • In one embodiment, the immunoglobulin light chains are derived from no more than one, or no more than two, rearranged immunoglobulin light chain V and J gene segments. In a specific embodiment, the light chains are derived from no more than one rearranged immunoglobulin light chain V and J gene segments.
  • In one aspect, a mouse as described herein is provided that expresses an immunoglobulin light chain derived from no more than one, or no more than two, human Vκ/Jκ sequences, wherein the mouse comprises a replacement of all or substantially all endogenous mouse heavy chain variable region gene segments with one or more human heavy chain variable region gene segments, and the mouse exhibits a ratio of (a) CD19+ B cells that express an immunoglobulin having a λ light chain, to (b) CD19+ B cells that express an immunoglobulin having a κ light chain, of about 1 to about 20.
  • In one embodiment, the mouse expresses a single κ light chain derived from a human Vκ1-39Jκ5 sequence, and the ratio of CD19+ B cells that express an immunoglobulin having a 2 light chain to CD19+ B cells that express an immunoglobulin having a κ light chain is about 1 to about 20; in one embodiment, the ratio is about 1 to at least about 66; in a specific embodiment, the ratio is about 1 to 66.
  • In one embodiment, the mouse expresses a single κ light chain derived from a human Vκ3-20Jκ5 sequence, and the ratio of CD19+ B cells that express an immunoglobulin having a λ light chain to CD19+ B cells that express an immunoglobulin having a κ light chain is about 1 to about 20; in one embodiment, the ratio is about 1 to about 21. In specific embodiments, the ratio is 1 to 20, or 1 to 21.
  • In one aspect, a genetically modified mouse is provided that expresses a single rearranged κ light chain, wherein the mouse comprises a functional λ light chain locus, and wherein the mouse expresses a B cell population that comprises Igκ+ cells that express a κ light chain derived from the same single rearranged κ light chain. In one embodiment, the percent of Igκ+Igλ+ B cells in the mouse is about the same as in a wild type mouse. In a specific embodiment, the percent of Igκ+Igλ+ B cells in the mouse is about 2 to about 6 percent. In a specific embodiment, the percent of Igκ+Igλ+ B cells in a mouse wherein the single rearranged κ light chain is derived from a Vκ1-39Jκ5 sequence is about 2 to about 3; in a specific embodiment, the percent is about 2.6. In a specific embodiment, the percent of Igκ+Igλ+ B cells in a mouse wherein the single rearranged κ light chain is derived from a Vκ3-20Jκ1 sequence is about 4 to about 8; in a specific embodiment, about 6.
  • In one aspect, a genetically modified mouse is provided, wherein the mouse expresses a single rearranged κ light chain derived from a human Vκ and Jκ gene segment, wherein the mouse expresses a B cell population that comprises a single κ light chain derived from the single rearranged κ light chain sequence, wherein the genetically modified mouse has not been rendered resistant to somatic hypermutations. In one embodiment, at least 90% of the κ light chains expressed on a B cell of the mouse exhibit from at least one to about five somatic hypermutations.
  • In one aspect, a genetically modified mouse is provided that is modified to express a single κ light chain derived from no more than one, or no more than two, rearranged κ light chain sequences, wherein the mouse exhibits a κ light chain usage that is about two-fold or more, at least about three-fold or more, or at least about four-fold or more greater than the κ light chain usage exhibited by a wild type mouse, or greater than the κ light chain usage exhibited by a mouse of the same strain that comprises a wild type repertoire of κ light chain gene segments. In a specific embodiment, the mouse expresses the single κ light chain from no more than one rearranged κ light chain sequence. In a more specific embodiment, the rearranged κ light chain sequence is selected from a Vκ1-39Jκ5 and Vκ3-20Jκ1 sequence. In one embodiment, the rearranged κ light chain sequence is a Vκ1-39Jκ5 sequence. In one embodiment, the rearranged κ light chain sequence is a Vκ3-20Jκ1 sequence.
  • In one aspect, a genetically modified mouse is provided that expresses a single κ light chain derived from no more than one, or no more than two, rearranged κ light chain sequences, wherein the mouse exhibits a κ light chain usage that is about 100-fold or more, at least about 200-fold or more, at least about 300-fold or more, at least about 400-fold or more, at least about 500-fold or more, at least about 600-fold or more, at least about 700-fold or more, at least about 800-fold or more, at least about 900-fold or more, at least about 1000-fold or more greater than the same κ light chain usage exhibited by a mouse bearing a complete or substantially complete human κ light chain locus. In a specific embodiment, the mouse bearing a complete or substantially complete human κ light chain locus lacks a functional unrearranged mouse κ light chain sequence. In a specific embodiment, the mouse expresses the single κ light chain from no more than one rearranged κ light chain sequence. In one embodiment, the mouse comprises one copy of a rearranged κ light chain sequence (e.g., a heterozygote). In one embodiment, the mouse comprises two copies of a rearranged κ light chain sequence (e.g., a homozygote). In a more specific embodiment, the rearranged κ light chain sequence is selected from a Vκ1-39Jκ5 and Vκ3-20Jκ1 sequence. In one embodiment, the rearranged K light chain sequence is a Vκ1-39Jκ5 sequence. In one embodiment, the rearranged κ light chain sequence is a Vκ3-20Jκ1 sequence.
  • In one aspect, a genetically modified mouse is provided that expresses a single light chain derived from no more than one, or no more than two, rearranged light chain sequences, wherein the light chain in the genetically modified mouse exhibits a level of expression that is at least 10-fold to about 1,000-fold, 100-fold to about 1,000-fold, 200-fold to about 1,000-fold, 300-fold to about 1,000-fold, 400-fold to about 1,000-fold, 500-fold to about 1,000-fold, 600-fold to about 1,000-fold, 700-fold to about 1,000-fold, 800-fold to about 1,000-fold, or 900-fold to about 1,000-fold higher than expression of the same rearranged light chain exhibited by a mouse bearing a complete or substantially complete light chain locus. In one embodiment, the light chain comprises a human sequence. In a specific embodiment, the human sequence is a κ sequence. In one embodiment, the human sequence is a λ sequence. In one embodiment, the light chain is a fully human light chain.
  • In one embodiment, the level of expression is characterized by quantitating mRNA of transcribed light chain sequence, and comparing it to transcribed light chain sequence of a mouse bearing a complete or substantially complete light chain locus.
  • In one aspect, a genetically modified mouse is provided that expresses a single κ light chain derived from no more than one, or no more than two, rearranged κ light chain sequences, wherein the mouse, upon immunization with antigen, exhibits a serum titer that is comparable to a wild type mouse immunized with the same antigen. In a specific embodiment, the mouse expresses a single κ light chain from no more than one rearranged κ light chain sequence. In one embodiment, the serum titer is characterized as total immunoglobulin. In a specific embodiment, the serum titer is characterized as IgM specific titer. In a specific embodiment, the serum titer is characterized as IgG specific titer. In a more specific embodiment, the rearranged κ light chain sequence is selected from a Vκ1-39Jκ5 and Vκ3-20Jκ1 sequence. In one embodiment, the rearranged κ light chain sequence is a Vκ1-39Jκ5 sequence. In one embodiment, the rearranged κ light chain sequence is a Vκ3-20Jκ1 sequence.
  • In one aspect, a genetically modified mouse is provided that expresses a population of antigen-specific antibodies, wherein all of the immunoglobulin light chains of the population of antigen-specific antibodies comprise a human light chain variable (VL) region derived from the same single human VL gene segment and the immunoglobulin heavy chains comprise a human heavy chain variable (VH) region derived from one of a plurality of human VH gene segments.
  • In various embodiments, the human VH gene segments are selected from VH1-2, VH1-3, VH1-8, VH1-18, VH1-24, VH1-46, VH1-58, VH1-69, VH2-5, VH2-26, VH2-70, VH3-7, VH3-9, VH3-11, VH3-13, VH3-15, VH3-20, VH3-21, VH3-23, VH3-30, VH3-33, VH3-43, VH3-48, VH3-53, VH3-64, VH3-72, VH3-73, VH4-31, VH4-34, VH4-39, VH4-59, VH5-51, and VH6-1.
  • In various embodiments, same single human VL gene segment is selected from a human Vκ1-39 gene segment and a human Vκ3-20 gene segment. In various embodiments, all of the immunoglobulin light chains comprise a human light chain J (JL) gene segment selected from a Jκ and a Jλ gene segment. In a specific embodiment, the human JL gene segment is selected from a human Jκ1 and a Jκ5 gene segment. In various embodiments, the mouse lacks a sequence selected from a mouse immunoglobulin VL gene segment, a mouse immunoglobulin JL gene segment, and a combination thereof. In various embodiments, the human VL region is operably linked to a human, mouse, or rat immunoglobulin light chain constant (CL) region. In a specific embodiment, the human VL region is operably linked to a mouse Cκ region. In a specific embodiment, the human VL region is operably linked to a rat Cκ region.
  • In various embodiments, the human VL region is expressed from an endogenous immunoglobulin light chain locus. In various embodiments, the human VH region is operably linked to a human, mouse, or rat immunoglobulin heavy chain constant (CH) region. In various embodiments the (CH) region comprises a human sequence selected from a C H1, a hinge, a CH2, a CH3, a CH4, and/or a combination thereof. In various embodiments, the human VH region is expressed from an endogenous immunoglobulin heavy chain locus.
  • In one aspect, a genetically modified mouse is provided that expresses a plurality of immunoglobulin heavy chains associated with a single light chain. In one embodiment, the heavy chain comprises a human sequence. In various embodiments, the human sequence is selected from a variable sequence, a C H1, a hinge, a CH2, a CH3, and a combination thereof. In one embodiment, the single light chain comprises a human sequence. In various embodiments, the human sequence is selected from a variable sequence, a constant sequence, and a combination thereof. In one embodiment, the mouse comprises a disabled endogenous immunoglobulin locus and expresses the heavy chain and/or the light chain from a transgene or extrachromosomal episome. In one embodiment, the mouse comprises a replacement at an endogenous mouse locus of some or all endogenous mouse heavy chain gene segments (i.e., V, D, J), and/or some or all endogenous mouse heavy chain constant sequences (e.g., C H1, hinge, CH2, CH3, or a combination thereof), and/or some or all endogenous mouse light chain sequences (e.g., V, J, constant, or a combination thereof), with one or more human immunoglobulin sequences.
  • In one aspect, a mouse suitable for making antibodies that have the same light chain is provided, wherein all or substantially all antibodies made in the mouse are expressed with the same light chain. In one embodiment, the light chain is expressed from an endogenous light chain locus.
  • In one aspect, a method for making a light chain for a human antibody is provided, comprising obtaining from a mouse as described herein a light chain sequence and a heavy chain sequence, and employing the light chain sequence and the heavy chain sequence in making a human antibody. In one embodiment, the human antibody is a bispecific antibody.
  • In one aspect, a method for identifying a human heavy chain variable domain that is capable of binding an antigen of interest with an engineered light chain as described herein is provided, wherein the method comprises providing a heavy chain variable domain derived from a first antibody that is capable of binding the antigen, repairing the heavy chain variable domain with a germline light chain sequence and transfecting a cell so that each are expressed to form a second antibody, exposing the second antibody to the antigen, and measuring binding of the second antibody to the antigen.
  • In one embodiment, the light chain of the first antibody comprises a human Vκ1-39 sequence. In one embodiment, the light chain of the first antibody comprises a human Vκ3-20 sequence. In one embodiment, the germline light chain sequence comprises a human Vκ1-39 or Vκ3-20 sequence. In various embodiments, binding of the second antibody to the antigen is determined by comparison of binding of the first antibody to the antigen.
  • Any of the embodiments and aspects described herein can be used in conjunction with one another, unless otherwise indicated or apparent from the context. Other embodiments will become apparent to those skilled in the art from a review of the ensuing description.
  • BRIEF DESCRIPTION OF FIGURES
  • FIG. 1 illustrates a targeting strategy for replacing endogenous mouse immunoglobulin light chain variable region gene segments with a human Vκ1-39Jκ5 gene region.
  • FIG. 2 illustrates a targeting strategy for replacing endogenous mouse immunoglobulin light chain variable region gene segments with a human Vκ3-20Jκ1 gene region.
  • FIG. 3 illustrates a targeting strategy for replacing endogenous mouse immunoglobulin light chain variable region gene segments with a human VpreB/Jλ5 gene region.
  • FIG. 4 shows the percent of CD19+ B cells (y-axis) from peripheral blood for wild type mice (WT), mice homozygous for an engineered human rearranged Vκ1-39Jκ5 light chain region (Vκ1-39Jκ5 HO) and mice homozygous for an engineered human rearranged Vκ3-20Jκ1 light chain region (Vκ3-20Jκ1 HO).
  • FIG. 5A shows the relative mRNA expression (y-axis) of a Vκ1-39-derived light chain in a quantitative PCR assay using probes specific for the junction of an engineered human rearranged Vκ1-39Jκ5 light chain region (Vκ1-39Jκ5 Junction Probe) and the human Vκ1-39 gene segment (Vκ1-39 Probe) in a mouse homozygous for a replacement of the endogenous Vκ and Jκ gene segments with human Vκ and Jκ gene segments (Hκ), a wild type mouse (WT), and a mouse heterozygous for an engineered human rearranged Vκ1-39Jκ5 light chain region (Vκ1-39Jκ5 HET). Signals are normalized to expression of mouse CK. N.D.: not detected.
  • FIG. 5B shows the relative mRNA expression (y-axis) of a Vκ1-39-derived light chain in a quantitative PCR assay using probes specific for the junction of an engineered human rearranged Vκ1-39Jκ5 light chain region (Vκ1-39Jκ5 Junction Probe) and the human Vκ1-39 gene segment (Vκ1-39 Probe) in a mouse homozygous for a replacement of the endogenous Vκ and Jκ gene segments with human Vκ and Jκ gene segments (Hκ), a wild type mouse (WT), and a mouse homozygous for an engineered human rearranged Vκ1-39Jκ5 light chain region (Vκ1-39Jκ5 HO). Signals are normalized to expression of mouse Cκ.
  • FIG. 5C shows the relative mRNA expression (y-axis) of a Vκ3-20-derived light chain in a quantitative PCR assay using probes specific for the junction of an engineered human rearranged Vκ3-20Jκ1 light chain region (Vκ3-20Jκ1 Junction Probe) and the human Vκ3-20 gene segment (Vκ3-20 Probe) in a mouse homozygous for a replacement of the endogenous Vκ and Jκ gene segments with human Vκ and Jκ gene segments (Hκ), a wild type mouse (WT), and a mouse heterozygous (HET) and homozygous (HO) for an engineered human rearranged Vκ3-20Jκ1 light chain region. Signals are normalized to expression of mouse CK.
  • FIG. 6A shows IgM (left) and IgG (right) titer in wild type (WT; N=2) and mice homozygous for an engineered human rearranged Vκ1-39Jκ5 light chain region (Vκ1-39Jκ5 HO; N=2) immunized with β-galatosidase.
  • FIG. 6B shows total immunoglobulin (IgM, IgG, IgA) titer in wild type (WT; N=5) and mice homozygous for an engineered human rearranged Vκ3-20Jκ1 light chain region (Vκ3-20Jκ1 HO; N=5) immunized with β-galatosidase.
  • DETAILED DESCRIPTION
  • This invention is not limited to particular methods, and experimental conditions described, as such methods and conditions may vary. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only, and is not intended to be limiting, since the scope of the present invention is defined by the claims.
  • Unless defined otherwise, all terms and phrases used herein include the meanings that the terms and phrases have attained in the art, unless the contrary is clearly indicated or clearly apparent from the context in which the term or phrase is used. Although any methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention, particular methods and materials are now described. All publications mentioned are hereby incorporated by reference in their entirety.
  • The term “antibody”, as used herein, includes immunoglobulin molecules comprising four polypeptide chains, two heavy (H) chains and two light (L) chains inter-connected by disulfide bonds. Each heavy chain comprises a heavy chain variable (VH) region and a heavy chain constant region (CH). The heavy chain constant region comprises three domains, C H1, CH2 and CH3. Each light chain comprises a light chain variable (VL) region and a light chain constant region (CL). The VH and VL regions can be further subdivided into regions of hypervariability, termed complementarity determining regions (CDR), interspersed with regions that are more conserved, termed framework regions (FR). Each VH and VL comprises three CDRs and four FRs, arranged from amino-terminus to carboxy-terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4 (heavy chain CDRs may be abbreviated as HCDR1, HCDR2 and HCDR3; light chain CDRs may be abbreviated as LCDR1, LCDR2 and LCDR3. The term “high affinity” antibody refers to an antibody that has a KD with respect to its target epitope about of 10−9 M or lower (e.g., about 1×10−9 M, 1×10−10 M, 1×10−11 M, or about 1×10−12 M). In one embodiment, KD is measured by surface plasmon resonance, e.g., BIACORE™; in another embodiment, KD is measured by ELISA.
  • The phrase “bispecific antibody” includes an antibody capable of selectively binding two or more epitopes. Bispecific antibodies generally comprise two nonidentical heavy chains, with each heavy chain specifically binding a different epitope—either on two different molecules (e.g., different epitopes on two different immunogens) or on the same molecule (e.g., different epitopes on the same immunogen). If a bispecific antibody is capable of selectively binding two different epitopes (a first epitope and a second epitope), the affinity of the first heavy chain for the first epitope will generally be at least one to two or three or four or more orders of magnitude lower than the affinity of the first heavy chain for the second epitope, and vice versa. Epitopes specifically bound by the bispecific antibody can be on the same or a different target (e.g., on the same or a different protein). Bispecific antibodies can be made, for example, by combining heavy chains that recognize different epitopes of the same immunogen. For example, nucleic acid sequences encoding heavy chain variable sequences that recognize different epitopes of the same immunogen can be fused to nucleic acid sequences encoding the same or different heavy chain constant regions, and such sequences can be expressed in a cell that expresses an immunoglobulin light chain. A typical bispecific antibody has two heavy chains each having three heavy chain CDRs, followed by (N-terminal to C-terminal) a C H1 domain, a hinge, a CH2 domain, and a CH3 domain, and an immunoglobulin light chain that either does not confer epitope-binding specificity but that can associate with each heavy chain, or that can associate with each heavy chain and that can bind one or more of the epitopes bound by the heavy chain epitope-binding regions, or that can associate with each heavy chain and enable binding or one or both of the heavy chains to one or both epitopes.
  • The term “cell” includes any cell that is suitable for expressing a recombinant nucleic acid sequence. Cells include those of prokaryotes and eukaryotes (single-cell or multiple-cell), bacterial cells (e.g., strains of E. coli, Bacillus spp., Streptomyces spp., etc.), mycobacteria cells, fungal cells, yeast cells (e.g., S. cerevisiae, S. pombe, P. pastoris, P. methanolica, etc.), plant cells, insect cells (e.g., SF-9, SF-21, baculovirus-infected insect cells, Trichoplusia ni, etc.), non-human animal cells, human cells, or cell fusions such as, for example, hybridomas or quadromas. In some embodiments, the cell is a human, monkey, ape, hamster, rat, or mouse cell. In some embodiments, the cell is eukaryotic and is selected from the following cells: CHO (e.g., CHO K1, DXB-11 CHO, Veggie-CHO), COS (e.g., COS-7), retinal cell, Vero, CV1, kidney (e.g., HEK293, 293 EBNA, MSR 293, MDCK, HaK, BHK), HeLa, HepG2, WI38, MRC 5, Colo205, HB 8065, HL-60, (e.g., BHK21), Jurkat, Daudi, A431 (epidermal), CV-1, U937, 3T3, L cell, C127 cell, SP2/0, NS-0, MMT 060562, Sertoli cell, BRL 3A cell, HT1080 cell, myeloma cell, tumor cell, and a cell line derived from an aforementioned cell. In some embodiments, the cell comprises one or more viral genes, e.g., a retinal cell that expresses a viral gene (e.g., a PER.C6™ cell).
  • The phrase “complementarity determining region,” or the term “CDR,” includes an amino acid sequence encoded by a nucleic acid sequence of an organism's immunoglobulin genes that normally (i.e., in a wild type animal) appears between two framework regions in a variable region of a light or a heavy chain of an immunoglobulin molecule (e.g., an antibody or a T cell receptor). A CDR can be encoded by, for example, a germline sequence or a rearranged or unrearranged sequence, and, for example, by a naive or a mature B cell or a T cell. A CDR can be somatically mutated (e.g., vary from a sequence encoded in an animal's germline), humanized, and/or modified with amino acid substitutions, additions, or deletions. In some circumstances (e.g., for a CDR3), CDRs can be encoded by two or more sequences (e.g., germline sequences) that are not contiguous (e.g., in an unrearranged nucleic acid sequence) but are contiguous in a B cell nucleic acid sequence, e.g., as the result of splicing or connecting the sequences (e.g., V-D-J recombination to form a heavy chain CDR3).
  • The term “conservative,” when used to describe a conservative amino acid substitution, includes substitution of an amino acid residue by another amino acid residue having a side chain R group with similar chemical properties (e.g., charge or hydrophobicity). In general, a conservative amino acid substitution will not substantially change the functional properties of interest of a protein, for example, the ability of a variable region to specifically bind a target epitope with a desired affinity. Examples of groups of amino acids that have side chains with similar chemical properties include aliphatic side chains such as glycine, alanine, valine, leucine, and isoleucine; aliphatic-hydroxyl side chains such as serine and threonine; amide-containing side chains such as asparagine and glutamine; aromatic side chains such as phenylalanine, tyrosine, and tryptophan; basic side chains such as lysine, arginine, and histidine; acidic side chains such as aspartic acid and glutamic acid; and, sulfur-containing side chains such as cysteine and methionine. Conservative amino acids substitution groups include, for example, valine/leucine/isoleucine, phenylalanine/tyrosine, lysine/arginine, alanine/valine, glutamate/aspartate, and asparagine/glutamine. In some embodiments, a conservative amino acid substitution can be substitution of any native residue in a protein with alanine, as used in, for example, alanine scanning mutagenesis. In some embodiments, a conservative substitution is made that has a positive value in the PAM250 log-likelihood matrix disclosed in Gonnet et al. (1992) Exhaustive Matching of the Entire Protein Sequence Database, Science 256:1443-45, hereby incorporated by reference. In some embodiments, the substitution is a moderately conservative substitution wherein the substitution has a nonnegative value in the PAM250 log-likelihood matrix.
  • In some embodiments, residue positions in an immunoglobulin light chain or heavy chain differ by one or more conservative amino acid substitutions. In some embodiments, residue positions in an immunoglobulin light chain or functional fragment thereof (e.g., a fragment that allows expression and secretion from, e.g., a B cell) are not identical to a light chain whose amino acid sequence is listed herein, but differs by one or more conservative amino acid substitutions.
  • The phrase “epitope-binding protein” includes a protein having at least one CDR and that is capable of selectively recognizing an epitope, e.g., is capable of binding an epitope with a KD that is at about one micromolar or lower (e.g., a KD that is about 1×10−6 M, 1×10−7 M, 1×10−9 M, 1×10−9 M, 1×10−10 M, 1×10−11 M, or about 1×10−12 M). Therapeutic epitope-binding proteins (e.g., therapeutic antibodies) frequently require a KD that is in the nanomolar or the picomolar range.
  • The phrase “functional fragment” includes fragments of epitope-binding proteins that can be expressed, secreted, and specifically bind to an epitope with a KD in the micromolar, nanomolar, or picomolar range. Specific recognition includes having a KD that is at least in the micromolar range, the nanomolar range, or the picomolar range.
  • The term “germline” includes reference to an immunoglobulin nucleic acid sequence in a non-somatically mutated cell, e.g., a non-somatically mutated B cell or pre-B cell or hematopoietic cell.
  • The phrase “heavy chain,” or “immunoglobulin heavy chain” includes an immunoglobulin heavy chain constant region sequence from any organism. Heavy chain variable domains include three heavy chain CDRs and four FR regions, unless otherwise specified. Fragments of heavy chains include CDRs, CDRs and FRs, and combinations thereof. A typical heavy chain has, following the variable domain (from N-terminal to C-terminal), a C H1 domain, a hinge, a CH2 domain, and a CH3 domain. A functional fragment of a heavy chain includes a fragment that is capable of specifically recognizing an epitope (e.g., recognizing the epitope with a KD in the micromolar, nanomolar, or picomolar range), that is capable of expressing and secreting from a cell, and that comprises at least one CDR.
  • The term “identity” when used in connection with sequence includes identity as determined by a number of different algorithms known in the art that can be used to measure nucleotide and/or amino acid sequence identity. In some embodiments described herein, identities are determined using a ClustalW v. 1.83 (slow) alignment employing an open gap penalty of 10.0, an extend gap penalty of 0.1, and using a Gonnet similarity matrix (MACVECTOR™ 10.0.2, MacVector Inc., 2008). The length of the sequences compared with respect to identity of sequences will depend upon the particular sequences, but in the case of a light chain constant domain, the length should contain sequence of sufficient length to fold into a light chain constant domain that is capable of self-association to form a canonical light chain constant domain, e.g., capable of forming two beta sheets comprising beta strands and capable of interacting with at least one C H1 domain of a human or a mouse. In the case of a C H1 domain, the length of sequence should contain sequence of sufficient length to fold into a C H1 domain that is capable of forming two beta sheets comprising beta strands and capable of interacting with at least one light chain constant domain of a mouse or a human.
  • The phrase “immunoglobulin molecule” includes two immunoglobulin heavy chains and two immunoglobulin light chains. The heavy chains may be identical or different, and the light chains may be identical or different.
  • The phrase “light chain” includes an immunoglobulin light chain sequence from any organism, and unless otherwise specified includes human κ and λ. light chains and a VpreB, as well as surrogate light chains. Light chain variable (VL) domains typically include three light chain CDRs and four framework (FR) regions, unless otherwise specified. Generally, a full-length light chain includes, from amino terminus to carboxyl terminus, a VL domain that includes FR1-CDR1-FR2-CDR2-FR3-CDR3-FR4, and a light chain constant domain. Light chains include those, e.g., that do not selectively bind either a first or a second epitope selectively bound by the epitope-binding protein in which they appear. Light chains also include those that bind and recognize, or assist the heavy chain with binding and recognizing, one or more epitopes selectively bound by the epitope-binding protein in which they appear. Common light chains are those derived from a rearranged human Vκ1-39Jκ5 sequence or a rearranged human Vκ3-20Jκ1 sequence, and include somatically mutated (e.g., affinity matured) versions.
  • The phrase “micromolar range” is intended to mean 1-999 micromolar; the phrase “nanomolar range” is intended to mean 1-999 nanomolar; the phrase “picomolar range” is intended to mean 1-999 picomolar.
  • The phrase “somatically mutated” includes reference to a nucleic acid sequence from a B cell that has undergone class-switching, wherein the nucleic acid sequence of an immunoglobulin variable region (e.g., a heavy chain variable domain or including a heavy chain CDR or FR sequence) in the class-switched B cell is not identical to the nucleic acid sequence in the B cell prior to class-switching, such as, for example, a difference in a CDR or framework nucleic acid sequence between a B cell that has not undergone class-switching and a B cell that has undergone class-switching. “Somatically mutated” includes reference to nucleic acid sequences from affinity-matured B cells that are not identical to corresponding immunoglobulin variable region sequences in B cells that are not affinity-matured (i.e., sequences in the genome of germline cells). The phrase “somatically mutated” also includes reference to an immunoglobulin variable region nucleic acid sequence from a B cell after exposure of the B cell to an epitope of interest, wherein the nucleic acid sequence differs from the corresponding nucleic acid sequence prior to exposure of the B cell to the epitope of interest. The phrase “somatically mutated” refers to sequences from antibodies that have been generated in an animal, e.g., a mouse having human immunoglobulin variable region nucleic acid sequences, in response to an immunogen challenge, and that result from the selection processes inherently operative in such an animal.
  • The term “unrearranged,” with reference to a nucleic acid sequence, includes nucleic acid sequences that exist in the germline of an animal cell.
  • The phrase “variable domain” includes an amino acid sequence of an immunoglobulin light or heavy chain (modified as desired) that comprises the following amino acid regions, in sequence from N-terminal to C-terminal (unless otherwise indicated): FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4.
  • Common Light Chain
  • Prior efforts to make useful multispecific epitope-binding proteins, e.g., bispecific antibodies, have been hindered by variety of problems that frequently share a common paradigm: in vitro selection or manipulation of sequences to rationally engineer, or to engineer through trial-and-error, a suitable format for pairing a heterodimeric bispecific human immunoglobulin. Unfortunately, most if not all of the in vitro engineering approaches provide largely ad hoc fixes that are suitable, if at all, for individual molecules. On the other hand, in vivo methods for employing complex organisms to select appropriate pairings that are capable of leading to human therapeutics have not been realized.
  • Generally, native mouse sequences are frequently not a good source for human therapeutic sequences. For at least that reason, generating mouse heavy chain immunoglobulin variable regions that pair with a common human light chain is of limited practical utility. More in vitro engineering efforts would be expended in a trial-and-error process to try to humanize the mouse heavy chain variable sequences while hoping to retain epitope specificity and affinity while maintaining the ability to couple with the common human light chain, with uncertain outcome. At the end of such a process, the final product may maintain some of the specificity and affinity, and associate with the common light chain, but ultimately immunogenicity in a human would likely remain a profound risk.
  • Therefore, a suitable mouse for making human therapeutics would include a suitably large repertoire of human heavy chain variable region gene segments in place of endogenous mouse heavy chain variable region gene segments. The human heavy chain variable region gene segments should be able to rearrange and recombine with an endogenous mouse heavy chain constant domain to form a reverse chimeric heavy chain (i.e., a heavy chain comprising a human variable domain and a mouse constant region). The heavy chain should be capable of class switching and somatic hypermutation so that a suitably large repertoire of heavy chain variable domains are available for the mouse to select one that can associate with the limited repertoire of human light chain variable regions.
  • A mouse that selects a common light chain for a plurality of heavy chains has a practical utility. In various embodiments, antibodies that express in a mouse that can only express a common light chain will have heavy chains that can associate and express with an identical or substantially identical light chain. This is particularly useful in making bispecific antibodies. For example, such a mouse can be immunized with a first immunogen to generate a B cell that expresses an antibody that specifically binds a first epitope. The mouse (or a mouse genetically the same) can be immunized with a second immunogen to generate a B cell that expresses an antibody that specifically binds the second epitope. Variable heavy regions can be cloned from the B cells and expresses with the same heavy chain constant region, and the same light chain, and expressed in a cell to make a bispecific antibody, wherein the light chain component of the bispecific antibody has been selected by a mouse to associate and express with the light chain component.
  • The inventors have engineered a mouse for generating immunoglobulin light chains that will suitably pair with a rather diverse family of heavy chains, including heavy chains whose variable regions depart from germline sequences, e.g., affinity matured or somatically mutated variable regions. In various embodiments, the mouse is devised to pair human light chain variable domains with human heavy chain variable domains that comprise somatic mutations, thus enabling a route to high affinity binding proteins suitable for use as human therapeutics.
  • The genetically engineered mouse, through the long and complex process of antibody selection within an organism, makes biologically appropriate choices in pairing a diverse collection of human heavy chain variable domains with a limited number of human light chain options. In order to achieve this, the mouse is engineered to present a limited number of human light chain variable domain options in conjunction with a wide diversity of human heavy chain variable domain options. Upon challenge with an immunogen, the mouse maximizes the number of solutions in its repertoire to develop an antibody to the immunogen, limited largely or solely by the number or light chain options in its repertoire. In various embodiments, this includes allowing the mouse to achieve suitable and compatible somatic mutations of the light chain variable domain that will nonetheless be compatible with a relatively large variety of human heavy chain variable domains, including in particular somatically mutated human heavy chain variable domains.
  • To achieve a limited repertoire of light chain options, the mouse is engineered to render nonfunctional or substantially nonfunctional its ability to make, or rearrange, a native mouse light chain variable domain. This can be achieved, e.g., by deleting the mouse's light chain variable region gene segments. The endogenous mouse locus can then be modified by an exogenous suitable human light chain variable region gene segment of choice, operably linked to the endogenous mouse light chain constant domain, in a manner such that the exogenous human variable region gene segments can combine with the endogenous mouse light chain constant region gene and form a rearranged reverse chimeric light chain gene (human variable, mouse constant). In various embodiments, the light chain variable region is capable of being somatically mutated. In various embodiments, to maximize ability of the light chain variable region to acquire somatic mutations, the appropriate enhancer(s) is retained in the mouse. For example, in modifying a mouse κ light chain locus to replace endogenous mouse κ light chain gene segments with human κ light chain gene segments, the mouse κ intronic enhancer and mouse κ 3′ enhancer are functionally maintained, or undisrupted.
  • A genetically engineered mouse is provided that expresses a limited repertoire of reverse chimeric (human variable, mouse constant) light chains associated with a diversity of reverse chimeric (human variable, mouse constant) heavy chains. In various embodiments, the endogenous mouse κ light chain gene segments are deleted and replaced with a single (or two) rearranged human light chain region, operably linked to the endogenous mouse Cκ gene. In embodiments for maximizing somatic hypermutation of the rearranged human light chain region, the mouse κ intronic enhancer and the mouse κ 3′ enhancer are maintained. In various embodiments, the mouse also comprises a nonfunctional λ light chain locus, or a deletion thereof or a deletion that renders the locus unable to make a λ light chain.
  • A genetically engineered mouse is provided that, in various embodiments, comprises a light chain variable region locus lacking endogenous mouse light chain VL and JL gene segments and comprising a rearranged human light chain variable region, in one embodiment a rearranged human VL/JL sequence, operably linked to a mouse constant region, wherein the locus is capable of undergoing somatic hypermutation, and wherein the locus expresses a light chain comprising the human VL/JL sequence linked to a mouse constant region. Thus, in various embodiments, the locus comprises a mouse κ 3′ enhancer, which is correlated with a normal, or wild type, level of somatic hypermutation.
  • The genetically engineered mouse in various embodiments when immunized with an antigen of interest generates B cells that exhibit a diversity of rearrangements of human immunoglobulin heavy chain variable regions that express and function with one or with two rearranged light chains, including embodiments where the one or two light chains comprise human light chain variable regions that comprise, e.g., 1 to 5 somatic mutations. In various embodiments, the human light chains so expressed are capable of associating and expressing with any human immunoglobulin heavy chain variable region expressed in the mouse.
  • Epitope-binding Proteins Binding More Than One Epitope
  • The compositions and methods of described herein can be used to make binding proteins that bind more than one epitope with high affinity, e.g., bispecific antibodies. Advantages of the invention include the ability to select suitably high binding (e.g., affinity matured) heavy chain immunoglobulin chains each of which will associate with a single light chain.
  • Synthesis and expression of bispecific binding proteins has been problematic, in part due to issues associated with identifying a suitable light chain that can associate and express with two different heavy chains, and in part due to isolation issues. The methods and compositions described herein allow for a genetically modified mouse to select, through otherwise natural processes, a suitable light chain that can associate and express with more than one heavy chain, including heavy chains that are somatically mutated (e.g., affinity matured). Human VL and VH sequences from suitable B cells of immunized mice as described herein that express affinity matured antibodies having reverse chimeric heavy chains (i.e., human variable and mouse constant) can be identified and cloned in frame in an expression vector with a suitable human constant region gene sequence (e.g., a human IgG1). Two such constructs can be prepared, wherein each construct encodes a human heavy chain variable domain that binds a different epitope. One of the human VLs (e.g., human Vκ1-39Jκ5 or human Vκ3-20Jκ1), in germline sequence or from a B cell wherein the sequence has been somatically mutated, can be fused in frame to a suitable human constant region gene (e.g., a human κ constant gene). These three fully human heavy and light constructs can be placed in a suitable cell for expression. The cell will express two major species: a homodimeric heavy chain with the identical light chain, and a heterodimeric heavy chain with the identical light chain. To allow for a facile separation of these major species, one of the heavy chains is modified to omit a Protein A-binding determinant, resulting in a differential affinity of a homodimeric binding protein from a heterodimeric binding protein. Compositions and methods that address this issue are described in U.S. Ser. No. 12/832,838, filed 25 Jun. 2010, entitled “Readily Isolated Bispecific Antibodies with Native Immunoglobulin Format,” published as US 2010/0331527A1, hereby incorporated by reference.
  • In one aspect, an epitope-binding protein as described herein is provided, wherein human VL and VH sequences are derived from mice described herein that have been immunized with an antigen comprising an epitope of interest.
  • In one embodiment, an epitope-binding protein is provided that comprises a first and a second polypeptide, the first polypeptide comprising, from N-terminal to C-terminal, a first epitope-binding region that selectively binds a first epitope, followed by a constant region that comprises a first CH3 region of a human IgG selected from IgG1, IgG2, IgG4, and a combination thereof; and, a second polypeptide comprising, from N-terminal to C-terminal, a second epitope-binding region that selectively binds a second epitope, followed by a constant region that comprises a second CH3 region of a human IgG selected from IgG1, IgG2, IgG4, and a combination thereof, wherein the second CH3 region comprises a modification that reduces or eliminates binding of the second CH3 domain to protein A.
  • In one embodiment, the second CH3 region comprises an H95R modification (by IMGT exon numbering; H435R by EU numbering). In another embodiment, the second CH3 region further comprises a Y96F modification (IMGT; Y436F by EU).
  • In one embodiment, the second CH3 region is from a modified human IgG1, and further comprises a modification selected from the group consisting of D16E, L18M, N44S, K52N, V57M, and V82I (IMGT; D356E, L358M, N384S, K392N, V397M, and V422I by EU).
  • In one embodiment, the second CH3 region is from a modified human IgG2, and further comprises a modification selected from the group consisting of N44S, K52N, and V82I (IMGT; N384S, K392N, and V422I by EU).
  • In one embodiment, the second CH3 region is from a modified human IgG4, and further comprises a modification selected from the group consisting of Q15R, N44S, K52N, V57M, R69K, E79Q, and V82I (IMGT; Q355R, N384S, K392N, V397M, R409K, E419Q, and V422I by EU).
  • One method for making an epitope-binding protein that binds more than one epitope is to immunize a first mouse in accordance with the invention with an antigen that comprises a first epitope of interest, wherein the mouse comprises an endogenous immunoglobulin light chain variable region locus that does not contain an endogenous mouse VL that is capable of rearranging and forming a light chain, wherein at the endogenous mouse immunoglobulin light chain variable region locus is a single rearranged human VL region operably linked to the mouse endogenous light chain constant region gene, and the rearranged human VL region is selected from a human Vκ1-39Jκ5 and a human Vκ3-20Jκ1, and the endogenous mouse VH gene segments have been replaced in whole or in part with human VH gene segments, such that immunoglobulin heavy chains made by the mouse are solely or substantially heavy chains that comprise human variable domains and mouse constant domains. When immunized, such a mouse will make a reverse chimeric antibody, comprising only one of two human light chain variable domains (e.g., one of human Vκ1-39Jκ5 or human Vκ3-20Jκ1). Once a B cell is identified that encodes a VH that binds the epitope of interest, the nucleotide sequence of the VH (and, optionally, the VL) can be retrieved (e.g., by PCR) and cloned into an expression construct in frame with a suitable human immunoglobulin constant domain. This process can be repeated to identify a second VH domain that binds a second epitope, and a second VH gene sequence can be retrieved and cloned into an expression vector in frame to a second suitable immunoglobulin constant domain. The first and the second immunoglobulin constant domains can the same or different isotype, and one of the immunoglobulin constant domains (but not the other) can be modified as described herein or in US 2010/0331527A1, and epitope-binding protein can be expressed in a suitable cell and isolated based on its differential affinity for Protein A as compared to a homodimeric epitope-binding protein, e.g., as described in US 2010/0331527A1.
  • In one embodiment, a method for making a bispecific epitope-binding protein is provided, comprising identifying a first affinity-matured (e.g., comprising one or more somatic hypermutations) human VH nucleotide sequence (VH1) from a mouse as described herein, identifying a second affinity-matured (e.g., comprising one or more somatic hypermutations) human VH nucleotide sequence (VH2) from a mouse as described herein, cloning VH1 in frame with a human heavy chain lacking a Protein A-determinant modification as described in US 2010/0331527A1 for form heavy chain 1 (HC1), cloning VH2 in frame with a human heavy chain comprising a Protein A-determinant as described in US 2010/0331527A1 to form heavy chain 2 (HC2), introducing an expression vector comprising HC1 and the same or a different expression vector comprising HC2 into a cell, wherein the cell also expresses a human immunoglobulin light chain that comprises a human Vκ1-39/human Jκ5 or a human Vκ3-20/human Jκ1 fused to a human light chain constant domain, allowing the cell to express a bispecific epitope-binding protein comprising a VH domain encoded by VH1 and a VH domain encoded by VH2, and isolating the bispecific epitope-binding protein based on its differential ability to bind Protein A as compared with a monospecific homodimeric epitope-binding protein. In a specific embodiment, HC1 is an IgG1, and HC2 is an IgG1 that comprises the modification H95R (IMGT; H435R by EU) and further comprises the modification Y96F (IMGT; Y436F by EU). In one embodiment, the VH domain encoded by V H1, the VH domain encoded by VH2, or both, are somatically mutated.
  • Human VH Genes That Express with a Common Human VL
  • A variety of human variable regions from affinity-matured antibodies raised against four different antigens were expressed with either their cognate light chain, or at least one of a human light chain selected from human Vκ1-39Jκ5, human Vκ3-20Jκ1, or human VpreBJλ5 (see Example 1). For antibodies to each of the antigens, somatically mutated high affinity heavy chains from different gene families paired successfully with rearranged human germline Vκ1-39Jκ5 and Vκ3-20Jκ1 regions and were secreted from cells expressing the heavy and light chains. For Vκ1-39Jκ5 and Vκ3-20Jκ1, VH domains derived from the following human VH gene families expressed favorably: 1-2, 1-8, 1-24, 2-5, 3-7, 3-9, 3-11, 3-13, 3-15, 3-20, 3-23, 3-30, 3-33, 3-48, 4-31, 4-39, 4-59, 5-51, and 6-1. Thus, a mouse that is engineered to express a limited repertoire of human VL domains from one or both of Vκ1-39Jκ5 and Vκ3-20Jκ1 will generate a diverse population of somatically mutated human VH domains from a VH locus modified to replace mouse VH gene segments with human VH gene segments.
  • Mice genetically engineered to express reverse chimeric (human variable, mouse constant) immunoglobulin heavy chains associated with a single rearranged light chain (e.g., a Vκ1-39/J or a Vκ3-20/J), when immunized with an antigen of interest, generated B cells that comprised a diversity of human VH rearrangements and expressed a diversity of high-affinity antigen-specific antibodies with diverse properties with respect to their ability to block binding of the antigen to its ligand, and with respect to their ability to bind variants of the antigen (see Examples 5 through 10).
  • Thus, the mice and methods described herein are useful in making and selecting human immunoglobulin heavy chain variable domains, including somatically mutated human heavy chain variable domains, that result from a diversity of rearrangements, that exhibit a wide variety of affinities (including exhibiting a KD of about a nanomolar or less), a wide variety of specificities (including binding to different epitopes of the same antigen), and that associate and express with the same or substantially the same human immunoglobulin light chain variable region.
  • The following examples are provided so as to describe to those of ordinary skill in the art how to make and use methods and compositions of the invention, and are not intended to limit the scope of what the inventors regard as their invention. Efforts have been made to ensure accuracy with respect to numbers used (e.g., amounts, temperature, etc.) but some experimental errors and deviations should be accounted for. Unless indicated otherwise, parts are parts by weight, molecular weight is average molecular weight, temperature is indicated in Celsius, and pressure is at or near atmospheric.
  • EXAMPLES
  • The following examples are provided so as to describe to those of ordinary skill in the art how to make and use methods and compositions of the invention, and are not intended to limit the scope of what the inventors regard as their invention. Efforts have been made to ensure accuracy with respect to numbers used (e.g., amounts, temperature, etc.) but some experimental errors and deviations should be accounted for. Unless indicated otherwise, temperature is indicated in Celsius, pressure is at or near atmospheric, parts are by parts by weight, and molecular weight is average molecular weight.
  • Example 1 Identification of Human Heavy Chain Variable Regions That Associate With Selected Human Light Chain Variable Regions
  • An in vitro expression system was constructed to determine if a single rearranged human germline light chain could be co-expressed with human heavy chains from antigen specific human antibodies.
  • Methods for generating human antibodies in genetically modified mice are known (see e.g., U.S. Pat. No. 6,596,541, Regeneron Pharmaceuticals, VELOCIMMUNE®). The VELOCIMMUNE® technology involves generation of a genetically modified mouse having a genome comprising human heavy and light chain variable regions operably linked to endogenous mouse constant region loci such that the mouse produces an antibody comprising a human variable region and a mouse constant region in response to antigenic stimulation. The DNA encoding the variable regions of the heavy and light chains of the antibodies produced from a VELOCIMMUNE® mouse are fully human. Initially, high affinity chimeric antibodies are isolated having a human variable region and a mouse constant region. As described below, the antibodies are characterized and selected for desirable characteristics, including affinity, selectivity, epitope, etc. The mouse constant regions are replaced with a desired human constant region to generate a fully human antibody containing a non-IgM isotype, for example, wild type or modified IgG1, IgG2, IgG3 or IgG4. While the constant region selected may vary according to specific use, high affinity antigen-binding and target specificity characteristics reside in the variable region.
  • A VELOCIMMUNE® mouse was immunized with a growth factor that promotes angiogenesis (Antigen C) and antigen-specific human antibodies were isolated and sequenced for V gene usage using standard techniques recognized in the art. Selected antibodies were cloned onto human heavy and light chain constant regions and 69 heavy chains were selected for pairing with one of three human light chains: (1) the cognate κ light chain linked to a human κ constant region, (2) a rearranged human germline Vκ1-39Jκ5 linked to a human κ constant region, or (3) a rearranged human germline Vκ3-20Jκ1 linked to a human κ constant region. Each heavy chain and light chain pair were co-transfected in CHO-K1 cells using standard techniques. Presence of antibody in the supernatant was detected by anti-human IgG in an ELISA assay. Antibody titer (ng/ml) was determined for each heavy chain/light chain pair and titers with the different rearranged germline light chains were compared to the titers obtained with the parental antibody molecule (i.e., heavy chain paired with cognate light chain) and percent of native titer was calculated (Table 1). VH: Heavy chain variable gene. ND: no expression detected under current experimental conditions.
  • TABLE 1
    Antibody Titer (ng/mL) Percent of Native Titer
    Cognate Vκ3-
    VH LC Vκ1-39Jκ5 Vκ3-20Jκ1 Vκ1-39Jκ5 20Jκ1
    3-15 63 23 11 36.2 17.5
    1-2 103 53 ND 51.1
    3-23 83 60 23 72.0 27.5
    3-33 15 77 ND 499.4
    4-31 22 69 17 309.4 76.7
    3-7 53 35 28 65.2 53.1
    22 32 19 148.8 89.3
    1-24 3 13 ND 455.2
    3-33 1 47 ND 5266.7
    3-33 58 37 ND 63.1
    110 67 18 60.6 16.5
    3-23 127 123 21 96.5 16.3
    3-33 28 16 2 57.7 7.1
    3-23 32 50 38 157.1 119.4
    18 45 18 254.3 101.7
    3-9 1 30 23 2508.3 1900.0
    3-11 12 26 6 225.9 48.3
    1-8 16 ND 13 81.8
    3-33 54 81 10 150.7 19.1
    34 9 ND 25.9
    3-20 7 14 54 203.0 809.0
    3-33 19 38 ND 200.5
    3-11 48 ND 203 423.6
    11 23 8 212.7 74.5
    3-33 168 138 182 82.0 108.2
    3-20 117 67 100 57.5 86.1
    3-23 86 61 132 70.7 154.1
    3-33 20 12 33 60.9 165.3
    4-31 69 92 52 133.8 75.0
    3-23 87 78 62 89.5 71.2
    1-2 31 82 51 263.0 164.6
    3-23 53 93 151 175.4 285.4
    11 8 17 75.7 151.4
    3-33 114 36 27 31.6 23.4
    3-15 73 39 44 53.7 59.6
    3-33 1 34 16 5600.0 2683.3
    3-9 58 112 57 192.9 97.6
    3-33 67 20 105 30.1 157.0
    3-33 34 21 24 62.7 70.4
    3-20 10 49 91 478.4 888.2
    3-33 66 32 25 48.6 38.2
    3-23 17 59 56 342.7 329.8
    58 108 19 184.4 32.9
    68 54 20 79.4 29.9
    3-33 42 35 32 83.3 75.4
    29 19 13 67.1 43.9
    3-9 24 34 29 137.3 118.4
    3-30/33 17 33 7 195.2 43.1
    3-7 25 70 74 284.6 301.6
    3-33 87 127 ND 145.1
    6-1 28 56 ND 201.8
    3-33 56 39 20 69.9 36.1
    3-33 10 53 1 520.6 6.9
    3-33 20 67 10 337.2 52.3
    3-33 11 36 18 316.8 158.4
    3-23 12 42 32 356.8 272.9
    3-33 66 95 15 143.6 22.5
    3-15 55 68 ND 123.1
    32 68 3 210.9 10.6
    1-8 28 48 ND 170.9
    3-33 124 192 21 154.3 17.0
    3-33 0 113 ND 56550.0
    3-33 10 157 1 1505.8 12.5
    3-33 6 86 15 1385.5 243.5
    3-23 70 115 22 163.5 31.0
    3-7 71 117 21 164.6 29.6
    3-33 82 100 47 122.7 57.1
    3-7 124 161 41 130.0 33.5
  • In a similar experiment, VELOCIMMUNE® mice were immunized with several different antigens and selected heavy chains of antigen specific human antibodies were tested for their ability to pair with different rearranged human germline light chains (as described above). The antigens used in this experiment included an enzyme involved in cholesterol homeostasis (Antigen A), a serum hormone involved in regulating glucose homeostasis (Antigen B), a growth factor that promotes angiogenesis (Antigen C) and a cell-surface receptor (Antigen D). Antigen specific antibodies were isolated from mice of each immunization group and the heavy chain and light chain variable regions were cloned and sequenced. From the sequence of the heavy and light chains, V gene usage was determined and selected heavy chains were paired with either their cognate light chain or a rearranged human germline Vκ1-39Jκ5 region. Each heavy/light chain pair was co-transfected in CHO-K1 cells and the presence of antibody in the supernatant was detected by anti-human IgG in an ELISA assay. Antibody titer (μg/ml) was determined for each heavy chain/light chain pairing and titers with the different rearranged human germline light chains were compared to the titers obtained with the parental antibody molecule (i.e., heavy chain paired with cognate light chain) and percent of native titer was calculated (Table 2). VH: Heavy chain variable gene. Vκ: κ light chain variable gene. ND: no expression detected under current experimental conditions.
  • TABLE 2
    Titer (μg/ml) Percent of
    Anti- VH VH + VH + Native
    Antigen body VH Alone Vκ1-39Jκ5 Titer
    A 320 1-18 2-30 0.3 3.1 2.0 66
    321 2-5  2-28 0.4 0.4 1.9 448
    334 2-5  2-28 0.4 2.7 2.0 73
    313 3-13 3-15 0.5 0.7 4.5 670
    316 3-23 4-1  0.3 0.2 4.1 2174
    315 3-30 4-1  0.3 0.2 3.2 1327
    318 4-59 1-17 0.3 4.6 4.0 86
    B 257 3-13 1-5  0.4 3.1 3.2 104
    283 3-13 1-5  0.4 5.4 3.7 69
    637 3-13 1-5  0.4 4.3 3.0 70
    638 3-13 1-5  0.4 4.1 3.3 82
    624 3-23 1-17 0.3 5.0 3.9 79
    284 3-30 1-17 0.3 4.6 3.4 75
    653 3-33 1-17 0.3 4.3 0.3 7
    268 4-34 1-27 0.3 5.5 3.8 69
    633 4-34 1-27 0.6 6.9 3.0 44
    C 730 3-7  1-5  0.3 1.1 2.8 249
    728 3-7  1-5  0.3 2.0 3.2 157
    691 3-9  3-20 0.3 2.8 3.1 109
    749 3-33 3-15 0.3 3.8 2.3 62
    750 3-33 1-16 0.3 3.0 2.8 92
    724 3-33 1-17 0.3 2.3 3.4 151
    706 3-33 1-16 0.3 3.6 3.0 84
    744 1-18 1-12 0.4 5.1 3.0 59
    696 3-11 1-16 0.4 3.0 2.9 97
    685 3-13 3-20 0.3 0.5 3.4 734
    732 3-15 1-17 0.3 4.5 3.2 72
    694 3-15 1-5  0.4 5.2 2.9 55
    743 3-23 1-12 0.3 3.2 0.3 10
    742 3-23 2-28 0.4 4.2 3.1 74
    693 3-23 1-12 0.5 4.2 4.0 94
    D 136 3-23 2-28 0.4 5.0 2.7 55
    155 3-30 1-16 0.4 1.0 2.2 221
    163 3-30 1-16 0.3 0.6 3.0 506
    171 3-30 1-16 0.3 1.0 2.8 295
    145 3-43 1-5  0.4 4.4 2.9 65
    49 3-48 3-11 0.3 1.7 2.6 155
    51 3-48 1-39 0.1 1.9 0.1 4
    159 3-7  6-21 0.4 3.9 3.6 92
    169 3-7  6-21 0.3 1.3 3.1 235
    134 3-9  1-5  0.4 5.0 2.9 58
    141 4-31 1-33 2.4 4.2 2.6 63
    142 4-31 1-33 0.4 4.2 2.8 67
  • The results obtained from these experiments demonstrate that somatically mutated, high affinity heavy chains from different gene families are able to pair with rearranged human germline Vκ1-39κ5 and Vκ3-20Jκ1 regions and be secreted from the cell as a normal antibody molecule. As shown in Table 1, antibody titer was increased for about 61% (42 of 69) heavy chains when paired with the rearranged human Vκ1-39Jκ5 light chain and about 29% (20 of 69) heavy chains when paired with the rearranged human Vκ3-20Jκ1 light chain as compared to the cognate light chain of the parental antibody. For about 20% (14 of 69) of the heavy chains, both rearranged human germline light chains conferred an increase in expression as compared to the cognate light chain of the parental antibody. As shown in Table 2, the rearranged human germline Vκ1-39Jκ5 region conferred an increase in expression of several heavy chains specific for a range of different classes of antigens as compared to the cognate light chain for the parental antibodies. Antibody titer was increased by more than two-fold for about 35% (15/43) of the heavy chains as compared to the cognate light chain of the parental antibodies. For two heavy chains (315 and 316), the increase was greater than ten-fold as compared to the parental antibody. Within all the heavy chains that showed increase expression relative to the cognate light chain of the parental antibody, family three (VH3) heavy chains are over represented in comparison to other heavy chain variable region gene families. This demonstrates a favorable relationship of human VH3 heavy chains to pair with rearranged human germline Vκ1-39Jκ5 and Vκ3-20Jκ1 light chains.
  • Example 2 Generation of a Rearranged Human Germline Light Chain Locus
  • Various rearranged human germline light chain targeting vectors were made using VELOCIGENE® technology (see, e.g., U.S. Pat. No. 6,586,251 and Valenzuela et al. (2003) High-throughput engineering of the mouse genome coupled with high-resolution expression analysis, Nature Biotech. 21(6):652-659) to modify mouse genomic Bacterial Artificial Chromosome (BAC) clones 302g12 and 254m04 (Invitrogen). Using these two BAC clones, genomic constructs were engineered to contain a single rearranged human germline light chain region and inserted into an endogenous κ light chain locus that was previously modified to delete the endogenous κ variable and joining gene segments.
  • Construction of Rearranged Human Germline Light Chain Targeting Vectors. Three different rearranged human germline light chain regions were made using standard molecular biology techniques recognized in the art. The human variable gene segments used for constructing these three regions included rearranged human Vκ1-39Jκ5 sequence, a rearranged human Vκ3-20Jκ1 sequence and a rearranged human VpreBJλ5 sequence.
  • A DNA segment containing exon 1 (encoding the leader peptide) and intron 1 of the mouse Vκ3-7 gene was made by de novo DNA synthesis (Integrated DNA Technologies). Part of the 5′ untranslated region up to a naturally occurring Blpl restriction enzyme site was included. Exons of human Vκ1-39 and Vκ3-20 genes were PCR amplified from human genomic BAC libraries. The forward primers had a 5′ extension containing the splice acceptor site of intron 1 of the mouse Vκ3-7 gene. The reverse primer used for PCR of the human Vκ1-39 sequence included an extension encoding human Jκ5, whereas the reverse primer used for PCR of the human Vκ3-20 sequence included an extension encoding human Jκ1. The human VpreBJλ5 sequence was made by de novo DNA synthesis (Integrated DNA Technologies). A portion of the human Jκ-Cκ intron including the splice donor site was PCR amplified from plasmid pBS-296-HA18-PIScel. The forward PCR primer included an extension encoding part of either a human Jκ5, Jκ1, or Jκ5 sequence. The reverse primer included a PI-Scel site, which was previously engineered into the intron.
  • The mouse Vκ3-7 exon1/intron 1, human variable light chain exons, and human Jκ-Cκ intron fragments were sewn together by overlap extension PCR, digested with Blpl and PI-Scel, and ligated into plasmid pBS-296-HA18-PIScel, which contained the promoter from the human Vκ3-15 variable gene segment. A loxed hygromycin cassette within plasmid pBS-296-HA18-PIScel was replaced with a FRTed hygromycin cassette flanked by NotI and AscI sites. The NotI/PI-Scel fragment of this plasmid was ligated into modified mouse BAC 254m04, which contained part of the mouse Jκ-Cκ intron, the mouse CK exon, and about 75 kb of genomic sequence downstream of the mouse κ locus, which provided a 3′ homology arm for homologous recombination in mouse ES cells. The Notl/Ascl fragment of this BAC was then ligated into modified mouse BAC 302g12, which contained a FRTed neomycin cassette and about 23 kb of genomic sequence upstream of the endogenous κ locus for homologous recombination in mouse ES cells.
  • Rearranged Human Germline Vκ1-39Jκ5 Targeting Vector (FIG. 1). Restriction enzyme sites were introduced at the 5′ and 3′ ends of an engineered light chain insert for cloning into a targeting vector: an Ascl site at the 5′ end and a PI-Scel site at the 3′ end. Within the 5′ AscI site and the 3′ PI-Scel site the targeting construct from 5′ to 3′ included a 5′ homology arm containing sequence 5′ to the endogenous mouse κ light chain locus obtained from mouse BAC clone 302g12, a FRTed neomycin resistance gene, an genomic sequence including the human Vκ3-15 promoter, a leader sequence of the mouse Vκ3-7 variable gene segment, a intron sequence of the mouse Vκ3-7 variable gene segment, an open reading frame of a rearranged human germline Vκ1-39Jκ5 region, a genomic sequence containing a portion of the human Jκ-Cκ intron, and a 3′ homology arm containing sequence 3′ of the endogenous mouse Jκ5 gene segment obtained from mouse BAC clone 254m04 (FIG. 1, middle). Genes and/or sequences upstream of the endogenous mouse κ light chain locus and downstream of the most 3′ Jκ gene segment (e.g., the endogenous 3′ enhancer) were unmodified by the targeting construct (see FIG. 1). The sequence of the engineered human Vκ1-39Jκ5 locus is shown in SEQ ID NO:1.
  • Targeted insertion of the rearranged human germline Vκ1-39Jκ5 region into BAC DNA was confirmed by polymerase chain reaction (PCR) using primers located at sequences within the rearranged human germline light chain region. Briefly, the intron sequence 3′ to the mouse Vκ3-7 leader sequence was confirmed with primers ULC-m1F (AGGTGAGGGT ACAGATAAGT GTTATGAG; SEQ ID NO:2) and ULC-m1R (TGACAAATGC CCTAATTATA GTGATCA; SEQ ID NO:3). The open reading frame of the rearranged human germline Vκ1-39Jκ5 region was confirmed with primers 1633-h2F (GGGCAAGTCA GAGCATTAGC A; SEQ ID NO:4) and 1633-h2R (TGCAAACTGG ATGCAGCATA G; SEQ ID NO:5). The neomycin cassette was confirmed with primers neoF (GGTGGAGAGG CTATTCGGC; SEQ ID NO:6) and neoR (GAACACGGCG GCATCAG; SEQ ID NO:7). Targeted BAC DNA was then used to electroporate mouse ES cells to created modified ES cells for generating chimeric mice that express a rearranged human germline Vκ1-39Jκ5 region.
  • Positive ES cell clones were confirmed by TAQMAN™ screening and karyotyping using probes specific for the engineered Vκ1-39Jκ5 light chain region inserted into the endogenous locus. Briefly, probe neoP (TGGGCACAAC AGACAATCGG CTG; SEQ ID NO:8) which binds within the neomycin marker gene, probe ULC-m1P (CCATTATGAT GCTCCATGCC TCTCTGTTC; SEQ ID NO:9) which binds within the intron sequence 3′ to the mouse Vκ3-7 leader sequence, and probe 1633h2P (ATCAGCAGAA ACCAGGGAAA GCCCCT; SEQ ID NO:10) which binds within the rearranged human germline Vκ1-39Jκ5 open reading frame. Positive ES cell clones were then used to implant female mice to give rise to a litter of pups expressing the germline Vκ1-39Jκ5 light chain region.
  • Alternatively, ES cells bearing the rearranged human germline Vκ1-39Jκ5 light chain region are transfected with a construct that expresses FLP in order to remove the FRTed neomycin cassette introduced by the targeting construct. Optionally, the neomycin cassette is removed by breeding to mice that express FLP recombinase (e.g., U.S. Pat. No. 6,774,279). Optionally, the neomycin cassette is retained in the mice.
  • Rearranged Human Germline Vκ3-20Jκ1 Targeting Vector (FIG. 2). In a similar fashion, an engineered light chain locus expressing a rearranged human germline Vκ3-20Jκ1 region was made using a targeting construct including, from 5′ to 3′, a 5′ homology arm containing sequence 5′ to the endogenous mouse κ light chain locus obtained from mouse BAC clone 302g12, a FRTed neomycin resistance gene, a genomic sequence including the human Vκ3-15 promoter, a leader sequence of the mouse Vκ3-7 variable gene segment, an intron sequence of the mouse Vκ3-7 variable gene segment, an open reading frame of a rearranged human germline Vκ3-20Jκ1 region, a genomic sequence containing a portion of the human Jκ-Cκ intron, and a 3′ homology arm containing sequence 3′ of the endogenous mouse Jκ5 gene segment obtained from mouse BAC clone 254m04 (FIG. 2, middle). The sequence of the engineered human Vκ3-20Jκ1 locus is shown in SEQ ID NO:11.
  • Targeted insertion of the rearranged human germline Vκ3-20Jκ1 region into BAC DNA was confirmed by polymerase chain reaction (PCR) using primers located at sequences within the rearranged human germline Vκ3-20Jκ1 light chain region. Briefly, the intron sequence 3′ to the mouse Vκ3-7 leader sequence was confirmed with primers ULC-m1F (SEQ ID NO:2) and ULC-m1R (SEQ ID NO:3). The open reading frame of the rearranged human germline Vκ3-20Jκ1 region was confirmed with primers 1635-h2F (TCCAGGCACC CTGTCTTTG; SEQ ID NO:12) and 1635-h2R (AAGTAGCTGC TGCTAACACT CTGACT; SEQ ID NO:13). The neomycin cassette was confirmed with primers neoF (SEQ ID NO:6) and neoR (SEQ ID NO:7). Targeted BAC DNA was then used to electroporate mouse ES cells to created modified ES cells for generating chimeric mice that express the rearranged human germline Vκ3-20Jκ1 light chain.
  • Positive ES cell clones were confirmed by TAQMAN™ screening and karyotyping using probes specific for the engineered Vκ3-20Jκ1 light chain region inserted into the endogenous κ light chain locus. Briefly, probe neoP (SEQ ID NO:8) which binds within the neomycin marker gene, probe ULC-m1P (SEQ ID NO:9) which binds within the mouse Vκ3-7 leader sequence, and probe 1635h2P (AAAGAGCCAC CCTCTCCTGC AGGG; SEQ ID NO:14) which binds within the human Vκ3-20Jκ1 open reading frame. Positive ES cell clones were then used to implant female mice. A litter of pups expressing the human germline Vκ3-20Jκ1 light chain region.
  • Alternatively, ES cells bearing human germline Vκ3-20Jκ1 light chain region can be transfected with a construct that expresses FLP in order to remove the FRTed neomycin cassette introduced by the targeting construct. Optionally, the neomycin cassette may be removed by breeding to mice that express FLP recombinase (e.g., U.S. Pat. No. 6,774,279). Optionally, the neomycin cassette is retained in the mice.
  • Rearranged Human Germline VpreBJλ5 Targeting Vector (FIG. 3). In a similar fashion, an engineered light chain locus expressing a rearranged human germline VpreBJλ5 region was made using a targeting construct including, from 5′ to 3′, a 5′ homology arm containing sequence 5′ to the endogenous mouse κ light chain locus obtained from mouse BAC clone 302g12, a FRTed neomycin resistance gene, an genomic sequence including the human Vκ3-15 promoter, a leader sequence of the mouse Vκ3-7 variable gene segment, an intron sequence of the mouse Vκ3-7 variable gene segment, an open reading frame of a rearranged human germline VpreBJλ5 region, a genomic sequence containing a portion of the human Jκ-Cκ intron, and a 3′ homology arm containing sequence 3′ of the endogenous mouse Jκ5 gene segment obtained from mouse BAC clone 254m04 (FIG. 3, middle). The sequence of the engineered human VpreBJλ5 locus is shown in SEQ ID NO:15.
  • Targeted insertion of the rearranged human germline VpreBJλ5 region into BAC DNA was confirmed by polymerase chain reaction (PCR) using primers located at sequences within the rearranged human germline VpreBJλ5 region light chain region. Briefly, the intron sequence 3′ to the mouse Vκ3-7 leader sequence was confirmed with primers ULC-m1F (SEQ ID NO:2) and ULC-m1R (SEQ ID NO:3). The open reading frame of the rearranged human germline VpreBJλ5 region was confirmed with primers 1616-h1F (TGTCCTCGGC CCTTGGA; SEQ ID NO:16) and 1616-h1R (CCGATGTCAT GGTCGTTCCT; SEQ ID NO:17). The neomycin cassette was confirmed with primers neoF (SEQ ID NO:6) and neoR (SEQ ID NO:7). Targeted BAC DNA was then used to electroporate mouse ES cells to created modified ES cells for generating chimeric mice that express the rearranged human germline VpreBJλ5 light chain.
  • Positive ES cell clones are confirmed by TAQMAN™ screening and karyotyping using probes specific for the engineered VpreBJλ5 light chain region inserted into the endogenous κ light chain locus. Briefly, probe neoP (SEQ ID NO:8) which binds within the neomycin marker gene, probe ULC-m1P (SEQ ID NO:9) which binds within the mouse IgVκ3-7 leader sequence, and probe 1616h1P (ACAATCCGCC TCACCTGCAC CCT; SEQ ID NO:18) which binds within the human VpreBJλ5 open reading frame. Positive ES cell clones are then used to implant female mice to give rise to a litter of pups expressing a germline light chain region.
  • Alternatively, ES cells bearing the rearranged human germline VpreBJλ5 light chain region are transfected with a construct that expresses FLP in order to remove the FRTed neomycin cassette introduced by the targeting construct. Optionally, the neomycin cassette is removed by breeding to mice that express FLP recombinase (e.g., U.S. Pat. No. 6,774,279). Optionally, the neomycin cassette is retained in the mice.
  • Example 3 Generation of Mice Expressing a Single Rearranged Human Light Chain
  • Targeted ES cells described above were used as donor ES cells and introduced into an 8-cell stage mouse embryo by the VELOCIMOUSE® method (see, e.g., U.S. Pat. No. 7,294,754 and Poueymirou et al. (2007) F0 generation mice that are essentially fully derived from the donor gene-targeted ES cells allowing immediate phenotypic analyses Nature Biotech. 25(1):91-99. VELOCIMICE® independently bearing an engineered human germline Vκ1-39Jκ5 light chain region, a Vκ3-20Jκ1 light chain region or a VpreBJλ5 light chain region are identified by genotyping using a modification of allele assay (Valenzuela et al., supra) that detects the presence of the unique rearranged human germline light chain region.
  • Pups are genotyped and a pup heterozygous or homozygous for the unique rearranged human germline light chain region are selected for characterizing expression of the rearranged human germline light chain region.
  • Flow Cytometry. Expression of the rearranged human light chain region in the normal antibody repertoire of common light chain mice was validated by analysis of immunoglobulin κ and λ expression in splenocytes and peripheral blood of common light chain mice. Cell suspensions from harvested spleens and peripheral blood of wild type (n=5), Vκ1-39Jκ5 common light chain heterozygote (n=3), Vκ1-39Jκ5 common light chain homozygote (n=3), Vκ3-20Jκ1 common light chain heterozygote (n=2), and Vκ3-20Jκ1 common light chain homozygote (n=2) mice were made using standard methods and stained with CD19+, Igλ+ and Igκ+ using fluorescently labeled antibodies (BD Pharmigen).
  • Briefly, 1×106 cells were incubated with anti-mouse CD16/CD32 (clone 2.4G2, BD Pharmigen) on ice for 10 minutes, followed by labeling with the following antibody cocktail for 30 minutes on ice: APC conjugated anti-mouse CD19 (clone 1D3, BD Pharmigen), PerCP-Cy5.5 conjugated anti-mouse CD3 (clone 17A2, BioLegend), FITC conjugated anti-mouse Igκ (clone 187.1, BD Pharmigen), PE conjugated anti-mouse Igλ (clone RML-42, BioLegend). Following staining, cells were washed and fixed in 2% formaldehyde. Data acquisition was performed on an LSRII flow cytometer and analyzed with FlowJo. Gating: total B cells (CD19+CD3), Igκ+ B cells (Igκ+IgλCD19+CD3), Igλ+ B cells (IgκIgλ+CD19+CD3). Data gathered from blood and splenocyte samples demonstrated similar results. Table 3 sets forth the percent positive CD19+ B cells from peripheral blood of one representative mouse from each group that are Igλ+, Igκ+, or Igλ+Igκ+. Percent of CD19+ B cells in peripheral blood from wild type (WT) and mice homozygous for either the Vκ1-39Jκ5 or Vκ3-20Jκ1 common light chain are shown in FIG. 4.
  • TABLE 3
    CD19+ B cells
    Mouse Igλ+ Igκ+ Igλ+Igκ+
    wild type 4.8 93 0.53
    Vκ1-39Jκ5 1.4 93 2.6
    Vκ3-20Jκ1 4.2 88 6
  • Common Light Chain Expression. Expression of each common light chain (Vκ1-39Jκ5 and Vκ3-20Jκ1) was analyzed in heterozygous and homozygous mice using a quantitative PCR assay (e.g. TAQMAN™).
  • Briefly, CD19+ B cells were purified from the spleens of wild type, mice homozygous for a replacement of the mouse heavy chain and κ light chain variable region loci with corresponding human heavy chain and κ light chain variable region loci (Hκ), as well as mice homozygous and heterozygous for each rearranged human light chain region (Vκ1-39Jκ5 or Vκ3-20Jκ1) using mouse CD19 Microbeads (Miltenyi Biotec) according to manufacturer's specifications. Total RNA was purified from CD19+ B cells using RNeasy Mini kit (Qiagen) according to manufacturer's specifications and genomic RNA was removed using an RNase-free DNase on-column treatment (Qiagen). 200 ng mRNA was reverse-transcribed into cDNA using the First Stand cDNA Synthesis kit (Invitrogen) and the resulting cDNA was amplified with the Taqman Universal PCR Master Mix (Applied Biosystems). All reactions were performed using the ABI 7900 Sequence Detection System (Applied Biosystems) using primers and Taqman MGB probes spanning (1) the Vκ-Jκ junction for both common light chains, (2) the Vκ gene alone (i.e. Vκ1-39 and Vκ3-20), and (3) the mouse Cκ region. Table 4 sets forth the sequences of the primers and probes employed for this assay. Relative expression was normalized to expression of the mouse Cκ region. Results are shown in FIGS. 5A, 5B and 5C.
  • TABLE 4
    SEQ
    Primer/Probe Description  ID
    Region (5′-3′) NOs:
    Vκ1-39Jκ5 (sense) 19
    Junction AGCAGTCTGC AACCTGAAGA TTT
    (anti-sense) 20
    GTTTAATCTC CAGTCGTGTC CCTT
    (probe) 21
    CCTCCGATCA CCTTC
    Vκ1-39 (sense) 22
    AAACCAGGGA AAGCCCCTAA
    (anti-sense) 23
    ATGGGACCCC ACTTTGCA
    (probe) 24
    CTCCTGATCT ATGCTGCAT
    Vκ3-20Jκ1 (sense) 25
    CAGCAGACTG GAGCCTGAAG A
    Junction (anti-sense) 26
    TGATTTCCAC CTTGGTCCCT T
    (probe) 27
    TAGCTCACCT TGGACGTT
    Vκ3-20 (sense) 28
    CTCCTCATCT ATGGTGCATC CA
    (anti-sense) 29
    GACCCACTGC CACTGAACCT
    (probe) 30
    CCACTGGCAT CCC
    Mouse Cκ (sense) 31
    TGAGCAGCAC CCTCACGTT
    (anti-sense) 32
    GTGGCCTCAC AGGTATAGCT GTT
    (probe) 33
    ACCAAGGACG AGTATGAA
  • Antigen Specific Common Light Chain Antibodies. Common light chain mice bearing either a Vκ1-39Jκ5 or Vκ3-20Jκ1 common light chain at the endogenous mouse κlight chain locus were immunized with (3-galactosidase and antibody titer was measured.
  • Briefly, β-galactosidase (Sigma) was emulsified in titermax adjuvant (Sigma), as per manufacturers directions. Wild type (n=7), Vκ1-39Jκ5 common light chain homozygotes (n=2) and Vκ3-20Jκ1 common light chain homozygotes (n=5) were immunized by subcutaneous injection with 100 μg β-galactosidase/Titermax. Mice were boosted by subcutaneous injection two times, 3 weeks apart, with 50 μg β-galactosidase/Titermax. After the second boost, blood was collected from anaesthetized mice using a retro-orbital bleed into serum separator tubes (BD Biosciences) as per manufacturer's directions. To measure anti-β-galactosidase IgM or IgG antibodies, ELISA plates (Nunc) were coated with 1 μg/mL β-galactosidase overnight at 4° C. Excess antigen was washed off before blocking with PBS with 1% BSA for one hour at room temperature. Serial dilutions of serum were added to the plates and incubated for one hour at room temperature before washing. Plates were then incubated with HRP conjugated anti-IgM (Southern Biotech) or anti-IgG (Southern Biotech) for one hour at room temperature. Following another wash, plates were developed with TMB substrate (BD Biosciences). Reactions were stopped with 1N sulfuric acid and OD450 was read using a Victor X5 Plate Reader (Perkin Elmer). Data was analyzed with GraphPad Prism and signal was calculated as the dilution of serum that is two times above background. Results are shown in FIGS. 6A and 6B.
  • As shown in this Example, the ratio of κ/λ B cells in both the splenic and peripheral compartments of Vκ1-39Jκ5 and Vκ3-20Jκ1 common light chain mice demonstrated a near wild type pattern (Table 3 and FIG. 4). VpreBJλ5 common light chain mice, however, demonstrated fewer peripheral B cells, of which about 1-2% express the engineered human light chain region (data not shown). The expression levels of the Vκ1-39Jκ5 and Vκ3-20Jκ1 rearranged human light chain regions from the endogenous κ light chain locus were elevated in comparison to an endogenous κ light chain locus containing a complete replacement of mouse Vκ and Jκ gene segments with human Vκ and Jκ gene segments (FIGS. 5A, 5B and 5C). The expression levels of the VpreBJλ5 rearranged human light chain region demonstrated similar high expression from the endogenous κ light chain locus in both heterozygous and homozygous mice (data not shown). This demonstrates that in direct competition with the mouse λ, κ, or both endogenous light chain loci, a single rearranged human VL/JL sequence can yield better than wild type level expression from the endogenous κ light chain locus and give rise to normal splenic and blood B cell frequency. Further, the presence of an engineered κ light chain locus having either a human Vκ1-39Jκ5 or human Vκ3-20Jκ1 sequence was well tolerated by the mice and appear to function in wild type fashion by representing a substantial portion of the light chain repertoire in the humoral component of the immune response (FIGS. 6A and 6B).
  • Example 4 Breeding of Mice Expressing a Single Rearranged Human Germline Light Chain
  • This Example describes several other genetically modified mouse strains that can be bred to any one of the common light chain mice described herein to create multiple genetically modified mouse strains harboring multiple genetically modified immunoglobulin loci.
  • Endogenous Igλ Knockout (KO). To optimize the usage of the engineered light chain locus, mice bearing one of the rearranged human germline light chain regions are bred to another mouse containing a deletion in the endogenous λ light chain locus. In this manner, the progeny obtained will express, as their only light chain, the rearranged human germline light chain region as described in Example 2. Breeding is performed by standard techniques recognized in the art and, alternatively, by a commercial breeder (e.g., The Jackson Laboratory). Mouse strains bearing an engineered light chain locus and a deletion of the endogenous λ light chain locus are screened for presence of the unique light chain region and absence of endogenous mouse λ light chains.
  • Humanized Endogenous Heavy Chain Locus. Mice bearing an engineered human germline light chain locus are bred with mice that contain a replacement of the endogenous mouse heavy chain variable gene locus with the human heavy chain variable gene locus (see U.S. Pat. No. 6,596,541; the VELOCIMMUNE® mouse, Regeneron Pharmaceuticals, Inc.). The VELOCIMMUNE® mouse comprises a genome comprising human heavy chain variable regions operably linked to endogenous mouse constant region loci such that the mouse produces antibodies comprising a human heavy chain variable region and a mouse heavy chain constant region in response to antigenic stimulation. The DNA encoding the variable regions of the heavy chains of the antibodies is isolated and operably linked to DNA encoding the human heavy chain constant regions. The DNA is then expressed in a cell capable of expressing the fully human heavy chain of the antibody.
  • Mice bearing a replacement of the endogenous mouse VH locus with the human VH locus and a single rearranged human germline VL region at the endogenous κ light chain locus are obtained. Reverse chimeric antibodies containing somatically mutated heavy chains (human VH and mouse CH) with a single human light chain (human VL and mouse CL) are obtained upon immunization with an antigen of interest. VH and VL nucleotide sequences of B cells expressing the antibodies are identified and fully human antibodies are made by fusion the VH and VL nucleotide sequences to human CH and CL nucleotide sequences in a suitable expression system.
  • Example 5 Generation of Antibodies from Mice Expressing Human Heavy Chains and a Rearranged Human Germline Light Chain Region
  • After breeding mice that contain the engineered human light chain region to various desired strains containing modifications and deletions of other endogenous Ig loci (as described in Example 4), selected mice can be immunized with an antigen of interest.
  • Generally, a VELOCIMMUNE® mouse containing one of the single rearranged human germline light chain regions is challenged with an antigen, and lymphatic cells (such as B-cells) are recovered from serum of the animals. The lymphatic cells are fused with a myeloma cell line to prepare immortal hybridoma cell lines, and such hybridoma cell lines are screened and selected to identify hybridoma cell lines that produce antibodies containing human heavy chain variables and a rearranged human germline light chains which are specific to the antigen used for immunization. DNA encoding the variable regions of the heavy chains and the light chain are isolated and linked to desirable isotypic constant regions of the heavy chain and light chain. Due to the presence of the endogenous mouse sequences and any additional cis-acting elements present in the endogenous locus, the single light chain of each antibody may be somatically mutated. This adds additional diversity to the antigen-specific repertoire comprising a single light chain and diverse heavy chain sequences. The resulting cloned antibody sequences are subsequently expressed in a cell, such as a CHO cell. Alternatively, DNA encoding the antigen-specific chimeric antibodies or the variable domains of the light and heavy chains are identified directly from antigen-specific lymphocytes.
  • Initially, high affinity chimeric antibodies are isolated having a human variable region and a mouse constant region. As described above, the antibodies are characterized and selected for desirable characteristics, including affinity, selectivity, epitope, etc. The mouse constant regions are replaced with a desired human constant region to generate the fully human antibody containing a somatically mutated human heavy chain and a single light chain derived from a rearranged human germline light chain region of the invention. Suitable human constant regions include, for example wild type or modified IgG1 or IgG4.
  • Separate cohorts of VELOCIMMUNE® mice containing a replacement of the endogenous mouse heavy chain locus with human VH, DH, and JH gene segments and a replacement of the endogenous mouse κ light chain locus with either the engineered germline Vκ1-39Jκ5 human light chain region or the engineered germline Vκ3-20Jκ1 human light chain region (described above) were immunized with a human cell-surface receptor protein (Antigen E). Antigen E is administered directly onto the hind footpad of mice with six consecutive injections every 3-4 days. Two to three micrograms of Antigen E are mixed with 10 μg of CpG oligonucleotide (Cat # tlrl-modn—ODN1826 oligonucleotide; InVivogen, San Diego, Calif.) and 25 μg of Adju-Phos (Aluminum phosphate gel adjuvant, Cat# H-71639-250; Brenntag Biosector, Frederikssund, Denmark) prior to injection. A total of six injections are given prior to the final antigen recall, which is given 3-5 days prior to sacrifice. Bleeds after the 4th and 6th injection are collected and the antibody immune response is monitored by a standard antigen-specific immunoassay.
  • When a desired immune response is achieved splenocytes are harvested and fused with mouse myeloma cells to preserve their viability and form hybridoma cell lines. The hybridoma cell lines are screened and selected to identify cell lines that produce Antigen E-specific common light chain antibodies. Using this technique several anti-Antigen E-specific common light chain antibodies (i.e., antibodies possessing human heavy chain variable domains, the same human light chain variable domain, and mouse constant domains) are obtained.
  • Alternatively, anti-Antigen E common light chain antibodies are isolated directly from antigen-positive B cells without fusion to myeloma cells, as described in U.S. 2007/0280945A1, herein specifically incorporated by reference in its entirety. Using this method, several fully human anti-Antigen E common light chain antibodies (i.e., antibodies possessing human heavy chain variable domains, either an engineered human Vκ1-39Jκ5 light chain or an engineered human Vκ3-20Jκ1 light chain region, and human constant domains) were obtained.
  • The biological properties of the exemplary anti-Antigen E common light chain antibodies generated in accordance with the methods of this Example are described in detail in the sections set forth below.
  • Example 6 Heavy Chain Gene Segment Usage in Antigen-Specific Common Light Chain Antibodies
  • To analyze the structure of the human anti-Antigen E common light chain antibodies produced, nucleic acids encoding heavy chain antibody variable regions were cloned and sequenced. From the nucleic acid sequences and predicted amino acid sequences of the antibodies, gene usage was identified for the heavy chain variable region (HCVR) of selected common light chain antibodies obtained from immunized VELOCIMMUNE® mice containing either the engineered human Vκ1-39Jκ5 light chain or engineered human Vκ3-20Jκ1 light chain region. Results are shown in Tables 5 and 6, which demonstrate that mice according to the invention generate antigen-specific common light chain antibodies from a variety of human heavy chain gene segments, due to a variety of rearrangements, when employing either a mouse that expresses a light chain from only a human Vκ1-39- or a human Vκ3-20-derived light chain. Human VH gene segments of the 2, 3, 4, and 5 families rearranged with a variety of human DH segments and human JH segments to yield antigen-specific antibodies.
  • TABLE 5
    Vκ1-39Jκ5
    Common Light Chain Antibodies
    HCVR
    Antibody VH DH JH
    2952 2-5  6-6  1
    5978 2-5  6-6  1
    5981 2-5  3-22 1
    6027 3-13 6-6  5
    3022 3-23 3-10 4
    3028 3-23 3-3  4
    5999 3-23 6-6  4
    6009 3-23 2-8  4
    6011 3-23 7-27 4
    5980 3-30 1-1  4
    3014 3-30 1-7  4
    3015 3-30 1-7  4
    3023 3-30 1-7  4
    3024 3-30 1-7  4
    3032 3-30 1-7  4
    6024 3-30 1-7  4
    6025 3-30 1-7  4
    6031 3-30 1-7  4
    6007 3-30 3-3  4
    2982 3-30 3-22 5
    6001 3-30 3-22 5
    6005 3-30 3-22 5
    6035 3-30 5-5  2
    3013 3-30 5-12 4
    3042 3-30 5-12 4
    2955 3-30 6-6  1
    3043 3-30 6-6  3
    3018 3-30 6-6  4
    2949 3-30 6-6  5
    2950 3-30 6-6  5
    2954 3-30 6-6  5
    2978 3-30 6-6  5
    3016 3-30 6-6  5
    3017 3-30 6-6  5
    3033 3-30 6-6  5
    3041 3-30 6-6  5
    5979 3-30 6-6  5
    5998 3-30 6-6  5
    6004 3-30 6-6  5
    6010 3-30 6-6  5
    6019 3-30 6-6  5
    6021 3-30 6-6  5
    6022 3-30 6-6  5
    6023 3-30 6-6  5
    6030 3-30 6-6  5
    6032 3-30 6-6  5
    2985 3-30 6-13 4
    2997 3-30 6-13 4
    3011 3-30 6-13 4
    3047 3-30 6-13 4
    5982 3-30 6-13 4
    6002 3-30 6-13 4
    6003 3-30 6-13 4
    6012 3-30 6-13 4
    6013 3-30 6-13 4
    6014 3-30 6-13 4
    6015 3-30 6-13 4
    6016 3-30 6-13 4
    6017 3-30 6-13 4
    6020 3-30 6-13 4
    6034 3-30 6-13 4
    2948 3-30 7-27 4
    2987 3-30 7-27 4
    2996 3-30 7-27 4
    3005 3-30 7-27 4
    3012 3-30 7-27 4
    3020 3-30 7-27 4
    3021 3-30 7-27 4
    3025 3-30 7-27 4
    3030 3-30 7-27 4
    3036 3-30 7-27 4
    5997 3-30 7-27 4
    6033 3-30 7-27 4
    3004 3-30 7-27 5
    6028 3-30 7-27 6
    3010 4-59 3-16 3
    3019 4-59 3-16 3
    6018 4-59 3-16 3
    6026 4-59 3-16 3
    6029 4-59 3-16 3
    6036 4-59 3-16 3
    6037 4-59 3-16 3
    2964 4-59 3-22 3
    3027 4-59 3-16 4
    3046 5-51 5-5  3
    6000 1-69 6-13 4
    6006 1-69 6-6  5
    6008 1-69 6-13 4
  • TABLE 6
    Vκ3-20Jκ1
    Common Light Chain Antibodies
    HCVR
    Antibody VH DH JH
    5989 3-30 3-3  3
    5994 3-33 1-7  4
    5985 3-33 2-15 4
    5987 3-33 2-15 4
    5995 3-33 2-15 4
    2968 4-39 1-26 3
    5988 4-39 1-26 3
    5990 4-39 1-26 3
    5992 4-39 1-26 3
    2975 5-51 6-13 5
    2972 5-51 3-16 6
    5986 5-51 3-16 6
    5993 5-51 3-16 6
    5996 5-51 3-16 6
    5984 3-53 1-1  4
  • Example 7 Determination of Blocking Ability of Antigen-Specific Common Light Chain Antibodies by LUMINEX™ Assay
  • Ninety-eight human common light chain antibodies raised against Antigen E were tested for their ability to block binding of Antigen E's natural ligand (Ligand Y) to Antigen E in a bead-based assay.
  • The extracellular domain (ECD) of Antigen E was conjugated to two myc epitope tags and a 6× histidine tag (Antigen E-mmH) and amine-coupled to carboxylated microspheres at a concentration of 20 μg/mL in MES buffer. The mixture was incubated for two hours at room temperature followed by bead deactivation with 1M Tris pH 8.0 followed by washing in PBS with 0.05% (v/v) Tween-20. The beads were then blocked with PBS (Irvine Scientific, Santa Ana, Calif.) containing 2% (w/v) BSA (Sigma-Aldrich Corp., St. Louis, Mo.). In a 96-well filter plate, supernatants containing Antigen E-specific common light chain antibodies, were diluted 1:15 in buffer. A negative control containing a mock supernatant with the same media components as for the antibody supernatant was prepared. Antigen E-labeled beads were added to the supernatants and incubated overnight at 4° C. Biotinylated-Ligand Y protein was added to a final concentration of 0.06 nM and incubated for two hours at room temperature. Detection of biotinylated-Ligand Y bound to Antigen E-myc-myc-6His labeled beads was determined with R-Phycoerythrin conjugated to Streptavidin (Moss Inc, Pasadena, Md.) followed by measurement in a LUMINEX™ flow cytometry-based analyzer. Background Mean Fluorescence Intensity (MFI) of a sample without Ligand Y was subtracted from all samples. Percent blocking was calculated by division of the background-subtracted MFI of each sample by the adjusted negative control value, multiplying by 100 and subtracting the resulting value from 100.
  • In a similar experiment, the same 98 human common light chain antibodies raised against Antigen E were tested for their ability to block binding of Antigen E to Ligand Y-labeled beads.
  • Briefly, Ligand Y was amine-coupled to carboxylated microspheres at a concentration of 20 μg/mL diluted in MES buffer. The mixture and incubated two hours at room temperature followed by deactivation of beads with 1M Tris pH 8 then washing in PBS with 0.05% (v/v) Tween-20. The beads were then blocked with PBS (Irvine Scientific, Santa Ana, Calif.) containing 2% (w/v) BSA (Sigma-Aldrich Corp., St. Louis, Mo.). In a 96-well filter plate, supernatants containing Antigen E-specific common light chain antibodies were diluted 1:15 in buffer. A negative control containing a mock supernatant with the same media components as for the antibody supernatant was prepared. A biotinylated-Antigen E-mmH was added to a final concentration of 0.42 nM and incubated overnight at 4° C. Ligand Y-labeled beads were then added to the antibody/Antigen E mixture and incubated for two hours at room temperature. Detection of biotinylated-Antigen E-mmH bound to Ligand Y-beads was determined with R-Phycoerythrin conjugated to Streptavidin (Moss Inc, Pasadena, Md.) followed by measurement in a LUMINEX™ flow cytometry-based analyzer. Background Mean Fluorescence Intensity (MFI) of a sample without Antigen E was subtracted from all samples. Percent blocking was calculated by division of the background-subtracted MFI of each sample by the adjusted negative control value, multiplying by 100 and subtracting the resulting value from 100.
  • Tables 7 and 8 show the percent blocking for all 98 anti-Antigen E common light chain antibodies tested in both LUMINEX™ assays. ND: not determined under current experimental conditions.
  • TABLE 7
    Vκ1-39Jκ5
    Common Light Chain Antibodies
    % Blocking of % Blocking of
    Antibody Antigen E-Labeled Beads Antigen E In Solution
    2948 81.1 47.8
    2948G 38.6 ND
    2949 97.6 78.8
    2949G 97.1 73.7
    2950 96.2 81.9
    2950G 89.8 31.4
    2952 96.1 74.3
    2952G 93.5 39.9
    2954 93.7 70.1
    2954G 91.7 30.1
    2955 75.8 30.0
    2955G 71.8 ND
    2964 92.1 31.4
    2964G 94.6 43.0
    2978 98.0 95.1
    2978G 13.9 94.1
    2982 92.8 78.5
    2982G 41.9 52.4
    2985 39.5 31.2
    2985G 2.0 5.0
    2987 81.7 67.8
    2987G 26.6 29.3
    2996 87.3 55.3
    2996G 95.9 38.4
    2997 93.4 70.6
    2997G 9.7 7.5
    3004 79.0 48.4
    3004G 60.3 40.7
    3005 97.4 93.5
    3005G 77.5 75.6
    3010 98.0 82.6
    3010G 97.9 81.0
    3011 87.4 42.8
    3011G 83.5 41.7
    3012 91.0 60.8
    3012G 52.4 16.8
    3013 80.3 65.8
    3013G 17.5 15.4
    3014 63.4 20.7
    3014G 74.4 28.5
    3015 89.1 55.7
    3015G 58.8 17.3
    3016 97.1 81.6
    3016G 93.1 66.4
    3017 94.8 70.2
    3017G 87.9 40.8
    3018 85.4 54.0
    3018G 26.1 12.7
    3019 99.3 92.4
    3019G 99.3 88.1
    3020 96.7 90.3
    3020G 85.2 41.5
    3021 74.5 26.1
    3021G 81.1 27.4
    3022 65.2 17.6
    3022G 67.2 9.1
    3023 71.4 28.5
    3023G 73.8 29.7
    3024 73.9 32.6
    3024G 89.0 10.0
    3025 70.7 15.6
    3025G 76.7 24.3
    3027 96.2 61.6
    3027G 98.6 75.3
    3028 92.4 29.0
    3028G 87.3 28.8
    3030 6.0 10.6
    3030G 41.3 14.2
    3032 76.5 31.4
    3032G 17.7 11.0
    3033 98.2 86.1
    3033G 93.6 64.0
    3036 74.7 32.7
    3036G 90.1 51.2
    3041 95.3 75.9
    3041G 92.4 51.6
    3042 88.1 73.3
    3042G 60.9 25.2
    3043 90.8 65.8
    3043G 92.8 60.3
  • TABLE 8
    Vκ3-20Jκ1
    Common Light Chain Antibodies
    % Blocking of % Blocking of
    Antibody Antigen E-Labeled Beads Antigen E In Solution
    2968 97.1 73.3
    2968G 67.1 14.6
    2969 51.7 20.3
    2969G 37.2 16.5
    2970 92.2 34.2
    2970G 92.7 27.2
    2971 23.4 11.6
    2971G 18.8 18.9
    2972 67.1 38.8
    2972G 64.5 39.2
    2973 77.7 27.0
    2973G 51.1 20.7
    2974 57.8 12.4
    2974G 69.9 17.6
    2975 49.4 18.2
    2975G 32.0 19.5
    2976 1.0 1.0
    2976G 50.4 20.4
  • In the first LUMINEX™ experiment described above, 80 common light chain antibodies containing the Vκ1-39Jκ5 engineered light chain were tested for their ability to block Ligand Y binding to Antigen E-labeled beads. Of these 80 common light chain antibodies, 68 demonstrated>50% blocking, while 12 demonstrated<50% blocking (6 at 25-50% blocking and 6 at <25% blocking). For the 18 common light chain antibodies containing the Vκ3-20Jκ1 engineered light chain, 12 demonstrated>50% blocking, while 6 demonstrated<50% blocking (3 at 25-50% blocking and 3 at <25% blocking) of Ligand Y binding to Antigen E-labeled beads.
  • In the second LUMINEX™ experiment described above, the same 80 common light chain antibodies containing the Vκ1-39Jκ5 engineered light chain were tested for their ability to block binding of Antigen E to Ligand Y-labeled beads. Of these 80 common light chain antibodies, 36 demonstrated>50% blocking, while 44 demonstrated<50% blocking (27 at 25-50% blocking and 17 at <25% blocking). For the 18 common light chain antibodies containing the Vκ3-20Jκ1 engineered light chain, 1 demonstrated>50% blocking, while 17 demonstrated<50% blocking (5 at 25-50% blocking and 12 at <25% blocking) of Antigen E binding to Ligand Y-labeled beads.
  • The data of Tables 7 and 8 establish that the rearrangements described in Tables 5 and 6 generated anti-Antigen E-specific common light chain antibodies that blocked binding of Ligand Y to its cognate receptor Antigen E with varying degrees of efficacy, which is consistent with the anti-Antigen E common light chain antibodies of Tables 5 and 6 comprising antibodies with overlapping and non-overlapping epitope specificity with respect to Antigen E.
  • Example 8 Determination of Blocking Ability of Antigen-Specific Common Light Chain Antibodies by ELISA
  • Human common light chain antibodies raised against Antigen E were tested for their ability to block Antigen E binding to a Ligand Y-coated surface in an ELISA assay.
  • Ligand Y was coated onto 96-well plates at a concentration of 2 pg/mL diluted in PBS and incubated overnight followed by washing four times in PBS with 0.05% Tween-20. The plate was then blocked with PBS (Irvine Scientific, Santa Ana, Calif.) containing 0.5% (w/v) BSA (Sigma-Aldrich Corp., St. Louis, Mo.) for one hour at room temperature. In a separate plate, supernatants containing anti-Antigen E common light chain antibodies were diluted 1:10 in buffer. A mock supernatant with the same components of the antibodies was used as a negative control. Antigen E-mmH (described above) was added to a final concentration of 0.150 nM and incubated for one hour at room temperature. The antibody/Antigen E-mmH mixture was then added to the plate containing Ligand Y and incubated for one hour at room temperature. Detection of Antigen E-mmH bound to Ligand Y was determined with Horse-Radish Peroxidase (HRP) conjugated to anti-Penta-His antibody (Qiagen, Valencia, Calif.) and developed by standard colorimetric response using tetramethylbenzidine (TMB) substrate (BD Biosciences, San Jose, Calif.) neutralized by sulfuric acid. Absorbance was read at OD450 for 0.1 sec. Background absorbance of a sample without Antigen E was subtracted from all samples. Percent blocking was calculated by division of the background-subtracted MFI of each sample by the adjusted negative control value, multiplying by 100 and subtracting the resulting value from 100.
  • Tables 9 and 10 show the percent blocking for all 98 anti-Antigen E common light chain antibodies tested in the ELISA assay. ND: not determined under current experimental conditions.
  • TABLE 9
    Vκ1-39Jκ5
    Common Light Chain Antibodies
    % Blocking of
    Antibody Antigen E In Solution
    2948 21.8
    2948G 22.9
    2949 79.5
    2949G 71.5
    2950 80.4
    2950G 30.9
    2952 66.9
    2952G 47.3
    2954 55.9
    2954G 44.7
    2955 12.1
    2955G 25.6
    2964 34.8
    2964G 47.7
    2978 90.0
    2978G 90.2
    2982 59.0
    2982G 20.4
    2985 10.5
    2985G ND
    2987 31.4
    2987G ND
    2996 29.3
    2996G ND
    2997 48.7
    2997G ND
    3004 16.7
    3004G 3.5
    3005 87.2
    3005G 54.3
    3010 74.5
    3010G 84.6
    3011 19.4
    3011G ND
    3012 45.0
    3012G 12.6
    3013 39.0
    3013G 9.6
    3014 5.2
    3014G 17.1
    3015 23.7
    3015G 10.2
    3016 78.1
    3016G 37.4
    3017 61.6
    3017G 25.2
    3018 40.6
    3018G 14.5
    3019 94.6
    3019G 92.3
    3020 80.8
    3020G ND
    3021 7.6
    3021G 20.7
    3022 2.4
    3022G 15.0
    3023 9.1
    3023G 19.2
    3024 7.5
    3024G 15.2
    3025 ND
    3025G 13.9
    3027 61.4
    3027G 82.7
    3028 40.3
    3028G 12.3
    3030 ND
    3030G 9.5
    3032 ND
    3032G 13.1
    3033 77.1
    3033G 32.9
    3036 17.6
    3036G 24.6
    3041 59.3
    3041G 30.7
    3042 39.9
    3042G 16.1
    3043 57.4
    3043G 46.1
  • TABLE 10
    Vκ3-20Jκ1
    Common Light Chain Antibodies
    % Blocking of
    Antibody Antigen E In Solution
    2968 68.9
    2968G 15.2
    2969 10.1
    2969G 23.6
    2970 34.3
    2970G 41.3
    2971 6.3
    2971G 27.1
    2972 9.6
    2972G 35.7
    2973 20.7
    2973G 23.1
    2974 ND
    2974G 22.0
    2975 8.7
    2975G 19.2
    2976 4.6
    2976G 26.7
  • As described in this Example, of the 80 common light chain antibodies containing the Vκ1-39Jκ5 engineered light chain tested for their ability to block Antigen E binding to a Ligand Y-coated surface, 22 demonstrated>50% blocking, while 58 demonstrated<50% blocking (20 at 25-50% blocking and 38 at <25% blocking). For the 18 common light chain antibodies containing the Vκ3-20Jκ1 engineered light chain, one demonstrated>50% blocking, while 17 demonstrated<50% blocking (5 at 25-50% blocking and 12 at <25% blocking) of Antigen E binding to a Ligand Y-coated surface.
  • These results are also consistent with the Antigen E-specific common light chain antibody pool comprising antibodies with overlapping and non-overlapping epitope specificity with respect to Antigen E.
  • Example 9 BIACORE™ Affinity Determination for Antigen-Specific Common Light Chain Antibodies
  • Equilibrium dissociation constants (KD) for selected antibody supernatants were determined by SPR (Surface Plasmon Resonance) using a BIACORE™ T100 instrument (GE Healthcare). All data was obtained using HBS-EP (10mM Hepes, 150 mM NaCl, 0.3 mM EDTA, 0.05% Surfactant P20, pH 7.4) as both the running and sample buffers, at 25° C. Antibodies were captured from crude supernatant samples on a CM5 sensor chip surface previously derivatized with a high density of anti-human Fc antibodies using standard amine coupling chemistry. During the capture step, supernatants were injected across the anti-human Fc surface at a flow rate of 3 μL/min, for a total of 3 minutes. The capture step was followed by an injection of either running buffer or analyte at a concentration of 100 nM for 2 minutes at a flow rate of 35 μL/min. Dissociation of antigen from the captured antibody was monitored for 6 minutes. The captured antibody was removed by a brief injection of 10 mM glycine, pH 1.5. All sensorgrams were double referenced by subtracting sensorgrams from buffer injections from the analyte sensorgrams, thereby removing artifacts caused by dissociation of the antibody from the capture surface. Binding data for each antibody was fit to a 1:1 binding model with mass transport using BIAcore T100 Evaluation software v2.1. Results are shown in Tables 11 and 12.
  • TABLE 11
    Vκ1-39Jκ5
    Common Light Chain Antibodies
    100 nM Antigen E
    Antibody KD (nM) T1/2 (min)
    2948 8.83 28
    2948G 95.0 1
    2949 3.57 18
    2949G 6.37 9
    2950 4.91 17
    2950G 13.6 5
    2952 6.25 7
    2952G 7.16 4
    2954 2.37 24
    2954G 5.30 9
    2955 14.4 6
    2955G 12.0 4
    2964 14.8 6
    2964G 13.0 9
    2978 1.91 49
    2978G 1.80 58
    2982 6.41 19
    2982G 16.3 9
    2985 64.4 9
    2985G 2.44 8
    2987 21.0 11
    2987G 37.6 4
    2996 10.8 9
    2996G 24.0 2
    2997 7.75 19
    2997G 151 1
    3004 46.5 14
    3004G 1.93 91
    3005 2.35 108
    3005G 6.96 27
    3010 4.13 26
    3010G 2.10 49
    3011 59.1 5
    3011G 41.7 5
    3012 9.71 20
    3012G 89.9 2
    3013 20.2 20
    3013G 13.2 4
    3014 213 4
    3014G 36.8 3
    3015 29.1 11
    3015G 65.9 0
    3016 4.99 17
    3016G 18.9 4
    3017 9.83 8
    3017G 55.4 2
    3018 11.3 36
    3018G 32.5 3
    3019 1.54 59
    3019G 2.29 42
    3020 5.41 39
    3020G 41.9 6
    3021 50.1 6
    3021G 26.8 4
    3022 25.7 17
    3022G 20.8 12
    3023 263 9
    3023G 103 5
    3024 58.8 7
    3024G 7.09 10
    3025 35.2 6
    3025G 42.5 8
    3027 7.15 6
    3027G 4.24 18
    3028 6.89 37
    3028G 7.23 22
    3030 46.2 7
    3030G 128 3
    3032 53.2 9
    3032G 13.0 1
    3033 4.61 17
    3033G 12.0 5
    3036 284 12
    3036G 18.2 10
    3041 6.90 12
    3041G 22.9 2
    3042 9.46 34
    3042G 85.5 3
    3043 9.26 29
    3043G 13.1 22
  • TABLE 12
    Vκ3-20Jκ1
    Common Light Chain Antibodies
    100 nM Antigen E
    Antibody KD (nM) T1/2 (min)
    2968 5.50 8
    2968G 305 0
    2969 34.9 2
    2969G 181 1
    2970G 12.3 3
    2971G 32.8 22
    2972 6.02 13
    2972G 74.6 26
    2973 5.35 39
    2973G 11.0 44
    2974 256 0
    2974G 138 0
    2975 38.0 2
    2975G 134 1
    2976 6.73 10
    2976G 656 8
  • The binding affinities of common light chain antibodies comprising the rearrangements shown in Tables 5 and 6 vary, with nearly all exhibiting a KD in the nanomolar range. The affinity data is consistent with the common light chain antibodies resulting from the combinatorial association of rearranged variable domains described in Tables 5 and 6 being high-affinity, clonally selected, and somatically mutated. Coupled with data previously shown, the common light chain antibodies described in Tables 5 and 6 comprise a collection of diverse, high-affinity antibodies that exhibit specificity for one or more epitopes on Antigen E.
  • Example 10 Determination of Binding Specificities of Antigen-Specific Common Light Chain Antibodies by LUMINEX™ Assay
  • Selected anti-Antigen E common light chain antibodies were tested for their ability to bind to the ECD of Antigen E and Antigen E ECD variants, including the cynomolgous monkey ortholog (Mf Antigen E), which differs from the human protein in approximately 10% of its amino acid residues; a deletion mutant of Antigen E lacking the last 10 amino acids from the C-terminal end of the ECD (Antigen E-ACT); and two mutants containing an alanine substitution at suspected locations of interaction with Ligand Y (Antigen E-Ala1 and AntigenE-Ala2). The Antigen E proteins were produced in CHO cells and each contained a myc-myc-His C-terminal tag.
  • For the binding studies, Antigen E ECD protein or variant protein (described above) from 1 mL of culture medium was captured by incubation for 2 hr at room temperature with 1×106 microsphere (LUMINEX™) beads covalently coated with an anti-myc monoclonal antibody (MAb 9E10, hybridoma cell line CRL-1729™; ATCC, Manassas, Va.). The beads were then washed with PBS before use. Supernatants containing anti-Antigen E common light chain antibodies were diluted 1:4 in buffer and added to 96-well filter plates. A mock supernatant with no antibody was used as negative control. The beads containing the captured Antigen E proteins were then added to the antibody samples (3000 beads per well) and incubated overnight at 4° C. The following day, the sample beads were washed and the bound common light chain antibody was detected with a R-phycoerythrin-conjugated anti-human IgG antibody. The fluorescence intensity of the beads (approximately 100 beads counted for each antibody sample binding to each Antigen E protein) was measured with a LUMINEX™ flow cytometry-based analyzer, and the median fluorescence intensity (MFI) for at least 100 counted beads per bead/antibody interaction was recorded. Results are shown in Tables 13 and 14.
  • TABLE 13
    Vκ1-39Jκ5 Common Light Chain Antibodies
    Mean Fluorescence Intensity (MFI)
    Antigen Antigen Antigen E- Antigen E- Mf
    Antibody E-ECD E-ΔCT Ala1 Ala2 Antigen E
    2948 1503 2746 4953 3579 1648
    2948G 537 662 2581 2150 863
    2949 3706 4345 8169 5678 5142
    2949G 3403 3318 7918 5826 5514
    2950 3296 4292 7756 5171 4749
    2950G 2521 2408 7532 5079 3455
    2952 3384 1619 1269 168 911
    2952G 3358 1001 108 55 244
    2954 2808 3815 7114 5039 3396
    2954G 2643 2711 7620 5406 3499
    2955 1310 2472 4738 3765 1637
    2955G 1324 1802 4910 3755 1623
    2964 5108 1125 4185 346 44
    2964G 4999 729 4646 534 91
    2978 6986 2800 14542 10674 8049
    2978G 5464 3295 11652 8026 6452
    2982 4955 2388 13200 9490 6772
    2982G 3222 2013 8672 6509 4949
    2985 1358 832 4986 3892 1669
    2985G 43 43 128 244 116
    2987 3117 1674 7646 5944 2546
    2987G 3068 1537 9202 6004 4744
    2996 4666 1917 12875 9046 6459
    2996G 2752 1736 8742 6150 4873
    2997 5164 2159 12167 8361 5922
    2997G 658 356 3392 2325 1020
    3004 2794 1397 8542 6268 3083
    3004G 2753 1508 8267 5808 4345
    3005 5683 2221 12900 9864 5868
    3005G 4344 2732 10669 7125 5880
    3010 4829 1617 2642 3887 44
    3010G 3685 1097 2540 3022 51
    3011 2859 2015 7855 5513 3863
    3011G 2005 1072 6194 4041 3181
    3012 3233 2221 8543 5637 3307
    3012G 968 378 3115 2261 1198
    3013 2343 1791 6715 4810 2528
    3013G 327 144 1333 1225 370
    3014 1225 1089 5436 3621 1718
    3014G 1585 851 5178 3705 2411
    3015 3202 2068 8262 5554 3796
    3015G 1243 531 4246 2643 1611
    3016 4220 2543 8920 5999 5666
    3016G 2519 1277 6344 4288 4091
    3017 3545 2553 8700 5547 5098
    3017G 1972 1081 5763 3825 3038
    3018 2339 1971 6140 4515 2293
    3018G 254 118 978 1020 345
    3019 5235 1882 7108 4249 54
    3019G 4090 1270 4769 3474 214
    3020 3883 3107 8591 6602 4420
    3020G 2165 1209 6489 4295 2912
    3021 1961 1472 6872 4641 2742
    3021G 2091 1005 6430 3988 2935
    3022 2418 793 7523 2679 36
    3022G 2189 831 6182 3051 132
    3023 1692 1411 5788 3898 2054
    3023G 1770 825 5702 3677 2648
    3024 1819 1467 6179 4557 2450
    3024G 100 87 268 433 131
    3025 1853 1233 6413 4337 2581
    3025G 1782 791 5773 3871 2717
    3027 4131 1018 582 2510 22
    3027G 3492 814 1933 2596 42
    3028 4361 2545 9884 5639 975
    3028G 2835 1398 7124 3885 597
    3030 463 277 1266 1130 391
    3030G 943 302 3420 2570 1186
    3032 2083 1496 6594 4402 2405
    3032G 295 106 814 902 292
    3033 4409 2774 8971 6331 5825
    3033G 2499 1234 6745 4174 4210
    3036 1755 1362 6137 4041 1987
    3036G 2313 1073 6387 4243 3173
    3041 3674 2655 8629 5837 4082
    3041G 2519 1265 6468 4274 3320
    3042 2653 2137 7277 5124 3325
    3042G 1117 463 4205 2762 1519
    3043 3036 2128 7607 5532 3366
    3043G 2293 1319 6573 4403 3228
  • TABLE 14
    Vκ3-20Jκ1 Common Light Chain Antibodies
    Mean Fluorescence Intensity (MFI)
    Antigen Antigen Antigen E- Antigen E- Mf Antigen
    Antibody E-ECD E-ΔCT Ala1 Ala2 E
    2968 6559 3454 14662 3388 29
    2968G 2149 375 9109 129 22
    2969 2014 1857 7509 5671 3021
    2969G 1347 610 6133 4942 2513
    2970 5518 1324 14214 607 32
    2970G 4683 599 12321 506 31
    2971 501 490 2506 2017 754
    2971G 578 265 2457 2062 724
    2972 2164 2158 8408 6409 3166
    2972G 1730 992 6364 4602 2146
    2973 3527 1148 3967 44 84
    2973G 1294 276 1603 28 44
    2974 1766 722 8821 241 19
    2974G 2036 228 8172 135 26
    2975 1990 1476 8669 6134 2468
    2975G 890 315 4194 3987 1376
    2976 147 140 996 1079 181
    2976G 1365 460 6024 3929 1625
  • The anti-Antigen E common light chain antibody supernatants exhibited high specific binding to the beads linked to Antigen E-ECD. For these beads, the negative control mock supernatant resulted in negligible signal (<10 MFI) when combined with the Antigen E-ECD bead sample, whereas the supernatants containing anti-Antigen E common light chain antibodies exhibited strong binding signal (average MFI of 2627 for 98 antibody supernatants; MFI>500 for 91/98 antibody samples).
  • As a measure of the ability of the selected anti-Antigen E common light chain antibodies to identify different epitopes on the ECD of Antigen E, the relative binding of the antibodies to the variants were determined. All four Antigen E variants were captured to the anti-myc LUMINEX™ beads as described above for the native Antigen E-ECD binding studies, and the relative binding ratios (MFIvariant/MFIAntigen E-ECD) were determined. For 98 tested common light chain antibody supernatants shown in Tables 12 and 13, the average ratios (MFIvariant/MFIAntigen E-ECD) differed for each variant, likely reflecting different capture amounts of proteins on the beads (average ratios of 0.61, 2.9, 2.0, and 1.0 for Antigen E-ΔCT, Antigen E-Ala1, Antigen E-Ala2, and Mf Antigen E, respectively). For each protein variant, the binding for a subset of the 98 tested common light chain antibodies showed greatly reduced binding, indicating sensitivity to the mutation that characterized a given variant. For example, 19 of the common light chain antibody samples bound to the Mf Antigen E with MFIvariant/MFIAntigen E-ECD of <8%. Since many in this group include high or moderately high affinity antibodies (5 with KD <5 nM, 15 with KD<50 nM), it is likely that the lower signal for this group results from sensitivity to the sequence (epitope) differences between native Antigen E-ECD and a given variant rather than from lower affinities.
  • These data establish that the common light chain antibodies described in Tables 5 and 6 represent a diverse group of Antigen-E-specific common light chain antibodies that specifically recognize more than one epitope on Antigen E.
  • Example 11 Light Chain Shuffling in Common Light Chain Antibodies
  • Heavy chains of selected antigen-specific common light chain antibodies were tested for binding to Antigen E after repairing the heavy chains with either a germline Vκ1-39Jκ5 or a germline Vκ3-20Jκ1 engineered light chain (as described in Example 1).
  • Briefly, 247 heavy chains of Antigen E-specific common light chain antibodies (Vκ1-39Jκ5 and Vκ3-20Jκ1) were transfected with either a germline Vκ1-39 or a germline Vκ3-20 engineered light chain and rescreened for binding to Antigen E by a LUMINEX™ assay (as described in Example 7 and Example 10). Binding to Antigen E was confirmed by BIACORE™ (as described in Example 9). The results are shown in Table 15.
  • As shown in this Example, twenty-eight common light chain antibodies specific for Antigen E were capable of binding to Antigen E when repaired with a germline form of the light chain.
  • TABLE 15
    Original Repaired
    Light Chain Light Chain No. Tested No. Confirmed Binders
    1-39 1-39 198 23
    3-20 3-20 49 5
  • Example 12 Heavy Chain Gene Usage and Somatic Hypermutation Frequency in Common Light Chain Antibodies
  • Heavy and light chain sequences (>6000) of antibodies raised in VELCOIMMUNE® mice (e.g., U.S. Pat. No. 6,596,541 and U.S. Pat. No. 7,105,348) were compiled with heavy and light chain sequences (>600) of common light chain antibodies obtained by a multi-antigen immunization scheme employing the engineered light chain mice (described above) to compare heavy chain gene segment usage and somatic hypermutation frequencies of the antibody chains.
  • Heavy Chain Gene Usage. Heavy and light chain sequences obtained from VELOCIMMUNE® mice containing a replacement of the endogenous mouse heavy chain locus with human VH, DH, and JH gene segments and a replacement of the endogenous mouse κ light chain locus with either the engineered germline Vκ1-39Jκ5 human light chain region or the engineered germline Vκ3-20Jκ1 human light chain region (as described in Example 2) immunized with a human cell-surface receptor (Antigen E), a heterodimer of two human cell-surface glycoproteins (Antigen F), a human cytokine receptor (Antigen G) and a human tumor differentiation antigen (Antigen H) were analyzed for heavy chain gene segment usage and VH and JH gene segments were recorded. Results are shown in Tables 16-18. Percentages in Tables 16-18 represent rounded values and in some cases may not equal 100% when added together.
  • Table 16 sets forth the percent heavy chain family usage for antibodies from VELCOIMMUNE® mice (VI), antibodies from VELCOIMMUNE® mice having a cognate W1-39 light chain (VI-Vκ1-39), antibodies from Vκ1-39 engineered light chain mice (Vκ1-39), antibodies from VELCOIMMUNE® mice having a cognate Vκ3-20 light chain (VI-Vκ3-20), and antibodies from Vκ3-20 engineered light chain mice (Vκ3-20). Table 17 sets forth the percent VH and JH gene usage for antibodies from VELCOIMMUNE® mice (VI), antibodies from VELCOIMMUNE® mice having a cognate Vκ1-39 light chain (VI-Vκ1-39), antibodies from Vκ1-39 engineered light chain mice (Vκ1-39), antibodies from VELCOIMMUNE® mice having a cognate Vκ3-20 light chain (VI-Vκ3-20), and antibodies from Vκ3-20 engineered light chain mice (Vκ3-20). Table 18 sets forth the percent VH gene usage for antibodies from Vκ1-39 engineered light chain mice (Vκ1-39 Mice) from each immunization group (Antigens E, F, G and H) and the percent VH gene usage for antibodies from Vκ3-20 engineered light chain mice (Vκ3-20 Mice) from selected immunization groups (Antigens E and G).
  • As shown in this Example, heavy chain gene usage for antigens tested in Vκ1-39Jκ5 engineered light chain mice was characterized by a preponderance of VH family III subgroups (VH3-7, VH3-9, VH3-11, VH3-13, VH3-20, VH3-23, VH3-30, VH3-33 and VH3-48). Notable usage of other VH family subgroups was characterized by usage of VH1-18, VH1-69, VH2-5, VH4-59 and VH6-1. For antigens tested in Vκ3-20Jκ1 engineered light chain mice, heavy chain gene usage was characterized by a preponderance of VH family III, VH family IV and VH family V subgroups (VH3-11, VH3-30, VH3-33, VH4-39, VH4-59 and VH5-51). Notable usage of other VH family subgroups was characterized by usage of VH1-18, VH1-69, VH2-70 and VH6-1.
  • Somatic Hypermutation Frequency. Heavy and light chains from antibodies raised in VELCOIMMUNE® mice and the engineered light chain mice (described above) were aligned to germline sequences according to the heavy and light chain gene usage demonstrated for each heavy and/or light chain. Amino acid changes for each framework region (FW) and complementarity determining region (CDR) for both heavy and light chain of each sequence were calculated. Results are shown in Tables 19-22. Percentages in Tables 21-24 represent rounded values and in some cases may not equal 100% when added together.
  • Table 19 sets forth the number of amino acid (AA) changes observed in each FW and CDR region of heavy chains of antibodies from VELCOIMMUNE® mice, heavy chains of antibodies from Vκ1-39 engineered light chain mice (Vκ1-39 Mice) and heavy chains of antibodies from Vκ3-20 engineered light chain mice (Vκ3-20 Mice). Table 20 sets forth the number of amino acid (AA) changes observed in each FW and CDR region of light chains of antibodies from VELCOIMMUNE® mice, the light chain of antibodies from Vκ1-39 engineered mice (Vκ1-39 Mice) and the light chain of antibodies from Vκ3-20 engineered mice (Vκ3-20 Mice). Table 21 sets forth the number of amino acid (AA) changes observed in each FW and CDR region of heavy chains of antibodies from Vκ1-39 engineered light chain mice (Vκ1-39 Mice) for selected immunization groups (Antigens E, F and H). Table 22 sets forth the number of amino acid (AA) changes observed in each FW and CDR region of heavy chains of antibodies from Vκ3-20 engineered light chain mice (Vκ3-20 Mice) for selected immunization groups (Antigens E and G).
  • TABLE 16
    VH Family VI VI-Vκ1-39 Vκ1-39 VI-Vκ3-20 Vκ3-20
    1 9.0 14.8 3.3 7.1 4.9
    2 2.2 1.8 4.6 0 1.6
    3 77.8 69.8 77.3 61.4 29.5
    4 8.4 8.3 11.2 27.1 39.3
    5 0.9 0 0.7 4.3 23.0
    6 1.7 5.3 3.0 0 1.6
  • TABLE 17
    VI VI-Vκ1-39 Vκ1-39 VI-Vκ3-20 Vκ3-20
    VH Gene
    1-2 3.9 8.3 0 2.9 0
    1-3 0 0 0 0 0
    1-8 1.3 0.6 0 1.4 0
    1-18 3.0 0.6 1.3 2.1 1.6
    1-24 0.4 3.6 0 0.7 0
    1-46 0.1 0 0 0 0
    1-58 0 0 0 0 0
    1-69 0.3 1.8 2.0 0 3.3
    2-5 1.9 0 4.6 0 0
    2-26 0.2 1.8 0.0 0 0
    2-70 0.1 0 0 0 1.6
    3-7 3.0 14.8 0 1.4 0
    3-9 8.5 3.6 29.6 16.4 0
    3-11 5.4 10.7 0 7.1 1.6
    3-13 3.2 1.8 0.7 2.1 0
    3-15 4.0 4.7 0.3 0.7 0
    3-20 1.0 0.6 0.3 5.0 0
    3-21 0.8 0.6 0 2.1 0
    3-23 20.4 8.9 3.3 8.6 0
    3-30 17.6 4.1 35.2 12.9 1.6
    3-33 12.6 14.8 0 5.0 26.2
    3-43 0.2 0.6 0 0 0
    3-48 0.8 1.2 7.2 0 0
    3-53 0.3 3.6 0.3 0 0
    3-64 0 0 0.3 0 0
    3-72 0 0 0 0 0
    3-73 0 0 0 0 0
    4-31 2.7 0 0.7 8.6 0
    4-34 1.8 0.6 0.3 14.3 0
    4-39 1.6 0.6 3.0 2.1 14.8
    4-59 2.3 7.1 7.2 2.1 24.6
    5-51 0.9 0 0.7 4.3 23.0
    6-1 1.7 5.3 3.0 0 1.6
    JH Gene
    1 1.5 1.2 7.1 0 0
    2 4.5 2.4 0.7 5.0 26.9
    3 10.5 16.6 13.1 13.6 26.9
    4 44.0 34.3 32.3 50.7 9.6
    5 9.6 10.1 16.8 7.9 1.9
    6 29.7 35.5 30.0 22.9 34.6
  • TABLE 18
    Vκ1-39 Mice Vκ3-20 Mice
    Antigen Antigen Antigen Antigen Antigen Antigen
    VH Gene E F G H E G
    1-2  0 0 0 0 0 0
    1-3  0 0 0 0 0 0
    1-8  0 0 0 0 0 0
    1-18 0 0 0 8.3 0 3.1
    1-24 0 0 0 0 0 0
    1-46 0 0 0 0 0 0
    1-58 0 0 0 0 0 0
    1-69 2.9 0 25.0 0 0 6.3
    2-5  8.2 0 0 0 0 0
    2-26 0 0 0 0 0 0
    2-70 0 0 0 0 0 3.1
    3-7  0 0 0 0 0 0
    3-9  1.2 98.8 0 14.6 0 0
    3-11 0 0 0 0 0 3.1
    3-13 0.6 0 25.0 0 0 0
    3-15 0 1.2 0 0 0 0
    3-20 0 0 25.0 0 0 0
    3-21 0 0 0 0 0 0
    3-23 4.1 0 25.0 4.2 0 0
    3-30 62.9 0 0 0 3.4 0
    3-33 0 0 0 0 13.8 37.5
    3-43 0 0 0 0 0 0
    3-48 0.6 0 0 43.8 0 0
    3-53 1.6 0 0 0 0 0
    3-64 1.6 0 0 0 0 0
    3-72 0 0 0 0 0 0
    3-73 0 0 0 0 0 0
    4-31 0 0 0 4.2 0 0
    4-34 0 0 0 2.1 0 0
    4-39 5.3 0 0 0 31.0 0
    4-59 11.8 0 0 4.2 3.4 43.8
    5-51 1.2 0 0 0 48.3 0
    6-1  0 0 0 18.8 0 3.1
  • TABLE 19
    Heavy Chains of Antibodies from
    VELCOIMMUNE ® Mice
    # AA Changes FW1 CDR1 FW2 CDR2 FW3 CDR3
    0 63 32 36 26 12 82
    1 23 32 41 31 22 17
    2 9 25 17 23 27 1
    3 4 10 5 16 13 0
    4 0 1 1 3 12 0
    >5 1 0 0 1 14 0
    Heavy Chains of Antibodies from Vκ1-39 Mice
    # AA Changes FW1 CDR1 FW2 CDR2 FW3 CDR3
    0 65 8 34 30 9 37
    1 25 26 35 34 19 54
    2 9 44 23 20 33 9
    3 1 19 8 12 22 0
    4 0 3 0 5 11 0
    >5 1 0 0 0 7 0
    Heavy Chains of Antibodies from Vκ3-20 Mice
    #AA Changes FW1 CDR1 FW2 CDR2 FW3 CDR3
    0 57 8 54 16 8 93
    1 41 43 34 39 21 7
    2 2 25 10 18 20 0
    3 0 15 2 21 13 0
    4 0 10 0 3 20 0
    >5 0 0 0 2 18 0
  • TABLE 20
    Light Chains of Antibodies from
    VELCOIMMUNE ® Mice
    # AA Changes FW1 CDR1 FW2 CDR2 FW3 CDR3
    0 65 24 49 60 33 23
    1 24 20 34 31 27 38
    2 9 27 16 9 18 28
    3 1 20 1 0 14 7
    4 0 7 0 0 4 3
    >5 1 1 0 0 3 0
    Light Chains of Antibodies from Vκ1-39 Mice
    # AA Changes FW1 CDR1 FW2 CDR2 FW3 CDR3
    0 91 75 80 90 71 63
    1 9 19 17 10 21 27
    2 0 5 1 1 5 8
    3 0 0 1 0 2 1
    4 0 0 0 0 2 1
    >5 0 0 0 0 0 0
    Light Chains of Antibodies from Vκ3-20 Mice
    #AA Changes FW1 CDR1 FW2 CDR2 FW3 CDR3
    0 90 47 93 97 63 57
    1 10 27 3 3 20 43
    2 0 27 3 0 17 0
    3 0 0 0 0 0 0
    4 0 0 0 0 0 0
    >5 0 0 0 0 0 0
  • TABLE 21
    Heavy Chains of Anti-Antigen E Antibodies
    from Vκ1-39 Mice
    # AA Changes FW1 CDR1 FW2 CDR2 FW3 CDR3
    0 75 8 49 41 14 36
    1 21 25 33 35 25 52
    2 4 43 14 18 28 12
    3 0 20 4 5 16 0
    4 0 5 0 1 12 0
    >5 1 0 0 0 5 0
    Heavy Chains of Anti-Antigen F Antibodies
    from Vκ1-39 Mice
    # AA Changes FW1 CDR1 FW2 CDR2 FW3 CDR3
    0 52 0 6 6 2 15
    1 35 24 32 35 15 78
    2 11 59 46 22 49 7
    3 0 17 16 24 29 0
    4 0 0 0 12 4 0
    >5 1 0 0 0 1 0
    Heavy Chains of Anti-Antigen H Antibodies
    from Vκ1-39 Mice
    # AA Changes FW1 CDR1 FW2 CDR2 FW3 CDR3
    0 54 21 29 33 4 77
    1 17 35 50 27 6 23
    2 23 21 15 21 25 0
    3 6 21 4 15 27 0
    4 0 2 2 2 15 0
    >5 0 0 0 2 23 0
  • TABLE 22
    Heavy Chains of Anti-Antigen E Antibodies
    from Vκ3-20 Mice
    # AA Changes FW1 CDR1 FW2 CDR2 FW3 CDR3
    0 79 17 62 24 17 90
    1 21 28 34 55 31 10
    2 0 28 3 21 24 0
    3 0 14 0 0 10 0
    4 0 14 0 0 3 0
    >5 0 0 0 0 14 0
    Heavy Chains of Anti-Antigen G Antibodies
    from Vκ3-20 Mice
    # AA Changes FW1 CDR1 FW2 CDR2 FW3 CDR3
    0 38 0 47 9 0 97
    1 59 56 34 25 13 3
    2 3 22 16 16 16 0
    3 0 16 3 41 16 0
    4 0 6 0 6 34 0
    >5 0 0 0 3 22 0

Claims (26)

1. A mouse that expresses a population of antigen-specific antibodies in response to challenge with an antigen, wherein all of the immunoglobulin light chains of the population of antigen-specific antibodies comprise a human light chain variable (VL) region derived from the same single human VL gene segment and the immunoglobulin heavy chains comprise a human heavy chain variable (VH) region derived from a human VH gene segment selected from VH1-2, VH1-3, VH1-8, VH1-18, VH1-24, VH1-46, VH1-58, VH1-69, VH2-5, VH2-26, VH2-70, VH3-7, VH3-9, VH3-11, VH3-13, VH3-15, VH3-20, VH3-21, VH3-23, VH3-30, VH3-33, VH3-43, VH3-48, VH3-53, VH3-64, VH3-72, VH3-73, VH4-31, VH4-34, VH4-39, VH4-59, VH5-51, VH6-1.
2. The mouse of claim 1, wherein the same single human VL gene segment is a human Vκ1-39 gene segment.
3. The mouse of claim 1, wherein the same single human VL gene segment is a human Vκ3-20 gene segment.
4. The mouse of claim 1, wherein all of the immunoglobulin light chains comprise a human light chain J (JL) gene segment.
5. The mouse of claim 4, wherein the human JL gene segment is selected from a human Jκ1 and a Jκ5 gene segment.
6. The mouse of claim 1, wherein the mouse lacks a sequence selected from a mouse immunoglobulin VL gene segment, a mouse immunoglobulin JL gene segment, and a combination thereof.
7. The mouse of claim 1, wherein the human VL region is operably linked to a human, mouse, or rat immunoglobulin light chain constant (CL) region.
8. The mouse of claim 1, wherein the human VL region is expressed from an endogenous immunoglobulin light chain locus.
9. The mouse of claim 1, wherein the human VH region is operably linked to a human, mouse, or rat immunoglobulin heavy chain constant (CH) region.
10. The mouse of claim 1, wherein the human VH region is expressed from an endogenous immunoglobulin heavy chain locus.
11. A mouse that expresses a population of antigen-specific antibodies in response to challenge with an antigen, wherein all of the immunoglobulin light chains of the population of antigen-specific antibodies comprise a human VL region derived from a human Vκ1-39 gene segment and the immunoglobulin heavy chains comprise a human heavy chain variable (VH) region derived from a human VH gene segment selected from VH1-18, VH1-69, VH2-5, VH3-9, VH3-13, VH3-15, VH3-20, VH3-23, VH3-30, VH3-48, VH3-53, VH3-64, VH4-31, VH4-34, VH4-39, VH4-59, VH5-51 and VH6-1.
12. The mouse of claim 11, wherein the human VL region comprises a Jκ5 sequence.
13. The mouse of claim 11, wherein the mouse lacks a mouse immunoglobulin VL and/or JL. gene segment.
14. The mouse of claim 11, wherein the human VL region is operably linked to a human, rat, or mouse immunoglobulin light chain constant (CL) region.
15. The mouse of claim 11, wherein the human VL region is expressed from an endogenous immunoglobulin light chain locus.
16. The mouse of claim 11, wherein the human VH region comprises a sequence that encodes a human JH region selected from a human JH1, JH2, JH3, JH4, JH5, and JH6 gene segment.
17. The mouse of claim 11, wherein the human VH region is operably linked to a human, rat, or mouse immunoglobulin heavy chain constant (CH) region.
18. The mouse of claim 11, wherein the human VH region is expressed from an endogenous immunoglobulin heavy chain locus.
19. A mouse that expresses a population of antigen-specific antibodies in response to challenge with an antigen, wherein all of the immunoglobulin light chains of the population of antigen-specific antibodies comprise a human VL region derived from a human Vκ3-20 gene segment and the immunoglobulin heavy chains comprise a human heavy chain variable (VH) region derived from a human VH gene segment selected from VH1-18, VH1-69, VH2-70, VH3-11, VH3-30, VH3-33, VH3-53, VH4-39, VH4-59, VH5-51 and VH6-1.
20. The mouse of claim 19, wherein the human VL region comprises a Jκ5 sequence.
21. The mouse of claim 19, wherein the mouse lacks a mouse immunoglobulin VL and/or JL gene segment.
22. The mouse of claim 19, wherein the human VL region is operably linked to a human, rat, or mouse immunoglobulin light chain constant (CL) region.
23. The mouse of claim 19, wherein the human VL region is expressed from an endogenous immunoglobulin light chain locus.
24. The mouse of claim 19, wherein the human VH region comprises a sequence that encodes a human JH region selected from a human JH2, JH3, JH4, JH5, and JH6 gene segment.
25. The mouse of claim 19, wherein the human VH region is operably linked to a human, rat, or mouse immunoglobulin heavy chain constant (CH) region.
26. The mouse of claim 19, wherein the human VH region is expressed from an endogenous immunoglobulin heavy chain locus.
US13/412,936 2010-02-08 2012-03-06 Common Light Chain Mouse Abandoned US20120192300A1 (en)

Priority Applications (32)

Application Number Priority Date Filing Date Title
US13/412,936 US20120192300A1 (en) 2010-02-08 2012-03-06 Common Light Chain Mouse
US13/488,628 US20130045492A1 (en) 2010-02-08 2012-06-05 Methods For Making Fully Human Bispecific Antibodies Using A Common Light Chain
CN201611201918.8A CN107090471A (en) 2012-03-06 2013-03-05 Common light chain mouse
IN8163DEN2014 IN2014DN08163A (en) 2012-03-06 2013-03-05
MX2014010794A MX353278B (en) 2012-03-06 2013-03-05 Common light chain mouse.
KR20147026624A KR20140136462A (en) 2012-03-06 2013-03-05 Common light chain mouse
PCT/US2013/029125 WO2013134263A1 (en) 2012-03-06 2013-03-05 Common light chain mouse
NZ630637A NZ630637A (en) 2012-03-06 2013-03-05 Common light chain mouse
CN201380012671.3A CN104244709B (en) 2012-03-06 2013-03-05 Common light chain mice
EP13712618.1A EP2822379A1 (en) 2012-03-06 2013-03-05 Common light chain mouse
SG11201405088QA SG11201405088QA (en) 2012-03-06 2013-03-05 Common light chain mouse
EP16203839.2A EP3165086A1 (en) 2012-03-06 2013-03-05 Common light chain mouse
RU2014140239A RU2683514C2 (en) 2012-03-06 2013-03-05 Common light chain mouse
SG10201610294RA SG10201610294RA (en) 2010-02-08 2013-03-05 Common light chain mouse
CA2865029A CA2865029A1 (en) 2012-03-06 2013-03-05 Common light chain mouse
AU2013204140A AU2013204140B2 (en) 2012-03-06 2013-03-05 Common light chain mouse
JP2014561037A JP2015509380A (en) 2012-03-06 2013-03-05 Common light chain mice
US13/798,310 US20130185821A1 (en) 2010-02-08 2013-03-13 Common Light Chain Mouse
US13/798,455 US9796788B2 (en) 2010-02-08 2013-03-13 Mice expressing a limited immunoglobulin light chain repertoire
IL234207A IL234207A0 (en) 2012-03-06 2014-08-19 Genetically modified mice expressing a common reverse chimeric light chain and reverse chimeric heavy chains
US14/473,970 US9969814B2 (en) 2010-02-08 2014-08-29 Methods for making fully human bispecific antibodies using a common light chain
HK15106625.3A HK1205873A1 (en) 2012-03-06 2015-07-10 Common light chain mouse
US15/056,713 US20160219847A1 (en) 2010-02-08 2016-02-29 Common light chain mouse
AU2016202488A AU2016202488C1 (en) 2012-03-06 2016-04-20 Common light chain mouse
US15/700,973 US10167344B2 (en) 2010-02-08 2017-09-11 Mice expressing a limited immunoglobulin light chain repertoire
US15/891,987 US20190021295A1 (en) 2010-02-08 2018-02-08 Common light chain mouse
US15/951,130 US20190071519A1 (en) 2010-02-08 2018-04-11 Methods for making fully human bispecific antibodies using a common light chain
JP2018088350A JP2018138047A (en) 2010-02-08 2018-05-01 Common light chain mouse
US16/128,360 US10412940B2 (en) 2010-02-08 2018-09-11 Mice expressing a limited immunoglobulin light chain repertoire
US16/159,496 US20190090462A1 (en) 2010-02-08 2018-10-12 Common light chain mouse
US16/530,030 US11026407B2 (en) 2010-02-08 2019-08-02 Mice expressing a limited immunoglobulin light chain repertoire
US17/238,710 US20210315189A1 (en) 2010-02-08 2021-04-23 Mice expressing a limited immunoglobulin light chain repertoire

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US30228210P 2010-02-08 2010-02-08
US13/022,759 US10143186B2 (en) 2010-02-08 2011-02-08 Common light chain mouse
US13/093,156 US20120021409A1 (en) 2010-02-08 2011-04-25 Common Light Chain Mouse
US13/412,936 US20120192300A1 (en) 2010-02-08 2012-03-06 Common Light Chain Mouse

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US13/093,156 Continuation-In-Part US20120021409A1 (en) 2010-02-08 2011-04-25 Common Light Chain Mouse

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US13/488,628 Continuation-In-Part US20130045492A1 (en) 2010-02-08 2012-06-05 Methods For Making Fully Human Bispecific Antibodies Using A Common Light Chain

Publications (1)

Publication Number Publication Date
US20120192300A1 true US20120192300A1 (en) 2012-07-26

Family

ID=43663643

Family Applications (5)

Application Number Title Priority Date Filing Date
US13/022,759 Active US10143186B2 (en) 2010-02-08 2011-02-08 Common light chain mouse
US13/412,936 Abandoned US20120192300A1 (en) 2010-02-08 2012-03-06 Common Light Chain Mouse
US13/948,818 Abandoned US20130302836A1 (en) 2010-02-08 2013-07-23 Common light chain mouse
US16/165,987 Active 2031-02-16 US10986820B2 (en) 2010-02-08 2018-10-19 Common light chain mouse
US17/209,964 Pending US20210204531A1 (en) 2010-02-08 2021-03-23 Common light chain mouse

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US13/022,759 Active US10143186B2 (en) 2010-02-08 2011-02-08 Common light chain mouse

Family Applications After (3)

Application Number Title Priority Date Filing Date
US13/948,818 Abandoned US20130302836A1 (en) 2010-02-08 2013-07-23 Common light chain mouse
US16/165,987 Active 2031-02-16 US10986820B2 (en) 2010-02-08 2018-10-19 Common light chain mouse
US17/209,964 Pending US20210204531A1 (en) 2010-02-08 2021-03-23 Common light chain mouse

Country Status (30)

Country Link
US (5) US10143186B2 (en)
EP (4) EP3095871B1 (en)
JP (8) JP5955781B2 (en)
KR (5) KR102283195B1 (en)
CN (2) CN102791866B (en)
AU (1) AU2011213585B2 (en)
BR (2) BR122014002928A2 (en)
CA (1) CA2789154A1 (en)
CY (2) CY1118241T1 (en)
DK (3) DK2501817T4 (en)
ES (3) ES2603559T5 (en)
HK (2) HK1231505A1 (en)
HR (3) HRP20151188T4 (en)
HU (3) HUE026229T2 (en)
IL (3) IL221265B (en)
LT (2) LT2505654T (en)
ME (2) ME02288B (en)
MX (3) MX350983B (en)
MY (1) MY166529A (en)
NZ (4) NZ601679A (en)
PL (3) PL3095871T3 (en)
PT (3) PT2505654T (en)
RS (3) RS55315B2 (en)
RU (2) RU2571205C2 (en)
SG (4) SG10201500821UA (en)
SI (3) SI2505654T2 (en)
SM (2) SMT201500283B (en)
TR (1) TR201906650T4 (en)
WO (1) WO2011097603A1 (en)
ZA (1) ZA201205944B (en)

Cited By (35)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100146647A1 (en) * 2008-06-27 2010-06-10 Merus B. V. Antibody producing non-human mammals
US20110195454A1 (en) * 2010-02-08 2011-08-11 Regeneron Pharmaceuticals, Inc. Common Light Chain Mouse
WO2013138712A1 (en) 2012-03-16 2013-09-19 Regeneron Pharmaceuticals, Inc. Non-human animals expressing ph-sensitive immunoglobulin sequences
WO2013138680A1 (en) 2012-03-16 2013-09-19 Regeneron Pharmaceuticals, Inc. Histidine engineered light chain antibodies and genetically modified non-human animals for generating the same
EP2147594B1 (en) * 2008-06-27 2013-11-13 Merus B.V. Antibody producing non-human mammals
WO2014022540A1 (en) 2012-08-02 2014-02-06 Regeneron Pharmaceuticals, Inc. Multivalent antigen-binding proteins
NL2012424A (en) * 2013-03-13 2014-09-16 Regeneron Pharma Common light chain mouse.
WO2015042250A1 (en) 2013-09-18 2015-03-26 Regeneron Pharmaceuticals, Inc. Histidine engineered light chain antibodies and genetically modified non-human animals for generating the same
WO2015143414A2 (en) 2014-03-21 2015-09-24 Regeneron Pharmaceuticals, Inc. Non-human animals that make single domain binding proteins
WO2015143406A2 (en) 2014-03-21 2015-09-24 Regeneron Pharmaceuticals, Inc. Vl antigen binding proteins exhibiting distinct binding characteristics
US9145588B2 (en) 2011-09-26 2015-09-29 Merus Biopharmaceuticals B.V. Generation of binding molecules
EP2967012A1 (en) 2013-03-14 2016-01-20 Erasmus University Medical Center Rotterdam Transgenic non-human mammal for antibody production
US9301510B2 (en) 2012-03-16 2016-04-05 Regeneron Pharmaceuticals, Inc. Mice that produce antigen-binding proteins with pH-dependent binding characteristics
US9332742B2 (en) 2012-03-16 2016-05-10 Regeneron Pharmaceuticals, Inc. Histidine engineered light chain antibodies and genetically modified non-human animals for generating the same
WO2017123804A1 (en) 2016-01-13 2017-07-20 Regeneron Pharmaceuticals, Inc. Rodents having an engineered heavy chain diversity region
US9738701B2 (en) 2003-05-30 2017-08-22 Merus N.V. Method for selecting a single cell expressing a heterogeneous combination of antibodies
US9758805B2 (en) 2012-04-20 2017-09-12 Merus N.V. Methods and means for the production of Ig-like molecules
US9796788B2 (en) 2010-02-08 2017-10-24 Regeneron Pharmaceuticals, Inc. Mice expressing a limited immunoglobulin light chain repertoire
WO2017201476A1 (en) 2016-05-20 2017-11-23 Regeneron Pharmaceuticals, Inc. Methods for breaking immunological tolerance using multiple guide rnas
WO2017210586A1 (en) 2016-06-03 2017-12-07 Regeneron Pharmaceuticals, Inc. Non-human animals expressing exogenous terminal deoxynucleotidyltransferase
US9969814B2 (en) 2010-02-08 2018-05-15 Regeneron Pharmaceuticals, Inc. Methods for making fully human bispecific antibodies using a common light chain
US10130081B2 (en) 2011-08-05 2018-11-20 Regeneron Pharmaceuticals, Inc. Humanized universal light chain mice
WO2019190922A1 (en) 2018-03-24 2019-10-03 Regeneron Pharmaceuticals, Inc. Genetically modified non-human animals for generating therapeutic antibodies against peptide-mhc complexes, methods of making and uses thereof
USRE47770E1 (en) 2002-07-18 2019-12-17 Merus N.V. Recombinant production of mixtures of antibodies
WO2019241692A1 (en) 2018-06-14 2019-12-19 Regeneron Pharmaceuticals, Inc. Non-human animals capable of dh-dh rearrangement in the immunoglobulin heavy chain coding sequences
WO2020172505A1 (en) 2019-02-22 2020-08-27 Regeneron Pharmaceuticals, Inc. Rodents having genetically modified sodium channels and methods of use thereof
WO2021026409A1 (en) 2019-08-08 2021-02-11 Regeneron Pharmaceuticals, Inc. Novel antigen binding molecule formats
US10934571B2 (en) 2002-07-18 2021-03-02 Merus N.V. Recombinant production of mixtures of antibodies
US10954310B2 (en) 2010-08-02 2021-03-23 Regeneran Pharmaceuticals, Inc. Mice that make VL binding proteins
WO2021113297A1 (en) 2019-12-02 2021-06-10 Regeneron Pharmaceuticals, Inc. Peptide-mhc ii protein constructs 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
WO2022140219A1 (en) 2020-12-23 2022-06-30 Regeneron Pharmaceuticals, Inc. Nucleic acids encoding anchor modified antibodies and uses thereof
WO2022140494A1 (en) 2020-12-23 2022-06-30 Regeneron Pharmaceuticals, Inc. Methods for obtaining antibodies that bind transmembrane proteins and cells that produce the same
US11773176B2 (en) 2020-01-24 2023-10-03 Aprilbio Co., Ltd. Multispecific antibodies, compositions comprising the same, and vectors and uses thereof
US11877565B2 (en) 2008-12-18 2024-01-23 Erasmus University Medical Center Antibody production

Families Citing this family (152)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6596541B2 (en) 2000-10-31 2003-07-22 Regeneron Pharmaceuticals, Inc. Methods of modifying eukaryotic cells
US20050144655A1 (en) 2000-10-31 2005-06-30 Economides Aris N. Methods of modifying eukaryotic cells
PL2064325T3 (en) 2006-09-01 2012-05-31 Therapeutic Human Polyclonals Inc Enhanced expression of human or humanized immunoglobulin in non-human transgenic animals
US9676845B2 (en) 2009-06-16 2017-06-13 Hoffmann-La Roche, Inc. Bispecific antigen binding proteins
EP2564695B1 (en) 2009-07-08 2015-04-15 Kymab Limited Animal models and therapeutic molecules
US9445581B2 (en) 2012-03-28 2016-09-20 Kymab Limited Animal models and therapeutic molecules
US20120204278A1 (en) 2009-07-08 2012-08-09 Kymab Limited Animal models and therapeutic molecules
US20120021409A1 (en) * 2010-02-08 2012-01-26 Regeneron Pharmaceuticals, Inc. Common Light Chain Mouse
DK2905338T3 (en) * 2010-06-22 2017-11-06 Regeneron Pharma TRANSGENE MICE WITH A MODIFIED ENDOGENT LAMBDA IMMUNGLOBULIN LOCUS
CN105950654B (en) * 2010-11-27 2020-03-20 朱坚 Humanized transgenic animal
ES2758974T5 (en) 2011-02-25 2023-06-08 Regeneron Pharma ADAM6 Mice
ES2549638T3 (en) 2011-02-28 2015-10-30 F. Hoffmann-La Roche Ag Antigen binding proteins
CN103403025B (en) 2011-02-28 2016-10-12 弗·哈夫曼-拉罗切有限公司 Monovalent antigen binding protein
WO2013041844A2 (en) 2011-09-19 2013-03-28 Kymab Limited Antibodies, variable domains & chains tailored for human use
JP2014531452A (en) * 2011-09-19 2014-11-27 カイマブ・リミテッド Animals, repertoire and methods
GB2495083A (en) * 2011-09-26 2013-04-03 Kymab Ltd Human VpreB and chimaeric surrogate light chains in transgenic non-human vertebrates
WO2013045916A1 (en) 2011-09-26 2013-04-04 Kymab Limited Chimaeric surrogate light chains (slc) comprising human vpreb
EP3628329B1 (en) * 2011-09-30 2022-08-10 Dana-Farber Cancer Institute, Inc. Therapeutic peptides comprising antibodies binding to mhc class 1 polypeptide related sequence a (mica)
ES2906462T3 (en) 2011-10-17 2022-04-18 Regeneron Pharma Restricted immunoglobulin heavy chain mice
US20140283153A1 (en) 2011-10-28 2014-09-18 Trianni, Inc. Transgenic animals and methods of use
US9253965B2 (en) 2012-03-28 2016-02-09 Kymab Limited Animal models and therapeutic molecules
KR101924805B1 (en) 2011-12-20 2018-12-04 리제너론 파마슈티칼스 인코포레이티드 Humanized light chain mice
RU2628305C2 (en) 2012-03-02 2017-08-15 Регенерон Фармасьютиказ, Инк. Human antibodies to clostridium difficile toxins
EP2822379A1 (en) * 2012-03-06 2015-01-14 Regeneron Pharmaceuticals, Inc. Common light chain mouse
AU2015227453B2 (en) * 2012-03-16 2017-05-25 Regeneron Pharmaceuticals, Inc. Non-human animals expressing ph-sensitive immunoglobulin sequences
GB2502127A (en) 2012-05-17 2013-11-20 Kymab Ltd Multivalent antibodies and in vivo methods for their production
US10251377B2 (en) 2012-03-28 2019-04-09 Kymab Limited Transgenic non-human vertebrate for the expression of class-switched, fully human, antibodies
JP6267689B2 (en) * 2012-05-10 2018-01-24 バイオアトラ、エルエルシー Multispecific monoclonal antibody
SG10201609535TA (en) * 2012-06-05 2017-01-27 Regeneron Pharma Methods for making fully human bispecific antibodies using a common light chain
KR102381716B1 (en) 2012-06-12 2022-04-01 리제너론 파마슈티칼스 인코포레이티드 Humanized non-human animals with restricted immunoglobulin heavy chain loci
JO3462B1 (en) 2012-08-22 2020-07-05 Regeneron Pharma Human Antibodies to GFR?3 and methods of use thereof
JOP20200236A1 (en) 2012-09-21 2017-06-16 Regeneron Pharma Anti-cd3 antibodies, bispecific antigen-binding molecules that bind cd3 and cd20, and uses thereof
SI2900694T1 (en) * 2012-09-27 2018-12-31 Merus N.V. Bispecific igg antibodies as t cell engagers
AU2014205086B2 (en) 2013-01-14 2019-04-18 Xencor, Inc. Novel heterodimeric proteins
SI2958938T1 (en) * 2013-02-20 2019-08-30 Regeneron Pharmaceuticals, Inc. Mice expressing humanized t-cell co-receptors
WO2014130690A1 (en) * 2013-02-20 2014-08-28 Regeneron Pharmaceuticals, Inc. Non-human animals with modified immunoglobulin heavy chain sequences
CN105164154B (en) * 2013-02-22 2019-06-07 瑞泽恩制药公司 Express the mouse of humanization major histocompatibility complex
CN105208855B (en) * 2013-03-11 2018-04-27 瑞泽恩制药公司 The transgenic mice of chimeric major histocompatibility complex (MHC) the II quasi-molecules of expression
WO2014160202A1 (en) * 2013-03-13 2014-10-02 Regeneron Pharmaceuticals, Inc. Mice expressing a limited immunoglobulin light chain repertoire
US10858417B2 (en) 2013-03-15 2020-12-08 Xencor, Inc. Heterodimeric proteins
US9788534B2 (en) 2013-03-18 2017-10-17 Kymab Limited Animal models and therapeutic molecules
KR102049990B1 (en) 2013-03-28 2019-12-03 삼성전자주식회사 Fusion protein comprising anti-c-Met antibody and VEGF binding fragment
EP2992012B1 (en) 2013-04-29 2019-07-17 F.Hoffmann-La Roche Ag Human fcrn-binding modified antibodies and methods of use
US9783618B2 (en) 2013-05-01 2017-10-10 Kymab Limited Manipulation of immunoglobulin gene diversity and multi-antibody therapeutics
US20150033372A1 (en) * 2013-05-01 2015-01-29 Kymab Limited Human VpreB & Chimaeric Surrogate Light Chains in Transgenic Non-Human Vertebrates
US11707056B2 (en) 2013-05-02 2023-07-25 Kymab Limited Animals, repertoires and methods
US9783593B2 (en) 2013-05-02 2017-10-10 Kymab Limited Antibodies, variable domains and chains tailored for human use
BR112016006502A2 (en) 2013-09-30 2017-09-19 Chugai Pharmaceutical Co Ltd METHOD FOR THE PRODUCTION OF ANTIGEN-BINDING MOLECULE USING MODIFIED HELPER PHAGUE
DE112014004537T5 (en) * 2013-10-01 2016-07-21 Kymab Limited Animal models and therapeutic molecules
MX2016003593A (en) 2013-10-11 2016-06-02 Hoffmann La Roche Multispecific domain exchanged common variable light chain antibodies.
EP3842455A1 (en) 2014-01-15 2021-06-30 F. Hoffmann-La Roche AG Fc-region variants with improved protein a-binding
WO2015107026A1 (en) 2014-01-15 2015-07-23 F. Hoffmann-La Roche Ag Fc-region variants with modified fcrn- and maintained protein a-binding properties
TWI680138B (en) 2014-01-23 2019-12-21 美商再生元醫藥公司 Human antibodies to pd-l1
TWI681969B (en) 2014-01-23 2020-01-11 美商再生元醫藥公司 Human antibodies to pd-1
IL301147A (en) 2014-02-28 2023-05-01 Merus Nv Antibody that binds erbb-2 and erbb-3
NZ724013A (en) 2014-02-28 2019-11-29 Merus Nv Antibodies that bind egfr and erbb3
TWI701042B (en) 2014-03-19 2020-08-11 美商再生元醫藥公司 Methods and antibody compositions for tumor treatment
MA44179B1 (en) * 2014-05-13 2020-10-28 Univ Pennsylvania Compositions comprising an adeno-associated virus expressing double antibody constructs and their uses
EA037006B1 (en) 2014-06-06 2021-01-26 Бристол-Майерс Сквибб Компани Antibodies against glucocorticoid-induced tumor necrosis factor receptor (gitr) and uses thereof
BR112017005245A2 (en) 2014-09-19 2017-12-12 Regeneron Pharma genetically modified nonhuman animal, methods for producing t cell, t cell hybridoma, nucleic acid, specific antibody, human cell, genetically modified nonhuman animal and for inducing an immune response, cell, t cell hybridoma, acid nucleic acid, specific antibody, chimeric antigen receptor, non-human embryo, chimeric antigen receptor locus, and nucleic acid composition.
AU2015327819B2 (en) 2014-10-03 2021-07-01 Massachusetts Institute Of Technology Antibodies that bind ebola glycoprotein and uses thereof
AR102522A1 (en) 2014-11-06 2017-03-08 Hoffmann La Roche FC REGION VARIATIONS WITH MODIFIED PROPERTIES OF UNION TO FCRN AND PROTEIN A
WO2016081490A1 (en) 2014-11-17 2016-05-26 Regeneron Pharmaceuticals, Inc. Methods for tumor treatment using cd3xcd20 bispecific antibody
RS60631B1 (en) 2014-11-21 2020-09-30 Bristol Myers Squibb Co Antibodies against cd73 and uses thereof
CA2967426A1 (en) 2014-11-26 2016-06-02 Xencor, Inc. Heterodimeric antibodies that bind cd3 and tumor antigens
US10259887B2 (en) 2014-11-26 2019-04-16 Xencor, Inc. Heterodimeric antibodies that bind CD3 and tumor antigens
TWI702229B (en) 2014-12-19 2020-08-21 美商再生元醫藥公司 Human antibodies to influenza hemagglutinin
PT3233912T (en) 2014-12-19 2021-08-09 Regenesance B V Antibodies that bind human c6 and uses thereof
TWI708786B (en) 2014-12-23 2020-11-01 美商必治妥美雅史谷比公司 Antibodies to tigit
CA2987410A1 (en) 2015-05-29 2016-12-08 Bristol-Myers Squibb Company Antibodies against ox40 and uses thereof
AU2016285920A1 (en) 2015-06-29 2018-02-01 Bristol-Myers Squibb Company Antibodies to CD40 with enhanced agonist activity
KR20180025865A (en) 2015-07-06 2018-03-09 리제너론 파마슈티칼스 인코포레이티드 Multispecific antigen binding molecules and uses thereof
CN108026174B (en) 2015-07-10 2023-02-17 美勒斯公司 Human CD3 binding antibodies
JOP20160154B1 (en) 2015-07-31 2021-08-17 Regeneron Pharma Anti-psma antibodies, bispecific antigen-binding molecules that bind psma and cd3, and uses thereof
IL258036B (en) 2015-09-23 2022-09-01 Regeneron Pharma Optimized anti-cd3 bispecific antibodies and uses thereof
TWI756187B (en) 2015-10-09 2022-03-01 美商再生元醫藥公司 Anti-lag3 antibodies and uses thereof
AU2016340764B2 (en) 2015-10-23 2023-06-01 Fundació lnstitut de Recerca Biomèdica (IRB Barcelona) Binding molecules that inhibit cancer growth
AU2016356780A1 (en) 2015-11-19 2018-06-28 Bristol-Myers Squibb Company Antibodies against glucocorticoid-induced tumor necrosis factor receptor (GITR) and uses thereof
SG11201807677YA (en) 2016-03-04 2018-10-30 Univ Rockefeller Antibodies to cd40 with enhanced agonist activity
EA201891983A8 (en) 2016-03-04 2020-05-28 Бристол-Майерс Сквибб Компани COMBINED THERAPY BY ANTIBODIES TO CD73
US11352446B2 (en) 2016-04-28 2022-06-07 Regeneron Pharmaceuticals, Inc. Methods of making multispecific antigen-binding molecules
EP3455256A1 (en) 2016-05-09 2019-03-20 Bristol-Myers Squibb Company Tl1a antibodies and uses thereof
TWI822521B (en) 2016-05-13 2023-11-11 美商再生元醫藥公司 Methods of treating skin cancer by administering a pd-1 inhibitor
US10633434B2 (en) 2016-06-14 2020-04-28 Regeneron Pharmaceuticals, Inc. Anti-C5 antibodies
AU2017281034B2 (en) * 2016-06-21 2024-03-14 Teneobio, Inc. CD3 binding antibodies
JP7027401B2 (en) 2016-07-14 2022-03-01 ブリストル-マイヤーズ スクイブ カンパニー Antibodies to TIM3 and its use
JP7366742B2 (en) 2016-08-26 2023-10-23 サノフイ Multispecific antibodies that facilitate selective light chain pairing
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
WO2018052503A1 (en) 2016-09-14 2018-03-22 Teneobio, Inc. Cd3 binding antibodies
WO2018067331A1 (en) 2016-09-23 2018-04-12 Regeneron Pharmaceuticals, Inc. Bi specific anti-muc16-cd3 antibodies and nti-muc16 drug conjugates
KR102520731B1 (en) 2016-09-23 2023-04-14 리제너론 파아마슈티컬스, 인크. Anti-STEAP2 antibodies, antibody-drug conjugates, and bispecific antigen-binding molecules that bind to STEAP2 and CD3, and uses thereof
WO2018071822A2 (en) 2016-10-13 2018-04-19 Massachusetts Institute Of Technology Antibodies that bind zika virus envelope protein and uses thereof
JP7167041B2 (en) 2017-02-10 2022-11-08 リジェネロン・ファーマシューティカルズ・インコーポレイテッド Radiolabeled anti-LAG3 antibody for immunoPET imaging
EP3583124A1 (en) 2017-02-17 2019-12-25 Bristol-Myers Squibb Company Antibodies to alpha-synuclein and uses thereof
WO2018158719A1 (en) 2017-03-02 2018-09-07 Novartis Ag Engineered heterodimeric proteins
EP3600413A1 (en) 2017-03-31 2020-02-05 Merus N.V. Erbb-2 and erbb3 binding bispecific antibodies for use in the treatment f cells that have an nrg1 fusion gene
US11603407B2 (en) 2017-04-06 2023-03-14 Regeneron Pharmaceuticals, Inc. Stable antibody formulation
TWI788340B (en) 2017-04-07 2023-01-01 美商必治妥美雅史谷比公司 Anti-icos agonist antibodies and uses thereof
AU2018288803A1 (en) 2017-06-20 2020-02-06 Teneoone, Inc. Anti-BCMA heavy chain-only antibodies
TW202348250A (en) 2017-07-24 2023-12-16 美商再生元醫藥公司 Stabilized antibody compositions and methods of producing same
MX2020001432A (en) 2017-08-09 2020-03-20 Merus Nv Antibodies that bind egfr and cmet.
CN111148758B (en) 2017-09-29 2022-12-09 瑞泽恩制药公司 Bispecific antigen binding molecules that bind to staphylococcal target antigens and complement components and uses thereof
AU2018383751A1 (en) 2017-12-13 2020-06-25 Regeneron Pharmaceuticals, Inc. Anti-C5 antibody combinations and uses thereof
SG11202005880XA (en) 2017-12-22 2020-07-29 Teneobio Inc Heavy chain antibodies binding to cd22
JP7358361B2 (en) 2018-01-12 2023-10-10 ブリストル-マイヤーズ スクイブ カンパニー Antibodies against TIM3 and their uses
EA202091563A1 (en) 2018-01-26 2020-10-08 Регенерон Фармасьютикалз, Инк. HUMAN ANTIBODIES TO HEMAGGLUTININ OF THE INFLUENZA VIRUS
TW202003565A (en) 2018-03-23 2020-01-16 美商必治妥美雅史谷比公司 Antibodies against MICA and/or MICB and uses thereof
CN112074538A (en) 2018-04-30 2020-12-11 瑞泽恩制药公司 Antibodies and bispecific antigen binding molecules that bind HER2 and/or APLP2, conjugates and uses thereof
CA3100021A1 (en) 2018-05-17 2019-11-21 Regeneron Pharmaceuticals, Inc. Anti-cd63 antibodies, conjugates, and uses thereof
US11548947B2 (en) 2018-06-21 2023-01-10 Regeneron Pharmaceuticals, Inc. Bispecific anti-PSMA X anti-CD28 antibodies and uses thereof
MX2021000488A (en) 2018-07-19 2021-04-12 Regeneron Pharma BISPECIFIC ANTI-BCMA x ANTI-CD3 ANTIBODIES AND USES THEREOF.
WO2020041537A1 (en) 2018-08-23 2020-02-27 Regeneron Pharmaceuticals, Inc. Anti-fc epsilon-r1 alpha (fcer1a) antibodies, bispecific antigen-binding molecules that bind fcer1a and cd3, and uses thereof
EP3844189A1 (en) 2018-08-31 2021-07-07 Regeneron Pharmaceuticals, Inc. Dosing strategy that mitigates cytokine release syndrome for cd3/c20 bispecific antibodies
CA3115102A1 (en) 2018-10-23 2020-04-30 Regeneron Pharmaceuticals, Inc. Anti-npr1 antibodies and uses thereof
PE20211284A1 (en) 2018-11-16 2021-07-19 Bristol Myers Squibb Co ANTI-NKG2A ANTIBODIES AND USES OF THEM
WO2020106358A1 (en) 2018-11-20 2020-05-28 Takeda Vaccines, Inc. Novel anti-zika virus antibodies and uses thereof
CA3123420A1 (en) 2018-12-19 2020-06-25 Regeneron Pharmaceuticals, Inc. Bispecific anti-muc16 x anti-cd28 antibodies and uses thereof
MA54540A (en) 2018-12-19 2021-10-27 Regeneron Pharma ANTI-CD28 X ANTI-CD22 BISPECIFIC ANTIBODIES AND THEIR USES
US20220090125A1 (en) 2018-12-21 2022-03-24 Compass Therapeutics Llc Transgenic mouse expressing common human light chain
CA3127236A1 (en) 2019-01-22 2020-07-30 Bristol-Myers Squibb Company Antibodies against il-7r alpha subunit and uses thereof
EP3927832A4 (en) 2019-02-18 2022-11-30 Biocytogen Pharmaceuticals (Beijing) Co., Ltd. Genetically modified non-human animals with humanized immunoglobulin locus
US11912767B2 (en) 2019-03-22 2024-02-27 Regeneron Pharmaceuticals, Inc. EGFR × CD28 multispecific antibodies
KR20220016880A (en) 2019-06-05 2022-02-10 리제너론 파마슈티칼스 인코포레이티드 Non-human animals with a restricted lambda light chain repertoire expressed at the kappa locus and uses thereof
CA3140075A1 (en) 2019-06-11 2020-12-17 Regeneron Pharmaceuticals, Inc. Anti-pcrv antibodies that bind pcrv, compositions comprising anti-pcrv antibodies, and methods of use thereof
JP2022537931A (en) 2019-06-14 2022-08-31 テネオバイオ, インコーポレイテッド A multispecific heavy chain antibody that binds to CD22 and CD3
EP3986934A1 (en) 2019-06-21 2022-04-27 Regeneron Pharmaceuticals, Inc. Use of bispecific antigen-binding molecules that bind muc16 and cd3 in combination with 4-1bb co-stimulation
MA56515A (en) 2019-06-21 2022-04-27 Regeneron Pharma USE OF B-SPECIFIC ANTIGEN-BINDING MOLECULES BINDING TO PSMA AND CD3 IN COMBINATION WITH 4-1BB CO-STIMULATION
KR20220044821A (en) 2019-08-15 2022-04-11 리제너론 파아마슈티컬스, 인크. Multispecific antigen-binding molecules for targeting cells and uses thereof
GB201912008D0 (en) 2019-08-21 2019-10-02 Cambridge Entpr Ltd Antibody
WO2021086899A1 (en) 2019-10-28 2021-05-06 Regeneron Pharmaceuticals, Inc. Anti-hemagglutinin antibodies and methods of use thereof
JP2023505218A (en) 2019-12-06 2023-02-08 リジェネロン・ファーマシューティカルズ・インコーポレイテッド Methods of treating multiple myeloma with bispecific anti-BCMA x anti-CD3 antibodies
WO2021163170A1 (en) 2020-02-11 2021-08-19 Regeneron Pharmaceuticals, Inc. Anti-acvr1 antibodies and uses thereof
BR112022021690A2 (en) 2020-04-29 2022-12-20 Teneobio Inc MULTISPECIFIC HEAVY CHAIN ANTIBODIES WITH MODIFIED HEAVY CHAIN CONSTANT REGIONS
WO2021231366A1 (en) 2020-05-12 2021-11-18 Regeneron Pharmaceuticals, Inc. Anti-glp1r antagonist antibodies and methods of use thereof
WO2021231976A1 (en) 2020-05-14 2021-11-18 Xencor, Inc. Heterodimeric antibodies that bind prostate specific membrane antigen (psma) and cd3
WO2021231732A1 (en) 2020-05-15 2021-11-18 Bristol-Myers Squibb Company Antibodies to garp
CN115667532A (en) * 2020-06-02 2023-01-31 百奥赛图(北京)医药科技股份有限公司 Genetically modified non-human animals with a common light chain immunoglobulin locus
AU2021329378A1 (en) 2020-08-19 2023-03-23 Xencor, Inc. Anti-CD28 compositions
AU2021342159A1 (en) 2020-09-11 2023-03-02 Regeneron Pharmaceuticals, Inc. Identification and production of antigen-specific antibodies
IL301037A (en) 2020-09-18 2023-05-01 Regeneron Pharma Antigen-binding molecules that bind cd38 and/or cd28, and uses thereof
US11866502B2 (en) 2020-10-22 2024-01-09 Regeneron Pharmaceuticals, Inc. Anti-FGFR2 antibodies and methods of use thereof
CN114524878A (en) 2020-11-23 2022-05-24 康诺亚生物医药科技(成都)有限公司 Bispecific antibody and application thereof
WO2022132943A1 (en) 2020-12-16 2022-06-23 Regeneron Pharmaceuticals, Inc. Mice expressing humanized fc alpha receptors
WO2022165171A1 (en) 2021-01-28 2022-08-04 Regeneron Pharmaceuticals, Inc. Compositions and methods for treating cytokine release syndrome
EP4305067A1 (en) 2021-03-09 2024-01-17 Xencor, Inc. Heterodimeric antibodies that bind cd3 and cldn6
WO2022192586A1 (en) 2021-03-10 2022-09-15 Xencor, Inc. Heterodimeric antibodies that bind cd3 and gpc3
US20230250170A1 (en) 2021-12-06 2023-08-10 Regeneron Pharmaceuticals, Inc. Antagonist anti-npr1 antibodies and methods of use thereof
TW202400228A (en) 2022-02-25 2024-01-01 美商再生元醫藥公司 Dosing regimens for mitigation of cytokine release syndrome
WO2023196903A1 (en) 2022-04-06 2023-10-12 Regeneron Pharmaceuticals, Inc. Bispecific antigen-binding molecules that bind and cd3 and tumor associated antigens (taas) and uses thereof
US20230357446A1 (en) 2022-04-11 2023-11-09 Regeneron Pharmaceuticals, Inc. Compositions and methods for universal tumor cell killing
WO2023225098A1 (en) 2022-05-18 2023-11-23 Regeneron Pharmaceuticals, Inc. Multispecific antigen binding molecules that bind cd38 and 4-1bb, and uses thereof
WO2024015816A1 (en) 2022-07-12 2024-01-18 Regeneron Pharmaceuticals, Inc. Antibodies to ciliary neurotrophic factor receptor (cntfr) and methods of use thereof

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6596541B2 (en) * 2000-10-31 2003-07-22 Regeneron Pharmaceuticals, Inc. Methods of modifying eukaryotic cells
US20060015957A1 (en) * 1991-08-28 2006-01-19 Genpharm International, Inc. Transgenic non-human animals for producing chimeric antibodies
US20080069822A1 (en) * 2005-12-05 2008-03-20 Symphogen A/S Anti-orthopoxvirus recombinant polyclonal antibody
US7435871B2 (en) * 2001-11-30 2008-10-14 Amgen Fremont Inc. Transgenic animals bearing human Igλ light chain genes
US20100146647A1 (en) * 2008-06-27 2010-06-10 Merus B. V. Antibody producing non-human mammals
US20100310586A1 (en) * 2007-07-27 2010-12-09 ARETA INTERNATIONAL S.r.l Idiotypic vaccine

Family Cites Families (135)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE68929167T2 (en) 1988-09-06 2000-11-16 Xoma Corp Gene expression elements and production of chimeric mouse-human antibodies
GB8823869D0 (en) 1988-10-12 1988-11-16 Medical Res Council Production of antibodies
WO1991000906A1 (en) 1989-07-12 1991-01-24 Genetics Institute, Inc. Chimeric and transgenic animals capable of producing human antibodies
US5574205A (en) 1989-07-25 1996-11-12 Cell Genesys Homologous recombination for universal donor cells and chimeric mammalian hosts
US5959177A (en) 1989-10-27 1999-09-28 The Scripps Research Institute Transgenic plants expressing assembled secretory antibodies
CA2351200A1 (en) 1989-12-01 1991-06-13 Gene Pharming Europe Bv Production of recombinant polypeptides by bovine species and transgenic methods
US6713610B1 (en) 1990-01-12 2004-03-30 Raju Kucherlapati Human antibodies derived from immunized xenomice
US6657103B1 (en) 1990-01-12 2003-12-02 Abgenix, Inc. Human antibodies derived from immunized xenomice
US6150584A (en) 1990-01-12 2000-11-21 Abgenix, Inc. Human antibodies derived from immunized xenomice
DE69120146T2 (en) 1990-01-12 1996-12-12 Cell Genesys Inc GENERATION OF XENOGENIC ANTIBODIES
US6673986B1 (en) 1990-01-12 2004-01-06 Abgenix, Inc. Generation of xenogeneic antibodies
US6075181A (en) 1990-01-12 2000-06-13 Abgenix, Inc. Human antibodies derived from immunized xenomice
US6255458B1 (en) 1990-08-29 2001-07-03 Genpharm International High affinity human antibodies and human antibodies against digoxin
US5770429A (en) 1990-08-29 1998-06-23 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
ES2246502T3 (en) 1990-08-29 2006-02-16 Genpharm International, Inc. TRANSGENIC NON-HUMAN ANIMALS ABLE TO PRODUCE HETEROLOGICAL ANTIBODIES.
US7084260B1 (en) 1996-10-10 2006-08-01 Genpharm International, Inc. High affinity human antibodies and human antibodies against human antigens
US5667988A (en) 1992-01-27 1997-09-16 The Scripps Research Institute Methods for producing antibody libraries using universal or randomized immunoglobulin light chains
US7067284B1 (en) 1992-01-27 2006-06-27 The Scripps Research Institute Methods for producing antibody libraries using universal or randomized immunoglobulin light chains
ATE381614T1 (en) 1992-07-24 2008-01-15 Amgen Fremont Inc FORMATION OF XENOGENE ANTIBODIES
ATE149570T1 (en) 1992-08-17 1997-03-15 Genentech Inc BISPECIFIC IMMUNOADHESINS
CA2161351C (en) 1993-04-26 2010-12-21 Nils Lonberg Transgenic non-human animals capable of producing heterologous antibodies
US5693506A (en) 1993-11-16 1997-12-02 The Regents Of The University Of California Process for protein production in plants
US5827690A (en) 1993-12-20 1998-10-27 Genzyme Transgenics Corporatiion Transgenic production of antibodies in milk
DE69430014D1 (en) 1993-12-23 2002-04-04 Infigen Inc EMBRYONIC STEM CELLS of ungulates AS CORE DONATORS AND CORE TRANSFER TECHNIQUES FOR THE PRODUCTION OF CHIMERIC AND TRANSGENIC ANIMALS
DK0744958T3 (en) 1994-01-31 2003-10-20 Univ Boston Polyclonal antibody libraries
US7119248B1 (en) 1994-04-12 2006-10-10 Miltenyi Biotec Gmbh Antibodies against epitopes with homology to self antigens, methods of preparation and applications thereof
US6080560A (en) 1994-07-25 2000-06-27 Monsanto Company Method for producing antibodies in plant cells
US5731168A (en) 1995-03-01 1998-03-24 Genentech, Inc. Method for making heteromultimeric polypeptides
ES2176574T3 (en) 1996-09-03 2002-12-01 Gsf Forschungszentrum Umwelt USE OF BI AND TRIESPECIFIC ANTIBODIES FOR INDUCTION OF TUMOR IMMUNITY.
DK1500329T3 (en) 1996-12-03 2012-07-09 Amgen Fremont Inc Human antibodies that specifically bind TNF-alpha
CA2282722A1 (en) 1997-03-06 1998-09-11 Infigen, Inc. Method of cloning animals
WO1998046645A2 (en) 1997-04-14 1998-10-22 Micromet Gesellschaft Für Biomedizinische Forschung Mbh Method for the production of antihuman antigen receptors and uses thereof
DE69830901T2 (en) 1997-05-02 2006-05-24 Genentech Inc., San Francisco A method for producing multispecific antibodies having heteromultimeric and common components
US20020062010A1 (en) * 1997-05-02 2002-05-23 Genentech, Inc. Method for making multispecific antibodies having heteromultimeric and common components
US6774279B2 (en) 1997-05-30 2004-08-10 Carnegie Institution Of Washington Use of FLP recombinase in mice
RU2221809C2 (en) 1997-10-03 2004-01-20 Тугаи Сейяку Кабусики Кайся Method for preparing natural humanized antibody
DE69831265T2 (en) 1997-11-18 2006-06-08 Pioneer Hi-Bred International, Inc. COMPOSITIONS AND METHODS FOR THE GENETIC MODIFICATION OF PLANTS
GB9823930D0 (en) 1998-11-03 1998-12-30 Babraham Inst Murine expression of human ig\ locus
US6914128B1 (en) 1999-03-25 2005-07-05 Abbott Gmbh & Co. Kg Human antibodies that bind human IL-12 and methods for producing
SI1161548T2 (en) 1999-04-15 2010-02-26 Crucell Holland Bv Recombinant protein production in a human cell using sequences encoding adenovirus e1 protein
EP1268832A4 (en) 2000-02-29 2003-04-02 Univ Auburn Production of antibodies in transgenic plastids
RU2262511C2 (en) 2000-05-18 2005-10-20 Джапан Тобакко, Инк. Humanized monoclonal antibody raised against ailim, co-stimulating molecule for signal transfer and its pharmaceutical applying
IL154063A0 (en) 2000-07-21 2003-07-31 Us Agriculture Methods for the replacement, translocation and stacking of dna in eukaryotic genomes
EA013564B1 (en) 2000-08-03 2010-06-30 Терапеутик Хьюман Поликлоналз Инк. Humanized immunoglobulin and pharmaceutical composition comprising thereof
EP1184458A1 (en) 2000-08-28 2002-03-06 U-BISys B.V. Differentially expressed CD46 epitopes, proteinaceous molecules capable of binding thereto, and uses thereof
US20020119148A1 (en) 2000-09-01 2002-08-29 Gerritsen Mary E. ErbB4 antagonists
US7105348B2 (en) 2000-10-31 2006-09-12 Regeneron Pharmaceuticals, Inc. Methods of modifying eukaryotic cells
US6586251B2 (en) 2000-10-31 2003-07-01 Regeneron Pharmaceuticals, Inc. Methods of modifying eukaryotic cells
AR032028A1 (en) 2001-01-05 2003-10-22 Pfizer ANTIBODIES AGAINST THE RECEIVER OF THE SIMILAR TO INSULIN GROWTH FACTOR
US6961875B2 (en) 2001-03-22 2005-11-01 International Business Machines Corporation Method and apparatus for capturing event traces for debug and analysis
GB0110029D0 (en) 2001-04-24 2001-06-13 Grosveld Frank Transgenic animal
DE60237282D1 (en) 2001-06-28 2010-09-23 Domantis Ltd DOUBLE-SPECIFIC LIGAND AND ITS USE
WO2004006955A1 (en) 2001-07-12 2004-01-22 Jefferson Foote Super humanized antibodies
JP2005522192A (en) 2001-07-19 2005-07-28 パーラン セラピューティクス, インコーポレイテッド Multimeric proteins and methods of making and using multimeric proteins
ES2350248T3 (en) 2001-10-01 2011-01-20 Deutsches Krebsforschungszentrum Stiftung des öffentlichen Rechts PROCEDURE FOR THE PRODUCTION OF PROTEIN LIBRARIES AND THE SELECTION OF PROTEINS FROM THE SAME.
US20060199204A1 (en) 2001-10-05 2006-09-07 U.S. Epa Genetic testing for male factor infertility
WO2003031656A1 (en) 2001-10-05 2003-04-17 United States Environmental Protection Agency Genetic testing for male factor infertility
GB0130267D0 (en) 2001-12-19 2002-02-06 Neutec Pharma Plc Focussed antibody technology
AU2003214842A1 (en) 2002-01-17 2003-09-02 Albert Einstein College Of Medicine Of Yeshiva University Mutations caused by activation-induced cytidine deaminase
KR20040099264A (en) 2002-01-18 2004-11-26 이노비오 에이에스 Bispecific antibody dna constructs for intramuscular administration
US7282567B2 (en) 2002-06-14 2007-10-16 Immunomedics, Inc. Monoclonal antibody hPAM4
AU2003235833A1 (en) 2002-04-26 2003-11-10 Chugai Seiyaku Kabushiki Kaisha Method of screening agonistic antibody
CN101962408A (en) 2002-07-12 2011-02-02 杰斐逊·富特 Super-humanized antibody
ES2368733T3 (en) * 2002-07-18 2011-11-21 Merus B.V. RECOMBINANT PRODUCTION OF MIXTURES OF ANTIBODIES.
CL2003002461A1 (en) 2002-11-27 2005-01-07 Dow Chemical Company Agroscien IMMUNOGLOBULIN THAT UNDERSTANDS AT LEAST ONE AFUCOSILATED GLICAN, COMPOSITION THAT CONTAINS IT, NUCLEOTIDIC SEQUENCE AND VECTOR THAT UNDERSTANDS IT, PROCEDURE TO PRODUCE IMMUNOGLOBULIN SAID IN PLANTS.
GB0228210D0 (en) 2002-12-03 2003-01-08 Babraham Inst Single chain antibodies
GB0230203D0 (en) 2002-12-27 2003-02-05 Domantis Ltd Fc fusion
EP1439234A1 (en) 2003-01-08 2004-07-21 ARTEMIS Pharmaceuticals GmbH Targeted transgenesis using the rosa26 locus
JP4477579B2 (en) 2003-01-21 2010-06-09 中外製薬株式会社 Antibody light chain screening method
US20060206949A1 (en) 2003-01-28 2006-09-14 Sylvain Arnould Custom-made meganuclease and use thereof
EP1606387A4 (en) 2003-03-04 2008-04-23 Alexion Pharma Inc Vectors used to create hybrid constant regions
CA2527694C (en) 2003-05-30 2015-07-14 Hendricus Renerus Jacobus Mattheus Hoogenboom Fab library for the preparation of anti vegf and anti rabies virus fabs
AU2004257292A1 (en) 2003-07-15 2005-01-27 Therapeutic Human Polyclonals, Inc. Humanized immunoglobulin loci
US20050153392A1 (en) 2003-08-11 2005-07-14 Roland Buelow Transgenesis with humanized immunoglobulin loci
WO2005038001A2 (en) 2003-10-14 2005-04-28 Therapeutic Human Polyclonals, Inc. Improved transgenesis by sperm-mediated gene transfer
AU2005206536B2 (en) 2004-01-16 2010-09-02 Regeneron Pharmaceuticals, Inc. Fusion polypeptides capable of activating receptors
CN1560081A (en) 2004-02-17 2005-01-05 大连帝恩生物工程有限公司 Preparing human source monoclone antibody by mouse capable of producing human IgGl weight chain-k light chain and application thereof
US7625549B2 (en) 2004-03-19 2009-12-01 Amgen Fremont Inc. Determining the risk of human anti-human antibodies in transgenic mice
CA2827654C (en) 2004-10-19 2019-04-02 Regeneron Pharmaceuticals, Inc. Method for generating an animal homozygous for a genetic modification
AU2005333602B2 (en) * 2004-10-22 2012-04-12 Medimmune, Llc High affinity antibodies against HMGB1 and methods of use thereof
CA2584814A1 (en) 2004-10-22 2006-05-04 Therapeutic Human Polyclonals, Inc. Suppression of endogenous immunoglobulin expression in non-human transgenic animals
EP1874817A2 (en) 2005-04-29 2008-01-09 Innate Pharma Transgenic animals and methods of making recombinant antibodies
CA2608481A1 (en) 2005-05-14 2006-11-23 Fudan University Piggybac as a tool for genetic manipulation and analysis in vertebrates
FR2888850B1 (en) 2005-07-22 2013-01-11 Pf Medicament NOVEL ANTI-IGF-IR ANTIBODIES AND THEIR APPLICATIONS
JP5184374B2 (en) 2006-01-25 2013-04-17 エラスムス・ユニヴァーシティ・メディカル・センター・ロッテルダム Allele exclusion
US7462759B2 (en) 2006-02-03 2008-12-09 Pioneer Hi-Bred International, Inc. Brittle stalk 2 gene family and related methods and uses
CA2638774C (en) 2006-03-31 2015-11-24 Medarex, Inc. Transgenic animals expressing chimeric antibodies for use in preparing human antibodies
MX2008014804A (en) 2006-06-02 2009-01-27 Regeneron Pharma High affinity antibodies to human il-6 receptor.
CN101501073A (en) 2006-08-22 2009-08-05 G2英弗勒美欣私人有限公司 Method for producing antibodies
CN101522716B (en) 2006-10-02 2013-03-20 瑞泽恩制药公司 High affinity human antibodies to human IL-4 receptor
US7608693B2 (en) 2006-10-02 2009-10-27 Regeneron Pharmaceuticals, Inc. High affinity human antibodies to human IL-4 receptor
RU2448979C2 (en) 2006-12-14 2012-04-27 Ридженерон Фармасьютикалз, Инк. Human antibodies to delta-like human ligand-4
GB0700194D0 (en) 2007-01-05 2007-02-14 Univ Edinburgh Humanisation of animals
AU2008224950B2 (en) 2007-03-13 2013-10-03 National Jewish Medical And Research Center Methods for generation of antibodies
EP2152880B1 (en) 2007-06-01 2011-08-31 Omt, Inc. Compositions and methods for inhibiting endogenous immunoglobulin genes and producing transgenic human idiotype antibodies
WO2009013620A2 (en) 2007-06-11 2009-01-29 Erasmus University Medical Center Rotterdam Homologous recombination
US9693539B2 (en) 2007-08-10 2017-07-04 E. R. Squibb & Sons, L.L.C. HCO32 and HCO27 and related examples
KR20100058541A (en) 2007-08-15 2010-06-03 아뮤닉스 인코포레이티드 Compositions and methods for modifying properties of biologically active polypeptides
EP2193146B1 (en) 2007-09-14 2016-05-25 Adimab, LLC Rationally designed, synthetic antibody libraries and uses therefor
WO2009051974A1 (en) 2007-10-17 2009-04-23 Nuvelo, Inc. Antibodes to cll-1
CN105001333B (en) 2007-12-14 2019-05-17 诺沃—诺迪斯克有限公司 Anti-human NKG2D antibody and application thereof
PT2235064E (en) 2008-01-07 2016-03-01 Amgen Inc Method for making antibody fc-heterodimeric molecules using electrostatic steering effects
EP2271758B1 (en) 2008-04-14 2018-09-12 Innovative Targeting Solutions Inc. Sequence diversity generation in immunoglobulins
EP2669298A3 (en) 2008-05-23 2014-02-26 Ablexis, LLC Single variable immunoglobulin domain comprising VL-DH-JL
EP2147594B1 (en) 2008-06-27 2013-11-13 Merus B.V. Antibody producing non-human mammals
NZ592308A (en) 2008-09-30 2012-11-30 Ablexis Llc Non-human mammals for the production of chimeric antibodies
DK2356270T3 (en) 2008-11-07 2016-12-12 Fabrus Llc Combinatorial antibody libraries and uses 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
CA2753287A1 (en) 2009-02-24 2010-09-02 Glaxo Group Limited Antigen-binding constructs
KR20160007685A (en) 2009-05-07 2016-01-20 볼보 컨스트럭션 이큅먼트 에이비 A working machine and a method for operating a working machine
US8685896B2 (en) 2009-05-29 2014-04-01 Morphosys Ag Collection and methods for its use
AU2010265933B2 (en) * 2009-06-26 2015-05-14 Regeneron Pharmaceuticals, Inc. Readily isolated bispecific antibodies with native immunoglobulin format
EP2564695B1 (en) 2009-07-08 2015-04-15 Kymab Limited Animal models and therapeutic molecules
US20120204278A1 (en) 2009-07-08 2012-08-09 Kymab Limited Animal models and therapeutic molecules
US20120233715A1 (en) 2009-11-17 2012-09-13 Kyowa Hakko Kirin Co., Ltd Human artificial chromosome vector
PT2954779T (en) 2009-12-10 2019-05-29 Regeneron Pharma Mice that make heavy chain antibodies
US20130185821A1 (en) 2010-02-08 2013-07-18 Regeneron Pharmaceuticals, Inc. Common Light Chain Mouse
EP3095871B1 (en) 2010-02-08 2019-04-10 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
US20130045492A1 (en) 2010-02-08 2013-02-21 Regeneron Pharmaceuticals, Inc. Methods For Making Fully Human Bispecific Antibodies Using A Common Light Chain
US20120021409A1 (en) 2010-02-08 2012-01-26 Regeneron Pharmaceuticals, Inc. Common Light Chain Mouse
US9527926B2 (en) 2010-05-14 2016-12-27 Rinat Neuroscience Corp. Heterodimeric proteins and methods for producing and purifying them
CN105695415A (en) 2010-06-17 2016-06-22 科马布有限公司 Animal models and therapeutic molecules
DK2905338T3 (en) 2010-06-22 2017-11-06 Regeneron Pharma TRANSGENE MICE WITH A MODIFIED ENDOGENT LAMBDA IMMUNGLOBULIN LOCUS
NZ707327A (en) 2010-08-02 2017-01-27 Regeneron Pharma Mice that make binding proteins comprising vl domains
ES2758974T5 (en) 2011-02-25 2023-06-08 Regeneron Pharma ADAM6 Mice
MY172718A (en) 2011-08-05 2019-12-11 Regeneron Pharma Humanized universal light chain mice
CA2791109C (en) 2011-09-26 2021-02-16 Merus B.V. Generation of binding molecules
ES2906462T3 (en) 2011-10-17 2022-04-18 Regeneron Pharma Restricted immunoglobulin heavy chain mice
GB201122047D0 (en) 2011-12-21 2012-02-01 Kymab Ltd Transgenic animals
EP2822379A1 (en) 2012-03-06 2015-01-14 Regeneron Pharmaceuticals, Inc. Common light chain mouse
MX360109B (en) 2012-04-20 2018-10-23 Merus Nv Methods and means for the production of ig-like molecules.
SG10201609535TA (en) 2012-06-05 2017-01-27 Regeneron Pharma Methods for making fully human bispecific antibodies using a common light chain
WO2014160202A1 (en) 2013-03-13 2014-10-02 Regeneron Pharmaceuticals, Inc. Mice expressing a limited immunoglobulin light chain repertoire
EP2970437A1 (en) 2013-03-13 2016-01-20 Regeneron Pharmaceuticals, Inc. Common light chain mouse

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060015957A1 (en) * 1991-08-28 2006-01-19 Genpharm International, Inc. Transgenic non-human animals for producing chimeric antibodies
US6596541B2 (en) * 2000-10-31 2003-07-22 Regeneron Pharmaceuticals, Inc. Methods of modifying eukaryotic cells
US7435871B2 (en) * 2001-11-30 2008-10-14 Amgen Fremont Inc. Transgenic animals bearing human Igλ light chain genes
US20080069822A1 (en) * 2005-12-05 2008-03-20 Symphogen A/S Anti-orthopoxvirus recombinant polyclonal antibody
US20100310586A1 (en) * 2007-07-27 2010-12-09 ARETA INTERNATIONAL S.r.l Idiotypic vaccine
US20100146647A1 (en) * 2008-06-27 2010-06-10 Merus B. V. Antibody producing non-human mammals

Non-Patent Citations (6)

* Cited by examiner, † Cited by third party
Title
Aucouturier et al. (1993) J. Immunol., Vol. 150(8), 3561-3568 *
de Wildt et al. (1999) J. Mol. Biol., Vol. 285, 895-901 *
Longo et al. (2008) J. Immunol., Vol. 181, 1299-1306. *
Mendez et al. (1997) Nat. Genetics, Vol. 15, 146-156 *
Sirac et al.(2006) Blood, Vol. 108, 536-543 *
V-BASE sequence directory (1997) www2.mrc-lmb.cam.ac.uk *

Cited By (81)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
USRE47770E1 (en) 2002-07-18 2019-12-17 Merus N.V. Recombinant production of mixtures of antibodies
US10934571B2 (en) 2002-07-18 2021-03-02 Merus N.V. Recombinant production of mixtures of antibodies
US9738701B2 (en) 2003-05-30 2017-08-22 Merus N.V. Method for selecting a single cell expressing a heterogeneous combination of antibodies
US10605808B2 (en) 2003-05-30 2020-03-31 Merus N.V. Antibody producing non-human animals
US10670599B2 (en) 2003-05-30 2020-06-02 Merus N.V. Method for selecting a single cell expressing a heterogeneous combination of antibodies
US11785924B2 (en) 2008-06-27 2023-10-17 Merus N.V. Antibody producing non-human animals
US9765133B2 (en) 2008-06-27 2017-09-19 Merus N.V. Antibody producing non-human mammals
US9944695B2 (en) 2008-06-27 2018-04-17 Merus N.V. Antibody producing non-human mammals
US9951124B2 (en) 2008-06-27 2018-04-24 Merus N.V. Antibody producing non-human mammals
US11237165B2 (en) 2008-06-27 2022-02-01 Merus N.V. Antibody producing non-human animals
US11559049B2 (en) 2008-06-27 2023-01-24 Merus N.V. Antibody producing non-human animals
US20100146647A1 (en) * 2008-06-27 2010-06-10 Merus B. V. Antibody producing non-human mammals
EP2556747A3 (en) * 2008-06-27 2016-08-03 Merus N.V. Antibody producing non-human mammals.
US10966411B2 (en) 2008-06-27 2021-04-06 Merus N.V. Antibody producing non-human mammals
US11925174B2 (en) 2008-06-27 2024-03-12 Merus N.V. Antibody producing non-human animals
EP2147594B1 (en) * 2008-06-27 2013-11-13 Merus B.V. Antibody producing non-human mammals
US11877565B2 (en) 2008-12-18 2024-01-23 Erasmus University Medical Center Antibody production
US11026407B2 (en) 2010-02-08 2021-06-08 Regeneran Pharmaceuticals, Inc. Mice expressing a limited immunoglobulin light chain repertoire
US9796788B2 (en) 2010-02-08 2017-10-24 Regeneron 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
US20110195454A1 (en) * 2010-02-08 2011-08-11 Regeneron Pharmaceuticals, Inc. Common Light Chain Mouse
US10167344B2 (en) 2010-02-08 2019-01-01 Regeneron Pharmaceuticals, Inc. Mice expressing a limited immunoglobulin light chain repertoire
US10143186B2 (en) 2010-02-08 2018-12-04 Regeneron Pharmaceuticals, Inc. Common light chain mouse
US10954310B2 (en) 2010-08-02 2021-03-23 Regeneran Pharmaceuticals, Inc. Mice that make VL binding proteins
US10130081B2 (en) 2011-08-05 2018-11-20 Regeneron Pharmaceuticals, Inc. Humanized universal light chain mice
US11357217B2 (en) 2011-08-05 2022-06-14 Regeneron Pharmaceuticals, Inc. Humanized universal light chain mice
US10647781B2 (en) 2011-09-26 2020-05-12 Merus N.V. Generation of binding molecules
US9908946B2 (en) 2011-09-26 2018-03-06 Merus N.V. Generation of binding molecules
US9145588B2 (en) 2011-09-26 2015-09-29 Merus Biopharmaceuticals B.V. Generation of binding molecules
US11408095B2 (en) 2011-09-26 2022-08-09 Merus N.V. Generation of binding molecules
EP3808175A1 (en) 2012-03-16 2021-04-21 Regeneron Pharmaceuticals, Inc. Histidine engineered light chain antibodies and genetically modified rodents for generating the same
EP2883449A1 (en) 2012-03-16 2015-06-17 Regeneron Pharmaceuticals, Inc. Histidine engineered light chain antibodies and genetically modified non-human animals for generating the same
WO2013138712A1 (en) 2012-03-16 2013-09-19 Regeneron Pharmaceuticals, Inc. Non-human animals expressing ph-sensitive immunoglobulin sequences
US9801362B2 (en) 2012-03-16 2017-10-31 Regeneron Pharmaceuticals, Inc. Non-human animals expressing pH-sensitive immunoglobulin sequences
US11192947B2 (en) 2012-03-16 2021-12-07 Regeneran Pharmaceuticals, Inc. Histidine engineered light chain antibodies and genetically modified non-human animals for generating same
EP3348140A2 (en) 2012-03-16 2018-07-18 Regeneron Pharmaceuticals, Inc. Histidine engineered light chain antibodies and genetically modified rodents for generating the same
US11224207B2 (en) 2012-03-16 2022-01-18 Regeneran Pharmaceuticals, Inc. Non-human animals expressing pH-sensitive immunoglobulin sequences
US9422370B2 (en) 2012-03-16 2016-08-23 Regeneron Pharmaceuticals, Inc. Histidine engineered light chain antibodies and genetically modified non-human animals for generating the same
US9648856B2 (en) 2012-03-16 2017-05-16 Regeneron Pharmaceuticals, Inc. Non-human animals expressing pH-sensitive immunoglobulin sequences
US9301510B2 (en) 2012-03-16 2016-04-05 Regeneron Pharmaceuticals, Inc. Mice that produce antigen-binding proteins with pH-dependent binding characteristics
US9334334B2 (en) 2012-03-16 2016-05-10 Regeneron Pharmaceuticals, Inc. Histidine engineered light chain antibodies and genetically modified non-human animals for generating the same
EP3539374A1 (en) 2012-03-16 2019-09-18 Regeneron Pharmaceuticals, Inc. Non-human animals expressing ph-sensitive immunoglobulin sequences
WO2013138680A1 (en) 2012-03-16 2013-09-19 Regeneron Pharmaceuticals, Inc. Histidine engineered light chain antibodies and genetically modified non-human animals for generating the same
US9332742B2 (en) 2012-03-16 2016-05-10 Regeneron Pharmaceuticals, Inc. Histidine engineered light chain antibodies and genetically modified non-human animals for generating the same
US10752929B2 (en) 2012-04-20 2020-08-25 Merus N.V. Methods and means for the production of ig-like molecules
US10337045B2 (en) 2012-04-20 2019-07-02 Merus N.V. Methods and means for the production of Ig-like molecules
US10329596B2 (en) 2012-04-20 2019-06-25 Merus N.V. Methods and means for the production of Ig-like molecules
US11926859B2 (en) 2012-04-20 2024-03-12 Merus N.V. Methods and means for the production of Ig-like molecules
US9758805B2 (en) 2012-04-20 2017-09-12 Merus N.V. Methods and means for the production of Ig-like molecules
WO2014022540A1 (en) 2012-08-02 2014-02-06 Regeneron Pharmaceuticals, Inc. Multivalent antigen-binding proteins
NL2012424A (en) * 2013-03-13 2014-09-16 Regeneron Pharma Common light chain mouse.
WO2014160179A1 (en) * 2013-03-13 2014-10-02 Regeneron Pharmaceuticals, Inc. Common light chain mouse
NL2015603A (en) * 2013-03-13 2016-03-16 Regeneron Pharma Common light chain mouse.
EP2967012A1 (en) 2013-03-14 2016-01-20 Erasmus University Medical Center Rotterdam Transgenic non-human mammal for antibody production
EP2967012B1 (en) * 2013-03-14 2020-09-16 Erasmus University Medical Center Rotterdam Transgenic non-human mammal for antibody production
US9980470B2 (en) 2013-03-14 2018-05-29 Erasmus University Medical Center Antibody production
EP3549437A1 (en) 2013-09-18 2019-10-09 Regeneron Pharmaceuticals, Inc. Histidine engineered light chain antibodies and genetically modified non-human animals for generating the same
WO2015042250A1 (en) 2013-09-18 2015-03-26 Regeneron Pharmaceuticals, Inc. Histidine engineered light chain antibodies and genetically modified non-human animals for generating the same
US10881085B2 (en) * 2014-03-21 2021-01-05 Regeneron Pharmaceuticals, Inc. Non-human animals that make single domain binding proteins
US20150289489A1 (en) * 2014-03-21 2015-10-15 Regeneron Pharmaceuticals, Inc. Non-human animals that make single domain binding proteins
WO2015143406A2 (en) 2014-03-21 2015-09-24 Regeneron Pharmaceuticals, Inc. Vl antigen binding proteins exhibiting distinct binding characteristics
WO2015143414A2 (en) 2014-03-21 2015-09-24 Regeneron Pharmaceuticals, Inc. Non-human animals that make single domain binding proteins
US10787522B2 (en) 2014-03-21 2020-09-29 Regeneron Pharmaceuticals, Inc. VL antigen binding proteins exhibiting distinct binding characteristics
EP3895528A1 (en) 2014-03-21 2021-10-20 Regeneron Pharmaceuticals, Inc. Non-human animals that make single domain binding proteins
US11111314B2 (en) 2015-03-19 2021-09-07 Regeneron Pharmaceuticals, Inc. Non-human animals that select for light chain variable regions that bind antigen
EP4257599A2 (en) 2016-01-13 2023-10-11 Regeneron Pharmaceuticals, Inc. Rodents having an engineered heavy chain diversity region
WO2017123804A1 (en) 2016-01-13 2017-07-20 Regeneron Pharmaceuticals, Inc. Rodents having an engineered heavy chain diversity region
WO2017201476A1 (en) 2016-05-20 2017-11-23 Regeneron Pharmaceuticals, Inc. Methods for breaking immunological tolerance using multiple guide rnas
EP4218408A1 (en) 2016-06-03 2023-08-02 Regeneron Pharmaceuticals, Inc. Rodents expressing exogenous terminal deoxynucleotidyltransferase
US10980221B2 (en) 2016-06-03 2021-04-20 Regeneron Pharmaceuticals, Inc. Non-human animals expressing exogenous terminal deoxynucleotidyltransferase
WO2017210586A1 (en) 2016-06-03 2017-12-07 Regeneron Pharmaceuticals, Inc. Non-human animals expressing exogenous terminal deoxynucleotidyltransferase
WO2019190922A1 (en) 2018-03-24 2019-10-03 Regeneron Pharmaceuticals, Inc. Genetically modified non-human animals for generating therapeutic antibodies against peptide-mhc complexes, methods of making and uses thereof
EP4023059A1 (en) 2018-06-14 2022-07-06 Regeneron Pharmaceuticals, Inc. Non-human animals capable of dh-dh rearrangement in the immunoglobulin heavy chain coding sequences
WO2019241692A1 (en) 2018-06-14 2019-12-19 Regeneron Pharmaceuticals, Inc. Non-human animals capable of dh-dh rearrangement in the immunoglobulin heavy chain coding sequences
WO2020172505A1 (en) 2019-02-22 2020-08-27 Regeneron Pharmaceuticals, Inc. Rodents having genetically modified sodium channels and methods of use thereof
US11464217B2 (en) 2019-02-22 2022-10-11 Regeneron Pharmaceuticals, Inc. Rodents having genetically modified sodium channels and methods of use thereof
WO2021026409A1 (en) 2019-08-08 2021-02-11 Regeneron Pharmaceuticals, Inc. Novel antigen binding molecule formats
WO2021113297A1 (en) 2019-12-02 2021-06-10 Regeneron Pharmaceuticals, Inc. Peptide-mhc ii protein constructs and uses thereof
US11773176B2 (en) 2020-01-24 2023-10-03 Aprilbio Co., Ltd. Multispecific antibodies, compositions comprising the same, and vectors and uses thereof
WO2022140494A1 (en) 2020-12-23 2022-06-30 Regeneron Pharmaceuticals, Inc. Methods for obtaining antibodies that bind transmembrane proteins and cells that produce the same
WO2022140219A1 (en) 2020-12-23 2022-06-30 Regeneron Pharmaceuticals, Inc. Nucleic acids encoding anchor modified antibodies and uses thereof

Also Published As

Publication number Publication date
EP3095871B1 (en) 2019-04-10
EP2501817B1 (en) 2015-08-26
IL221265A0 (en) 2012-10-31
ES2547142T3 (en) 2015-10-02
US10143186B2 (en) 2018-12-04
DK2501817T3 (en) 2015-12-07
SI2505654T2 (en) 2020-08-31
CA2789154A1 (en) 2011-08-11
SG183149A1 (en) 2012-09-27
RS55315B2 (en) 2020-08-31
IL221265B (en) 2020-04-30
KR102283195B1 (en) 2021-07-29
IL267362A (en) 2019-08-29
ES2728942T3 (en) 2019-10-29
EP2505654B1 (en) 2016-08-24
JP6470789B2 (en) 2019-02-13
PT2501817E (en) 2015-11-04
RS54367B2 (en) 2021-08-31
SMT201500283B (en) 2016-01-08
JP2019076108A (en) 2019-05-23
SG10201500821UA (en) 2015-04-29
ME02646B (en) 2017-06-20
ME02288B (en) 2016-02-20
RU2724663C2 (en) 2020-06-25
RS59001B1 (en) 2019-08-30
JP2022001066A (en) 2022-01-06
KR20180018838A (en) 2018-02-21
NZ631363A (en) 2016-05-27
AU2011213585B2 (en) 2014-02-06
EP2501817A1 (en) 2012-09-26
HRP20151188T4 (en) 2021-10-01
KR20200060534A (en) 2020-05-29
KR20190077630A (en) 2019-07-03
RS54367B1 (en) 2016-04-28
HRP20161551T1 (en) 2016-12-30
MX350983B (en) 2017-09-27
SI2501817T1 (en) 2015-12-31
EP2501817B2 (en) 2021-04-21
KR101995735B1 (en) 2019-07-03
CN105010238A (en) 2015-11-04
MX2012009168A (en) 2012-08-31
BR112012019887A2 (en) 2015-09-15
SG10201913463QA (en) 2020-03-30
KR102116296B1 (en) 2020-05-28
CN102791866B (en) 2015-07-29
SG10201610294RA (en) 2017-02-27
HUE026229T2 (en) 2016-06-28
HRP20161551T4 (en) 2020-10-02
US20210204531A1 (en) 2021-07-08
JP2020096633A (en) 2020-06-25
EP3540066A1 (en) 2019-09-18
BR122014002928A2 (en) 2015-12-29
RU2015147726A (en) 2019-01-15
PL2505654T5 (en) 2020-11-30
RU2015147726A3 (en) 2019-06-25
EP2505654A1 (en) 2012-10-03
EP2505654B2 (en) 2020-05-13
KR102432611B1 (en) 2022-08-16
KR101829691B1 (en) 2018-02-19
ES2547142T5 (en) 2021-12-09
PL3095871T3 (en) 2019-10-31
SI2505654T1 (en) 2016-12-30
JP2013518597A (en) 2013-05-23
JP6960005B2 (en) 2021-11-05
SMT201600432B (en) 2017-01-10
RU2571205C2 (en) 2015-12-20
DK3095871T3 (en) 2019-06-11
DK2501817T4 (en) 2021-07-26
JP2015171386A (en) 2015-10-01
JP7233504B2 (en) 2023-03-06
PT3095871T (en) 2019-06-12
TR201906650T4 (en) 2019-05-21
CN105010238B (en) 2018-07-20
US20190040123A1 (en) 2019-02-07
DK2505654T4 (en) 2020-07-27
IL290085A (en) 2022-03-01
CY1118241T1 (en) 2017-06-28
HRP20151188T1 (en) 2015-12-04
NZ601679A (en) 2014-01-31
US20110195454A1 (en) 2011-08-11
US10986820B2 (en) 2021-04-27
LT3095871T (en) 2019-05-27
JP2018138047A (en) 2018-09-06
ZA201205944B (en) 2013-06-26
RU2012138431A (en) 2014-03-20
JP5955781B2 (en) 2016-07-20
PL2501817T5 (en) 2021-08-16
HK1231505A1 (en) 2017-12-22
PT2505654T (en) 2016-11-18
JP2017140050A (en) 2017-08-17
HK1174358A1 (en) 2013-06-07
CN102791866A (en) 2012-11-21
HUE029785T2 (en) 2017-04-28
JP6666483B2 (en) 2020-03-13
SI2501817T2 (en) 2021-09-30
PL2505654T3 (en) 2017-02-28
RS55315B1 (en) 2017-03-31
ES2603559T3 (en) 2017-02-28
WO2011097603A1 (en) 2011-08-11
IL267362B (en) 2022-02-01
KR20130010110A (en) 2013-01-25
HRP20191191T1 (en) 2019-10-04
LT2505654T (en) 2016-12-12
BR112012019887B1 (en) 2021-06-08
EP3095871A1 (en) 2016-11-23
JP6215264B2 (en) 2017-10-18
ES2603559T5 (en) 2021-02-22
AU2011213585A1 (en) 2012-08-30
HUE045591T2 (en) 2019-12-30
CY1121794T1 (en) 2020-07-31
MX2021000286A (en) 2022-06-08
SI3095871T1 (en) 2019-06-28
KR20210095730A (en) 2021-08-02
US20130302836A1 (en) 2013-11-14
NZ719253A (en) 2022-08-26
MY166529A (en) 2018-07-10
NZ619512A (en) 2014-10-31
JP2023062142A (en) 2023-05-02
PL2501817T3 (en) 2016-02-29

Similar Documents

Publication Publication Date Title
US20210204531A1 (en) Common light chain mouse
AU2016202488C1 (en) Common light chain mouse
US20200024368A1 (en) Mice expressing a limited immunoglobulin light chain repertoire
US20190071519A1 (en) Methods for making fully human bispecific antibodies using a common light chain
US20190090462A1 (en) Common light chain mouse
EP2701499B9 (en) Non-human animals expressing antibodies having a common light chain
AU2013204019B2 (en) Common light chain mouse

Legal Events

Date Code Title Description
AS Assignment

Owner name: REGENERON PHARMACEUTICALS, INC., NEW YORK

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:BABB, ROBERT;MCWHIRTER, JOHN;MACDONALD, LYNN;AND OTHERS;SIGNING DATES FROM 20120402 TO 20120409;REEL/FRAME:028016/0970

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STCB Information on status: application discontinuation

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