US20120204278A1 - Animal models and therapeutic molecules - Google Patents

Animal models and therapeutic molecules Download PDF

Info

Publication number
US20120204278A1
US20120204278A1 US13/310,431 US201113310431A US2012204278A1 US 20120204278 A1 US20120204278 A1 US 20120204278A1 US 201113310431 A US201113310431 A US 201113310431A US 2012204278 A1 US2012204278 A1 US 2012204278A1
Authority
US
United States
Prior art keywords
human
regions
mammal
mouse
cell
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
US13/310,431
Other languages
English (en)
Inventor
Allan Bradley
E-Chiang Lee
Qi Liang
Wei Wang
Anais Legent
Ian Kirby
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.)
Kymab Ltd
Original Assignee
Kymab Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=43428834&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20120204278(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority claimed from GBGB0911846.4A external-priority patent/GB0911846D0/en
Priority claimed from GB0913102A external-priority patent/GB0913102D0/en
Priority claimed from PCT/GB2011/050019 external-priority patent/WO2011158009A1/fr
Application filed by Kymab Ltd filed Critical Kymab Ltd
Priority to US13/310,431 priority Critical patent/US20120204278A1/en
Assigned to KYMAB LIMITED reassignment KYMAB LIMITED ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: LIANG, Qi, WANG, WEI, BRADLEY, ALLAN, MR., KIRBY, IAN, MR., LEE, E-CHIANG, MR., LEGENT, ANAIS
Priority to US13/416,684 priority patent/US9447177B2/en
Publication of US20120204278A1 publication Critical patent/US20120204278A1/en
Priority to PCT/GB2012/052960 priority patent/WO2013061098A2/fr
Priority to PCT/GB2012/052956 priority patent/WO2013079953A1/fr
Priority to CA2857569A priority patent/CA2857569A1/fr
Priority to EP12795606.8A priority patent/EP2649184B1/fr
Priority to EP12795841.1A priority patent/EP2785845A2/fr
Priority to EP15188522.5A priority patent/EP2989894B9/fr
Priority to CN201280059193.7A priority patent/CN104160031B/zh
Priority to JP2014543973A priority patent/JP2015502149A/ja
Priority to EP23201805.1A priority patent/EP4282879A3/fr
Priority to EP17196214.5A priority patent/EP3298889A1/fr
Priority to DE202012013369.1U priority patent/DE202012013369U1/de
Priority to ES15188522T priority patent/ES2816899T3/es
Priority to AU2012343587A priority patent/AU2012343587B2/en
Priority to BR112014013121A priority patent/BR112014013121A2/pt
Priority to DK12795606.8T priority patent/DK2649184T3/en
Priority to US13/740,727 priority patent/US9505827B2/en
Priority to US14/040,427 priority patent/US20140201854A1/en
Priority to US14/040,405 priority patent/US20140201856A1/en
Priority to US14/056,707 priority patent/US20140150126A1/en
Priority to US14/056,434 priority patent/US20140182003A1/en
Priority to US14/056,700 priority patent/US11812731B2/en
Priority to HK13112514.7A priority patent/HK1185100A1/zh
Priority to US14/080,630 priority patent/US10165763B2/en
Priority to US14/137,902 priority patent/US9434782B2/en
Priority to US14/497,054 priority patent/US20150113669A1/en
Priority to US14/516,461 priority patent/US10064398B2/en
Priority to US14/750,870 priority patent/US20160044900A1/en
Priority to US14/818,162 priority patent/US11564380B2/en
Priority to US14/935,010 priority patent/US9504236B2/en
Priority to US15/016,211 priority patent/US20160150768A1/en
Priority to US15/214,963 priority patent/US20160345551A1/en
Priority to US15/232,122 priority patent/US11606941B2/en
Priority to US15/251,969 priority patent/US20170051045A1/en
Priority to US15/360,502 priority patent/US20170071174A1/en
Priority to US15/369,595 priority patent/US20170081423A1/en
Priority to US15/383,101 priority patent/US20170099815A1/en
Priority to US15/383,353 priority patent/US20170094956A1/en
Priority to US15/383,188 priority patent/US20170101482A1/en
Priority to US15/383,342 priority patent/US20170099816A1/en
Priority to US15/385,372 priority patent/US20170105396A1/en
Priority to US15/385,348 priority patent/US20170099817A1/en
Priority to JP2017235119A priority patent/JP2018038428A/ja
Priority to US15/955,216 priority patent/US20180295821A1/en
Priority to US16/725,707 priority patent/US20200205384A1/en
Priority to US16/870,365 priority patent/US20200352144A1/en
Priority to US16/870,413 priority patent/US20200375158A1/en
Priority to JP2020092295A priority patent/JP2020124228A/ja
Priority to US16/905,557 priority patent/US20200352145A1/en
Priority to US16/905,537 priority patent/US20200337280A1/en
Priority to JP2022128160A priority patent/JP2022159413A/ja
Priority to US17/943,533 priority patent/US20240057572A1/en
Priority to US18/162,043 priority patent/US20230225302A1/en
Priority to US18/166,813 priority patent/US20230263143A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K67/00Rearing or breeding animals, not otherwise provided for; New or modified breeds of animals
    • A01K67/027New or modified breeds of vertebrates
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K67/00Rearing or breeding animals, not otherwise provided for; New or modified breeds of animals
    • A01K67/027New or modified breeds of vertebrates
    • A01K67/0271Chimeric vertebrates, e.g. comprising exogenous cells
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K67/00Rearing or breeding animals, not otherwise provided for; New or modified breeds of animals
    • A01K67/027New or modified breeds of vertebrates
    • A01K67/0275Genetically modified vertebrates, e.g. transgenic
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K67/00Rearing or breeding animals, not otherwise provided for; New or modified breeds of animals
    • A01K67/027New or modified breeds of vertebrates
    • A01K67/0275Genetically modified vertebrates, e.g. transgenic
    • A01K67/0276Knock-out vertebrates
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K67/00Rearing or breeding animals, not otherwise provided for; New or modified breeds of animals
    • A01K67/027New or modified breeds of vertebrates
    • A01K67/0275Genetically modified vertebrates, e.g. transgenic
    • A01K67/0278Knock-in vertebrates, e.g. humanised vertebrates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/02Bacterial antigens
    • A61K39/107Vibrio
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/35Allergens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/12Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from bacteria
    • C07K16/1203Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from bacteria from Gram-negative bacteria
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/12Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from bacteria
    • C07K16/1203Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from bacteria from Gram-negative bacteria
    • C07K16/1239Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from bacteria from Gram-negative bacteria from Vibrionaceae (G)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/46Hybrid immunoglobulins
    • C07K16/461Igs containing Ig-regions, -domains or -residues form different species
    • C07K16/462Igs containing a variable region (Fv) from one specie and a constant region (Fc) from another
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/8509Vectors or expression systems specially adapted for eukaryotic hosts for animal cells for producing genetically modified animals, e.g. transgenic
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; 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; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • A01K2217/05Animals comprising random inserted nucleic acids (transgenic)
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • A01K2217/05Animals comprising random inserted nucleic acids (transgenic)
    • A01K2217/052Animals comprising random inserted nucleic acids (transgenic) inducing gain of function
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; 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; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • A01K2217/07Animals genetically altered by homologous recombination
    • A01K2217/075Animals genetically altered by homologous recombination inducing loss of function, i.e. knock out
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; 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; AVICULTURE; APICULTURE; PISCICULTURE; 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; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2267/00Animals characterised by purpose
    • A01K2267/01Animal expressing industrially exogenous proteins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/10Immunoglobulins specific features characterized by their source of isolation or production
    • C07K2317/14Specific host cells or culture conditions, e.g. components, pH or temperature
    • 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
    • C07K2317/51Complete heavy chain or Fd fragment, i.e. VH + CH1
    • 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
    • C07K2317/52Constant or Fc region; Isotype
    • 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
    • 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
    • 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
    • C12N2015/8518Vectors or expression systems specially adapted for eukaryotic hosts for animal cells for producing genetically modified animals, e.g. transgenic expressing industrially exogenous proteins, e.g. for pharmaceutical use, human insulin, blood factors, immunoglobulins, pseudoparticles

Definitions

  • the present invention relates inter alia to non-human animals and cells that are engineered to contain exogenous DNA, such as human immunoglobulin gene DNA, their use in medicine and the study of disease, methods for production of non-human animals and cells, and antibodies and antibody chains produced by such animals and derivatives thereof.
  • exogenous DNA such as human immunoglobulin gene DNA
  • mice with human immune systems In order to get around the problems of humanizing antibodies a number of companies set out to generate mice with human immune systems. The strategy used was to knockout the heavy and light chain loci in ES cells and complement these genetic lesions with transgenes designed to express the human heavy and light chain genes. Although fully human antibodies could be generated, these models have several major limitations:
  • WO2007117410 discloses chimaeric constructs for expressing chimaeric antibodies.
  • WO2010039900 discloses knock in cells and mammals having a genome encoding chimaeric antibodies.
  • the present invention provides, inter alia, a process for the generation in non-human mammals of antibodies that comprise a human Ig variable region, and further provides non-human animal models for the generation of such antibodies.
  • NCBI build 37 e.g. UCSC version mm9 see World Wide Web (www) genome.ucsc.edu and World Wide Web (www) genome.ucsc.edu/FAQ/FAQreleases.html
  • Human nucleotides coordinates are those corresponding to GRCh37 (e.g.
  • Rat nucleotides are those corresponding to RGSC 3.4 December 2004 ENSEMBL release 55.34w, or Baylor College of Medicine HGSC v3.4 November 2004 (e.g., UCSC rn4, see World Wide Web (www) genome.ucsc.edu and World Wide Web (www) genome.ucsc.edu/FAQ/FAQreleases.html) unless otherwise specified.
  • mice are disclosed for constructing a chimaeric human heavy and light chain loci in a non-human mammal, for example a mouse.
  • a non-human mammal for example a mouse.
  • Reference to work in mice herein is by way of example only, and reference to mice is taken to include reference to all non-human mammals unless otherwise apparent from the disclosure, with mice being preferred as the non-human mammal.
  • the invention relates to a non-human mammal whose genome comprises:
  • non-human mammal is able to produce a repertoire of chimaeric antibodies, or chimaeric light or heavy chains, having a non-human mammal constant region and a human variable region.
  • the invention relates to non-human mammal whose genome comprises
  • non-human mammal is able to produce a repertoire of chimaeric antibodies, or chimaeric light or heavy chains, having a non-human mammal constant region and a human variable region.
  • the invention relates to non-human mammalian cell whose genome comprises
  • the invention relates to a non-human mammalian cell whose genome comprises
  • the invention relates to a method for producing a non-human cell or mammal comprising inserting into a non-human mammal cell genome, such as an ES cell genome;
  • the invention relates to a method for producing an antibody or antibody chain specific to a desired antigen the method comprising immunizing a transgenic non-human mammal as disclosed herein with the desired antigen and recovering the antibody or antibody chain.
  • the invention relates to a method for producing a fully humanised antibody comprising immunizing a transgenic non-human mammal as disclosed herein with the desired antigen, recovering the antibody or cells producing the antibody and then replacing the non-human mammal constant region with a human constant region, for example by protein or DNA engineering.
  • the invention relates to humanised antibodies and antibody chains produced according to the present invention, both in chimaeric (for example, mouse-human) and fully humanised form, as well as fragments and derivatives of said antibodies and chains, and use of said antibodies, chains and fragments in medicine, including diagnosis.
  • chimaeric for example, mouse-human
  • fully humanised form as well as fragments and derivatives of said antibodies and chains, and use of said antibodies, chains and fragments in medicine, including diagnosis.
  • the invention relates to use of a non-human mammal as described herein as a model for the testing of drugs and vaccines.
  • the invention relates to a non-human mammal whose genome comprises:
  • non-human mammal is able to produce a repertoire of chimaeric antibodies or antibody chains having a non-human mammal constant region and a human variable region.
  • the invention relates to a non-human mammal whose genome comprises:
  • non-human mammal is able to produce a repertoire of chimaeric antibodies having a non-human mammal constant region and a human variable region.
  • non-human mammal genome is modified to prevent expression of fully host-species specific antibodies.
  • the inserted human DNA comprises at least 50% of the human heavy chain variable (V) genes, such as at least 60%, at least 70%, at least 80%, at least 90%, and in one aspect all of the human V genes.
  • V human heavy chain variable
  • the inserted human DNA comprises at least 50% of the human heavy chain diversity (D) genes, such as at least 60%, at least 70%, at least 80%, at least 90%, and in one aspect all of the human D genes.
  • D human heavy chain diversity
  • the inserted human DNA comprises at least 50% of the human heavy chain joining (J) genes, such as at least 60%, at least 70%, at least 80%, at least 90%, and in one aspect all of the human J genes.
  • J human heavy chain joining
  • the inserted human DNA comprises at least 50% of the human light chain Variable (V) genes, such as at least 60%, at least 70%, at least 80%, at least 90%, and in one aspect all of the human light chain V genes.
  • V human light chain Variable
  • the inserted human DNA comprises at least 50% of the human light chain joining (J) genes, such as at least 60%, at least 70%, at least 80%, at least 90%, and in one aspect all of the human light chain J genes.
  • J human light chain joining
  • the inserted human genes may be derived from the same individual or different individuals, or be synthetic or represent human consensus sequences.
  • V D and J regions are variable between human individuals, in one aspect there are considered to be 51 human V genes, 27 D and 6 J genes on the heavy chain, 40 human V genes and 5 J genes on the kappa light chain and 29 human V genes and 4 J genes on the lambda light chain (Janeway and Travers, Immunobiology, Third edition)
  • the human heavy chain locus inserted into the non-human mammal contains the full repertoire of human V, D and J regions, which in the genome is in functional arrangement with the non-human mammal constant regions such that functional chimaeric antibodies can be produced between the human variable and non-human mammal constant regions.
  • This total inserted human heavy chain genetic material is referred to herein as the human IgH VDJ region, and comprises DNA from a human genome that encodes all the exons encoding human V,D and J portions and suitably also the associated introns.
  • reference to the human Ig light chain kappa V and J regions herein refers to human DNA comprising all the exons encoding V and J regions and suitably also the associated introns of the human genome.
  • Reference to the human Ig light chain lambda V and J regions herein refers to human DNA comprising all the exons encoding V and J regions and suitably also the associated introns of the human genome.
  • Human variable regions are suitably inserted upstream of a non-human mammal constant region, the latter comprising all of the DNA required to encode the full constant region or a sufficient portion of the constant region to allow the formation of an effective chimaeric antibody capable of specifically recognising an antigen.
  • the chimaeric antibodies or antibody chains have a part of a host constant region sufficient to provide one or more effector functions seen in antibodies occurring naturally in a host mammal, for example that they are able interact with Fc receptors, and/or bind to complement.
  • references to a chimaeric antibody or antibody chain having a host non mammal constant region herein therefore is not limited to the complete constant region but also includes chimaeric antibodies or chains which have all of the host constant region, or a part thereof sufficient to provide one or more effector functions.
  • This also applies to non-human mammals and cells and methods of the invention in which human variable region DNA may be inserted into the host genome such that it forms a chimaeric antibody chain with all or part of a host constant region.
  • the whole of a host constant region is operably linked to human variable region DNA.
  • the host non-human mammal constant region herein is preferably the endogenous host wild-type constant region located at the wild type locus, as appropriate for the heavy or light chain.
  • the human heavy chain DNA is suitably inserted on mouse chromosome 12, suitably adjacent the mouse heavy chain constant region.
  • the insertion of the human DNA is targeted to the region between the J4 exon and the C ⁇ locus in the mouse genome IgH locus, and in one aspect is inserted between co-ordinates 114,667,090 and 114,665,190, or at co-ordinate 114,667,091, after 114,667,090.
  • the insertion of the human DNA such as the human light chain kappa VJ is targeted into mouse chromosome 6 between co-ordinates 70,673,899 and 70,675,515, suitably at position 70,674,734, or an equivalent position in the lambda mouse locus on chromosome 16.
  • the host non-human mammal constant region for forming the chimaeric antibody may be at a different (non endogenous) chromosomal locus.
  • the inserted human DNA such as the human variable VDJ or VJ region(s) may then be inserted into the non-human genome at a site which is distinct from that of the naturally occurring heavy or light constant region.
  • the native constant region may be inserted into the genome, or duplicated within the genome, at a different chromosomal locus to the native position, such that it is in a functional arrangement with the human variable region such that chimaeric antibodies of the invention can still be produced.
  • the human DNA is inserted at the endogenous host wild-type constant region located at the wild type locus between the host constant region and the host VDJ region.
  • variable region upstream of the non-human mammal constant region means that there is a suitable relative location of the two antibody portions, variable and constant, to allow the variable and constant regions to form a chimaeric antibody or antibody chain in vivo in the mammal.
  • the inserted human DNA and host constant region are in functional arrangement with one another for antibody or antibody chain production.
  • the inserted human DNA is capable of being expressed with different host constant regions through isotype switching.
  • isotype switching does not require or involve trans switching. Insertion of the human variable region DNA on the same chromosome as the relevant host constant region means that there is no need for trans-switching to produce isotype switching.
  • the transgenic loci used for the prior art models were of human origin, thus even in those cases when the transgenes were able to complement the mouse locus so that the mice produced B-cells producing fully human antibodies, individual antibody affinities rarely reached those which could be obtained from intact (non-transgenic) animals.
  • the principal reason for this is the fact that the control elements of the locus are human.
  • the signalling components for instance to activate hyper-mutation and selection of high affinity antibodies are compromised.
  • host non-human mammal constant regions are maintained and it is preferred that at least one non-human mammal enhancer or other control sequence, such as a switch region, is maintained in functional arrangement with the non-human mammal constant region, such that the effect of the enhancer or other control sequence, as seen in the host mammal, is exerted in whole or in part in the transgenic animal.
  • at least one non-human mammal enhancer or other control sequence such as a switch region
  • This approach above is designed to allow the full diversity of the human locus to be sampled, to allow the same high expression levels that would be achieved by non-human mammal control sequences such as enhancers, and is such that signalling in the B-cell, for example isotype switching using switch recombination sites, would still use non-human mammal sequences.
  • a mammal having such a genome would produce chimaeric antibodies with human variable and non-human mammal constant regions, but these could be readily humanized, for example in a cloning step. Moreover the in vivo efficacy of these chimaeric antibodies could be assessed in these same animals.
  • the inserted human IgH VDJ region comprises, in germline configuration, all of the V, D and J regions and intervening sequences from a human.
  • 800-1000 kb of the human IgH VDJ region is inserted into the non-human mammal IgH locus, and in one aspect a 940, 950 or 960 kb fragment is inserted.
  • this includes bases 105,400,051 to 106,368,585 from human chromosome 14.
  • the inserted IgH human fragment consists of bases 105,400,051 to 106,368,585 from chromosome 14.
  • the inserted human heavy chain DNA such as DNA consisting of bases 105,400,051 to 106,368,585 from chromosome 14 is inserted into mouse chromosome 12 between the end of the mouse J4 region and the E ⁇ region, suitably between co-ordinates 114,667,090 and 114,665,190, or at co-ordinate 114,667,091, after 114,667,090.
  • the insertion is between co-ordinates 114,667,089 and 114,667,090 (co-ordinates refer to NCBI m37, for the mouse C57BL/6J strain), or at equivalent position in another non-human mammal genome.
  • the inserted human kappa VJ region comprises, in germline configuration, all of the V and J regions and intervening sequences from a human.
  • this includes bases 88,940,356 to 89,857,000 from human chromosome 2, suitably approximately 917 kb.
  • the light chain VJ insert may comprise only the proximal clusters of V segments and J segments. Such an insert would be of approximately 473 kb.
  • the human light chain kappa DNA such as the human IgK fragment of bases 88,940,356 to 89,857,000 from human chromosome 2 is suitably inserted into mouse chromosome 6 between co-ordinates 70,673,899 and 70,675,515, suitably at position 70,674,734.
  • co-ordinates refer to NCBI36 for the human genome, ENSEMBL Release 54 and NCBIM37 for the mouse genome, relating to mouse strain C57BL/6J.
  • the human lambda VJ region comprises, in germline configuration, all of the V and J regions and intervening sequences from a human.
  • this includes analogous bases to those selected for the kappa fragment, from human chromosome 2.
  • a cell or non-human mammal of the invention in one embodiment, comprises an insertion of human heavy chain variable region DNA between co-ordinates 114, 666, 183 and 114, 666, 725, such as between 114 666 283 and 114 666 625, optionally between co-ordinates 114,666,335 and 114,666,536, optionally between 114,666,385 and 114,666,486, or between 114,666,425 and 114,666,446, or between 114,666,435 and 114,666,436 of mouse chromosome 12 with reference to NCBIM37 for the mouse genome, relating to mouse strain C57BL/6J or an equivalent position of mouse chromosome 12 from a different mouse strain or an equivalent position in the genome of another non-human vertebrate, e.g., a rat.
  • human heavy chain variable region DNA between co-ordinates 114, 666, 183 and 114, 666, 725, such as between 114 666 283
  • the insertion between co-ordinates 114,666,435 and 114,666,436 relating to mouse strain C57BL/6J is equivalent to an insertion between co-ordinates 1207826 and 1207827 on chromosome 12 with reference to the 129/SvJ genomic sequence of the GenBank® access number NT114985.2.
  • An insertion may be made at equivalent position in another genome, such as another mouse genome.
  • the cell or mammal of the invention comprises a human IgH VDJ region which comprises or consists of nucleotides 106,328,851-107,268,544, such as nucleotides 106,328,901-107,268,494, such as nucleotides 106,328,941-107,268,454, such as nucleotides 106,328,951-107,268,444 of human Chromosome 14, with reference to the GRCH37/hg19 sequence database, or insertion of equivalent nucleotides relating to chromosome 14 from a different human sequence or database.
  • the human insertion may be made between the regions indicated above.
  • a cell or mammal of the invention in one embodiment, comprises an insertion of the human kappa VJ region, suitably comprising or consisting of, in germline configuration, all of the V and J regions and intervening sequences from a human, the insertion of the human DNA being made between co-ordinates 70,673,918-70,675,517, such as between co-ordinates 70, 674,418 and 70 675, 017, such as between co-ordinates 70,674, 655-70,674,856, such as between co-ordinates 70,674, 705-70,674,906, such as between co-ordinates 70,674, 745-70,674,766, such as between co-ordinates 70,674,755 and 70,674,756 of mouse chromosome 6, numbering with reference to NCBIM37 for the mouse genome, relating to mouse strain C57BL/6J, or an insertion at an equivalent position in another genome, such as another mouse genome.
  • a cell or mammal of the invention comprises an insertion of nucleotides 89,159,079-89,630,437 and/or 89,941,714-90,266,976 of human chromosome 2 with reference to the GRCH37/hg19 sequence database (or equivalent nucleotides relating to chromosome 2 from a different human sequence or database), such as an insertion of these 2 discrete fragments without the intervening sequence, or an insertion of the complete 89,159,079-90,266,976 region.
  • the insertion may comprise, or consist, of:
  • the human insertion may be made between the regions indicated above.
  • a cell or mammal of the invention comprises an insertion of a human lambda region which comprises at least one human J ⁇ region (eg, a germline region) and at least one human C ⁇ region (eg, a germline region), optionally C ⁇ 6 and/or C ⁇ 7.
  • the cell or mammal comprises a plurality of human J ⁇ regions, optionally two or more of J ⁇ 1, J ⁇ 2, J ⁇ 6 and J ⁇ 7, optionally all of J ⁇ 1, J ⁇ 2, J ⁇ 6 and J ⁇ 7.
  • the cell or mammal comprises at least one human J ⁇ -C ⁇ cluster, optionally at least J ⁇ 7-C ⁇ 7.
  • the human JC cluster is inserted 3′ of the last endogenous J lambda or is inserted 3′ of the last endogenous J kappa region, suitably immediately 3′ of these sequences, or substantially immediately 3′ of these sequences.
  • the insertion into the mouse lambda locus is made downstream of the endogenous C1 gene segment, for example where there is a 3′ J1C1 cluster, suitably immediately 3′ of the C1 segment, or substantially immediately 3′ of the segment.
  • a human JC cluster is inserted into a kappa locus and any resulting cell or animal is heterozygous at that locus, such that the cell has one chromosome with human lambda DNA inserted into the kappa locus, and another chromosome with human kappa DNA at the endogenous kappa locus.
  • a cell or mammal of the invention comprises a human EA enhancer.
  • a cell or mammal may of the invention comprise an inserted human lambda VJ region, suitably comprising or consisting of, in germline configuration, all of the V and J regions and intervening sequences from a human, the inserted region comprises or consisting of nucleotides 22,375,509-23,327,984, such as nucleotides 22,375,559-23,327,934, such as nucleotides 22,375,599-23,327,894, such as nucleotides 22,375,609-23,327,884 from human Chromosome 22, with reference to the GRCH37/hg19 sequence database, or equivalent DNA from another human sequence or database.
  • the insertion into the mouse genome may be made between co-ordinates 19,027,763 and 19,061,845, such as between co-ordinates 19, 037, 763 and 19, 051, 845, such as between co-ordinates 19,047,451 and 19,047,652, such as between co-ordinates 19,047,491 and 19,047,602, such as between co-ordinates 19,047,541 and 19,047,562, such as between co-ordinates 19,047,551 and 19,047,552 of mouse Chromosome 16 (with reference to NCBIM37 for the mouse genome, relating to mouse strain C57BL/6J, equivalent to co-ordinates 1,293,646-1,293,647 of the 129 SvJ genomic sequence in the sequence file of NT — 039630.4), or may be an insertion at an equivalent position in other genome, such as another mouse genome.
  • the insertion of the human lambda nucleic acid into the mouse genome may alternatively be made between co-ordinates 70,673,918 and 70,675,517, such as between co-ordinates 70, 674,418 and 70 675, 017, such as between co-ordinates 70,674,655 and 70,674,856, such as between co-ordinates 70,674,705 and 70,674,806, such as between co-ordinates 70,674,745 and 70,674,766, such as between co-ordinates 70,674,755 and 70,674,756 of mouse Chromosome 6 (with reference to NCBIM37 for the mouse genome, relating to mouse strain C57BL/6J) or equivalent in another genome.
  • the human insertion may be made between the regions indicated above.
  • All specific human fragments described above may vary in length, and may for example be longer or shorter than defined as above, such as 500 bases, 1 KB, 2K, 3K, 4K, 5 KB, 10 KB, 20 KB, 30 KB, 40 KB or 50 KB or more, which suitably comprise all or part of the human V(D)J region, whilst preferably retaining the requirement for the final insert to comprise human genetic material encoding the complete heavy chain region and light chain region, as appropriate, as described above.
  • the 5′ end of the human insert described above is increased in length. Where the insert is generated in a stepwise fashion then the increase in length is generally in respect of the upstream (5′) clone.
  • the 3′ end of the last inserted human gene generally the last human J gene to be inserted is less than 2 kb, preferably less than 1 KB from the human-mouse join region.
  • the non-human mammal comprises some or all of the human light chain kappa VJ region as disclosed herein but not the human light chain lambda VJ region.
  • the cell or non-human mammal comprises a fully human lambda locus (lambda VJC regions from a human), a chimaeric kappa locus (human kappa VJ regions operatively linked to a host kappa constant region) and a chimaeric heavy chain locus, having a human VDJ region operatively linked to a host heavy chain constant region.
  • the genome comprises an insertion of V, D (heavy chain only) and J genes as described herein at the heavy chain locus and one light chain locus, or at the heavy chain locus and both light chain loci.
  • the genome is homozygous at one, or both, or all three loci.
  • the genome may be heterozygous at one or more of the loci, such as heterozygous for DNA encoding a chimaeric antibody chain and native (host cell) antibody chain.
  • the genome may be heterozygous for DNA capable of encoding 2 different antibody chains of the invention, for example, comprising 2 different chimaeric heavy chains or 2 different chimaeric light chains.
  • the invention relates to a non-human mammal or cell, and methods for producing said mammal or cell, as described herein, wherein the inserted human DNA, such as the human IgH VDJ region and/or light chain V, J regions are found on only one allele and not both alleles in the mammal or cell.
  • a mammal or cell has the potential to express both an endogenous host antibody heavy or light chain and a chimaeric heavy or light chain.
  • the human VDJ region, or light chain VJ region is not used in its entirety, but parts of the equivalent human VDJ or VJ region, such as the exons, from other species may be used, such as one or more V, D, or J exons from other species, or regulatory sequences from other species.
  • the sequences used in place of the human sequences are not human or mouse.
  • the sequences used may be from rodent, or, primate such as chimp.
  • 1, 2, 3, 4, or more, or all of the J regions from a primate other than a human may be used to replace, one, 2, 3, 4, or more or all of the human J exons in the VDJ/VJ region of the cells and animals of the invention.
  • the inserted human DNA such as the human IgH VDJ region, and/or light chain VJ regions, may be inserted such that they are operably linked in the genome with a mu constant region from a non-human, non-mouse species, such as a rodent or primate sequence, such as a rat sequence.
  • Non-human, non-mouse species from which DNA elements may be used in the present invention include rabbits, lamas, dromedary, alpacas, camels and sharks.
  • the inserted human DNA such as the human VDJ or VJ region, is not operably linked to the endogenous host mu sequence but rather to a non-host mu sequence.
  • Operable linkage suitably allows production of an antibody heavy or light chain comprising the human variable region.
  • the inserted human DNA such as the human IgH VDJ region (and/or light chain VJ regions) may be inserted into the host chromosome together with mu constant region nucleic acid which is not host mu constant region nucleic acid, and preferably is a mu constant region from a non-mouse, non-human species.
  • the inserted human DNA such as the human VDJ region (and/or light chain VJ regions) is operably linked to a non-human, non-mouse mu, and is able to form a chimaeric antibody heavy or light chain.
  • a non-mouse, non-human mu may be inserted into the host chromosome on a separate genetic element to that of the human variable region, or at a different location in the genome, suitably operably linked to the variable region such that a chimaeric antibody heavy or light can be formed.
  • the invention in an additional aspect relates to a non-human mammal or a cell whose genome comprises a plurality of human IgH V regions, one or more human D regions and one or more human J regions upstream of a host non-human mammal light chain constant region, arranged such that the cell or mammal is able to express a chimaeric antibody chain.
  • the invention also relates to a non-human mammal or a cell whose genome additionally or alternatively comprises a plurality of human Ig light chain V regions, and one or more human J regions upstream of a host non-human mammal heavy chain constant region, such that the cell or mammal is able to express a chimaeric antibody chain.
  • the cell or mammal may be able to express an antibody having both heavy and light chains, including at least one chimaeric antibody chain, as disclosed above.
  • the inserted human heavy chain variable regions may be any of those described herein, and may be inserted at the positions described above for insertion 5′ of the lambda and kappa constant regions.
  • the inserted human light chain variable regions may be those described above, and may be inserted at the positions described above for insertion 5′ of the heavy chain constant region.
  • the genome or the cell or non-human mammal of the invention may encode an antibody comprising an antibody chain having a human heavy chain variable region upstream of a mouse light chain constant region, or an antibody chain having a human light chain variable region upstream of a mouse heavy chain constant region, in combination with one of:
  • the invention also relates to a transgene encoding a plurality of human IgH V regions, one or more human D regions and one or more human J regions upstream of a host non-human mammal light chain constant region, optionally comprised within a vector.
  • the invention also relates to a transgene encoding a plurality of human Ig light chain V regions, and one or more human light chain J regions upstream of a host non-human mammal heavy chain constant region, optionally comprised within a vector.
  • the invention relates to a cell, or non-human mammal, the genome of which comprises: one or more human Ig light chain kappa V regions and one or more human Ig light chain kappa J regions upstream of all or part of the human kappa constant region.
  • the invention in another aspect relates to a cell, or non-human mammal, the genome of which comprises: one or more human Ig light chain lambda V regions and one or more human Ig light chain lambda J regions upstream of all or part of the human lambda constant region.
  • the light chain VJ and C regions are able to form antibody chains in vivo capable of specifically reacting with an antigen.
  • a human kappa and/or lambda region is inserted into the genome, in combination with insertion of the heavy chain VDJ region or part thereof, upstream of the host heavy chain constant region as disclosed herein.
  • the cell or non-human mammal of the invention may comprise:
  • the cell or non-human mammal of the invention may comprise
  • the insertion of the human VJC light chain DNA, or part thereof as disclosed above is made at the equivalent mouse locus.
  • the human light chain kappa VJC DNA, or part thereof is inserted immediately upstream or downstream of the mouse kappa VJC region.
  • the human light chain lambda VJC region or part thereof is inserted immediately upstream or downstream of the mouse lambda VJC region.
  • the human kappa VJC locus is inserted and not the human lambda VJC locus.
  • only the human lambda VJC locus is inserted and not the human kappa VJC locus. Insertions may be made using the techniques disclosed herein, and suitably do not remove the host sequences from the genome.
  • non-human mammal host VJC sequences may be inactivated in some way, by mutation, or inversion, or by insertion of the human variable region DNA, or by any other means.
  • the cell or non-human mammal of the invention may comprise an insertion of the complete VJC human region.
  • human kappa variable region DNA might be inserted into the genome in functional arrangement with a lambda constant region, for example inserted upstream of a lambda constant region.
  • human lambda region variable DNA might be inserted in functional arrangement with a kappa constant region, for example inserted upstream of a kappa constant region.
  • one or more non-human mammal control sequences such as the enhancer sequence(s) is maintained upstream of the nonhuman mammal Mu constant region, suitably in its native position with respect to the distance from the constant region.
  • one or more non-human mammal control sequences such as an enhancer sequence(s) are maintained downstream of the nonhuman mammal Mu constant region, suitably in its native position with respect to the distance from the constant region.
  • a non-human mammal switch sequence is maintained upstream of the non-human mammal Mu constant region, suitably in its native position with respect to distance from the constant region.
  • the host enhancer or switch sequences are operative in vivo with the host constant region sequence(s).
  • a switch sequence is neither human, nor native in the non-human mammal, for example in one aspect a non-human mammal switch sequence is not a mouse or human switch sequence.
  • the switch sequence may be, for example, a rodent or primate sequence, or a synthetic sequence.
  • the switch sequence may be a rat sequence where the non-human mammal is a mouse.
  • a mouse or human constant mu sequence may be placed under the control of a switch sequence from a rat, or chimp, or other switch sequence, suitably capable of allowing isotype switching to occur in vivo.
  • the switch sequence of the invention is a switch sequence comprising 3, 4, 5, 6 or more (up to 82) contiguous repeats of the repeat sequence GGGCT (SEQ ID no 46-50), such as a rat switch sequence.
  • rat switch herein it is meant that the switch is a wild-type switch corresponding to a switch from a rat genome or derived from such a switch.
  • the switch sequence of the invention is a rat switch sequence comprising the following repeats: GAGCT (296 repeats; SEQ ID No 18), GGGGT (50 repeats; SEQ ID No 19), and GGGCT (83 repeats; SEQ ID No 20).
  • the rat switch sequence comprises or consists of the sequence of SEQ ID no 1.
  • the switch is optionally a rat switch as described herein.
  • the switch sequence present in cells or mammal of the invention is a mouse switch, eg, is from a mouse such as a mouse 129 strain or mouse C57 strain, or from a strain derived therefrom, optionally comprising or consisting of the sequence of SEQ ID no 4 or 5.
  • mouse switch herein it is meant that the switch is a wild-type switch corresponding to a switch from a mouse genome or derived from such a switch.
  • the mouse switch sequence is optionally the endogenous switch or is a mouse switch from another mouse strain.
  • the cell or mammal of the invention may therefore comprise a human or non-human mammal switch sequence and a human or non-human mammal enhancer region or regions. They may be upstream of a human or non-human mammal constant region.
  • the control sequences are able to direct expression or otherwise control the production of antibodies comprising a constant region with which they are associated.
  • One combination envisaged is a rat switch with mouse enhancer sequences and mouse constant regions in a mouse cell.
  • the invention relates to a cell, preferably a non-human cell, or non-human mammal comprising an immunoglobulin heavy chain or light chain locus having DNA from 3 or more species.
  • the cell or animal may comprise host cell constant region DNA, one or more human V, D or J coding sequences and one or more non-human, non-host DNA regions that are able to control a region of the immunoglobulin locus, such as a switch sequence, promoter or enhancer which are able to control expression or isotype switching in vivo of the Ig DNA.
  • the cell or animal is a mouse and comprises additionally human DNA from the human Ig locus and additionally a non-mouse DNA sequence, such as a rat DNA sequence, capable of regulation of the mouse or human DNA.
  • the invention in another aspect relates to a cell, preferably non-human cell, or non-human mammal comprising an immunoglobulin heavy chain or light chain locus having DNA from 2 or more different human genomes.
  • a cell preferably non-human cell, or non-human mammal comprising an immunoglobulin heavy chain or light chain locus having DNA from 2 or more different human genomes.
  • it could comprise heavy chain V(D)J sequences from more than one human genome within a heavy or light chain, or heavy chain VDJ DNA from one genome and light chain VJ sequences from a different genome.
  • the invention relates to a DNA fragment or cell or non-human mammal comprising an immunoglobulin heavy chain or light chain locus, or part thereof, having DNA from 2 or more species, where one species contributes a non-coding region such as a regulatory region, and the other species coding regions such as V, D, J or constant regions.
  • the human promoter and/or other control elements that are associated with the different human V, D or J regions are maintained after insertion of the human VDJ into the mouse genome.
  • one or more of the promoter elements, or other control elements, of the human regions, such as the human V regions, are optimised to interact with the transcriptional machinery of a non-human mammal.
  • a human coding sequence may be placed under the control of an appropriate non-human mammal promoter, which allows the human DNA to be transcribed efficiently in the appropriate non-human animal cell.
  • the human region is a human V region coding sequence, and a human V region is placed under the control of a non-human mammal promoter.
  • the functional replacement of human promoter or other control regions by non-human mammal promoter or control regions may be carried out by use of recombineering, or other recombinant DNA technologies, to insert a part of the human Ig region (such as a human V region) into a vector (such as a BAC) containing a non-human Ig region.
  • the recombineering/recombinant technique suitably replaces a portion of the non-human (e.g. mouse) DNA with the human Ig region, and thus places the human Ig region under control of the non-human mammal promoter or other control region.
  • the human coding region for a human V region replaces a mouse V region coding sequence.
  • the human coding region for a human D region replaces a mouse D region coding sequence.
  • the human coding region for a human J region replaces a mouse J region coding sequence.
  • human V, D or J regions may be placed under the control of a non-human mammal promoter, such as a mouse promoter.
  • the only human DNA inserted into the non-human mammalian cell or animal are V, D or J coding regions, and these are placed under control of the host regulatory sequences or other (non-human, non-host) sequences,
  • reference to human coding regions includes both human introns and exons, or in another aspect simply exons and no introns, which may be in the form of cDNA.
  • recombineering or other recombinant DNA technologies, to insert a non-human-mammal (e.g. mouse) promoter or other control region, such as a promoter for a V region, into a BAC containing a human Ig region.
  • a recombineering step then places a portion of human DNA under control of the mouse promoter or other control region.
  • V, D and J regions may also be used to insert some or all of the V, D and J regions from the human heavy chain upstream of a light chain constant region, rather than upstream of the heavy chain constant region.
  • some or all of the human light chain V and J regions may be inserted upstream of the heavy chain constant region. Insertion may be at the endogenous constant region locus, for example between the endogenous constant and J region, and may be of some, or all, of the V, D or J genes alone, excluding promoter or enhancer sequences, or may be of some, or all, of the V, D or J genes with one or more or all respective promoter or enhancer sequences.
  • the full repertoire of V, D or J fragments in germline orientation may be inserted upstream and in functional arrangement with a host constant region.
  • the present invention allows V and/or D and/or J regions from a human, or any species, to be inserted into a chromosome of a cell from a different species that comprises a constant region, allowing a chimaeric antibody chain to be expressed.
  • the invention requires only that some human variable region DNA is inserted into the genome of a non-human mammal in operable arrangement with some, or all, of the human heavy chain constant region at the region of the endogenous heavy chain constant region locus such that an antibody chain can be produced.
  • the light chain DNA insertion can be in the form of a completely human construct, having both human variable DNA and human constant region DNA, or have human variable region DNA and constant region DNA from a non-human, non-host species.
  • Other variations are also possible, such as insertion of both of the light chain human variable region and host genome constant region.
  • the insertion of said light chain transgenes need not be at the equivalent endogenous locus, but may be anywhere in the genome.
  • the cell or mammal may produce chimaeric heavy chains (comprising human variable region DNA and mouse constant region DNA) and light chains comprising human variable and human constant region DNA.
  • the lambda and or kappa human variable region DNA can be inserted upstream of the endogenous locus, or downstream, or indeed on a different chromosome to the endogenous locus, and inserted with or without constant region DNA.
  • a further aspect of the invention relates to insertion of one or both light chain human variable regions downstream of the equivalent endogenous locus constant region, or elsewhere in the genome.
  • insertion of human variable region DNA at or close to the equivalent endogenous locus in the recipient genome is preferred, for example within 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 kb of the boundary (upstream or downstream) of a host immunoglobulin locus.
  • the invention can relate to a cell or non-human mammal whose genome comprises:
  • non-human mammal is able to produce a repertoire of chimaeric antibodies, or chimaeric light or heavy chains, having a non-human mammal constant region and a human variable region.
  • the genome of the cell or non-human mammal comprises:
  • the use of the methods of the invention allows a locus to be built up in a stepwise manner by sequential insertions, and thus allows for the insertion of human variable DNA together with human or non-human constant region DNA at any suitable location in the genome of a non-human host cell.
  • methods of the invention can be used to insert human immunoglobulin variable region DNA together with constant region DNA from the host genome anywhere in the genome of a non-human host cell, allowing a chimaeric antibody chain to be produced from a site other than the endogenous heavy region.
  • Any human heavy chain or light chain DNA construct contemplated above can be inserted into any desired position into the genome of a non-human host cell using the techniques described herein.
  • the present invention thus also relates to cells and mammals having genomes comprising such insertions.
  • the invention also relates to a vector, such as a BAC, comprising a human V, D or J region in a functional arrangement with a non-human mammal promoter, or other control sequence, such that the expression of the human V, D or J region is under the control of the non-human mammal promoter in a cell of the non-human mammal, such as an ES cell, in particular once inserted into the genome of that cell.
  • a vector such as a BAC, comprising a human V, D or J region in a functional arrangement with a non-human mammal promoter, or other control sequence, such that the expression of the human V, D or J region is under the control of the non-human mammal promoter in a cell of the non-human mammal, such as an ES cell, in particular once inserted into the genome of that cell.
  • the invention also relates to cells and non-human mammals containing said cells, which cells or mammals have a human V, D or J region in a functional arrangement with a non-human mammal promoter, or other control sequence, such that the expression of the human V, D or J region is under the control of the non-human mammal promoter in the cells or mammal.
  • one aspect of the invention thus relates to a non-human mammal host cell capable of expression of a human V, D or J coding sequence under the control of a host promoter or control region, the expression capable of producing a humanised antibody having a human variable domain and non-human mammal constant region.
  • the invention relates to a cell, such as a non mammalian cell, such as an ES cell, the genome of which comprises
  • the invention relates to a cell, such as a non-human mammal cells, such as ES cells whose genome comprises
  • the cell is an ES cell is capable of developing into a non-human mammal able to produce a repertoire of antibodies which are chimaeric, said chimaeric antibodies having a non-human mammal constant region and a human variable region.
  • the genome of the cell is modified to prevent expression of fully host-species specific antibodies.
  • the cell is an induced pluripotent stem cell (iPS cell).
  • iPS cell induced pluripotent stem cell
  • cells are isolated non-human mammalian cells.
  • a cell as disclosed herein is preferably a non-human mammalian cell.
  • the cell is a cell from a mouse strain selected from C57BL/6, M129 such as 129/SV, BALB/c, and any hybrid of C57BL/6, M129 such as 129/SV, or BALB/c.
  • the invention also relates to a cell line which is grown from or otherwise derived from cells as described herein, including an immortalised cell line.
  • the cell line may comprise inserted human V, D or J genes as described herein, either in germline configuration or after rearrangement following in vivo maturation.
  • the cell may be immortalised by fusion (eg, electrofusion or using PEG according to standard procedures) to a tumour cell (eg, P3X63-Ag8.653 (obtainable from LGC Standards; CRL-1580), SP2/0-Ag14 (obtainable from ECACC), NSI or NS0), to provide an antibody producing cell and cell line, or be made by direct cellular immortalisation.
  • the present invention also relates to vectors for use in the invention.
  • vectors for use in the invention.
  • such vectors are BACs (bacterial artificial chromosomes). It will be appreciated that other cloning vectors may be used in the invention, and therefore reference to BACs herein may be taken to refer generally to any suitable vector.
  • BACs used for generation of human DNA to be inserted such as the VDJ or VJ regions are trimmed so that in the final human VDJ or VJ region or part thereof in the non-human mammal, no sequence is duplicated or lost when compared to the original human genomic sequence.
  • the invention relates to a vector comprising an insert, preferably comprising a region of human DNA from some of the human VDJ or VJ locus, flanked by DNA which is not from that locus.
  • the flanking DNA may comprise one or more selectable markers or one or more site specific recombination sites.
  • the vector comprises 2 or more, such as 3, heterospecific and incompatible site specific recombination sites.
  • the site specific recombination sites may be loxP sites, or variants thereof, or FRT sites or variants thereof.
  • the vector comprises one or more transposon ITR (inverted terminal repeat) sequences.
  • non-human animals of the invention suitably do not produce any fully humanised antibodies. In one aspect this is because there is no DNA inserted from the human constant region. Alternatively there is no human constant region DNA in the genome capable of forming an antibody in conjunction with the inserted human variable region DNA component, for example due to mutation within any human constant region DNA or distance from any constant region human DNA and human variable region DNA.
  • human light chain constant region DNA may be included in the cell genome, such that a fully human lambda or kappa human antibody chain might be generated, but this would only be able to form an antibody with a chimaeric heavy chain, and not produce a fully human antibody having human variable and constant regions.
  • non-human mammal genome is modified to prevent expression of fully host-species specific antibodies.
  • Fully host species specific antibodies are antibodies that have both variable and constant regions from the host organism.
  • specific is not intended to relate to the binding of the antibodies produced by the cells or animals of the invention but rather to the origin of the DNA which encodes those antibodies.
  • the non-human mammal genome is modified to prevent expression of the native (fully host species specific) antibodies in the mammal by inactivation of all or a part of the host non-human mammal Ig loci.
  • inactivation or prevention of endogenous antibody or gene segment usage is, for example, substantially complete inactivation or prevention (substantially 100%, ie, essentially none (eg, less than 10, 5, 4, 3, 2, 1 or 0.5%) of the endogenous antibody chain (eg, no endogenous heavy chains) is expressed).
  • inactivation is more than 50% (ie, 50% or less of the antibodies or transcripts are of an endogenous antibody chain), 60%, 70%, 80%, 85%, 90%, 95%, 96%, 97%, 98% or 99%.
  • endogenous heavy chain expression is substantially inactivated such that no more than 85%, 90%, 95%, 96%, 97%, 98% or 99% of the heavy chain repertoire of the vertebrate (mammal) is provided by endogenous heavy chains.
  • endogenous heavy chain expression is substantially inactivated such that substantially none of the heavy chain repertoire of the vertebrate (mammal) is provided by endogenous heavy chains.
  • endogenous heavy chain expression is substantially inactivated such that no more than 85%, 90%, 95%, 96%, 97%, 98% or 99% of the kappa chain repertoire of the vertebrate (mammal) is provided by endogenous kappa chains.
  • endogenous kappa chain expression is substantially inactivated such that substantially none of the kappa chain repertoire of the vertebrate (mammal) is provided by endogenous kappa chains.
  • endogenous heavy chain expression is substantially inactivated such that no more than 85%, 90%, 95%, 96%, 97%, 98% or 99% of the lambda chain repertoire of the vertebrate (mammal) is provided by endogenous lambda chains.
  • endogenous lambda chain expression is substantially inactivated such that substantially none of the lambda chain repertoire of the vertebrate (mammal) is provided by endogenous lambda chains.
  • this is achieved by inversion of all or part of the non-human mammal VDJ region, or VJ region, optionally by insertion of one or more site specific recombinase sites into the genome and then use of these sites in recombinase-mediated excision or inversion of all or a part of the non-human mammal Ig locus.
  • a double inversion may be employed, the first to move the V(D)Js away from the endogenous locus and then a more local inversion which puts them in the correct orientation.
  • a single loxP site is used to invert the non-human mammal VDJ region to a centromeric locus or telomeric locus.
  • a mouse or mouse cell of the invention comprises inverted endogenous heavy chain gene segments (eg, VH, D and JH, such as the entire endogenous heavy chain VDJ region) that are immediately 3′ of position 119753123, 119659458 or 120918606 on an endogenous mouse chromosome 12.
  • VH, D and JH such as the entire endogenous heavy chain VDJ region
  • the genome of the mouse or cell is homozygous for said chromosome 12.
  • the invention also provides:—
  • a cassette for inversion and inactivation of endogenous non-human vertebrate (eg, mouse or rat) antibody chain gene segments the segments being part of an antibody chain locus sequence on a chromosome of a non-human vertebrate (eg, mouse or rat) cell (eg, ES cell) wherein the sequence is flanked at its 3′ end by a site-specific recombination site (eg, lox, rox or frt), the cassette comprising a nucleotide sequence encoding an expressible label or selectable marker and a compatible site-specific recombination site (eg, lox, rox or frt) flanked by a 5′ and a 3′ homology arm, wherein the homology arms correspond to or are homologous to adjacent stretches of sequence in the cell genome on a different chromosome or on said chromosome at least 10, 15, 20, 25, 30, 35, 40, 45 or 50 mb away from the endogenous gene segments.
  • the invention also provides:—
  • a cassette for inversion and inactivation of endogenous mouse antibody heavy chain gene segments the segments being part of a heavy chain locus sequence on chromosome 12 of a mouse cell (eg, ES cell) wherein the sequence is flanked at its 3′ end by a site-specific recombination site (eg, lox, rox or frt), the cassette comprising a nucleotide sequence encoding an expressible label or selectable marker and a compatible site-specific recombination site (eg, lox, rox or frt) flanked by a 5′ and a 3′ homology arm, wherein the homology arms correspond to or are homologous to adjacent stretches of sequence in the mouse cell genome on a different chromosome or on chromosome 12 at least 10, 15, 20, 25, 30, 35, 40, 45 or 50 mb away from the endogenous gene segments.
  • a site-specific recombination site eg, lox, rox or frt
  • the invention provides:—
  • a cassette for inversion and inactivation of endogenous mouse antibody heavy chain gene segments the segments being part of a heavy chain locus sequence on chromosome 12 of a mouse cell (eg, ES cell) wherein the sequence is flanked at its 3′ end by a site-specific recombination site (eg, lox, rox or frt), the cassette comprising a nucleotide sequence encoding an expressible label or selectable marker and a compatible site-specific recombination site (eg, lox, rox or frt) flanked by a 5′ and a 3′ homology arm, wherein (i) the 5′ homology arm is mouse chromosome 12 DNA from coordinate 119753124 to coordinate 119757104 and the 3′ homology arm is mouse chromosome 12 DNA from coordinate 119749288 to 119753123; or (ii) the 5′ homology arm is mouse chromosome 12 DNA from coordinate 119659459 to coordinate 119663126 and the 3′ homology arm is mouse
  • the invention provides a mouse or mouse cell whose genome comprises an inversion of a chromosome 12, wherein the inversion comprises inverted endogenous heavy chain gene segments (eg, VH, D and JH, such as the entire endogenous heavy chain VDJ region); wherein the mouse comprises a transgenic heavy chain locus comprising a plurality of human VH gene segments, a plurality of human D segments and a plurality of human JH segments operably connected upstream of an endogenous constant region (eg, C mu) so that the mouse or cell (optionally following differentiation into a B-cell) is capable of expressing an antibody comprising a variable region comprising sequences derived from the human gene segments; and wherein the inversion is (i) an inversion of mouse chromosome 12 from coordinate 119753123 to coordinate 114666436; (ii) an inversion of mouse chromosome 12 from coordinate 119659458 to coordinate 114666436; or (iii) an inversion of mouse chromosome 12 from coordinate 12
  • the endogenous gene segments are from a 129-derived mouse cell (eg, segments from an AB2.1 cell) and the homology arms are isogenic DNA (ie, identical to 129-derived endogenous sequences demarcated by the respective coordinates stated in (i) to (iii) above). Thus, no new sequence is created by homologous recombination using these homology arms.
  • the arms are from a mouse strain that is different from the endogenous strain.
  • the site-specific recombination sites are mutually compatible and mutually inverted such that, on expression of an associated recombinase enzyme (eg, Cre, Dre or Flp), recombination between the site in the inserted inversion cassette and the site flanking the endogenous gene segments is carried out, thereby inverting and moving the endogenous gene segments far upstream (5′) of their original location in the heavy chain locus. This inactivates endogenous heavy chain expression.
  • an associated recombinase enzyme eg, Cre, Dre or Flp
  • light chain inactivation can be performed by choosing the homology arms of the inversion cassette with reference to a chromosomal region spaced at least 10, 15, 20, 25, 30, 35, 40, 45 or 50 mb away from the endogenous light chain locus, the latter comprising a site-specific recombination site that is compatible with the site in the inversion cassette.
  • the expressible label is a fluorescent label, eg, GFP or a variant thereof (eg, YFP, CFP or RFP).
  • a label is used instead of a selection marker, such as one that confers resistance to allow for selection of transformants.
  • the invention provides a method of inactivating gene segments of an endogenous antibody locus, the method comprising
  • the genome of the progeny cell or vertebrate can comprise transgenic heavy and/or light chain loci, each capable of expressing antibody chains comprising human variable regions.
  • endogenous heavy and kappa light chain expression is inactivated by inverting endogenous heavy and kappa variable region gene segments according to the method of the invention.
  • endogenous lambda chain expression is also inactivated in this way.
  • endogenous regulatory elements eg, Smu and/or Emu
  • endogenous constant regions eg, Cmu and/or Cgamma
  • Sites that “flank” in the above contexts of the invention can be provided such that a site-specific recombination site immediately flanks the endogenous sequence or is spaced therefrom, eg, by no more than 250, 200, 250, 100, 50 or 20 kb in the 3′ direction.
  • non-human mammal genome into which human DNA is inserted comprises endogenous V, (D) and J regions, and the endogenous sequences have not been deleted.
  • the invention comprises a method for insertion of multiple DNA fragments into a DNA target, suitably to form a contiguous insertion in which the inserted fragments are joined together directly without intervening sequences.
  • the method is especially applicable to the insertion of a large DNA fragment into a host chromosome which can be carried out in a stepwise fashion.
  • the method comprises insertion of a first DNA sequence into a target, the sequence having a DNA vector portion and a first sequence of interest (X1); insertion of a second DNA sequence into the vector portion of the first sequence, the second DNA sequence having a second sequence of interest (X2) and a second vector portion; and then excising any vector sequence DNA separating X1 and X2 to provide a contiguous X1X2, or X2X1 sequence within the target.
  • the DNA target for insertion of the first DNA sequence may be a specific site or any point in the genome of a particular cell.
  • the general method is described herein in relation to the insertion of elements of the human VDJ region, but is applicable to insertion of any DNA region, from any organism, and in particular insertion of large DNA fragments of >100 kB, such as 100-250 kb, or even larger, such as that of the TCR or HLA.
  • Features and approaches described herein in respect of the VDJ insertion may be equally applied to the any of the methods disclosed
  • the inserted DNA is human DNA, such as the human VDJ or VJ region, is built up in the genome of a cell, such as an ES cell, in a stepwise manner using 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20 or more separate insertions for each heavy chain or light chain region. Fragments are suitably inserted at the same or substantially the same cell locus, e.g. ES cell locus, one after another, to form the complete VDJ or VJ region, or part thereof.
  • the present invention also relates to cells and non-human animals comprising intermediates in the process whose genomes may comprise only a partial VDJ region, such as only human variable region DNA.
  • the method for producing a transgenic non-human mammal comprises the insertion of human VDJ or VJ regions upstream of the host non-human mammal constant region by step-wise insertion of multiple fragments by homologous recombination, preferably using an iterative process.
  • fragments of approximately 100 KB from the human VDJ and VJ locus are inserted, suitably to form part of, or a complete, VDJ or VJ region after the final iteration of the insertion process, as disclosed herein.
  • the insertion process commences at a site where an initiation cassette has been inserted into the genome of a cell, such as an ES cell, providing a unique targeting region.
  • the initiation cassette is inserted in the non-human mammal heavy chain locus, for use in insertion of human heavy chain DNA.
  • an initiation cassette may be inserted in the non-human mammal light chain locus, for use in insertion of human light chain VJ DNA
  • the initiation cassette suitably comprises a vector backbone sequence with which a vector having a human DNA fragment in the same backbone sequence can recombine to insert the human DNA into the cell (e.g. ES) cell genome, and suitably a selection marker, such as a negative selection marker.
  • the vector backbone sequence is that of a BAC library, to allow BACs to be used in the construction of the ES cells and mammals.
  • the vector backbone sequence may however be any sequence which serves as a target site into which a homologous sequence can insert, for example by homologous recombination, for example RMCE, and is preferably not DNA encoding any of the VDJ or constant region.
  • the insertion of the first DNA fragment into an initiation cassette is followed by insertion of a second DNA fragment into a portion of the first DNA fragment, suitably a part of the vector backbone of the second DNA fragment.
  • an inserted DNA fragment comprises a part of the human VDJ region flanked by 5′ and/or 3′ sequences that are not from the human VDJ region.
  • the 5′ and/or 3′ flanking sequences may each contain one or more selectable markers, or be capable of creating a selectable system once inserted into the genome.
  • one or both flanking sequences may be removed from the genome in vitro, or in vivo, following insertion.
  • the method comprises insertion of a DNA fragment followed by selection of both 5′ and 3′ ends of the inserted fragment flanking the human VDJ DNA.
  • iterative insertion is made by insertion of DNA fragments at the 5′ end of the previous inserted fragment, and in this aspect there may be deletion in vivo of the vector DNA which separates the inserted human DNA sequences, to provide a contiguous human DNA sequence.
  • insertion of human VDJ DNA into a genome may be achieved without leaving any flanking DNA in the genome, for example by transposase mediate DNA excision.
  • transposase is the Piggybac transposase.
  • the first human variable region fragment is inserted by homologous recombination at the initiation cassette backbone sequence and then the DNA of any negative selection marker and initiation cassette are subsequently removed by recombination between recombinase target sequences, such as FRT using in this example, FLPase expression.
  • recombinase target sequences such as FRT using in this example, FLPase expression.
  • targeted insertions at the (e.g. BAC) backbone initiation sequence and subsequent removal by rearrangement between recombinase target sequences are repeated to build up the entire human VDJ region upstream of the host non-mammal constant region.
  • a selectable marker or system may be used in the method.
  • the marker may be generated upon insertion of a DNA fragment into a genome, for example forming a selectable marker in conjunction with a DNA element already present in the genome.
  • the cell (e.g. ES) cell genome does not contain 2 identical selectable markers at the same time during the process. It can be seen that the iterative process of insertion and selection can be carried out using only 2 different selection markers, as disclosed in the examples herein, and for example the third selectable marker may be identical to the first marker, as by the time of insertion of the third vector fragment the first vector fragment and the first marker has been removed.
  • a correct insertion event is confirmed before moving to the next step of any multistep cloning process, for example by confirmation of BAC structure using high density genomic arrays to screen ES cells to identify those with intact BAC insertions, sequencing and PCR verification.
  • the invention also relates to a polynucleotide ‘landing pad’ sequence, the polynucleotide comprising nucleic acid regions homologous to regions of a target chromosome to allow for insertion by homologous recombination into the target chromosome, and comprising a nucleic acid site which permits recombinase-driven insertion of nucleic acid into the landing pad.
  • the invention also relates to vectors, cells and mammals of the invention comprising a landing pad as disclosed herein inserted into the genome of the cell.
  • the landing pad optionally comprises a non-endogenous S-mu, e.g. a rat S-mu switch
  • the landing pad optionally comprises (in 5′ to 3′ orientation) a mouse E ⁇ sequence, a non-human, non-mouse (e.g. rat) Switch ⁇ and at least a portion of a mouse C ⁇ or the entire mouse C ⁇ .
  • a non-human, non-mouse e.g. rat
  • the rat switch sequence optionally comprises or consists of SEQ ID NO 1.
  • the landing pad optionally comprises the 5′ homology arm of SEQ ID NO 6.
  • the landing pad optionally has the sequence of SEQ ID 2 or SEQ ID NO 3.
  • the landing pad comprises an expressible label.
  • the label is a fluorescent label, eg, GFP or a variant thereof (eg, YFP, CFP or RFP).
  • a label is used instead of a selection marker (such as one that confers resistance to allow for selection of transformants).
  • the landing pad comprises 5′ and 3′ homology arms for insertion into the cell genome using homologous recombination.
  • the homology arms can be isogenic DNA (eg, identical to 129-derived endogenous sequences of when a 129-derived ES cell is used). Thus, no new sequence is created by homologous recombination using these homology arms.
  • the arms are from a mouse strain that is different from the endogenous strain (ES cell strain).
  • the methods of the invention include methods wherein the landing pad sequence comprises any of the configurations or sequences as disclosed herein.
  • Another method of the invention comprises the step of insertion of the landing pad into a mouse chromosome by homologous recombination between mouse J1-4 and mouse C mu sequences.
  • Another method of the invention comprises the step of insertion of the landing pad into the mouse chromosome 12 by homologous recombination between mouse J1-4 and E mu.
  • the method uses site specific recombination for insertion of one or more vectors into the genome of a cell, such as an ES cell.
  • Site specific recombinase systems are well known in the art and may include Cre-lox, and FLP/FRT or combinations thereof, in which recombination occurs between 2 sites having sequence homology.
  • recombinases and sites that may be used in the present invention include Dre recombinase, rox sites, and PhiC31 recombinase.
  • BACs are available from the Sanger centre, see “A genome-wide, end-sequenced 129Sv BAC library resource for targeting vector construction”. Adams D J, Quail M A, Cox T, van der Weyden L, Gorick B D, Su Q, Chan W I, Davies R, Bonfield J K, Law F, Humphray S, Plumb B, Liu P, Rogers J, Bradley A. Genomics. 2005 December; 86 (6):753-8. Epub 2005 Oct. 27. The Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire CB10 1SA, UK. BACs containing human DNA are also available from, for example, InvitrogenTM. A suitable library is described in Osoegawa K et al, Genome Research 2001. 11: 483-496.
  • a method of the invention specifically comprises:
  • the invention comprises
  • the non-human mammal is able to generate a diversity of at least 1 ⁇ 10 6 different functional chimaeric immunoglobulin sequence combinations.
  • the targeting is carried out in ES cells derived from the mouse C57BL/6N, C57BL/6J, 129S5 or 129Sv strain.
  • non-human animals such as mice are generated in a RAG-1-deficient or a RAG-2-deficient background, or other suitable genetic background which prevents the production of mature host B and T lymphocytes.
  • non-human mammal is a rodent, suitably a mouse
  • cells of the invention are rodent cells or ES cells, suitably mouse ES cells.
  • the ES cells of the present invention can be used to generate animals using techniques well known in the art, which comprise injection of the ES cell into a blastocyst followed by implantation of chimaeric blastocystys into females to produce offspring which can be bred and selected for homozygous recombinants having the required insertion.
  • the invention relates to a chimeric animal comprised of ES cell-derived tissue and host embryo derived tissue.
  • the invention relates to genetically-altered subsequent generation animals, which include animals having a homozygous recombinants for the VDJ and/or VJ regions.
  • the invention relates to a method for producing an antibody specific to a desired antigen the method comprising immunizing a transgenic non-human mammal as above with the desired antigen and recovering the antibody (see e.g. Harlow, E. & Lane, D. 1998, 5 th edition, Antibodies: A Laboratory Manual, Cold Spring Harbor Lab. Press, Plainview, N.Y.; and Pasqualini and Arap, Proceedings of the National Academy of Sciences (2004) 101:257-259).
  • an immunogenic amount of the antigen is delivered.
  • the invention also relates to a method for detecting a target antigen comprising detecting an antibody produced as above with a secondary detection agent which recognises a portion of that antibody.
  • the invention relates to a method for producing a fully humanised antibody comprising immunizing a transgenic non-human mammal as above with the desired antigen, recovering the antibody or cells expressing the antibody, and then replacing the non-human mammal constant region with a human constant region.
  • This can be done by standard cloning techniques at the DNA level to replace the non-human mammal constant region with an appropriate human constant region DNA sequence—see e.g. Sambrook, J and Russell, D. (2001, 3'd edition) Molecular Cloning: A Laboratory Manual (Cold Spring Harbor Lab. Press, Plainview, N.Y.).
  • the invention relates to humanised antibodies and antibody chains produced according to the present invention, both in chimaeric and fully humanised form, and use of said antibodies in medicine.
  • the invention also relates to a pharmaceutical composition comprising such an antibodies and a pharmaceutically acceptable carrier or other excipient.
  • Antibody chains containing human sequences such as chimaeric human—non-human antibody chains, are considered humanised herein by virtue of the presence of the human protein coding regions region.
  • Fully humanised antibodies may be produced starting from DNA encoding a chimaeric antibody chain of the invention using standard techniques.
  • chimaeric antibodies or antibody chains generated in the present invention may be manipulated, suitably at the DNA level, to generate molecules with antibody-like properties or structure, such as a human variable region from a heavy or light chain absent a constant region, for example a domain antibody; or a human variable region with any constant region from either heavy or light chain from the same or different species; or a human variable region with a non-naturally occurring constant region; or human variable region together with any other fusion partner.
  • the invention relates to all such chimaeric antibody derivatives derived from chimaeric antibodies identified according to the present invention.
  • the invention relates to use of animals of the present invention in the analysis of the likely effects of drugs and vaccines in the context of a quasi-human antibody repertoire.
  • the invention also relates to a method for identification or validation of a drug or vaccine, the method comprising delivering the vaccine or drug to a mammal of the invention and monitoring one or more of: the immune response, the safety profile; the effect on disease.
  • the invention also relates to a kit comprising an antibody or antibody derivative as disclosed herein and either instructions for use of such antibody or a suitable laboratory reagent, such as a buffer, antibody detection reagent.
  • a suitable laboratory reagent such as a buffer, antibody detection reagent.
  • the invention also relates to a method for making an antibody, or part thereof, the method comprising providing:
  • the present invention also relates to a chimaeric antibody comprising a human variable region and a non-human vertebrate or mammal (optionally a rat or mouse) constant region (optionally a C gamma or C mu), wherein the antibody is encoded by a nucleotide sequence corresponding to the nucleotide sequence of a chimaeric heavy chain locus of a cell (optionally a B-cell, ES cell or hybridoma), the locus comprising a non-human vertebrate constant region nucleotide sequence and a rearranged VDJ nucleotide sequence produced by the in vivo rearrangement of a human V region, a human D region and a human J region, the V region being selected from one of a V1-3 region, V2-5 region, V4-4 region, V1-2 region or V6-1 region, and optionally a V1-3 or V6-1 segment.
  • a chimaeric antibody comprising a human variable region and a non-
  • the J region is any of JH1, JH2, JH3, JH4, JH5 or JH6, and in one aspect is JH4 or JH6.
  • the D region is, in one aspect, any D3-9, D3-10, D6-13 or D6-19.
  • rearranged VDJ nucleotide sequence is produced by the in vivo rearrangement of human V1-3 and JH4 (optionally with D3-9, D3-10, D6-13 or D-19); or V1-3 and JH6 (optionally with D3-9, D3-10, D6-13 or D-19); or V6-1 and JH4 (optionally with D3-9, D3-10, D6-13 or D-19); or V6-1 and JH6 (optionally with D3-9, D3-10, D6-13 or D-19).
  • the rearranged VDJ nucleotide sequence is produced by the in vivo rearrangement of human V6-1 DH3-10, V1-3 DH3-10, V1-3 DH6-19, V1-3 Dh3-9 or V6-1 DH6-19.
  • the antibody comprises any combination exemplified in the Examples and Figures herein.
  • the in vivo rearrangement is in a cell (eg, B cell or ES cell) derived from the same non-human vertebrate species as the constant region sequence (eg, a mouse B cell or ES cell).
  • the invention also relates to a non-human vertebrate or mammal cell (eg, a B-cell or ES cell or hybridoma) whose genome comprises a chimaeric heavy chain locus as described above in this paragraph.
  • a non-human vertebrate or mammal eg, a mouse or rat
  • a non-human vertebrate or mammal eg, a mouse or rat
  • the present invention also relates to a non-human vertebrate or mammal having a genome encoding a chimaeric antibody, the chimaeric antibody comprising a human variable region and a non-human vertebrate or mammal (optionally a rat or mouse) constant region (optionally a C gamma or C mu), the mammal:
  • the invention also relates to a chimaeric antibody comprising a human variable region and a non-human vertebrate or mammal (optionally a rat or mouse) constant region (optionally a light chain constant region), wherein the antibody is obtainable from a mammal (optionally a rat or mouse) whose genome comprises an antibody chain locus comprising a germline human kappa V1-8 and germline human kappa J1 sequence, and wherein the antibody is obtainable by in vivo recombination in said mammal of the V1-8 and J1 sequences and wherein the antibody has a variable region sequence which is different from that which is encoded by germline human kappa V1-8 and germline human kappa J1 sequences.
  • the human germline sequences are able to undergo productive rearrangement to form a coding sequence which, in conjunction with the non-human constant region sequence, can be expressed as a chimaeric antibody chain having at least a complete human variable region and a non-human constant region.
  • a chimaeric antibody chain having at least a complete human variable region and a non-human constant region.
  • the combination of the germline human kappa V1-8 and germline human kappa J1 sequences per se which do not provide for an antibody coding sequence (due to the inclusion of stop codons).
  • the rearranged sequence of the chimaeric antibody is a result of somatic hypermutation.
  • the antibody is a kappa antibody; in another aspect the antibody comprises a non-human heavy chain constant region (eg, a rat or mouse C gamma or C mu).
  • Such motifs are not found in the equivalent position in the germline sequence as shown in the examples.
  • the invention also relates to a non-human vertebrate or mammal cell (eg, a B-cell or ES cell or hybridoma) whose genome comprises a chimaeric antibody chain locus as described above in this paragraph.
  • a non-human vertebrate or mammal eg, a mouse or rat
  • a non-human vertebrate or mammal eg, a mouse or rat
  • the invention also relates to a chimaeric antibody comprising a human variable region and a non-human vertebrate or mammal (optionally a rat or mouse) constant region (optionally a light chain constant region), wherein the antibody is obtainable from a mammal (optionally a rat or mouse) whose genome comprises an antibody chain locus comprising a germline human kappa V1-6 and germline human kappa J1 sequence, and wherein the antibody is obtainable by in vivo recombination in said mammal of the V1-6 and J1 sequences and wherein the antibody has a variable region sequence which is different from that which is encoded by germline human kappa V1-6 and germline human kappa J1 sequences.
  • the human germline sequences are able to undergo productive rearrangement to form a coding sequence which, in conjunction with the non-human constant region sequence, can be expressed as a chimaeric antibody chain having at least a complete human variable region and a non-human constant region.
  • a chimaeric antibody chain having at least a complete human variable region and a non-human constant region.
  • the combination of the germline human kappa V1-6 and germline human kappa J1 sequences per se which do not provide for an antibody coding sequence (due to the inclusion of stop codons).
  • the rearranged sequence of the chimaeric antibody is a result of somatic hypermutation.
  • the antibody is a kappa antibody; in another aspect the antibody comprises a non-human heavy chain constant region (eg, a rat or mouse C gamma or C mu).
  • motifs are not found in the equivalent position in the germline sequence as shown in the examples.
  • the invention also relates to a non-human vertebrate or mammal cell (eg, a B-cell or ES cell or hybridoma) whose genome comprises a chimaeric antibody chain locus as described above in this paragraph.
  • a non-human vertebrate or mammal eg, a mouse or rat
  • a non-human vertebrate or mammal eg, a mouse or rat
  • the invention also relates to a chimaeric antibody comprising a human variable region and a non-human (optionally a rat or mouse) constant region (optionally a C gamma or C mu or a C kappa), wherein the antibody is obtainable from a mammal (optionally a rat or mouse) whose genome comprises an antibody chain locus comprising a germline human kappa V1-5 and germline human kappa J1 sequence, and wherein the antibody is obtainable by in vivo recombination in said mammal of the V1-5 and J1 sequences.
  • the invention also relates to a non-human vertebrate or mammal cell (eg, a B-cell or ES cell or hybridoma) whose genome comprises a chimaeric antibody chain locus as described above in this paragraph.
  • a non-human vertebrate or mammal eg, a mouse or rat
  • a non-human vertebrate or mammal eg, a mouse or rat
  • the invention also relates to a chimaeric antibody comprising a human variable region and a non-human (optionally a rat or mouse) constant region (optionally a C gamma or C mu or a C kappa), wherein the antibody is obtainable from a mammal (optionally a rat or mouse) whose genome comprises an antibody chain locus comprising a germline human kappa V1-5 and germline human kappa J4 sequence, and wherein the antibody is obtainable by in vivo recombination in said mammal of the V1-5 and J4 sequences.
  • the invention also relates to a non-human vertebrate or mammal cell (eg, a B-cell or ES cell or hybridoma) whose genome comprises a chimaeric antibody chain locus as described above in this paragraph.
  • a non-human vertebrate or mammal eg, a mouse or rat
  • a non-human vertebrate or mammal eg, a mouse or rat
  • Antibodies of the invention may be isolated, in one aspect being isolated from the cell or organism in which they are expressed.
  • a non-human mammal whose genome comprises:
  • non-human mammal is able to produce a repertoire of chimaeric antibodies having a non-human mammal constant region and a human variable region
  • non-human mammal genome is modified to prevent expression of fully host-species specific antibodies.
  • a non-human mammal ES cell whose genome comprises:
  • the ES cell is capable of developing into a non-human mammal, being able to produce a repertoire of antibodies which are chimaeric, having a non-human mammal constant region and a human variable region.
  • a method for producing a transgenic non-human mammal able to produce a repertoire of chimaeric antibodies, the antibodies having a non-human mammal constant region and a human variable region comprising inserting by homologous recombination into a non-human mammal ES cell genome
  • steps (a) and (b) can be carried out in either order and each of steps (a) and (b) can be carried out in a stepwise manner or as a single step.
  • the insertion of human VDJ or VJ regions upstream of the host non-human mammal constant region is accomplished by step-wise insertion of multiple fragments by homologous recombination.
  • the step-wise insertions commence at a site where an initiation cassette has been inserted into the genome of an ES cell providing a unique targeting region consisting of a BAC backbone sequence and a negative selection marker.
  • the first human variable region fragment is inserted by homologous recombination at the initiation cassette BAC backbone sequence and said negative selection marker and initiation cassette are subsequently removed by recombination between recombinase target sequences.
  • repeated targeted insertions at the BAC backbone initiation sequence and subsequent removal of the backbone by rearrangement between recombinase target sequences is repeated to build up the entire human VDJ region upstream of the host non-mammal constant region.
  • a cell or non human mammal wherein the mammal is a mouse or the cell is a mouse cell and wherein the insertion of the human heavy chain DNA is made in a mouse genome between coordinates 114,667,091 and 114,665,190 of mouse chromosome 12.
  • the human IgH VDJ region comprises nucleotides 105,400,051 to 106,368,585 from human chromosome 14 (coordinates refer to NCBI36 for the human genome).
  • a method, cell or non human mammal wherein a human coding region DNA sequence is in a functional arrangement with a non-human mammal control sequence, such that transcription of the human DNA is controlled by the non-human mammal control sequence.
  • the initiation cassette is inserted between the mouse J4 and C alpha exons.
  • an initiation cassette suitable for use in the method comprising a vector backbone sequence and a selection marker.
  • the invention provides the following aspects (starting at aspect number 103):—
  • the invention further provides:—
  • said inserted DNA sequence comprises a human nucleotide sequence comprising said human antibody gene segments, wherein the nucleotide sequence is at least 110, 130, 150, 170, 190, 210, 230, 250, 270 or 290 kb.
  • the nucleotide sequence corresponds to a stretch of DNA sequence of human chromosome 14, comprising the gene segments and intervening sequences in germline configuration, eg, at least a sequence corresponding to the nucleotide sequence from coordinate 106328951 to coordinate 106601551 of a human chromosome 14, eg, a sequence in the GRCH37/hg19 sequence database.
  • the transgenic locus is a light chain kappa locus and the human antibody gene segments are between the 3′-most endogenous Jk gene segment and endogenous Ck; optionally wherein the human antibody gene segments comprise five functional human J ⁇ -C ⁇ clusters and at least one human VA gene segment, eg, at least a sequence corresponding to the nucleotide sequence from coordinate 23217291 to 23327884 of a lambda locus found on a human chromosome 22.
  • transgenic locus is a heavy chain locus and the human antibody gene segments are inserted between the 3′-most endogenous JH gene segment (eg, JH4 in a mouse genome) and endogenous Cmu.
  • Regeneration of the capture surface can be carried out with 10 mM glycine at pH1.7. This removes the captured antibody and allows the surface to be used for another interaction.
  • the binding data can be fitted to 1:1 model inherent using standard techniques, eg, using a model inherent to the ProteOn XPR36TM analysis software.
  • the invention also relates to an scFv, diabody or other antibody fragment comprising a VH and VL domain from an antibody or fragment of aspect 126 (optionally following affinity maturation, eg, by phage display).
  • the antigen is a serpin, eg, ovalbumin, antithrombin or antitrypsin.
  • Serpins are a group of proteins with similar structures that were first identified as a set of proteins able to inhibit proteases. The acronym serpin was originally coined because many serpins inhibit chymotrypsin-like serine proteases (serine protease inhibitors). The first members of the serpin superfamily to be extensively studied were the human plasma proteins antithrombin and antitrypsin, which play key roles in controlling blood coagulation and inflammation, respectively.
  • a method for producing an antibody specific to a desired antigen comprising immunizing a non-human mammal as disclosed herein with the desired antigen and recovering the antibody or a cell producing the antibody.
  • a method for producing a fully humanised antibody comprising immunizing a non-human mammal as disclosed herein and then replacing the non-human mammal constant region of an antibody specifically reactive with the antigen with a human constant region, suitably by engineering of the nucleic acid encoding the antibody.
  • the human coding region V, D or J region is in a functional arrangement with a mouse promoter sequence.
  • the invention also relates to a humanised antibody produced according to any methods disclosed herein and use of a humanised antibody so produced in medicine.
  • FIGS. 1-8 show an iterative process for insertion of a series of human BACs into a mouse Ig locus
  • FIGS. 9-18 show in more detail the process of FIGS. 1-8 for the IgH and kappa locus
  • FIGS. 19 and 20 show the principles behind antibody generation in chimaeric mice
  • FIG. 21 shows a possible insertion site for the human DNA in a mouse chromosome
  • FIGS. 22-26 disclose an alternative iterative process for insertion of a series of human BACs into a mouse Ig locus
  • FIGS. 27-29 illustrate a mechanism for inversion of the host VDJ region
  • FIG. 30 illustrates proof of principle for insertion of a plasmid using an RMCE approach
  • FIG. 31 illustrates sequential RMCE—Integration into Landing Pad
  • FIG. 32 illustrates confirmation of Successful Insertion into Landing Pad
  • FIG. 33 illustrates PCR Confirmation of 3′ End Curing
  • FIG. 34 illustrates insertion of BAC#1 and PCR Diagnostics
  • FIG. 35 illustrates JH and JK usage
  • FIG. 36 illustrates DH usage
  • FIG. 37 illustrates the distribution of CDR-H3 length in human VDJC ⁇ transcripts from chimera mice
  • FIG. 38 illustrates the distribution of nucleotide numbers of deletion and insertion in IGH-VDI or IGK-VJ junctions
  • FIG. 39 illustrates Distribution of JH Usage Within Each VHs
  • FIG. 40 illustrates Distribution of DH Usage Within Each VHs
  • FIG. 41 illustrates Nucleotide Gain or Loss at VJ Joints Generates IGK Variants
  • FIG. 42 illustrates Hypermutaion in J Regions Generates IGK Variants
  • FIG. 43 illustrates Joint Diversity Produces Functional CDS
  • FIG. 44 illustrates a plot of identity of J H gene segment use a 5′-RACE C ⁇ -specific library generated from the splenic B lymphocytes of transgenic mice according to the invention in which endogenous gene segment use has been inactivated by inversion
  • FIG. 45 illustrates the ratio of mouse V H to human V H usage as determined from antibody sequences from splenic B lymphocytes of transgenic mice according to the invention in which endogenous gene segment use has been inactivated by inversion
  • FIG. 46 illustrates inversion strategy schematic
  • FIG. 47 illustrates targeting construct R57 for inversion
  • FIG. 48 illustrates sequence analysis from a C ⁇ -specific 5′-RACE library of splenic B lymphocytes of S1 inv1 (one human IGH BAC (ie, multiple human VH, all functional human D and JH) with an inverted endogenous IGH locus) mouse shows that practically all the transcripts came from rearranged human V H -D-J H gene segments
  • FIG. 49 illustrates that the S1 inv1 mouse shows a similar usage of both D and J H gene segments to human
  • FIG. 50 illustrates that mouse V H usage is further significantly reduced following insertion of the 2 nd human BAC into the endogenous heavy chain locus
  • FIG. 51 illustrates a gel showing that normal class-switching (to IgG-type) was observed in transcripts from mice of the invention.
  • the rearranged transcripts were detected using RT-PCR with human VH-specific and mouse C ⁇ -specific primers for amplification from peripheral blood cells of immunized transgenic mice
  • FIG. 52 illustrates sequence analysis amplified fragments demonstrate hypermutation occurred within the human variable regions of these IG ⁇ chains from mice of the invention
  • FIG. 53 illustrates Flow cytometric analysis showing normal B-cell compartments in transgenic mice of the invention
  • FIGS. 54 & 55 illustrate normal IgH isotypes and serum levels are obtained in transgenic animals of the invention following immunisation with antigens
  • SEQ ID No 1 is a Rat switch sequence
  • SEQ ID No 2 is a landing pad targeting vector (long version)
  • SEQ ID No 3 is a landing pad targeting vector (shorter version)
  • SEQ ID No 4 is the mouse strain 129 switch
  • SEQ ID No 5 is the mouse strain C57 switch
  • SEQ ID No 6 is the 5′ homology arm of a landing pad
  • SEQ ID No 7 is oligo HV2-5
  • SEQ ID No 8 is oligo HV4-4
  • SEQ ID No 9 is oligo HV1-3
  • SEQ ID No 10 is oligo HV1-2
  • SEQ ID No 11 is oligo HV6-1
  • SEQ ID No 12 is oligo C ⁇
  • SEQ ID No 13 is oligo KV1-9
  • SEQ ID No 14 is oligo KV1-8
  • SEQ ID No 15 is oligo KV1-6
  • SEQ ID No 16 is oligo KV1-5
  • SEQ ID No 17 is oligo OK
  • SEQ ID Nos 18-20 are rat switch sequences
  • SEQ ID No 25 is Primer E1554
  • SEQ ID No 26 is Primer E1555
  • SEQ ID No 27 is Primer ELP1352_C ⁇ 1
  • SEQ ID No 28 is Primer ELP1353_C ⁇ 2b
  • SEQ ID No 29 is Primer ELP1354_C ⁇ 2a
  • SEQ ID No 30 is Primer ELP1356_VH4-4
  • SEQ ID No 31 is Primer ELP1357_VH1-2,3
  • SEQ ID No 32 is Primer ELP1358_VH6-1
  • SEQ ID No 33 is Primer mIgG1 — 2 rev
  • SEQ ID No 34 is Primer mIgG2b rev
  • SEQ ID No 35 is Primer mIgG2a — 2 rev
  • SEQ ID No 36 is Primer mCH1 unirev
  • SEQ ID No 37 is Primer mCH1 unirev — 2
  • SEQ ID Nos 38-45 are CDRH3 sequences
  • SEQ ID Nos 46-50 is 3, 4, 5, 6 or more (up to 82) repeats of GGGCT
  • SEQ ID Nos 51-55 are heavy chain CDR1 sequences against CTB (cloned and reference)
  • SEQ ID Nos 56-60 are heavy chain CDR2 sequences against CTB (cloned and reference)
  • SEQ ID NOs 61-63 are heavy chain CDR3 sequences against CTB (cloned and reference)
  • SEQ ID Nos 64-68 are J Region sequences against CTB (cloned and reference)
  • the words “comprising” (and any form of comprising, such as “comprise” and “comprises”), “having” (and any form of having, such as “have” and “has”), “including” (and any form of including, such as “includes” and “include”) or “containing” (and any form of containing, such as “contains” and “contain”) are inclusive or open-ended and do not exclude additional, unrecited elements or method steps
  • A, B, C, or combinations thereof refers to all permutations and combinations of the listed items preceding the term.
  • A, B, C, or combinations thereof is intended to include at least one of: A, B, C, AB, AC, BC, or ABC, and if order is important in a particular context, also BA, CA, CB, CBA, BCA, ACB, BAC, or CAB.
  • expressly included are combinations that contain repeats of one or more item or term, such as BB, AAA, MB, BBC, AAABCCCC, CBBAAA, CABABB, and so forth.
  • BB BB
  • AAA AAA
  • MB BBC
  • AAABCCCCCC CBBAAA
  • CABABB CABABB
  • the Kabat Database (G. Johnson and T. T. Wu, 2002; World Wide Web (www) kabatdatabase.com). Created by E. A. Kabat and T. T. Wu in 1966, the Kabat database publishes aligned sequences of antibodies, T-cell receptors, major histocompatibility complex (MHC) class I and II molecules, and other proteins of immunological interest.
  • MHC major histocompatibility complex
  • a searchable interface is provided by the SeqhuntII
  • IMGT the International ImMunoGeneTics Information System®; M.-P. Lefranc, 2002; World Wide Web (www) imgt.cines.fr).
  • IMGT is an integrated information system that specializes in antibodies, T cell receptors, and MHC molecules of all vertebrate species. It provides a common portal to standardized data that include nucleotide and protein sequences, oligonucleotide primers, gene maps, genetic polymorphisms, specificities, and two-dimensional (2D) and three-dimensional (3D) structures.
  • IMGT includes three sequence databases (IMGT/LIGM-DB, IMGT/MHC-DB, IMGT/PRIMERDB), one genome database (IMGT/GENE-DB), one 3D structure database (IMGT/3Dstructure-DB), and a range of web resources (“IMGT Marie-Paule page”) and interactive tools.
  • V-BASE (I. M. Tomlinson, 2002; World Wide Web (www) mrc-cpe.cam.ac.uk/vbase).
  • V-BASE is a comprehensive directory of all human antibody germline variable region sequences compiled from more than one thousand published sequences. It includes a version of the alignment software DNAPLOT (developed by Hans-Helmar Althaus and Werner Müller) that allows the assignment of rearranged antibody V genes to their closest germline gene segments.
  • Antibodies Structure and Sequence (A. C. R. Martin, 2002; World Wide Web (www) bioinf.org.uk/abs). This page summarizes useful information on antibody structure and sequence. It provides a query interface to the Kabat antibody sequence data, general information on antibodies, crystal structures, and links to other antibody-related information. It also distributes an automated summary of all antibody structures deposited in the Protein Databank (PDB). Of particular interest is a thorough description and comparison of the various numbering schemes for antibody variable regions.
  • PDB Protein Databank
  • AAAAA A Ho's Amazing Atlas of Antibody Anatomy; A. Honegger, 2001; World Wide Web (www) unizh.ch/ ⁇ antibody.
  • This resource includes tools for structural analysis, modeling, and engineering. It adopts a unifying scheme for comprehensive structural alignment of antibody and T-cell-receptor sequences, and includes Excel macros for antibody analysis and graphical representation.
  • WAM Web Antibody Modeling; N. Whitelegg and A. R. Rees, 2001; World Wide Web (www) antibody.bath.ac.uk). Hosted by the Centre for Protein Analysis and Design at the University of Bath, United Kingdom. Based on the AbM package (formerly marketed by Oxford Molecular) to construct 3D models of antibody Fv sequences using a combination of established theoretical methods, this site also includes the latest antibody structural information.
  • the Antibody Resource Page (The Antibody Resource Page, 2000; World Wide Web (www) antibodyresource.com). This site describes itself as the “complete guide to antibody research and suppliers.” Links to amino acid sequencing tools, nucleotide antibody sequencing tools, and hybridoma/cell-culture databases are provided.
  • compositions and/or methods disclosed and claimed herein can be made and executed without undue experimentation in light of the present disclosure. While the compositions and methods of this invention have been described in terms of preferred embodiments, it will be apparent to those of skill in the art that variations may be applied to the compositions and/or methods and in the steps or in the sequence of steps of the method described herein without departing from the concept, spirit and scope of the invention. All such similar substitutes and modifications apparent to those skilled in the art are deemed to be within the spirit, scope and concept of the invention as defined by the appended claims.
  • a mouse model of the invention can be achieved by inserting ⁇ 960 kb of the human heavy chain locus containing all the V, D and J-regions upstream of the mouse constant region and 473 kb of the human kappa region upstream of the mouse constant region.
  • the human lambda region is inserted upstream of the mouse constant region. This insertion is achieved by gene targeting in ES cells using techniques well known in the art.
  • High fidelity insertion of intact V-D-J regions into each locus in their native (wild-type) configuration is suitably achieved by insertion of human bacterial artificial chromosomes (BACs) into the locus.
  • BACs human bacterial artificial chromosomes
  • the BACs are trimmed so that in the final locus no sequence is duplicated or lost compared to the original. Such trimming can be carried out by recombineering.
  • the relevant human BACs, suitably trimmed covering these loci are on average 90 kb in size.
  • the first BACs to be inserted in the IgH and IgK loci may contain the following V-regions. IgH: V6-1, VII-1-1, V1-2, VIII-2-1, V1-3, V4-4, V2-5 and IgK: V4-1, V5-2, V7-3, V2-4, V1-5, V1-6, V3-7, V1-8.
  • each locus is assessed after the first BAC insertion using chimaeric mice and also after each subsequent BAC addition. See below for detailed description of this performance test.
  • BACs retain their wild-type configuration when inserted into the ES cell genome.
  • high density genomic arrays were deployed to screen ES cells to identify those with intact BAC insertions (Barrett, M. T., Scheffer, A., Ben-Dor, A., Sampas, N., Lipson, D., Kincaid, R., Tsang, P., Curry, B., Baird, K., Meltzer, P. S., et al. (2004). Comparative genomic hybridization using oligonucleotide microarrays and total genomic DNA. Proceedings of the National Academy of Sciences of the United States of America 101, 17765-17770).
  • This screen also enables one to identify and select against ES clones in which the ES cell genome is compromised and thus not able to populate the germ line of chimeric animals.
  • Other suitable genomic tools to facilitate this assessment include sequencing and PCR verification.
  • the correct BAC structure is confirmed before moving to the next step.
  • the cell lines currently in use for the KOMP and EUCOMM global knockout projects have been modified twice prior to their use for this project and their germ line transmission rates are unchanged from the parental cells (these lines are publicly available, see World Wide Web (www) komp.org and World Wide Web (www) eucomm.org).
  • This cell line, called JM8 can generate 100% ES cell-derived mice under published culture conditions (Pettitt, S. J., Liang, Q., Rairdan, X. Y., Moran, J. L., Prosser, H. M., Beier, D.
  • the ability to generate chimeras with such high percentage of ES cell-derived tissue has other advantages.
  • HPRT-ve cell line such as AB2.1, as disclosed in Ram ⁇ rez-Solis R, Liu P and Bradley A, “Chromosome engineering in mice,” Nature, 1995; 378; 6558; 720-4.
  • RAG-1 complementation While many clones will generate 100% ES derived mice some will not. Thus, at every step mice are generated in a RAG-1-deficient background. This provides mice with 100% ES-derived B- and T-cells which can be used directly for immunization and antibody production. Cells having a RAG-2 deficient background, or a combined RAG-1/RAG-2 deficient background may be used, or equivalent mutations in which mice produce only ES cell-derived B cells and/or T cells.
  • the human-mouse IgH and IgK loci can be engineered in a cell line in which one allele of the IgH or IgK locus has already been inactivated.
  • the inactivation of the host Ig locus, such as the IgH or IgK locus can be carried out after insertion.
  • mice that have the RAG-1 gene mutated are immunodeficient as they have no mature B- or T-lymphocytes (U.S. Pat. No. 5,859,307). T- and B-lymphocytes only differentiate if proper V(D)J recombination occurs. Since RAG-1 is an enzyme that is crucial for this recombination, mice lacking RAG-1 are immunodeficient. If host embryos are genetically RAG-1 homozygous mutant, a chimera produced by injecting such an embryo will not be able to produce antibodies if the animal's lymphoid tissues are derived from the host embryo.
  • JM8 cells and AB2.1 cells generally contribute in excess of 80% of the somatic tissues of the chimeric animal and would therefore usually populate the lymphoid tissue.
  • JM8 cells have wild-type RAG-1 activity and therefore antibodies produced in the chimeric animal would be encoded by the engineered JM8 ES cell genome only. Therefore, the chimeric animal can be challenged with an antigen by immunization and subsequently produce antibodies to that antigen. This allows one skilled in the art to test the performance of the engineered human/mouse IgH and IgK loci as described in the present invention. See FIGS. 19 and 20 .
  • chimeric animal as described to determine the extent of antibody diversity (see e.g. Harlow, E. & Lane, D. 1998, 5 th edition, Antibodies: A Laboratory Manual , Cold Spring Harbor Lab. Press, Plainview, N.Y.).
  • the existence in the chimeric animal's serum of certain antibody epitopes could be ascertained by binding to specific anti-idiotype antiserum, for example, in an ELISA assay.
  • One skilled in the art could also sequence the genomes of B-cell clones derived from the chimeric animal and compare said sequence to wild-type sequence to ascertain the level of hypermutation, such hypermutation indicative of normal antibody maturation.
  • chimeric animal would also use said chimeric animal to examine antibody function wherein said antibodies are encoded from the engineered Ig loci (see e.g. Harlow, E. & Lane, D. 1998, 5 th edition, Antibodies: A Laboratory Manual, Cold Spring Harbor Lab. Press, Plainview, N.Y.).
  • antisera could be tested for binding an antigen, said antigen used to immunize the chimeric animal. Such a measurement could be made by an ELISA assay.
  • one skilled in the art could test for neutralization of the antigen by addition of the antisera collected from the appropriately immunized chimeric animal.
  • Recombineering for the production of vectors for use in homologous recombination in ES cells is disclosed in, for example, WO9929837 and WO0104288, and the techniques are well known in the art.
  • the recombineering of the human DNA takes place using BACs as a source of said human DNA.
  • Human BAC DNA will be isolated using QIAGEN®, BAC purification kit.
  • the backbone of each human BAC will be modified using recombineering to the exact same or similar configuration as the BAC already inserted into the mouse IgH region.
  • each human BAC will be trimmed using recombineering so that once the BACs are inserted, a seamless contiguous part of the human V(D)J genomic region will form at the mouse IgH or IgK locus.
  • BAC DNA transfection by electroporation and genotyping will be performed accordingly to standard protocols (Prosser, H. M., Rzadzinska, A. K., Steel, K. P., and Bradley, A. (2008). “Mosaic complementation demonstrates a regulatory role for myosin VIIa in actin dynamics of stereocilia.” Molecular and Cellular Biology 28, 1702-1712; Ramirez-Solis, R., Davis, A. C., and Bradley, A. (1993).
  • C57BL/6N-derived cell lines such as the JM8 male ES cells will follow standard techniques.
  • the JM8 ES cells have been shown to be competent in extensively contributing to somatic tissues and to the germline, and are being used for large mouse mutagenesis programs at the Sanger Institute such as EUCOMM and KOMP (Pettitt, S. J., Liang, Q., Rairdan, X. Y., Moran, J. L., Prosser, H. M., Beier, D. R., Lloyd, K. C., Bradley, A., and Skarnes, W. C. (2009). “Agouti C57BL/6N embryonic stem cells for mouse genetic resources.” Nature Methods).
  • JM8 ES cells (1.0 ⁇ 10 7 ) will be electroporated (500 ⁇ F, 230V; Bio-Rad®) with 10 ⁇ g I-SceI linearized human BAC DNA.
  • the transfectants will be selected with either Puromycin (3 ⁇ g/ml) or G418 (150 ⁇ g/ml). The selection will begin either 24 hours (with G418) or 48 hours (with Puromycin) post electroporation and proceed for 5 days.
  • 10 ⁇ g linearized human BAC DNA can yield up to 500 Puromycin or G418 resistant ES cell colonies. The antibiotic resistant ES cell colonies will be picked into 96-well cell culture plates for genotyping to identify the targeted clones.
  • BAC integrity will be examined by PCR-amplifying each known functional V gene in the BAC.
  • the first human BAC chosen for the IgH locus has 6 functional V genes.
  • at least 14 pairs of PCR primers will be designed and used to PCR-amplify genomic DNA from the targeted ES cells. The human wild-type size and sequence of these fragments will ensure that the inserted BAC has not been rearranged.
  • More detailed CGH will also confirm the integrity of the inserted BACs.
  • an oligo aCGH platform which is developed by Agilent Technologies, Inc. This platform not only enables one to study genome-wide DNA copy number variation at high resolution (Barrett, M. T., Scheffer, A., Ben-Dor, A., Sampas, N., Lipson, D., Kincaid, R., Tsang, P., Curry, B., Baird, K., Meltzer, P. S., et al. (2004).
  • the targeted ES cell genomic DNA and normal human individual genomic DNA will be labelled separately with dyes and hybridized to the array.
  • Log 2 ratio between ⁇ 0.29 and +0.29 for the signal from any oligo probe are regarded as no copy number change.
  • the log 2 ratio threshold for “Duplication” is usually >0.29999, and for deletion is ⁇ 0.29999.
  • the FRT-flanked BAC backbone will be excised by using Flp site-specific recombinase. If regular Flp-catalyzed FRT recombination is not high enough, one can use Flo, an improved version of Flpo recombinase which in certain tests is 3-4 times more efficient than the original Flp in ES cells.
  • ES cells will become sensitive to Puromycin (or G418) and resistant to FIAU (for loss of the TK cassette). The excision events will be further characterized by PCR amplification of the junction fragment using human genomic DNA primers.
  • Targeting of the genome of an ES cell to produce a transgenic mouse may be carried out using a protocol as explained by reference to the attached FIGS. 1-18 .
  • FIG. 1 illustrates three basic backbone vectors; an initiating cassette and 2 large insert vectors 1 and 2 respectively.
  • the initiating cassette comprises sequences homologous to the desired site of insertion into the mouse genome, those sites flanking a selectable marker and stuffer primer sequence for PCR based genotyping to confirm correct insertion of BACs.
  • the Stuffer-primer sequence provides the basis for genotyping each BAC addition step. This sequence is considered to provide a robust well validated sequence template for PCR primer and may be located at the ISceI site, ideally ⁇ 1 kb from the BAC insert.
  • the large insert vectors comprise human DNA on plasmids with selectable markers and a unique restriction site for linearisation of the plasmid to aid in homologous recombination into the genome of the ES cell.
  • FIG. 2 illustrates insertion of an initiating cassette into the mouse genome by Homologous recombination between the mouse J4 and C alpha exons.
  • Puromycin selection allows identification of ES cells with insertion of the cassette.
  • pu(Delta)tk is a bifunctional fusion protein between puromycin N-acetyltransferase (Puro) and a truncated version of herpes simplex virus type 1 thymidine kinase (DeltaTk).
  • Murine embryonic stem (ES) cells transfected with pu(Delta)tk become resistant to puromycin and sensitive to 1-( ⁇ 2-deoxy-2-fluoro-1-beta-D-arabino-furanosyl)-5-iodouracil (FIAU).
  • FIAU 1-( ⁇ 2-deoxy-2-fluoro-1-beta-D-arabino-furanosyl)-5-iodouracil
  • puDeltatk is readily transmitted through the male germ line.
  • pu(Delta)tk is a convenient positive/negative selectable marker that can be widely used in many ES cell applications.
  • FIG. 3 illustrates targeting of the large insert vector 1 to the mouse ES cell genome.
  • Linearisation of the vector is made at the same position as the stuffer primer sequence which allows for a gap repair genotyping strategy, well known in the art—see Zheng et al NAR 1999, Vol 27, 11, 2354-2360.
  • a gap repair genotyping strategy well known in the art—see Zheng et al NAR 1999, Vol 27, 11, 2354-2360.
  • Juxtaposition of appropriate PCR primer sequences allows colonies to be screened individually for a positive PCR fragment indicating proper insertion.
  • Positive selection using G418 allows for identification of mouse ES cells containing the neo selection marker.
  • PCR verification can be made of all critical V, D and J regions.
  • Array comparative genomic hybridization can be used to validate the BAC structure.
  • FIG. 4 illustrates the puro-delta-tk cassette and the BAC plasmid backbone is deleted using Flpe and select in FIAU. Since Flpe works inefficiently in mouse ES cells (5% deletion with transient Flpe expression), it is expected that in most cases, the recombination occurs between the two FRT sites flanking the BAC backbone. Flpo can also be tested to find out the recombination efficiency between two FRT sites that are 10 kb away.
  • FIG. 5 illustrates a second large insert vector is targeted into the ES cell chromosome.
  • the human BAC is targeted to the mouse IgH locus using the same initiation cassette insertion followed by Isce1 BAC linearization, BAC targeting to the initiation cassette and gap-repair genotyping strategy. Verification of the BAC insertion is carried out as before.
  • FIG. 6 illustrates the FRTY flanked BAC backbone of large insert vector 2 and the neo marker are deleted via Flpo. Note that this is not selectable, thus it will be necessary for clonal analysis at this point. This will enable confirmation of the juxtaposition of the human 2 insert with human 1 and other validation efforts.
  • FIG. 7 illustrates the next large insert vector targeted to the mouse IgH locus.
  • the pu-delta TK cassette is then removed, as for FIG. 4 .
  • the process can be repeated to incorporate other BACs.
  • FIG. 8 illustrates the final predicted ES cell construct.
  • FIGS. 9-18 provide a further level of detail of this process.
  • site specific recombination can also be employed.
  • Site-specific recombination has been widely used in the last 20-years for the integration of transgenes into defined chromosomal loci. SSR involves recombination between homologous DNA sequences.
  • the first generation of SSR-based chromosomal targeting involved recombination between (i) a single recombination target site (RT) such as loxP or FRT in a transfected plasmid with (ii) a chromosomal RT site provided by a previous integration.
  • RT recombination target site
  • RMCE recombinase-mediated cassette exchange
  • This approach has been successfully exploited in a variety of efficient chromosomal targeting, including integration of BAC inserts of greater than 50 kb (Wallace, H. A. C. et al. (2007). “Manipulating the mouse genome to engineering precise functional syntenic replacements with human sequence”. Cell 128: 197-209; Prosser, H. M. et al. (2008). “Mosaic complementation demonstrates a regulatory role for myosin VIIa in actin dynamics of Stereocilia”. Mol. Cell Biol. 28: 1702-12).
  • the largest insert size of a BAC is about 300-kb and therefore this places an upper limit on cassette size for RMCE.
  • SRMCE sequential RMCE
  • the method comprises the steps of
  • insertion of at least one DNA fragment uses site specific recombination.
  • the approach utilizes three heterospecific and incompatible loxP sites.
  • the method is comprised of the steps as follows, and illustrated in FIGS. 22-26 :
  • the endogenous VDJ or VJ sequences can be inactivated through an inversion via chromosomal engineering as follows (see FIGS. 27-29 ):
  • the methods of insertion of the invention suitably provide one or more of:
  • FIG. 30 Proof of concept of the approach is disclosed in FIG. 30 .
  • a landing pad as shown in FIG. 22 was inserted into the genome of a mouse by homologous recombination, followed by insertion of the R21 plasmid into that landing pad via cre-mediated site specific recombination.
  • the insertion event generated a number of general insertion events, 360 G418 resistant colonies, of which ⁇ 220 were inserted into the desired locus, as demonstrated by disruption of the HRPT minilocus.
  • the R21 vector mimics the 1 st BAC insertion vector at the 5′ and 3′ ends, including all selection elements and recombinase target sites. In place of BAC sequences, there is a small ‘stuffer’ sequence. This vector will both test all the principals designed in the invention and allow easy testing of the results in that PCR across the stuffer is feasible and therefore allows both ends of the insertion to be easily tested.
  • R21 was co-electroporated with a cre-expressing vector into the ES cells harbouring the landing pad in the IGH locus. Four sets of transformed cells were transfected in parallel and then placed under different selection regimes as indicated in FIG. 30 .
  • G418 selection resulted in the largest number of colonies due to there being no requirement for specific landing-pad integration. Any integration of R21 into the genome will provide neo expression leading to G418-resistance. Puro selection resulted in a similar colony number to Puro+6TG or G418+6TG, suggesting that the stringency of Puro selection is due to the Puro ⁇ TK lacking a promoter in the vector. Puro expression is only acquired when an integration occurs near a promoter element—in this design most likely specifically in the landing pad.
  • the next step in the invention is to ‘cure’ the 3′ end of the integrated BAC vector, leaving a seamless transition between the insertion and the flanking genome.
  • This curing was demonstrated by expanding an individual clone from above (R21 inserted into the landing pad) and expressing piggyBac recombinase in this clone via transfection of an expressing plasmid.
  • FIAU was used to select colonies in which the 3′ modification was excised—ie, through loss of the ‘PGK-puro ⁇ TK’ element between the piggyBac terminal repeats.
  • Fifty such clones resulted from a transfection of 10 6 cells; of these six were tested for the expected genomic structure.
  • Successful curing resulted in positive PCR between the primer set labelled “3” in FIG. 32 .
  • 4 had correct excisions 1 clone remained in the original configuration and 1 other had a deletion.
  • Example 3 demonstrated that the design of the claimed invention was capable of providing for the insertion of a test vector into the genome at a defined location, in this case the R21 vector into the mouse IGH locus.
  • the use of the appropriate selection media and the expression of cre-recombinase resulted in a genomic alteration with the predicted structure.
  • BAC insert comprised human sequences from the IGH locus and was approximately 166-kb.
  • This engineered BAC was electroporated along with a cre-expressing plasmid DNA into mouse ES cells harbouring the landing pad at the mouse IGH locus.
  • the transfected cell population was grown in puro-containing media to select for appropriate insertion events.
  • FIG. 33 The expected recombination event and resulting structure are depicted in FIG. 33 .
  • a stringent selection for correct clones was expected when the transfected population was selected in puro-containing media. This is because the puro-coding region requires a promoter element and this is preferentially supplied by the landing pad after recombination. Accordingly, the majority of the 7 isolated clones had inserted correctly into the genome at the landing pad as determined by the diagnostic PCR.
  • the primers for diagnosing a correct insertion are depicted in FIG. 33 .
  • BACs Bacterial artificial chromosomes
  • human Ig gene segments human V, D and/or J gene segments.
  • landing pads were used in a method to construct chimaeric Ig loci in mouse embryonic stem cells (ES cells), such that chimaeric IgH and IgK loci were provided in which human gene segments are functionally inserted upstream of endogenous constant regions.
  • RT-PCR was performed for the RNA samples of white blood cells from those mice with the primer pairs of human variable (V) region and mouse constant (C) region.
  • V human variable
  • C mouse constant
  • the sequences of oligos are shown as follows (Table 1). Each V oligo is paired with C oligo (HV with C ⁇ ; KV with C ⁇ ) for PCR reaction.
  • RT-PCR results showed most of the human IGH-VDJ or IGK-VJ gene segments appropriately rearrange and express in the chimaera mice.
  • those specific RT-PCR fragments were cloned into a common vector for sequencing.
  • FIG. 41 shows an analysis of kappa mRNA from mice B-cells bearing rearranged VJ, the VJ having been rearranged from human germline kappa V1-8 and J1, and demonstrates that both that productive VJ rearrangement and somatic hypermutation can be obtained, the latter as seen from the changes in antibodies encoded by mRNA with respect to the germline sequences.
  • the same is displayed for V1-6 and J1 in FIG. 42 .
  • the recombination eliminates stop codons that are encoded by the combination of (unmutated) human germline gene segments, thereby allowing for antibody-encoding mRNA sequences.
  • FIG. 43 demonstrates that inserted human kappa V1-5 J1 and V1-5 J4 can produce functional coding sequences in vivo and junctional diversity.
  • a 5′-RACE C ⁇ -specific library was generated from the splenic B lymphocytes of transgenic mice, denoted S1 mice. These mice comprise transgenic heavy chain loci, each locus containing the six most 3′ functional human V H gene segments (V H 2-5, 7-4-1, 4-4, 1-3, 1-2, 6-1), and all the human D and J H gene segments inserted into the endogenous heavy chain locus between endogenous IGHJ4 and E ⁇ (mouse chromosome 12: between coordinates 114666435 and 114666436).
  • the human DNA was obtained from a bacterial artificial chromosome (BAC) containing the sequence of human chromosome 14 from coordinate 106328951 to coordinate 106494908.
  • BAC bacterial artificial chromosome
  • transgenic antibody loci using sRMCE is given elsewhere herein and in WO2011004192 (which is incorporated herein by reference). 4 ⁇ 96-well plates of clones were randomly picked for sequencing to determine the usage of the gene segments. All detected immunoglobulin heavy chains were rearranged from mouse V H or human V H with human D-J H . No mouse D and J H segments were detected in rearranged products ( FIG. 44 ).
  • the ratio of mouse V H to human V H usage was around 3 to 1 ( FIG. 45 ).
  • the endogenous mouse V H -D-J H was inverted and moved to a distant region of the same chromosome.
  • the rearrangement of mouse V H s to human D-J H segments was totally blocked by effects of inversion and distance from the heavy chain locus.
  • the inversion strategy included three steps: (a) targeting of an inversion cassette, (b) inversion of endogenous VDJ and (c) excision of markers ( FIG. 46 ).
  • the inversion cassette consists of four components: a CAGGS promoter-driven puromycin-resistant-delta-thymidine kinase (puro ⁇ tk) gene, a 5′ HPRT gene segment under the PGK promoter control, a loxP site between them and inversely oriented to another loxP site already in the heavy chain locus, and two flanking piggyback LTRs (PB3′LTRs).
  • the inversion targeting cassette was inserted to a region that is 5′ and distant to the endogenous IGH locus at chromosome 12 as shown in FIG. 46 .
  • the targeted ES clones were identified and confirmed by PCR.
  • transient expression of cre from a transfected plasmid resulted in inversion of a section of chromosome 12 fragment including the endogenous V H -D-J H locus and intervening sequences through recombination of two inverted loxP sites, ie, those in the inversion cassette and the landing pad for the BAC insertion respectively.
  • the invertants were selected by HAT and confirmed by junction PCRs cross the two recombined loxP sites.
  • the inversion rearranged the relative orientation of the PB3′LTRs from the inversion cassette and PB5′LTR from the landing pad to generate two piggyBac transposon structures flanking the inverted region.
  • PBase piggyBac transposase
  • these two transposons were excised from the chromosome (and thus the mouse cell genome).
  • the cured ES clones were selected by 1-( ⁇ 2-deoxy-2-fluoro-1-b-D-arabinofuranosyl)-5-iodouracil (FIAU) and 6TG, and confirmed by junction PCRs cross the excised regions.
  • Tissue culture The procedures for ES cell culture, electroporation and drug selection have been described previously (Ramirez-Solis, R., A. C. Davis, and A. Bradley. 1993. Gene targeting in mouse embryonic stem cells. Methods Enzymol. 225:855-878).
  • S1 cell line (S1.11.1) was cultured in M15 medium (KnockoutTM DMEM supplemented with 15% fetal bovine serum, 2 mM glutamine, antibiotics, and 0.1 mM 2-mercaptoethonal).
  • Targeting construct R57 ( FIG. 47 ) was linearized outside the region of homology by NotI.
  • a total of 20 ⁇ g of the linearized construct was electroporated into S1 cell lines (AB2.1-derived) with a Bio-Rad® Gene PulserTM, and 107 cells were plated onto three 90-mm-diameter SNL76/7 feeder plates containing M15 medium.
  • M15 containing puromycin (3 ⁇ g of the active ingredient per ml) was added to each 90-mm-diameter plate, and the cells were maintained under selection for 9 days. 96 puromycin-resistant clones were then picked and expanded in 96-well plates. The targeting events were identified by long-range PCR.
  • Cre-loxP mediated inversion 12 positive clones were pooled together and cultured in a 6-well tissue culture plate with M15 medium. The cells were transfected with 10 ⁇ g of pCAGGS-Cre plasmid for the inversion of mouse endogenous locus and then plated onto three 90-mm-diameter SNL76/7 feeder plates containing M15 medium. At 24 h after electroporation, M15 containing 1XHAT (hypoxanthine-aminopterin-thymidine) was added to each 90-mm-diameter plate, and the cells were maintained under selection for 7 days and then treated with 1 ⁇ HT (hypoxanthine-thymidine) for 2 days. 48 HAT resistant colonies were picked and genotyped by PCR amplification of the junctions after Cre-loxP mediated inversion.
  • 1XHAT hypoxanthine-aminopterin-thymidine
  • HyPBase-mediated marker excision 12 positive clones were pooled together and cultured in E-well tissue culture plate using M15 medium. The cells were transfected with 5 ⁇ g of HyPBase plasmid to activate the PB transposon LTRs flanking two selection markers (Hprt-mini gene and PGK-puro ⁇ tk gene) and plated onto one 90-mm-diameter SNL76/7 feeder plates containing M15 medium.
  • a serial dilution of the cells was then plated onto three 90-mm-diameter SNL76/7 feeder plates containing M15 supplemented with 1-( ⁇ 2-deoxy-2-fluoro-1-b-D-arabinofuranosyl)-5-iodouracil (FIAU).
  • FIAU 1-( ⁇ 2-deoxy-2-fluoro-1-b-D-arabinofuranosyl)-5-iodouracil
  • Mouse chimaeras were generated by microinjection of ES cells into C57/BL6 blastocysts and transferred into pseudopregnant recipients. Male chimaeras were test-crossed with C57/BL6 mice. Agouti F1 offspring were genotyped by S1 3′ junction PCR. Test-cross positive heterozygotes were further intercrossed to generate homozygotes.
  • the synthesised first cDNA strand was purified using High Pure PCR Product Purification Kit (Roche). Poly(A) tail was added following the protocol supplied with the 5′/3′ RACE kit (2nd Generation, Roche). The 5′ end of the V H -D-J H rearranged transcript was amplified by nested PCR with forward primers Oligo dT, which is included in the kit, and nested C ⁇ -specific reverse primers E1555 (5′-CACCAGATTCTTATCAGAC-3′; SEQ ID No 26).
  • the 5′ RACE PCR product was checked on a 1% agarose gel and purified using QIAquick® Gel Extraction Kit (QIAGEN) as the protocol supplied with the kit, then cloned into pDrive vector using QIAGEN PCR Cloning Kit (QIAGEN) for sequencing analysis.
  • QIAGEN QIAquick® Gel Extraction Kit
  • the S1 inv1 mouse also shows a similar usage of both D and J H gene segments to human ( FIG. 49 ) (Link, J M et al. Mol. Immunol. 2005. 42, 943-955).
  • a mouse was produced that comprises a transgenic heavy chain locus that expresses heavy chains comprising human variable regions, but no mouse variable regions, and furthermore the human variable regions demonstrated a normal, human sequence distribution corresponding to human D and J usage observed in humans.
  • Insertion of human DNA from a 1 st human BAC (BAC comprising a the sequence of mouse Chromosome 14 from coordinate 106328951 to coordinate 106494908; containing six most 3′ functional V H gene segments (V H 2-5, 7-4-1, 4-4, 1-3, 1-2, 6-1), and all the human D and J H gene segments) into the heavy chain endogenous locus of a AB2.1 ES cell genome between endogenous IGHJ4 and E ⁇ (at mouse chromosome 12: between coordinates 114666435 and 114666436) effectively inactivates the use of endogenous D and J H gene segments for expressed rearranged immunoglobulin heavy chain ( FIG. 44 ).
  • the rearranged transcripts with mouse V H gene segments are reduced in the resulting S1 mouse.
  • the proportion of transcripts using mouse V H is around 75% of all observed sequences ( FIG. 45 ).
  • human DNA from a 2 nd human BAC (Chr14: 106494909-106601551) (BAC comprising a the sequence of mouse Chromosome 14 from coordinate 106494909 to coordinate 106601551; containing 5 more functional VH gene segments (V H 3-13, 3-11, 3-9, 1-8, 3-7)) was inserted into the landing pad left behind after curing following the 1 st BAC insertion (see, eg, FIG. 24 ).
  • the mouse V H usage is further significantly reduced following this insertion of the 2 nd BAC into the locus.
  • the proportion of transcripts using mouse VH was further reduced to 35% of all observed sequences ( FIG. 50 ).
  • the B cell arm of the immune system has evolved to produce high affinity, antigen-specific antibodies in response to antigenic challenge.
  • Antibodies are generated in B lymphocytes by a process of gene rearrangement in which variable (V), diversity (D; for the IGH locus) and joining (J) gene segments are recombined, transcribed and spliced to a C ⁇ (for IGH) or a C ⁇ or C ⁇ (for IGL) constant region gene segment to form an IgM antibody.
  • V variable
  • D diversity
  • J joining
  • IgM is either located on the cell surface or secreted.
  • the recombination process generates a primary antibody repertoire with sufficient germ line diversity to bind a wide range of antigens.
  • the immune system adopts a two-stage diversification process to increase diversity further.
  • B cells When challenged with antigens, B cells undergo selection and maturation by a process called somatic mutation.
  • B cells expressing antibodies which bind to antigen undergo multiple rounds of diversification, clonal expansion and antigen selection in the germinal centres (GCs) of the secondary lymphoid organs.
  • GCs germinal centres
  • the rearranged variable regions of the immunoglobulin genes acquire somatic hypermutation through nucleotide substitution, addition or deletion.
  • This stepwise process creates a secondary repertoire from the weak binders selected originally from the primary repertoire and combines rapid proliferation of antigen-reactive B cells with intense selection for quality of binding, eventually giving rise to high affinity antibodies with broad epitope coverage.
  • antibodies undergo class switching in which the C ⁇ constant region is replaced by C ⁇ , C ⁇ or C ⁇ to produce respectively IgG, A or E classes of antibody with different effector functions.
  • RT-PCR and sequence analysis Wild type or S1 chimera mice at 6-8 weeks of age were primed by intraperitoneal injection of 10 6 sheep RBCs suspended in phosphate buffer saline (PBS). The immunized mice were boosted twice with the same amount of sheep RBCs two and four weeks after priming. Four days after the last boost, peripheral blood cells were collected from the immunized mice. Total RNA was isolated from peripheral blood cells with TRIzol® reagent (InvitrogenTM) and treated with DNase I. Reverse transcription polymerase chain reaction (RT-PCR) was performed using SuperScript® III First-Strand Synthesis System (InvitrogenTM) following the protocol supplied by the manufacturer.
  • TRIzol® reagent InvitrogenTM
  • DNase I Reverse transcription polymerase chain reaction
  • the 1st strand cDNA was synthesized with the specific C ⁇ primers (C ⁇ 1, C ⁇ 2a, C ⁇ 2b), following by PCR with specific human V primers (VH1-2,3, VH4-4, VH6-1) and C ⁇ primers (Table 2). Following reaction, the RT-PCR product was checked on a 1% agarose gel and purified using QiAquick® Gel Extraction Kit (QIAGEN) as the protocol supplied with the kit, then cloned into pDrive vector using QIAGEN PCR Cloning Kit (QIAGEN) for sequencing analysis.
  • QIAGEN QiAquick® Gel Extraction Kit
  • ELP1352_C ⁇ 1 5′-AGAGCGGCCGCTGGGCAACGTTGCAGGTGACGGTC-3′ SEQ ID No 27
  • ELP1353_C ⁇ 2b 5′-AGAGCGGCCGCTTTGTCCACCGTGGTGCTGCTGG-3′ SEQ ID No 28
  • ELP1354_C ⁇ 2a 5′-AGAGCGGCCGCACATTGCAGGTGATGGACTGGC-3′ SEQ ID No 29
  • ELP1356_VH4-4 5′-AGGACGCGTGAAACACCTGTGGTTCTTCCTCCTGC-3′ SEQ ID No 30 ELP1357_VH1-2,3 5′-AGGACGCGTCACCATGGACTGGACCTGGAGGAT-3′ SEQ ID No 31
  • ELP1358_VH6-1 5′-AGGACGCGTATGTCTGTCCTTCCTCATCTTCC-3′ SEQ ID No 32
  • the rearranged transcripts were detected using RT-PCR with human VH-specific and mouse C ⁇ -specific primers for amplification from peripheral blood cells of immunized transgenic mice ( FIG. 51 ). Further sequence analysis of these amplified fragments demonstrated hypermutation happened within the human variable regions of these IG ⁇ chains ( FIG. 52 ). These results indicate that loci of the invention comprising insertion of human IGH BAC containing V H , D and J H gene segments into the locus between endogenous IGHJ4 and E ⁇ regions has normal class switching and hypermutation functionality (IgM to IgG) following antigen challenge.
  • IgM to IgG normal class switching and hypermutation functionality
  • T1 cells transitional 1 (T1) and transitional 2 (T2) B cells.
  • T2 cells transitional 2 B cells.
  • T1 cells give rise to T2 cells
  • T2 cells can further differentiate into mature (M) B cells.
  • immature B cells 3-4 days old
  • mature B cells are long-lived (15-20 weeks old) and are ready to respond to antigens (Pillai S et al; Immunol. Reviews. 2004. 197: 206-218).
  • the component of mature B cell population is directly linked to the efficiency of humoral immune response.
  • the T1, T2 and M cell populations can be categorized by their cell surface IgM and IgD levels.
  • a normal phenotype of splenic B cell compartment is required to mount a robust immune response.
  • Flow cytometric analysis of mature B lymphocytes To obtain a single cell suspension from spleen, the spleens of mice listed below were gently passaged through a 30 ⁇ m cell strainer. Single cells were resuspended in PBS supplemented with 3% heat inactivated foetal calf serum (FCS; Gibco®). The following antibodies were used for staining:
  • mice Four different genotypes of mice were generated:—
  • mice Spleens from these na ⁇ ve mice were collected and analysed for their B cell compartments. The number and percentages of T1, T2 and M cells among those mice are similar ( FIG. 53 ), indicating that genetic manipulation of endogenous IG loci in transgenic mice according to the invention do not compromise their B cell development. These data help to establish that animals according to the invention provide a robust platform for antibody discovery.
  • mice (H1) carrying all human JH, all human DH and human Vh2-5 under control of a rat switch region or mice (S1) carrying all human JH, all human DH and human Vh2-5, Vh7-41, Vh-4-4, Vh1-3, Vh1-2 and Vh6-1 under control of a mouse switch region were immunised with 100 ⁇ g Cholera Toxin B subunit (CTB; Sigma-Aldrich® C9903) emulsified in Complete Freund's Adjuvant CFA; Sigma-Aldrich® F 5881).
  • CTB Cholera Toxin B subunit
  • At least three animals were injected sc or ip and then boosted with 25 ⁇ g antigen in Incomplete Freund's Adjuvant (IFA; Sigma-Aldrich® F 5506) at (i) 14 days and 21 days or (ii) 28 days after priming. Blood was taken before priming at day “ ⁇ 1” (pre-bleeds) and on the day the spleens were taken (usually 4 d after last boost). Serum was analysed by ELISA using an antigen independent assessment of Ig isotypes. This assay detects total serum antibodies of all species.
  • IFA Incomplete Freund's Adjuvant
  • mice IgG1, IgG2a, IgG2b and IgM were used ((Anti-mouse IgG1 HRP AbD Serotec STAR132P, Anti-mouse IgG2a HRP AbD Serotec STAR133P, Anti-mouse IgG2b HRP AbD Serotec STAR134P, Anti-mouse IgM HRP Abcam® ab97230) and concentrations were read off a standard curve produced for each isotype using polyclonal isotype controls (IgG1, Kappa murine myeloma Sigma-Aldrich® M9269, IgG2a, Kappa murine myeloma Sigma-Aldrich® M9144, IgG2b, Kappa from murine myeloma Sigma-Aldrich® M8894, IgM, Kappa from murine myeloma Sigma-Aldrich® M3795).
  • mice showed an increase in overall IgG levels after immunisation over pre-bleeds.
  • mice (+/+) not carrying any human immunoglobulin genes were included and immunised, these mice showed comparable changes in total observed Ig levels ( FIG. 54 ).
  • Individual isotype levels were more variable between animals possibly showing various stages of class switching. IgM levels never exceeded 800 ⁇ g/ml whereas IgG levels reached more than 6 mg/ml in some animals. Non-immunised controls showed no such increases in switched isotype Ig levels.
  • mice comprising multiple human VDJ gene segments under the control of a rat S ⁇ rat or mouse switch are able to undergo productive recombination and class switching in response to antigen challenge and that the mice produce antibody levels that are broadly comparable to unmodified mice
  • the transgenic mice are able to produce antibodies of each of the IgG1, IgG2a, IgG2b and IgM isotypes after immunisation. Titers for CTB-specific Ig in pre-bleeds and terminal bleeds were determined and all immunised animals showed at CTB-specific titres of at least 1/100 000.
  • Transgenic mice carrying all human JH, all human DH and human Vh2-5 under control of a rat Sp switch region were immunised with 25 ⁇ g ovalbumin (OVA; Sigma-Aldrich® A7641) in Sigma-Aldrich® adjuvant (Sigma Adjuvant System® S6322) ip and then boosted with the same amount of OVA in adjuvant at day 14 and day 21.
  • Spleenocytes were taken 4 days later and fused using 1 ml polyethyleneglycol (PEG Average MW1450; Sigma-Aldrich® P7306) with a myeloma line.
  • Fused hybridoma cells were plated on 5 96-well plates and after selection with hypoxanthine-aminopterin-thymidine (HAT) wells tested for expression of OVA-specific antibodies by ELISA.
  • Clones positive by ELISA were re-tested by surface plasmon resonance (SPR) and binding kinetics determined using the ProteOnTM XPR36 (Bio-Rad®). Briefly, anti-mouse IgG (GE BiacoreTM BR-1008-38) was coupled to a GLM biosensor chip by primary amine coupling, this was used to capture the antibodies to be tested directly from tissue culture supernatants.
  • Ovalbumin was used as the analyte and passed over the captured antibody surface at 1024 nM, 256 nM, 64 nM, 16 nM, 4 nM with a 0 nM (i.e. buffer alone) used to double reference the binding data.
  • Regeneration of the anti-mouse IgG capture surface was by 10 mM glycine pH1.7, this removed the captured antibody and allowed the surface to be used for another interaction.
  • the binding data was fitted to 1:1 model inherent to the ProteOnTM XPR36 analysis software.
  • the run was carried out 1 ⁇ HBS-EP (10 mM Hepes, 150 mM NaCl, 3 mM EDTA, 0.05% polysorbate, pH7.6 (Teknova H8022)) used as running buffer and carried out at 25° C.
  • heavy chain V-regions were recovered by RT-PCR (Access RT-PCR System, A1250, Promega) using forward primers specific for Ig signal sequences (Wardemann et al Science 301, 1374 (2003)) and the following reverse primers for the constant regions of mouse IgG (Table 3):
  • RT-PCR products were either directly sequenced using the same primer pairs or cloned in to TA plasmids (TOPO® TA Cloning® Kit for Sequencing, K4595-40, InvitrogenTM) and submitted for plasmid sequencing.
  • Results show that CDRH3 sequences had variable CDRs except for two identical clones (16C9 and 20B5) that also had near identical KD kinetic values.
  • the determined equilibrium binding constant KD ranged from 0.38 nM to 40.60 nM, as determined by SPR at 25° C.
  • mice comprising multiple human VDJ gene segments under the control of a rat C ⁇ switch are able to undergo productive recombination and produce high affinity antigen-specific antibodies whose CDR3 regions have sequences encoded by human gene segments (human JH was separately identified by V-Quest, IMGT).
  • mice carrying all human JH, all human DH and human Vh2-5, Vh7-41, Vh-4-4, Vh1-3, Vh1-2 and Vh6-1 under control of a mouse Sp switch region were immunised and fused as described in Example 11.
  • Fused hybridoma cells were plated on 5 96-well plates and after selection with hypoxanthine-aminopterin-thymidine (HAT) or G418 (Gibco® Cat No 10131-027, Lot 503317) and wells tested for expression of CTB-specific antibodies by ELISA. Clones positive by ELISA were re-tested by surface plasmon resonance SPR and binding kinetics determined using the ProteOn XPR36TM (Bio-Rad®).
  • anti-mouse IgG (GE BiacoreTM BR-1008-38) was coupled to a GLM biosensor chip by primary amine coupling, this was used to capture the antibodies to be tested directly from tissue culture supernatants.
  • Cholera toxin B was used as analyte and passed over the captured antibody surface at 256 nM, 64 nM, 16 nM, 4 nM and 1 nM, with a 0 nM (i.e. buffer alone) used to double reference the binding data.
  • Regeneration of the anti-mouse IgG capture surface was by 10 mM glycine pH1.7, this removed the captured antibody and allowed the surface to be used for another interaction.
  • the binding data was fitted to 1:1 model inherent to the ProteOn XPR36TM analysis software.
  • the run was carried out 1 ⁇ HBS-EP (10 mM Hepes, 150 mM NaCl, 3 mM EDTA, 0.05% polysorbate, pH7.6 (Teknova H8022)) used as running buffer and carried out at 37° C.

Landscapes

  • Life Sciences & Earth Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Immunology (AREA)
  • Environmental Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • Zoology (AREA)
  • Veterinary Medicine (AREA)
  • Biotechnology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Animal Behavior & Ethology (AREA)
  • Animal Husbandry (AREA)
  • Biodiversity & Conservation Biology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biomedical Technology (AREA)
  • Wood Science & Technology (AREA)
  • General Engineering & Computer Science (AREA)
  • Microbiology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • Physics & Mathematics (AREA)
  • Plant Pathology (AREA)
  • Epidemiology (AREA)
  • Mycology (AREA)
  • Cell Biology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
US13/310,431 2009-07-08 2011-12-02 Animal models and therapeutic molecules Abandoned US20120204278A1 (en)

Priority Applications (55)

Application Number Priority Date Filing Date Title
US13/310,431 US20120204278A1 (en) 2009-07-08 2011-12-02 Animal models and therapeutic molecules
US13/416,684 US9447177B2 (en) 2009-07-08 2012-03-09 Transgenic mouse homozygous for chimeric IgH locus
DK12795606.8T DK2649184T3 (en) 2011-12-02 2012-11-30 USE OF FERTILIZED TRANSGENE ANIMALS FOR THE MANUFACTURE OF ANTIBODIES CARRYING HUMAN VARIABLE REGIONS
EP15188522.5A EP2989894B9 (fr) 2011-12-02 2012-11-30 Utilisation des souris et des rats transgéniques fertiles pour la production d'anticorps portant des régions variables humaines
ES15188522T ES2816899T3 (es) 2011-12-02 2012-11-30 Uso de ratones o ratas transgénicos fértiles para la producción de anticuerpos que llevan regiones variables humanas
CA2857569A CA2857569A1 (fr) 2011-12-02 2012-11-30 Animaux transgeniques
EP12795606.8A EP2649184B1 (fr) 2011-12-02 2012-11-30 Utilisation des animaux transgéniques fertiles pour la production d'anticorps portant des régions variables humaines
EP12795841.1A EP2785845A2 (fr) 2011-12-02 2012-11-30 Commutation isotypique fonctionnelle de chaînes d'anticorps chimères et animaux chimères exprimant différents isotypes igh
PCT/GB2012/052960 WO2013061098A2 (fr) 2011-12-02 2012-11-30 Commutation isotypique fonctionnelle de chaînes d'anticorps chimères et animaux chimères exprimant différents isotypes igh
CN201280059193.7A CN104160031B (zh) 2011-12-02 2012-11-30 用于生产具有人可变区的抗体的可育的转基因动物
JP2014543973A JP2015502149A (ja) 2011-12-02 2012-11-30 ヒト可変領域を保有している抗体を産生するために有用な繁殖可能遺伝子導入動物
EP23201805.1A EP4282879A3 (fr) 2011-12-02 2012-11-30 Utilisation d'animaux transgéniques fertiles pour produire des régions variables humaines d'anticorps
EP17196214.5A EP3298889A1 (fr) 2011-12-02 2012-11-30 Utilisation d'animaux transgéniques fertiles pour produire des régions variables humaines d'anticorps
DE202012013369.1U DE202012013369U1 (de) 2011-12-02 2012-11-30 Fertile transgene Tiere, brauchbar zum Herstellen von Antikörpern, die humane variable Regionen tragen
PCT/GB2012/052956 WO2013079953A1 (fr) 2011-12-02 2012-11-30 Animaux transgéniques fertiles utiles dans la production d'anticorps portant des régions variables humaines
AU2012343587A AU2012343587B2 (en) 2011-12-02 2012-11-30 Fertile transgenic animals useful for producing antibodies bearing human variable regions
BR112014013121A BR112014013121A2 (pt) 2011-12-02 2012-11-30 animais transgênicos férteis úteis para produção de antibióticos portando regiões variáveis humanas
US13/740,727 US9505827B2 (en) 2009-07-08 2013-01-14 Animal models and therapeutic molecules
US14/040,427 US20140201854A1 (en) 2009-07-08 2013-09-27 Animal models and therapeutic molecules
US14/040,405 US20140201856A1 (en) 2009-07-08 2013-09-27 Animal models and therapeutic molecules
US14/056,707 US20140150126A1 (en) 2009-07-08 2013-10-17 Animal models and therapeutic molecules
US14/056,434 US20140182003A1 (en) 2009-07-08 2013-10-17 Animal models and therapeutic molecules
US14/056,700 US11812731B2 (en) 2009-07-08 2013-10-17 Animal models and therapeutic molecules
HK13112514.7A HK1185100A1 (zh) 2011-12-02 2013-11-07 用於生產帶有人抗體可變區的可繁殖轉殖基因動物
US14/080,630 US10165763B2 (en) 2009-07-08 2013-11-14 Animal models and therapeutic molecules
US14/137,902 US9434782B2 (en) 2009-07-08 2013-12-20 Animal models and therapeutic molecules
US14/497,054 US20150113669A1 (en) 2009-07-08 2014-09-25 Animal Models and Therapeutic Molecules
US14/516,461 US10064398B2 (en) 2009-07-08 2014-10-16 Animal models and therapeutic molecules
US14/750,870 US20160044900A1 (en) 2009-07-08 2015-06-25 Animal models and therapeutic molecules
US14/818,162 US11564380B2 (en) 2009-07-08 2015-08-04 Animal models and therapeutic molecules
US14/935,010 US9504236B2 (en) 2009-07-08 2015-11-06 Animal models and therapeutic molecules
US15/016,211 US20160150768A1 (en) 2009-07-08 2016-02-04 Animal Models and Therapeutic Molecules
US15/214,963 US20160345551A1 (en) 2009-07-08 2016-07-20 Animal models and therapeutic molecules
US15/232,122 US11606941B2 (en) 2009-07-08 2016-08-09 Animal models and therapeutic molecules
US15/251,969 US20170051045A1 (en) 2009-07-08 2016-08-30 Animal models and therapeutic molecules
US15/360,502 US20170071174A1 (en) 2009-07-08 2016-11-23 Animal models and therapeutic molecules
US15/369,595 US20170081423A1 (en) 2009-07-08 2016-12-05 ANIMAL MODELS AND THERAPEUTIC MOLECULES - Track 1
US15/383,342 US20170099816A1 (en) 2009-07-08 2016-12-19 Animal models and therapeutic molecules
US15/383,188 US20170101482A1 (en) 2009-07-08 2016-12-19 Animal Models and Therapeutic Molecules
US15/383,353 US20170094956A1 (en) 2009-07-08 2016-12-19 Animal models and therapeutic molecules
US15/383,101 US20170099815A1 (en) 2009-07-08 2016-12-19 Animal Models and Therapeutic Molecules
US15/385,372 US20170105396A1 (en) 2009-07-08 2016-12-20 Animal Models and Therapeutic Molecules
US15/385,348 US20170099817A1 (en) 2009-07-08 2016-12-20 Animal Models and Therapeutic Molecules
JP2017235119A JP2018038428A (ja) 2011-12-02 2017-12-07 ヒト可変領域を保有している抗体を産生するために有用な繁殖可能遺伝子導入動物
US15/955,216 US20180295821A1 (en) 2011-12-02 2018-04-17 Transgenic Animals
US16/725,707 US20200205384A1 (en) 2011-12-02 2019-12-23 Transgenic Animals
US16/870,413 US20200375158A1 (en) 2009-07-08 2020-05-08 Animal Models and Therapeutic Molecules
US16/870,365 US20200352144A1 (en) 2009-07-08 2020-05-08 Animal Models and Therapeutic Molecules
JP2020092295A JP2020124228A (ja) 2011-12-02 2020-05-27 ヒト可変領域を保有している抗体を産生するために有用な繁殖可能遺伝子導入動物
US16/905,557 US20200352145A1 (en) 2009-07-08 2020-06-18 Animal Models and Therapeutic Molecules
US16/905,537 US20200337280A1 (en) 2009-07-08 2020-06-18 Animal Models and Therapeutic Molecules
JP2022128160A JP2022159413A (ja) 2011-12-02 2022-08-10 ヒト可変領域を保有している抗体を産生するために有用な繁殖可能遺伝子導入動物
US17/943,533 US20240057572A1 (en) 2009-07-08 2022-09-13 Animal Models and Therapeutic Molecules
US18/162,043 US20230225302A1 (en) 2009-07-08 2023-01-31 Animal Models and Therapeutic Molecules
US18/166,813 US20230263143A1 (en) 2009-07-08 2023-02-09 Animal Models and Therapeutic Molecules

Applications Claiming Priority (9)

Application Number Priority Date Filing Date Title
US22396009P 2009-07-08 2009-07-08
GBGB0911846.4A GB0911846D0 (en) 2009-07-08 2009-07-08 Animal models and therapeutic molecules
GB0911846.4 2009-07-08
GB0913102.0 2009-07-28
GB0913102A GB0913102D0 (en) 2009-07-28 2009-07-28 Animal models and therapeutic molesules
US35566610P 2010-06-17 2010-06-17
PCT/GB2010/051122 WO2011004192A1 (fr) 2009-07-08 2010-07-07 Modèles d'animaux et molécules thérapeutiques
PCT/GB2011/050019 WO2011158009A1 (fr) 2010-06-17 2011-01-07 Modèles animaux et molécules thérapeutiques
US13/310,431 US20120204278A1 (en) 2009-07-08 2011-12-02 Animal models and therapeutic molecules

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
PCT/GB2010/051122 Continuation-In-Part WO2011004192A1 (fr) 2009-07-08 2010-07-07 Modèles d'animaux et molécules thérapeutiques
PCT/GB2011/050019 Continuation-In-Part WO2011158009A1 (fr) 2009-07-08 2011-01-07 Modèles animaux et molécules thérapeutiques

Related Child Applications (21)

Application Number Title Priority Date Filing Date
US13/416,684 Division US9447177B2 (en) 2009-07-08 2012-03-09 Transgenic mouse homozygous for chimeric IgH locus
US13/433,084 Continuation-In-Part US9445581B2 (en) 2009-07-08 2012-03-28 Animal models and therapeutic molecules
PCT/GB2012/052956 Continuation WO2013079953A1 (fr) 2011-12-02 2012-11-30 Animaux transgéniques fertiles utiles dans la production d'anticorps portant des régions variables humaines
US13/740,727 Division US9505827B2 (en) 2009-07-08 2013-01-14 Animal models and therapeutic molecules
US14/040,405 Division US20140201856A1 (en) 2009-07-08 2013-09-27 Animal models and therapeutic molecules
US14/040,427 Division US20140201854A1 (en) 2009-07-08 2013-09-27 Animal models and therapeutic molecules
US14/056,707 Division US20140150126A1 (en) 2009-07-08 2013-10-17 Animal models and therapeutic molecules
US14/056,700 Division US11812731B2 (en) 2009-07-08 2013-10-17 Animal models and therapeutic molecules
US14/056,434 Division US20140182003A1 (en) 2009-07-08 2013-10-17 Animal models and therapeutic molecules
US14/497,054 Continuation US20150113669A1 (en) 2009-07-08 2014-09-25 Animal Models and Therapeutic Molecules
US14/818,162 Division US11564380B2 (en) 2009-07-08 2015-08-04 Animal models and therapeutic molecules
US15/016,211 Continuation US20160150768A1 (en) 2009-07-08 2016-02-04 Animal Models and Therapeutic Molecules
US15/214,963 Continuation US20160345551A1 (en) 2009-07-08 2016-07-20 Animal models and therapeutic molecules
US15/232,122 Continuation US11606941B2 (en) 2009-07-08 2016-08-09 Animal models and therapeutic molecules
US15/385,348 Continuation US20170099817A1 (en) 2009-07-08 2016-12-20 Animal Models and Therapeutic Molecules
US15/385,372 Continuation US20170105396A1 (en) 2009-07-08 2016-12-20 Animal Models and Therapeutic Molecules
US15/955,216 Continuation US20180295821A1 (en) 2011-12-02 2018-04-17 Transgenic Animals
US16/870,365 Continuation US20200352144A1 (en) 2009-07-08 2020-05-08 Animal Models and Therapeutic Molecules
US16/870,413 Continuation US20200375158A1 (en) 2009-07-08 2020-05-08 Animal Models and Therapeutic Molecules
US16/905,537 Continuation US20200337280A1 (en) 2009-07-08 2020-06-18 Animal Models and Therapeutic Molecules
US16/905,557 Continuation US20200352145A1 (en) 2009-07-08 2020-06-18 Animal Models and Therapeutic Molecules

Publications (1)

Publication Number Publication Date
US20120204278A1 true US20120204278A1 (en) 2012-08-09

Family

ID=43428834

Family Applications (33)

Application Number Title Priority Date Filing Date
US13/310,431 Abandoned US20120204278A1 (en) 2009-07-08 2011-12-02 Animal models and therapeutic molecules
US13/416,684 Active US9447177B2 (en) 2009-07-08 2012-03-09 Transgenic mouse homozygous for chimeric IgH locus
US13/740,727 Active 2032-07-15 US9505827B2 (en) 2009-07-08 2013-01-14 Animal models and therapeutic molecules
US14/040,427 Abandoned US20140201854A1 (en) 2009-07-08 2013-09-27 Animal models and therapeutic molecules
US14/040,405 Abandoned US20140201856A1 (en) 2009-07-08 2013-09-27 Animal models and therapeutic molecules
US14/056,700 Active US11812731B2 (en) 2009-07-08 2013-10-17 Animal models and therapeutic molecules
US14/056,707 Abandoned US20140150126A1 (en) 2009-07-08 2013-10-17 Animal models and therapeutic molecules
US14/056,434 Abandoned US20140182003A1 (en) 2009-07-08 2013-10-17 Animal models and therapeutic molecules
US14/080,630 Active US10165763B2 (en) 2009-07-08 2013-11-14 Animal models and therapeutic molecules
US14/137,902 Active US9434782B2 (en) 2009-07-08 2013-12-20 Animal models and therapeutic molecules
US14/497,054 Abandoned US20150113669A1 (en) 2009-07-08 2014-09-25 Animal Models and Therapeutic Molecules
US14/516,461 Active US10064398B2 (en) 2009-07-08 2014-10-16 Animal models and therapeutic molecules
US14/750,870 Abandoned US20160044900A1 (en) 2009-07-08 2015-06-25 Animal models and therapeutic molecules
US14/818,162 Active US11564380B2 (en) 2009-07-08 2015-08-04 Animal models and therapeutic molecules
US15/016,211 Abandoned US20160150768A1 (en) 2009-07-08 2016-02-04 Animal Models and Therapeutic Molecules
US15/214,963 Abandoned US20160345551A1 (en) 2009-07-08 2016-07-20 Animal models and therapeutic molecules
US15/232,122 Active US11606941B2 (en) 2009-07-08 2016-08-09 Animal models and therapeutic molecules
US15/251,969 Abandoned US20170051045A1 (en) 2009-07-08 2016-08-30 Animal models and therapeutic molecules
US15/360,502 Abandoned US20170071174A1 (en) 2009-07-08 2016-11-23 Animal models and therapeutic molecules
US15/383,101 Abandoned US20170099815A1 (en) 2009-07-08 2016-12-19 Animal Models and Therapeutic Molecules
US15/383,188 Abandoned US20170101482A1 (en) 2009-07-08 2016-12-19 Animal Models and Therapeutic Molecules
US15/383,353 Abandoned US20170094956A1 (en) 2009-07-08 2016-12-19 Animal models and therapeutic molecules
US15/383,342 Abandoned US20170099816A1 (en) 2009-07-08 2016-12-19 Animal models and therapeutic molecules
US15/385,348 Abandoned US20170099817A1 (en) 2009-07-08 2016-12-20 Animal Models and Therapeutic Molecules
US15/385,372 Abandoned US20170105396A1 (en) 2009-07-08 2016-12-20 Animal Models and Therapeutic Molecules
US16/870,413 Abandoned US20200375158A1 (en) 2009-07-08 2020-05-08 Animal Models and Therapeutic Molecules
US16/870,365 Pending US20200352144A1 (en) 2009-07-08 2020-05-08 Animal Models and Therapeutic Molecules
US16/905,557 Abandoned US20200352145A1 (en) 2009-07-08 2020-06-18 Animal Models and Therapeutic Molecules
US16/905,537 Pending US20200337280A1 (en) 2009-07-08 2020-06-18 Animal Models and Therapeutic Molecules
US17/943,533 Pending US20240057572A1 (en) 2009-07-08 2022-09-13 Animal Models and Therapeutic Molecules
US18/059,809 Pending US20230270088A1 (en) 2009-07-08 2022-11-29 Animal Models and Therapeutic Molecules
US18/162,043 Pending US20230225302A1 (en) 2009-07-08 2023-01-31 Animal Models and Therapeutic Molecules
US18/166,813 Pending US20230263143A1 (en) 2009-07-08 2023-02-09 Animal Models and Therapeutic Molecules

Family Applications After (32)

Application Number Title Priority Date Filing Date
US13/416,684 Active US9447177B2 (en) 2009-07-08 2012-03-09 Transgenic mouse homozygous for chimeric IgH locus
US13/740,727 Active 2032-07-15 US9505827B2 (en) 2009-07-08 2013-01-14 Animal models and therapeutic molecules
US14/040,427 Abandoned US20140201854A1 (en) 2009-07-08 2013-09-27 Animal models and therapeutic molecules
US14/040,405 Abandoned US20140201856A1 (en) 2009-07-08 2013-09-27 Animal models and therapeutic molecules
US14/056,700 Active US11812731B2 (en) 2009-07-08 2013-10-17 Animal models and therapeutic molecules
US14/056,707 Abandoned US20140150126A1 (en) 2009-07-08 2013-10-17 Animal models and therapeutic molecules
US14/056,434 Abandoned US20140182003A1 (en) 2009-07-08 2013-10-17 Animal models and therapeutic molecules
US14/080,630 Active US10165763B2 (en) 2009-07-08 2013-11-14 Animal models and therapeutic molecules
US14/137,902 Active US9434782B2 (en) 2009-07-08 2013-12-20 Animal models and therapeutic molecules
US14/497,054 Abandoned US20150113669A1 (en) 2009-07-08 2014-09-25 Animal Models and Therapeutic Molecules
US14/516,461 Active US10064398B2 (en) 2009-07-08 2014-10-16 Animal models and therapeutic molecules
US14/750,870 Abandoned US20160044900A1 (en) 2009-07-08 2015-06-25 Animal models and therapeutic molecules
US14/818,162 Active US11564380B2 (en) 2009-07-08 2015-08-04 Animal models and therapeutic molecules
US15/016,211 Abandoned US20160150768A1 (en) 2009-07-08 2016-02-04 Animal Models and Therapeutic Molecules
US15/214,963 Abandoned US20160345551A1 (en) 2009-07-08 2016-07-20 Animal models and therapeutic molecules
US15/232,122 Active US11606941B2 (en) 2009-07-08 2016-08-09 Animal models and therapeutic molecules
US15/251,969 Abandoned US20170051045A1 (en) 2009-07-08 2016-08-30 Animal models and therapeutic molecules
US15/360,502 Abandoned US20170071174A1 (en) 2009-07-08 2016-11-23 Animal models and therapeutic molecules
US15/383,101 Abandoned US20170099815A1 (en) 2009-07-08 2016-12-19 Animal Models and Therapeutic Molecules
US15/383,188 Abandoned US20170101482A1 (en) 2009-07-08 2016-12-19 Animal Models and Therapeutic Molecules
US15/383,353 Abandoned US20170094956A1 (en) 2009-07-08 2016-12-19 Animal models and therapeutic molecules
US15/383,342 Abandoned US20170099816A1 (en) 2009-07-08 2016-12-19 Animal models and therapeutic molecules
US15/385,348 Abandoned US20170099817A1 (en) 2009-07-08 2016-12-20 Animal Models and Therapeutic Molecules
US15/385,372 Abandoned US20170105396A1 (en) 2009-07-08 2016-12-20 Animal Models and Therapeutic Molecules
US16/870,413 Abandoned US20200375158A1 (en) 2009-07-08 2020-05-08 Animal Models and Therapeutic Molecules
US16/870,365 Pending US20200352144A1 (en) 2009-07-08 2020-05-08 Animal Models and Therapeutic Molecules
US16/905,557 Abandoned US20200352145A1 (en) 2009-07-08 2020-06-18 Animal Models and Therapeutic Molecules
US16/905,537 Pending US20200337280A1 (en) 2009-07-08 2020-06-18 Animal Models and Therapeutic Molecules
US17/943,533 Pending US20240057572A1 (en) 2009-07-08 2022-09-13 Animal Models and Therapeutic Molecules
US18/059,809 Pending US20230270088A1 (en) 2009-07-08 2022-11-29 Animal Models and Therapeutic Molecules
US18/162,043 Pending US20230225302A1 (en) 2009-07-08 2023-01-31 Animal Models and Therapeutic Molecules
US18/166,813 Pending US20230263143A1 (en) 2009-07-08 2023-02-09 Animal Models and Therapeutic Molecules

Country Status (23)

Country Link
US (33) US20120204278A1 (fr)
EP (19) EP2421357B1 (fr)
JP (7) JP5944312B2 (fr)
KR (1) KR101875233B1 (fr)
CN (2) CN105340834B (fr)
AU (4) AU2010269978B2 (fr)
BR (1) BR112012000536A2 (fr)
CA (1) CA2767436A1 (fr)
CY (1) CY1126083T1 (fr)
DK (12) DK2517557T4 (fr)
ES (8) ES2403087T3 (fr)
HK (5) HK1172207A1 (fr)
HR (2) HRP20130253T1 (fr)
HU (2) HUE055817T2 (fr)
LT (1) LT3241435T (fr)
NO (1) NO2792236T3 (fr)
NZ (1) NZ597481A (fr)
PL (4) PL2564695T3 (fr)
PT (4) PT2517556E (fr)
SG (2) SG2014010995A (fr)
SI (2) SI3241435T1 (fr)
SM (1) SMT201300046B (fr)
WO (1) WO2011004192A1 (fr)

Cited By (38)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20110195454A1 (en) * 2010-02-08 2011-08-11 Regeneron Pharmaceuticals, Inc. Common Light Chain Mouse
EP2421357A1 (fr) 2009-07-08 2012-02-29 Kymab Limited Modèles d'animaux et molécules thérapeutiques
US20120322108A1 (en) * 2011-02-25 2012-12-20 Regeneron Pharmaceuticals, Inc. Adam6 Mice
US20130160153A1 (en) * 2011-12-20 2013-06-20 Regeneron Pharmaceuticals, Inc. Humanized Light Chain Mice
US20130212719A1 (en) * 2012-02-01 2013-08-15 Regeneron Pharmaceuticals, Inc. Humanized Rodents that Express Heavy Chain Containing VL Domains
US20150033369A1 (en) * 2013-05-02 2015-01-29 Kymab Limited Animals, repertoires & methods
US9253965B2 (en) 2012-03-28 2016-02-09 Kymab Limited Animal models and therapeutic molecules
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
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
US9445581B2 (en) 2012-03-28 2016-09-20 Kymab Limited Animal models and therapeutic molecules
US9516868B2 (en) 2010-08-02 2016-12-13 Regeneron Pharmaceuticals, Inc. Mice that make VL binding proteins
US9648856B2 (en) 2012-03-16 2017-05-16 Regeneron Pharmaceuticals, Inc. Non-human animals expressing pH-sensitive immunoglobulin sequences
US9783618B2 (en) 2013-05-01 2017-10-10 Kymab Limited Manipulation of immunoglobulin gene diversity and multi-antibody therapeutics
US9783593B2 (en) 2013-05-02 2017-10-10 Kymab Limited Antibodies, variable domains and chains tailored for human use
US9788534B2 (en) 2013-03-18 2017-10-17 Kymab Limited Animal models and therapeutic molecules
US9796788B2 (en) 2010-02-08 2017-10-24 Regeneron Pharmaceuticals, Inc. Mice expressing a limited immunoglobulin light chain repertoire
US9932398B2 (en) 2011-10-17 2018-04-03 Regeneron Pharmaceuticals, Inc. Restricted immunoglobulin heavy chain mice
US9930871B2 (en) 2013-02-20 2018-04-03 Regeneron Pharmaceuticals, Inc. Non-human animals with modified immunoglobulin heavy chain sequences
EP3311661A1 (fr) 2011-09-19 2018-04-25 Kymab Limited Manipulation de la diversité de gènes d'immunoglobuline et multi-anticorps thérapeutiques
US9963716B2 (en) 2011-09-26 2018-05-08 Kymab Limited Chimaeric surrogate light chains (SLC) comprising human VpreB
US9969814B2 (en) 2010-02-08 2018-05-15 Regeneron Pharmaceuticals, Inc. Methods for making fully human bispecific antibodies using a common light chain
EP2877571B1 (fr) 2013-09-18 2018-05-30 Kymab Limited Procédés, cellules et organismes
CN108486125A (zh) * 2018-03-27 2018-09-04 重庆金迈博生物科技有限公司 一种核酸分子及其在制备人源单域抗体中的应用
WO2018189520A1 (fr) 2017-04-10 2018-10-18 Genome Research Limited Modèles animaux et molécules thérapeutiques
US10130081B2 (en) 2011-08-05 2018-11-20 Regeneron Pharmaceuticals, Inc. Humanized universal light chain mice
US10149462B2 (en) 2013-10-01 2018-12-11 Kymab Limited Animal models and therapeutic molecules
US10238093B2 (en) 2012-06-12 2019-03-26 Regeneron Pharmaceuticals, Inc. Humanized non-human animals with restricted immunoglobulin heavy chain loci
US10251377B2 (en) 2012-03-28 2019-04-09 Kymab Limited Transgenic non-human vertebrate for the expression of class-switched, fully human, antibodies
EP2931030B1 (fr) 2012-12-14 2020-04-29 Open Monoclonal Technology, Inc. Polynucléotides codant pour des anticorps de rongeur ayant des idiotypes humains, et animaux les comprenant
US10667501B2 (en) 2012-05-17 2020-06-02 Kymab Limited Transgenic non-human vertebrate for the in vivo production of dual specificity immunoglobulins or hypermutated heavy chain only immunoglobulins
US10711280B2 (en) 2013-12-11 2020-07-14 Regeneron Pharmaceuticals, Inc. Methods and compositions for the targeted modification of a mouse ES cell genome
US10787522B2 (en) 2014-03-21 2020-09-29 Regeneron Pharmaceuticals, Inc. VL antigen binding proteins exhibiting distinct binding characteristics
US10881085B2 (en) 2014-03-21 2021-01-05 Regeneron Pharmaceuticals, Inc. Non-human animals that make single domain binding proteins
US20210029978A1 (en) * 2016-11-04 2021-02-04 Regeneron Pharmaceuticals, Inc. Non-human animals having an engineered immunoglobulin lambda light chain locus
US10980221B2 (en) 2016-06-03 2021-04-20 Regeneron Pharmaceuticals, Inc. Non-human animals expressing exogenous terminal deoxynucleotidyltransferase
US11111314B2 (en) 2015-03-19 2021-09-07 Regeneron Pharmaceuticals, Inc. Non-human animals that select for light chain variable regions that bind antigen
US12016313B2 (en) 2017-01-19 2024-06-25 Omniab Operations, Inc. Human antibodies from transgenic rodents with multiple heavy chain immunoglobulin loci

Families Citing this family (84)

* 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
PL2346994T3 (pl) 2008-09-30 2022-04-19 Ablexis, Llc Myszy knock-in do wytwarzania chimerycznych przeciwciał
CA3006800C (fr) 2010-03-31 2022-10-04 Ablexis, Llc Genie genetique sur des animaux non humains pour la production d'anticorps chimeriques
EP2582230A1 (fr) * 2010-06-17 2013-04-24 Kymab Limited Modèles animaux et molécules thérapeutiques
DK2480676T3 (en) 2010-06-22 2016-06-20 Regeneron Pharma MICE EXPRESSING A HYDRIDE immunoglobulin light chain
US10662256B2 (en) 2010-07-26 2020-05-26 Trianni, Inc. Transgenic mammals and methods of use thereof
US10793829B2 (en) 2010-07-26 2020-10-06 Trianni, Inc. Transgenic mammals and methods of use thereof
JP6482757B2 (ja) 2010-07-26 2019-03-13 トリアンニ インコーポレイテッドTrianni,Inc. トランスジェニック動物および使用方法
HUE037095T2 (hu) 2011-03-09 2018-08-28 Cell Signaling Technology Inc Eljárások és reagensek monoklonális ellenanyagok elõállítására
CA2841376C (fr) 2011-07-05 2023-02-28 Duke University Immunogenes gp120 a extremite n-terminale deletee
EP3741862A1 (fr) 2011-09-19 2020-11-25 Kymab Limited Animaux, répertoires & procédés
GB2501753A (en) * 2012-05-04 2013-11-06 Kymab Ltd Human antibodies
GB2495083A (en) * 2011-09-26 2013-04-03 Kymab Ltd Human VpreB and chimaeric surrogate light chains in transgenic non-human vertebrates
US10092638B2 (en) 2011-10-03 2018-10-09 Duke University GP120 immunogens and methods inducing neutralizing antibodies to human immunodeficiency virus
SI3262932T1 (sl) * 2011-10-28 2019-09-30 Regeneron Pharmaceuticals, Inc. Genetsko modificirani poglavitni histokompatibilnostni kompleks miši
SG11201401030WA (en) * 2011-10-28 2014-04-28 Regeneron Pharma Genetically modified t cell receptor mice
PT3272214T (pt) 2011-10-28 2020-03-04 Regeneron Pharma Ratinhos geneticamente modificados que expressam moléculas quiméricas de complexo principal de histocompatibilidade (mhc) ii
GB2496375A (en) 2011-10-28 2013-05-15 Kymab Ltd A non-human assay vertebrate comprising human antibody loci and human epitope knock-in, and uses thereof
US9591835B2 (en) 2011-10-28 2017-03-14 Regeneron Pharmaceuticals, Inc. Genetically modified major histocompatibility complex animals
US9043996B2 (en) 2011-10-28 2015-06-02 Regeneron Pharmaceuticals, Inc. Genetically modified major histocompatibility complex animals
GB201122047D0 (en) * 2011-12-21 2012-02-01 Kymab Ltd Transgenic animals
US20180295821A1 (en) * 2011-12-02 2018-10-18 Kymab Limited Transgenic Animals
SG11201405088QA (en) * 2012-03-06 2014-11-27 Regeneron Pharma Common light chain mouse
WO2013144567A1 (fr) 2012-03-28 2013-10-03 Kymab Limited Vertébré non-humain transgénique pour l'expression d'anticorps entièrement humains à commutation isotypique
KR102159163B1 (ko) 2012-08-03 2020-09-23 에스에이비, 엘엘씨 유전자삽입 동물에서 인간 항체의 제조를 위한 복합 염색체 조작
WO2014124156A1 (fr) 2013-02-06 2014-08-14 Regeneron Pharmaceuticals, Inc. Conception d'un immunogène sur la base d'une lignée de lymphocytes b faisant appel à des animaux humanisés
ES2736052T3 (es) 2013-02-20 2019-12-23 Regeneron Pharma Ratones que expresan co-receptores humanizados de células T
EP3699190B9 (fr) 2013-02-22 2023-10-04 Regeneron Pharmaceuticals, Inc. Cellules murines exprimant des complexes majeurs d'histocompatibilite humanisées
US20150342163A1 (en) 2013-02-22 2015-12-03 Regeneron Pharmaceuticals, Inc. Genetically modified major histocompatibility complex mice
GB201403775D0 (en) 2014-03-04 2014-04-16 Kymab Ltd Antibodies, uses & methods
MX2016016133A (es) * 2014-06-06 2017-08-04 Regeneron Pharma Métodos y composiciones para modificar un locus dirigido.
DK3169778T3 (da) * 2014-07-14 2023-12-18 Univ Washington State Nanos-knockout, der eliminerer kimbaneceller
WO2016044745A1 (fr) 2014-09-19 2016-03-24 Regeneron Pharmaceuticals, Inc. Récepteurs d'antigènes chimériques
GB201418713D0 (en) 2014-10-21 2014-12-03 Kymab Ltd Bindings Proteins
EP3222718A4 (fr) * 2014-11-20 2018-05-30 Kyoto University Procédé pour le knock-in d'adn dans une région cible d'un génome mammifère et cellule
PT3265123T (pt) 2015-03-03 2023-02-01 Kymab Ltd Anticorpos, usos e métodos
MX2017012829A (es) 2015-04-06 2018-02-23 Regeneron Pharma Respuestas inmunitarias mediadas por células t humanizadas en animales no humanos.
EP3384030A4 (fr) 2015-12-03 2019-07-03 Trianni, Inc. Diversité améliorée d'immunoglobulines
US11053288B2 (en) 2016-02-04 2021-07-06 Trianni, Inc. Enhanced production of immunoglobulins
WO2017156349A1 (fr) 2016-03-10 2017-09-14 Cold Genesys, Inc. Méthodes de traitement de tumeurs solides ou lymphatiques par polythérapie
US11066464B2 (en) 2016-03-21 2021-07-20 Kymab Limited Anti-malarial antibodies that bind circumsporozoite protein
WO2017163049A1 (fr) 2016-03-21 2017-09-28 Kymab Limited Anticorps anti-paludéens qui se lient à la protéine circumsporozoïte
GB2550114A (en) 2016-05-03 2017-11-15 Kymab Ltd Methods, regimens, combinations & antagonists
KR102531889B1 (ko) 2016-06-20 2023-05-17 키맵 리미티드 항-pd-l1 및 il-2 사이토카인
US9567399B1 (en) 2016-06-20 2017-02-14 Kymab Limited Antibodies and immunocytokines
WO2018029474A2 (fr) 2016-08-09 2018-02-15 Kymab Limited Anticorps anti-icos
KR20230044038A (ko) 2016-08-09 2023-03-31 키맵 리미티드 항-icos 항체
WO2018065552A1 (fr) 2016-10-06 2018-04-12 Innate Pharma Anticorps anti-cd39
EP3534947A1 (fr) 2016-11-03 2019-09-11 Kymab Limited Anticorps, combinaisons comprenant des anticorps, biomarqueurs, utilisations et procédés
EP3558369A4 (fr) * 2016-12-21 2020-09-09 Cephalon, Inc. Anticorps se liant spécifiquement à l'il-15 humaine et leurs utilisations
US11680101B2 (en) 2017-01-27 2023-06-20 Kymab Limited Anti-OPG antibodies
US10457717B2 (en) 2017-02-17 2019-10-29 Denali Therapeutics Inc. Engineered polypeptides
WO2018152285A1 (fr) 2017-02-17 2018-08-23 Denali Therapeutics Inc. Modèles transgéniques de récepteur de transferrine
US10143187B2 (en) 2017-02-17 2018-12-04 Denali Therapeutics Inc. Transferrin receptor transgenic models
IL268660B2 (en) 2017-02-17 2024-03-01 Denali Therapeutics Inc Polypeptides that bind to a transgenic transferrin receptor, polynucleotides encoding them, methods for their preparation and use
AU2018251987A1 (en) 2017-04-14 2019-12-05 Cg Oncology, Inc. Methods of treating bladder cancer
GB201709808D0 (en) 2017-06-20 2017-08-02 Kymab Ltd Antibodies
GB201709970D0 (en) 2017-06-22 2017-08-09 Kymab Ltd Bispecific antigen-binding molecules
WO2019028190A1 (fr) * 2017-08-01 2019-02-07 City Of Hope Anticorps anti-il1rap
KR20200085781A (ko) * 2017-10-20 2020-07-15 프레드 헛친슨 켄서 리서치 센터 선택된 항체를 발현하도록 유전자 변형된 b 세포를 생산하기 위한 시스템 및 방법
AU2018381316A1 (en) 2017-12-05 2020-04-23 Regeneron Pharmaceuticals, Inc. Non-human animals having an engineered immunoglobulin lambda light chain and uses thereof
WO2019122882A1 (fr) 2017-12-19 2019-06-27 Kymab Limited Anticorps bispécifique pour icos et pd-l1
GB201721338D0 (en) 2017-12-19 2018-01-31 Kymab Ltd Anti-icos Antibodies
TWI842701B (zh) 2018-03-24 2024-05-21 美商再生元醫藥公司 用於產生抗肽-mhc複合物之治療性抗體的經基因修飾的非人類動物、其製法與用途
SG11202008620VA (en) 2018-03-26 2020-10-29 Regeneron Pharma Humanized rodents for testing therapeutic agents
CN108486126A (zh) * 2018-03-27 2018-09-04 重庆金迈博生物科技有限公司 一种核酸分子及其在人源化抗体中的应用
KR102444180B1 (ko) 2018-06-14 2022-09-16 리제너론 파마슈티칼스 인코포레이티드 면역글로불린 중쇄 암호화 서열에서 dh-dh 재배열 가능한 비인간 동물
GB2576914A (en) 2018-09-06 2020-03-11 Kymab Ltd Antigen-binding molecules comprising unpaired variable domains produced in mammals
GB201815629D0 (en) 2018-09-25 2018-11-07 Kymab Ltd Antagonists
GB201820687D0 (en) 2018-12-19 2019-01-30 Kymab Ltd Antagonists
CA3125380A1 (fr) 2019-02-18 2020-08-27 Biocytogen Pharmaceuticals (Beijing) Co., Ltd. Animaux non humains genetiquement modifies ayant un locus d'immunoglobuline humanisee
AU2020272939A1 (en) 2019-04-09 2021-11-11 Abcuro, Inc. Killer cell lectin-like receptor subfamily G member 1 (KLRG1) depleting antibodies
MX2022006495A (es) 2019-11-29 2022-09-07 Kymab Ltd Tratamiento para la sobrecarga fisiologica de hierro.
CA3163549A1 (fr) 2019-12-02 2021-06-10 Regeneron Pharmaceuticals, Inc. Constructions proteiques de peptide-cmh ii et leurs utilisations
KR20230018439A (ko) 2020-06-02 2023-02-07 바이오사이토젠 파마슈티컬스 (베이징) 컴퍼니 리미티드 공동 경쇄 면역글로불린 좌위를 갖는 유전자 변형 비인간 동물
AU2020454777A1 (en) * 2020-06-25 2023-02-02 Humab Co., Ltd. Heterozygous transgenic animal
KR20230066386A (ko) 2020-09-11 2023-05-15 리제너론 파마슈티칼스 인코포레이티드 항원 특이적 항체의 확인 및 제조
KR20230084157A (ko) 2020-10-08 2023-06-12 주식회사 휴맵 인간화 면역글로불린 유전자좌를 포함하는 게놈을 가지는 형질전환 비인간-동물 제조방법
WO2022132943A1 (fr) 2020-12-16 2022-06-23 Regeneron Pharmaceuticals, Inc. Souris exprimant des récepteurs humanisés fc alpha
US20220195014A1 (en) 2020-12-23 2022-06-23 Regeneron Pharmaceuticals, Inc. Nucleic acids encoding anchor modified antibodies and uses thereof
AU2022320627A1 (en) 2021-07-26 2024-02-08 Abcuro, Inc. Killer cell lectin-like receptor subfamily g member 1 (klrg1) depleting antibodies
WO2023097236A1 (fr) * 2021-11-24 2023-06-01 The Rockefeller University Compositions et procédés pour générer des souris knock-in d'immunoglobuline
GB202216503D0 (en) 2022-11-05 2022-12-21 Quadrucept Bio Ltd Non-human vertebrates & cells
US20240294651A1 (en) 2023-01-30 2024-09-05 Kymab Limited Antibodies

Citations (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US114985A (en) * 1871-05-16 Improvement in apparatus for cooling and preserving milk and other liquids
US5807715A (en) * 1984-08-27 1998-09-15 The Board Of Trustees Of The Leland Stanford Junior University Methods and transformed mammalian lymphocyte cells for producing functional antigen-binding protein including chimeric immunoglobulin
US20060015957A1 (en) * 1991-08-28 2006-01-19 Genpharm International, Inc. Transgenic non-human animals for producing chimeric antibodies
WO2007117410A2 (fr) * 2006-03-31 2007-10-18 Medarex, Inc. Animaux transgéniques exprimant des anticorps chimériques destinés à être utilisés pour la préparation d'anticorps humains
US20080076379A1 (en) * 2006-09-01 2008-03-27 Wanda Ying Li Outdoor umbrella with audio system
WO2008076379A2 (fr) * 2006-12-14 2008-06-26 Regeneron Pharmaceuticals, Inc. Anticorps humains au ligand delta-4 humain
US7932431B2 (en) * 2003-02-26 2011-04-26 Crescendo Biologics Limited Genetically modified non-human mammals and cells
US20110236378A1 (en) * 2008-09-30 2011-09-29 Ablexis, Llc Non-human mammals for the production of chimeric antibodies
US8771988B2 (en) * 2007-10-12 2014-07-08 Hoffmann-La Roche Inc. Protein expression from multiple nucleic acids
US9783618B2 (en) * 2013-05-01 2017-10-10 Kymab Limited Manipulation of immunoglobulin gene diversity and multi-antibody therapeutics

Family Cites Families (182)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5169939A (en) 1985-05-21 1992-12-08 Massachusetts Institute Of Technology & Pres. & Fellows Of Harvard College Chimeric antibodies
US4720449A (en) 1985-06-03 1988-01-19 Polaroid Corporation Thermal imaging method
GB8823869D0 (en) 1988-10-12 1988-11-16 Medical Res Council Production of antibodies
WO1991000906A1 (fr) 1989-07-12 1991-01-24 Genetics Institute, Inc. Animaux chimeriques et transgeniques pouvant produire des anticorps humains
US6673986B1 (en) 1990-01-12 2004-01-06 Abgenix, Inc. Generation of xenogeneic antibodies
US6657103B1 (en) 1990-01-12 2003-12-02 Abgenix, Inc. Human antibodies derived from immunized xenomice
US6713610B1 (en) 1990-01-12 2004-03-30 Raju Kucherlapati Human antibodies derived from immunized xenomice
DE69133566T2 (de) 1990-01-12 2007-12-06 Amgen Fremont Inc. Bildung von xenogenen Antikörpern
US7084260B1 (en) 1996-10-10 2006-08-01 Genpharm International, Inc. High affinity human antibodies and human antibodies against human antigens
US5545806A (en) 1990-08-29 1996-08-13 Genpharm International, Inc. Ransgenic non-human animals for producing heterologous antibodies
WO1994025585A1 (fr) 1993-04-26 1994-11-10 Genpharm International, Inc. Animaux transgeniques capables de produire des anticorps heterologues
US5633425A (en) 1990-08-29 1997-05-27 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
US6300129B1 (en) 1990-08-29 2001-10-09 Genpharm International Transgenic non-human animals for producing heterologous antibodies
US5770429A (en) 1990-08-29 1998-06-23 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
US6255458B1 (en) * 1990-08-29 2001-07-03 Genpharm International High affinity human antibodies and human antibodies against digoxin
EP0814159B1 (fr) 1990-08-29 2005-07-27 GenPharm International, Inc. Souris transgéniques capables de produire des anticorps hétérologues
AU2515992A (en) * 1991-08-20 1993-03-16 Genpharm International, Inc. Gene targeting in animal cells using isogenic dna constructs
CA2124967C (fr) 1991-12-17 2008-04-08 Nils Lonberg Animaux transgeniques non humains capables de produire des anticorps heterologues
US5859307A (en) 1992-02-04 1999-01-12 Massachusetts Institute Of Technology Mutant RAG-1 deficient animals having no mature B and T lymphocytes
EP0652950B1 (fr) 1992-07-24 2007-12-19 Amgen Fremont Inc. Production d'anticorps xenogeniques
DE4228162C1 (de) 1992-08-25 1994-01-13 Rajewsky Klaus Dr Verfahren zum Ersetzen homologer Genabschnitte aus Säugern in der Keimbahn von nicht-menschlichen Säugern
JP4242447B2 (ja) 1993-01-22 2009-03-25 イミュネックス・コーポレーション Cd40リガンド遺伝子の突然変異の検出および治療
DE4331162A1 (de) 1993-09-14 1995-03-16 Bayer Ag Verfahren zur Herstellung von Cyaninfarbstoffen
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
US6130364A (en) 1995-03-29 2000-10-10 Abgenix, Inc. Production of antibodies using Cre-mediated site-specific recombination
AU3507297A (en) 1996-06-26 1998-01-14 Baylor College Of Medicine Chromosomal rearrangement by insertion of two recombination substrates
EP0937140B1 (fr) 1996-06-27 2007-09-26 Vlaams Interuniversitair Instituut voor Biotechnologie vzw. Anticorps ayant une interaction spécifique avec le site actif ou la fissure d'une molécule cible
AU5702298A (en) 1996-12-03 1998-06-29 Abgenix, Inc. Transgenic mammals having human Ig loci including plural VH and VK regions nd antibodies produced therefrom
US6319906B1 (en) 1996-12-31 2001-11-20 Isis Pharmaceuticals Oligonucleotide compositions and methods for the modulation of the expression of B7 protein
EP0979281B1 (fr) 1997-05-02 2005-07-20 Genentech, Inc. Procede de preparation d'anticorps multispecifiques presentant des composants heteromultimeres
US20020088019A1 (en) 1997-09-02 2002-07-04 Oron Yacoby-Zeevi Methods of and pharmaceutical compositions for improving implantation of embryos
NZ504300A (en) 1997-11-18 2002-06-28 Pioneer Hi Bred Int A method for directional stable transformation of eukaryotic cells using non-identical recombination sites
US6509156B1 (en) 1997-12-05 2003-01-21 Europaisches Laboratorium Fur Molekularoiologie (Embl) DNA cloning method relying on the E. coli recE/recT recombination system
EP0939120A1 (fr) 1998-02-27 1999-09-01 Gesellschaft für biotechnologische Forschung mbH (GBF) Méthode pour l'échange répétifif, dépourvu de marqueurs, de cassettes d'expression d'ADN dans le génome de cellules ou de parties de cellules
NZ509241A (en) 1998-08-07 2003-08-29 Du Pont Pharm Co Succinoylamino lactams as inhibitors of alpha-beta protein production
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
AU5867100A (en) 1999-05-03 2000-12-12 Medarex, Inc. Human antibodies to staphylococcus aureus
US6833268B1 (en) 1999-06-10 2004-12-21 Abgenix, Inc. Transgenic animals for producing specific isotypes of human antibodies via non-cognate switch regions
US6355412B1 (en) 1999-07-09 2002-03-12 The European Molecular Biology Laboratory Methods and compositions for directed cloning and subcloning using homologous recombination
US7605238B2 (en) 1999-08-24 2009-10-20 Medarex, Inc. Human CTLA-4 antibodies and their uses
CA2307503A1 (fr) * 2000-05-02 2001-11-02 Carlos F. Barbas Iii Peptides utilises comme vaccin ou traitement contre l'infection par le vih
JP2004504055A (ja) * 2000-07-21 2004-02-12 アメリカ合衆国 真核生物ゲノムにおけるdnaの置換、転座およびスタッキングの方法
US6596541B2 (en) 2000-10-31 2003-07-22 Regeneron Pharmaceuticals, Inc. Methods of modifying eukaryotic cells
US6586251B2 (en) 2000-10-31 2003-07-01 Regeneron Pharmaceuticals, Inc. Methods of modifying eukaryotic cells
US7105348B2 (en) 2000-10-31 2006-09-12 Regeneron Pharmaceuticals, Inc. Methods of modifying eukaryotic cells
AU2002219841A1 (en) * 2000-11-16 2002-05-27 Cornell Research Foundation Inc. Vectors for conditional gene inactivation
EP1343880B1 (fr) 2000-11-17 2009-05-27 Kyowa Hakko Kirin Co., Ltd. Expression d'immunoglobulines (humaines) xenogenes dans des bovines transgeniques clones
JP3523245B1 (ja) 2000-11-30 2004-04-26 メダレックス,インコーポレーテッド ヒト抗体作製用トランスジェニック染色体導入齧歯動物
EP1409646B1 (fr) * 2000-12-19 2012-06-13 Altor BioScience Corporation Animaux transgeniques a systeme immunitaire humanise
PE20020801A1 (es) 2001-01-05 2002-09-06 Pfizer Anticuerpos contra el receptor del factor de crecimiento similar a insulina
AUPR446701A0 (en) * 2001-04-18 2001-05-17 Gene Stream Pty Ltd Transgenic mammals for pharmacological and toxicological studies
JP2003000243A (ja) 2001-06-25 2003-01-07 Takachika Azuma 変異体の製造法
FR2827302B1 (fr) * 2001-07-13 2003-10-10 Genoway Cellule et animal transgenique modelisant la presentation antigenique humaine et leurs utilisations
WO2003017336A2 (fr) 2001-08-13 2003-02-27 Amberwave Systems Corporation Condensateurs a tranchee pour memoire dynamique a acces aleatoire
US20060199204A1 (en) 2001-10-05 2006-09-07 U.S. Epa Genetic testing for male factor infertility
WO2003031656A1 (fr) 2001-10-05 2003-04-17 United States Environmental Protection Agency Evaluation genetique du facteur de sterilite masculine
CA2847885C (fr) 2001-11-30 2022-03-22 Amgen Fremont Inc. Animaux transgeniques porteurs de genes a chaine legere iglamba d'immunoglobuline humaine
US20050095712A1 (en) 2002-01-17 2005-05-05 Alberto Martin Mutations caused by activation-induced cytidine deaminase
US8877901B2 (en) 2002-12-13 2014-11-04 Immunomedics, Inc. Camptothecin-binding moiety conjugates
PT1523496E (pt) 2002-07-18 2011-09-29 Merus B V Produção de misturas de anticorpos de forma recombinante
AU2003278790A1 (en) 2002-09-09 2004-03-29 California Institute Of Technology Methods and compositions for the generation of humanized mice
CA2501981A1 (fr) * 2002-10-01 2004-04-15 Mitos Pharmaceuticals, Inc. Preparations de radioprotection a l'oxyde d'azote et leurs methodes d'utilisation
DE10251918A1 (de) * 2002-11-08 2004-05-19 Horn, Carsten, Dipl.-Biochem. Dr. Systeme zur Erzeugung stabiler genomischer Transgen-Insertionen
US7700356B2 (en) * 2002-11-08 2010-04-20 The United States Of America As Represented By The Secretary Of Agriculture System for gene targeting and producing stable genomic transgene insertions
AR042145A1 (es) 2002-11-27 2005-06-08 Dow Agrociences Llc Produccion de inmunoglobulinas en plantas con una fucocilacion reducida
CN1326878C (zh) * 2003-04-29 2007-07-18 中国抗体制药有限公司 抗人非何杰金淋巴瘤嵌合抗体及其衍生物与应用
US20100069614A1 (en) 2008-06-27 2010-03-18 Merus B.V. Antibody producing non-human mammals
WO2004106375A1 (fr) 2003-05-30 2004-12-09 Merus Biopharmaceuticals B.V. I.O. Banque de fab pour la preparation de fab anti-vegf et antirabique
US7205148B2 (en) 2003-06-11 2007-04-17 Regeneron Pharmaceuticals, Inc. Genome mutation by intron insertion into an embryonic stem cell genome
GB2403475B (en) * 2003-07-01 2008-02-06 Oxitec Ltd Stable integrands
WO2005007696A2 (fr) * 2003-07-15 2005-01-27 Therapeutic Human Polyclonals, Inc. Locus d'immunoglobuline humanises
US7663017B2 (en) * 2003-07-30 2010-02-16 Institut Pasteur Transgenic mice having a human major histocompatability complex (MHC) phenotype, experimental uses and applications
WO2005019463A1 (fr) 2003-08-11 2005-03-03 Therapeutic Human Polyclonals, Inc. Amelioration de la transgenese a l'aide de loci d'immunoglobuline humanises
US7604994B2 (en) 2003-09-03 2009-10-20 Morphotek, Inc. Genetically altered antibody-producing cell lines with improved antibody characteristics
US7205140B2 (en) 2003-10-20 2007-04-17 Campusgen Gmbh Nucleotide sequence for creatinine deiminase and method of use
PL1691837T3 (pl) 2003-12-10 2012-11-30 Squibb & Sons Llc IP-10 przeciwciała i ich zastosowanie
US7625549B2 (en) 2004-03-19 2009-12-01 Amgen Fremont Inc. Determining the risk of human anti-human antibodies in transgenic mice
CA2564989C (fr) 2004-03-19 2014-05-27 Amgen, Inc. Reduction du risque d'anticorps humains et anti-humains par la manipulation du gene v
EP1978032A3 (fr) 2004-07-22 2009-02-18 Erasmus University Medical Center Rotterdam Molécules de liaison
FR2875239B1 (fr) 2004-09-10 2007-07-20 Inst Necker Ass Loi De 1901 Procede pour l'acceleration des mutations somatiques et son application en proteomique
WO2006029459A1 (fr) 2004-09-13 2006-03-23 Evogenix, Inc Anticorps specifiques pour le carcinome hepatocellulaire et d'autres carcinomes et ses applications
WO2006044492A2 (fr) 2004-10-14 2006-04-27 Ingenious Targeting Laboratory, Inc. Methodes de generation de lignees cellulaires derivees d'embryons de rats et de modification genetique de genome de rats
PL1802193T3 (pl) 2004-10-19 2014-09-30 Regeneron Pharma Sposób wytwarzania myszy homozygotycznej pod względem modyfikacji genetycznej
JP2008517600A (ja) * 2004-10-22 2008-05-29 セラピューティック ヒューマン ポリクローナルズ, インコーポレイテッド ヒト以外のトランスジェニック動物における内因性免疫グロブリン発現の抑制
WO2006055704A2 (fr) 2004-11-17 2006-05-26 Curagen Corporation Anticorps diriges contre des proteines ten-m et utilisations associees
EP2284194A1 (fr) 2004-12-21 2011-02-16 AstraZeneca AB Anticorps dirigés contre l'Angiopoiétine 2 et leurs utilisations
WO2006117699A2 (fr) 2005-04-29 2006-11-09 Innate Pharma Animaux transgeniques et procedes de fabrication d'anticorps recombines
US20100154070A1 (en) * 2005-05-14 2010-06-17 Tian Xu PiggyBac as a Tool for Genetic Manipulation and Analysis in Vertebrates
EP1780272A1 (fr) 2005-10-27 2007-05-02 GSF-Forschungszentrum für Umwelt und Gesundheit GmbH Mèthode pour augmenter les hypermutations somatiques, la conversion gènique et le changement de classe par recombinaison
GB0601513D0 (en) 2006-01-25 2006-03-08 Univ Erasmus Medical Ct Binding molecules 3
KR101481843B1 (ko) 2006-01-25 2015-01-12 에라스무스 유니버시티 메디컬 센터 로테르담 형질전환 동물 내에서의 중쇄만의 항체의 생성
US7462759B2 (en) 2006-02-03 2008-12-09 Pioneer Hi-Bred International, Inc. Brittle stalk 2 gene family and related methods and uses
US7846724B2 (en) * 2006-04-11 2010-12-07 Hoffmann-La Roche Inc. Method for selecting CHO cell for production of glycosylated antibodies
MY159787A (en) 2006-06-02 2017-01-31 Regeneron Pharma High affinity antibodies to human il-6 receptor
AU2007345747A1 (en) * 2006-06-27 2008-08-07 Vaxdesign Corporation Models for vaccine assessment
EP1878342A1 (fr) 2006-07-13 2008-01-16 Institut Pasteur Souris immunodeficientes transgéniques pour la classe I et II de HLA, et leurs utilisations
US8084024B2 (en) 2006-08-22 2011-12-27 G2 Inflammation Pty Ltd Method for producing antibodies
MX2009003393A (es) 2006-10-02 2009-05-11 Regeneron Pharma Anticuerpos humanos de alta afinidad para el receptor de il-4 humano.
US7732195B2 (en) * 2006-11-01 2010-06-08 Facet Biotech Corporation Tethered vectors for cell surface immunoglobulin display
GB0700194D0 (en) 2007-01-05 2007-02-14 Univ Edinburgh Humanisation of animals
WO2008103474A1 (fr) 2007-02-20 2008-08-28 Anaptysbio, Inc. Procédés de production de pharmacothèques, et leurs utilisations
ES2456961T3 (es) 2007-02-21 2014-04-24 University Of Massachusetts Anticuerpos humanos contra el virus de la hepatitis C (VHC), usos de los mismos
EP2132312B1 (fr) 2007-03-27 2016-01-27 Sea Lane Biotechnologies,llc. Constructions et bibliothèques comprenant des séquences de chaînes légères de substitution d'anticorps
GB0706628D0 (en) 2007-04-04 2007-05-16 Univ Erasmus Germ-line manipulation 1
LT2602323T (lt) 2007-06-01 2018-04-10 Open Monoclonal Technology, Inc. Kompozicijos ir būdai endogeninių imunoglobulinų genams slopinti ir transgeniniams žmogaus idiotipo antikūnams gaminti
WO2009013620A2 (fr) 2007-06-11 2009-01-29 Erasmus University Medical Center Rotterdam Recombinaison homologue
EP2178916B1 (fr) 2007-07-31 2014-12-17 Regeneron Pharmaceuticals, Inc. Anticorps humains contre le cd20 humain et leur procede d'utilisation
US9693539B2 (en) 2007-08-10 2017-07-04 E. R. Squibb & Sons, L.L.C. HCO32 and HCO27 and related examples
MX2010001395A (es) 2007-08-10 2010-03-10 Regeneron Pharma Anticuerpos humanos de alta afinidad de factor de crecimiento de nervio humano.
EP2245155A4 (fr) 2007-12-10 2011-05-25 Aliva Biopharmaceuticals Inc Procédés pour le remplacement séquentiel d'une zone ciblée par recombinaison homologue
US8227577B2 (en) * 2007-12-21 2012-07-24 Hoffman-La Roche Inc. Bivalent, bispecific antibodies
BRPI0906711A2 (pt) 2008-01-25 2015-06-30 Cabot Corp Médoto para preparação de pigmento coloridos modificados
KR101409375B1 (ko) 2008-01-31 2014-06-18 삼성전자주식회사 반도체 메모리장치의 블록 디코딩 회로
WO2009118524A2 (fr) 2008-03-26 2009-10-01 Iti Scotland Limited Insertion efficace d’adn dans des cellules souches embryonnaires
US8012714B2 (en) 2008-04-14 2011-09-06 Innovative Targeting Solutions, Inc. Sequence diversity generation in immunoglobulins
PL2288623T3 (pl) * 2008-05-23 2014-01-31 Ablexis Llc Sposób wytwarzania przeciwciał z pojedynczą domeną VL w zwierzętach transgenicznych
KR20110029156A (ko) 2008-06-27 2011-03-22 메뤼스 베.페. 항체 생산 비-인간 포유동물
JO3672B1 (ar) 2008-12-15 2020-08-27 Regeneron Pharma أجسام مضادة بشرية عالية التفاعل الكيماوي بالنسبة لإنزيم سبتيليسين كنفرتيز بروبروتين / كيكسين نوع 9 (pcsk9).
US20110314563A1 (en) 2008-12-18 2011-12-22 Kingdon Craig R Antibody production
US9085795B2 (en) 2009-02-04 2015-07-21 Molecular Innovations, Inc. Methods for screening candidate agents for modulating prorenin and renin, assays for detecting prorenin and antibodies
US20110305692A1 (en) 2009-02-24 2011-12-15 Glaxo Group Limited Antigen-binding contructs
GB0905023D0 (en) 2009-03-24 2009-05-06 Univ Erasmus Medical Ct Binding molecules
CA2757178C (fr) 2009-04-03 2020-05-19 Medical Research Council Mutants de cytidine desaminase induite par activation (aid) et procedes d'utilisation
EP2446032B8 (fr) 2009-06-26 2017-03-15 i2 Pharmaceuticals, Inc. Expression de chaînes légères substituts
HUE055817T2 (hu) 2009-07-08 2021-12-28 Kymab Ltd Állatmodellek és terápiás molekulák
GB0913102D0 (en) 2009-07-28 2009-09-02 Genome Res Ltd Animal models and therapeutic molesules
US9445581B2 (en) 2012-03-28 2016-09-20 Kymab Limited Animal models and therapeutic molecules
GB0911846D0 (en) 2009-07-08 2009-08-19 Genome Res Ltd Animal models and therapeutic molecules
CA2768207C (fr) 2009-07-15 2019-12-03 Aimm Therapeutics B.V. Moyens et procedes de production d'anticorps a haute affinite
JO3182B1 (ar) 2009-07-29 2018-03-08 Regeneron Pharma مضادات حيوية بشرية عالية الالفة مع تولد الاوعية البشرية - 2
WO2011019844A1 (fr) 2009-08-13 2011-02-17 Crystal Bioscience Inc. Animal transgénique pour la production d'anticorps ayant des cdr minimaux
US20120251552A1 (en) 2009-11-05 2012-10-04 Anaptysbio, Inc. Methods of generating improved antigen-binding agents using chain shuffling and optionally somatic hypermutation
CN102741404B (zh) 2009-11-17 2017-03-08 Sab有限责任公司 人类人工染色体载体
WO2011071957A1 (fr) 2009-12-07 2011-06-16 Sea Lane Biotechnologies, Llc Conjugués comprenant un échafaudage de substituts d'anticorps présentant des propriétés pharmacocinétiques améliorées
CN102711449B (zh) 2009-12-10 2015-01-07 瑞泽恩制药公司 生产重链抗体的小鼠
US20120021409A1 (en) 2010-02-08 2012-01-26 Regeneron Pharmaceuticals, Inc. Common Light Chain Mouse
NZ719253A (en) 2010-02-08 2022-08-26 Regeneron Pharma Common light chain mouse
JP6208580B2 (ja) 2010-05-17 2017-10-04 サンガモ セラピューティクス, インコーポレイテッド 新規のdna結合タンパク質及びその使用
EP2582230A1 (fr) 2010-06-17 2013-04-24 Kymab Limited Modèles animaux et molécules thérapeutiques
DK2480676T3 (en) 2010-06-22 2016-06-20 Regeneron Pharma MICE EXPRESSING A HYDRIDE immunoglobulin light chain
EP3960865A1 (fr) 2010-08-02 2022-03-02 Regeneron Pharmaceuticals, Inc. Souris produisant des protéines de liaison comprenant des domaines vl
ES2537207T3 (es) 2010-08-16 2015-06-03 Novimmune S.A. Métodos para la generación de anticuerpos multiespecíficos y multivalentes
JO3375B1 (ar) 2010-11-08 2019-03-13 Regeneron Pharma أجسام مضادة بشرية للجين a1 الشبيه بعامل النخر الورمي (tl1a)
AU2011348232A1 (en) 2010-12-22 2013-07-18 Genentech, Inc. Anti-PCSK9 antibodies and methods of use
US8642835B2 (en) 2011-02-25 2014-02-04 Regeneron Pharmaceuticals, Inc. ADAM6 mice
SG2014007686A (en) 2011-08-05 2014-03-28 Regeneron Pharma Humanized universal light chain mice
EP3741862A1 (fr) 2011-09-19 2020-11-25 Kymab Limited Animaux, répertoires & procédés
CN103945689B (zh) 2011-09-19 2016-10-19 科马布有限公司 免疫球蛋白基因多样性的操纵及多抗体治疗剂
WO2013045916A1 (fr) 2011-09-26 2013-04-04 Kymab Limited Chaînes légères substituts (cls) chimères comprenant vpreb humain
US10246509B2 (en) 2011-10-17 2019-04-02 Regeneron Pharmaceuticals, Inc. Restricted immunoglobulin heavy chain mice
US20130102031A1 (en) 2011-10-25 2013-04-25 Anaptysbio, Inc. Use of somatic hypermutation to create insertion and deletion mutations in vitro
GB2496375A (en) 2011-10-28 2013-05-15 Kymab Ltd A non-human assay vertebrate comprising human antibody loci and human epitope knock-in, and uses thereof
US20180295821A1 (en) 2011-12-02 2018-10-18 Kymab Limited Transgenic Animals
GB201122047D0 (en) 2011-12-21 2012-02-01 Kymab Ltd Transgenic animals
US9253965B2 (en) 2012-03-28 2016-02-09 Kymab Limited Animal models and therapeutic molecules
BR112014015238B1 (pt) 2011-12-20 2022-11-16 Regeneron Pharmaceuticals, Inc Método ex vivo para preparar um anticorpo que se liga a um antígeno de interesse compreendendo identificar sequências de ácido nucleico a partir de linfócitos b de camundongo geneticamente modificado pela colocação de um gene adam6
DK3597038T3 (da) 2012-02-01 2021-06-28 Regeneron Pharma Humaniserede gnavere, der udtrykker tungkæder indeholdende VL-domæner
RU2017127772A (ru) 2012-03-02 2019-02-04 Регенерон Фармасьютикалз, Инк. Человеческие антитела к токсинам clostridium difficile
SG11201405088QA (en) 2012-03-06 2014-11-27 Regeneron Pharma Common light chain mouse
CA2865644A1 (fr) 2012-03-16 2013-09-19 Regeneron Pharmaceuticals, Inc. Souris produisant des proteines de liaison a l'antigene ayant des caracteristiques de liaison dependant du ph
WO2013144567A1 (fr) 2012-03-28 2013-10-03 Kymab Limited Vertébré non-humain transgénique pour l'expression d'anticorps entièrement humains à commutation isotypique
US10251377B2 (en) 2012-03-28 2019-04-09 Kymab Limited Transgenic non-human vertebrate for the expression of class-switched, fully human, antibodies
GB2502127A (en) 2012-05-17 2013-11-20 Kymab Ltd Multivalent antibodies and in vivo methods for their production
JO3820B1 (ar) 2012-05-03 2021-01-31 Regeneron Pharma أجسام مضادة بشرية لـ fel d1وطرق لاستخدامها
DK2800811T3 (en) 2012-05-25 2017-07-17 Univ Vienna METHODS AND COMPOSITIONS FOR RNA DIRECTIVE TARGET DNA MODIFICATION AND FOR RNA DIRECTIVE MODULATION OF TRANSCRIPTION
US8518765B1 (en) 2012-06-05 2013-08-27 Intermolecular, Inc. Aqua regia and hydrogen peroxide HCl combination to remove Ni and NiPt residues
PT2858487T (pt) 2012-06-12 2020-01-15 Regeneron Pharma Animais não humanos humanizados com locus de cadeias pesadas imunoglobulina restrito
US8962913B2 (en) * 2012-06-18 2015-02-24 Regeneron Pharmaceuticals, Inc. Humanized IL-7 rodents
CN105755009A (zh) 2012-11-09 2016-07-13 深圳市作物分子设计育种研究院 一种育性基因及其应用
ES2658401T3 (es) 2012-12-12 2018-03-09 The Broad Institute, Inc. Suministro, modificación y optimización de sistemas, métodos y composiciones para la manipulación de secuencias y aplicaciones terapéuticas
MY185881A (en) 2013-02-20 2021-06-14 Regeneron Pharma Non-human animals with modified immunoglobulin heavy chain sequences
WO2014141192A1 (fr) 2013-03-15 2014-09-18 Erasmus University Medical Center Génération d'anticorps à chaînes lourdes uniquement
US9788534B2 (en) 2013-03-18 2017-10-17 Kymab Limited Animal models and therapeutic molecules
US20150033372A1 (en) 2013-05-01 2015-01-29 Kymab Limited Human VpreB & Chimaeric Surrogate Light Chains in Transgenic Non-Human Vertebrates
US9783593B2 (en) 2013-05-02 2017-10-10 Kymab Limited Antibodies, variable domains and chains tailored for human use
US11707056B2 (en) 2013-05-02 2023-07-25 Kymab Limited Animals, repertoires and methods
US20140331344A1 (en) 2013-05-03 2014-11-06 Kymab Ltd. Transgenic Animals
US20140331339A1 (en) 2013-05-03 2014-11-06 Kymab Limited Transgenic Non-Human Assay Vertebrates, Assays and Kits
SG10201802295XA (en) 2013-10-01 2018-04-27 Kymab Ltd Animal Models and Therapeutic Molecules
GB201710984D0 (en) 2017-07-07 2017-08-23 Kymab Ltd Cells, vertebrates, populations & methods
JP7087648B2 (ja) * 2018-05-08 2022-06-21 トヨタ自動車株式会社 電池パック

Patent Citations (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US114985A (en) * 1871-05-16 Improvement in apparatus for cooling and preserving milk and other liquids
US5807715A (en) * 1984-08-27 1998-09-15 The Board Of Trustees Of The Leland Stanford Junior University Methods and transformed mammalian lymphocyte cells for producing functional antigen-binding protein including chimeric immunoglobulin
US20060015957A1 (en) * 1991-08-28 2006-01-19 Genpharm International, Inc. Transgenic non-human animals for producing chimeric antibodies
US7932431B2 (en) * 2003-02-26 2011-04-26 Crescendo Biologics Limited Genetically modified non-human mammals and cells
WO2007117410A2 (fr) * 2006-03-31 2007-10-18 Medarex, Inc. Animaux transgéniques exprimant des anticorps chimériques destinés à être utilisés pour la préparation d'anticorps humains
US20080076379A1 (en) * 2006-09-01 2008-03-27 Wanda Ying Li Outdoor umbrella with audio system
WO2008076379A2 (fr) * 2006-12-14 2008-06-26 Regeneron Pharmaceuticals, Inc. Anticorps humains au ligand delta-4 humain
US8771988B2 (en) * 2007-10-12 2014-07-08 Hoffmann-La Roche Inc. Protein expression from multiple nucleic acids
US20110236378A1 (en) * 2008-09-30 2011-09-29 Ablexis, Llc Non-human mammals for the production of chimeric antibodies
US9783618B2 (en) * 2013-05-01 2017-10-10 Kymab Limited Manipulation of immunoglobulin gene diversity and multi-antibody therapeutics

Non-Patent Citations (33)

* Cited by examiner, † Cited by third party
Title
Adams Genomics. 2005 December; 86 (6):753-8) *
Aguilera et al (EMBO 4(13B): 3689-3693, 1985 *
Bates et al, JEM, 204, 13, 3247-3256, 2007 *
Butler Rev Scientifique et Technique Office International Des Epizooties. 1998, 17, 1, 43-70, *
Clark et al (Nature Reviews: Genetics. 2003, 825-833 *
Gama Sosa et al (Brain Struct Funct, 2010, 214:91-109 *
Huber et al Clinical and Vaccine Immunology, 2006, 13(9), 981-990 *
Kaushik, Veterinary Immunology and irnmunopathology 87 (2002) 347-350 *
Kingzette et aL, PNAS, 95,11840-11845 (1998) *
Lefranc, et al (Immunoglobulin Facts Book. London: Academic Press, 2001, pages 3-44, 98-100, and 102, *
Lefranc, et al (Immunoglobulin Facts Book. London: Academic Press, 3-44, 98-100 (Year: 2001) *
Lonberg et al Nature Biotech, 2005, 23, 1117-1125 *
Murphy et al Slide Presentation dated November 3, 2009, at Wellcome Trust Advanced Course: Genetic Manipulation of ES Cells, in Hinxton, UK, entitled "BAC-based Modifications of the Mouse Genome: The Big and the Backward", slides 1-57), *
NCBI accession no NT_114985, page 1. *
Nicholson et al Journal of Immunology, 12, 6898-6906 (Year: 1999) *
Niemann et al (Rev. Sci, Tech. Off. Int. Spiz. 2005, (24), 285-298) *
Oberdoerffer et al Nucleic Acid Res. , 31, 1-7, (2003) *
Oberdoerffer et al Nucleic Acids Res. 2003, 31: e140, *
Parng, The Journal of Immunology, 1996, 157: 5478-5486 *
Qi et al Hypertension. 2005; 45:1004-1011. *
Ramirez-Solis et al Nature, 1995, 378, 14, 720-724 *
Ristevik etal, 2005, 29, 153-163 *
Rosner et al Immunology 103:179-187 (Year: 2001) *
Rosner et al Immunology 103:179-187, 2001 *
Sen and Baltimore Cell, 1986, 46, 705-716 *
Smith Journal of Biotechnology 99 (2002) 1-22 *
Torres and Kuhn, Laboratory Protocols for Conditional Gene Targeting, 1997, p37-40 *
Van Snick et al (Arthritis and Rheumatism, 1983, 1085-1090 *
Wagner et al abstract Nucleic Acids Res. 1994; 22(8):1389-93 *
Wagner Eur. J. Immunol. 24: 2672-2681 (Year: 1994) *
Wallace et al Cell, 2007, 197-209 *
Zemlin et al Journal of Mol. Biology, 2003, 733-749 *
Zheng et al Mol. Cell Biol. 2000, 20, 648-655, *

Cited By (105)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9434782B2 (en) 2009-07-08 2016-09-06 Kymab Limited Animal models and therapeutic molecules
EP2421357A1 (fr) 2009-07-08 2012-02-29 Kymab Limited Modèles d'animaux et molécules thérapeutiques
US10064398B2 (en) 2009-07-08 2018-09-04 Kymab Limited Animal models and therapeutic molecules
US10165763B2 (en) 2009-07-08 2019-01-01 Kymab Limited Animal models and therapeutic molecules
US11812731B2 (en) 2009-07-08 2023-11-14 Kymab Ltd. Animal models and therapeutic molecules
US11606941B2 (en) 2009-07-08 2023-03-21 Kymab Limited Animal models and therapeutic molecules
EP3622813B1 (fr) 2009-07-08 2021-02-17 Kymab Limited Modèles d'animaux et molécules thérapeutiques
EP3028564B1 (fr) 2009-07-08 2017-09-27 Kymab Limited Modèles animaux et molécules thérapeutiques
US11564380B2 (en) 2009-07-08 2023-01-31 Kymab Limited Animal models and therapeutic molecules
EP2517556B1 (fr) 2009-07-08 2015-11-18 Kymab Limited Méthode de Recombination à un Site Spécifique, Rongeurs & Cellules de Rongeurs Capables d'Exprimer des Anticorps ou des Chaînes Chimériques
EP2604110B1 (fr) 2009-07-08 2016-11-30 Kymab Limited Modèles animaux et molécules thérapeutiques
US9504236B2 (en) 2009-07-08 2016-11-29 Kymab Limited Animal models and therapeutic molecules
US9505827B2 (en) 2009-07-08 2016-11-29 Kymab Limited Animal models and therapeutic molecules
US9447177B2 (en) 2009-07-08 2016-09-20 Kymab Limited Transgenic mouse homozygous for chimeric IgH locus
US10412940B2 (en) 2010-02-08 2019-09-17 Regeneron Pharmaceuticals, Inc. Mice expressing a limited immunoglobulin light chain repertoire
US20110195454A1 (en) * 2010-02-08 2011-08-11 Regeneron Pharmaceuticals, Inc. Common Light Chain Mouse
US10143186B2 (en) 2010-02-08 2018-12-04 Regeneron Pharmaceuticals, Inc. Common light chain mouse
US11026407B2 (en) 2010-02-08 2021-06-08 Regeneran Pharmaceuticals, Inc. Mice expressing a limited immunoglobulin light chain repertoire
US9969814B2 (en) 2010-02-08 2018-05-15 Regeneron Pharmaceuticals, Inc. Methods for making fully human bispecific antibodies using a common light chain
US10167344B2 (en) 2010-02-08 2019-01-01 Regeneron Pharmaceuticals, Inc. Mice expressing a limited immunoglobulin light chain repertoire
US9796788B2 (en) 2010-02-08 2017-10-24 Regeneron Pharmaceuticals, Inc. Mice expressing a limited immunoglobulin light chain repertoire
US10986820B2 (en) 2010-02-08 2021-04-27 Regeneron Pharmaceuticals, Inc. Common light chain mouse
US9516868B2 (en) 2010-08-02 2016-12-13 Regeneron Pharmaceuticals, Inc. Mice that make VL binding proteins
US10954310B2 (en) 2010-08-02 2021-03-23 Regeneran Pharmaceuticals, Inc. Mice that make VL binding proteins
US9686970B2 (en) 2010-08-02 2017-06-27 Regeneron Pharmaceuticals, Inc. Mice that make VL binding proteins
US10905108B2 (en) 2011-02-25 2021-02-02 Regeneron Pharmaceuticals, Inc. ADAM6 mice
US20120322108A1 (en) * 2011-02-25 2012-12-20 Regeneron Pharmaceuticals, Inc. Adam6 Mice
US8642835B2 (en) * 2011-02-25 2014-02-04 Regeneron Pharmaceuticals, Inc. ADAM6 mice
US10905109B2 (en) 2011-02-25 2021-02-02 Regeneren Pharmaceuticals, Inc. ADAM6 mice
US8697940B2 (en) * 2011-02-25 2014-04-15 Regeneron Pharmaceuticals, Inc. ADAM6 mice
US9944716B2 (en) 2011-02-25 2018-04-17 Regeneron Pharmaceuticals, Inc. ADAM6 mice
US10694725B2 (en) 2011-02-25 2020-06-30 Regeneron Pharmaceuticals, Inc. ADAM6 mice
US10577430B2 (en) 2011-02-25 2020-03-03 Regeneron Pharmaceuticals, Inc. ADAM6 mice
US10072095B2 (en) 2011-02-25 2018-09-11 Regeneron Pharmaceuticals, Inc. ADAM6 mice
US9932408B2 (en) 2011-02-25 2018-04-03 Regeneron Pharmaceuticals, Inc. ADAM6 mice
US11950578B2 (en) 2011-02-25 2024-04-09 Regeneron Pharmaceuticals, Inc. ADAM6 mice
US11357217B2 (en) 2011-08-05 2022-06-14 Regeneron Pharmaceuticals, Inc. Humanized universal light chain mice
US10130081B2 (en) 2011-08-05 2018-11-20 Regeneron Pharmaceuticals, Inc. Humanized universal light chain mice
JP7067868B2 (ja) 2011-09-19 2022-05-16 カイマブ・リミテッド 免疫グロブリン遺伝子多様性の操作およびマルチ抗体治療薬
EP3311661B1 (fr) 2011-09-19 2022-04-20 Kymab Limited Manipulation de la diversité de gènes d'immunoglobuline et multi-anticorps thérapeutiques
EP3311661A1 (fr) 2011-09-19 2018-04-25 Kymab Limited Manipulation de la diversité de gènes d'immunoglobuline et multi-anticorps thérapeutiques
US11051497B2 (en) 2011-09-19 2021-07-06 Kymab Limited Manipulation of immunoglobulin gene diversity and multi-antibody therapeutics
US9963716B2 (en) 2011-09-26 2018-05-08 Kymab Limited Chimaeric surrogate light chains (SLC) comprising human VpreB
US11261248B2 (en) 2011-10-17 2022-03-01 Regeneron Pharmaceuticals, Inc. Restricted immunoglobulin heavy chain mice
US10246509B2 (en) 2011-10-17 2019-04-02 Regeneron Pharmaceuticals, Inc. Restricted immunoglobulin heavy chain mice
US9932398B2 (en) 2011-10-17 2018-04-03 Regeneron Pharmaceuticals, Inc. Restricted immunoglobulin heavy chain mice
US9706759B2 (en) * 2011-12-20 2017-07-18 Regeneron Pharmaceuticals, Inc. Humanized light chain mice
US11612151B2 (en) 2011-12-20 2023-03-28 Regeneron Pharmaceuticals, Inc. Humanized light chain mice
US20130160153A1 (en) * 2011-12-20 2013-06-20 Regeneron Pharmaceuticals, Inc. Humanized Light Chain Mice
US9622459B2 (en) * 2011-12-20 2017-04-18 Regeneron Pharmaceuticals, Inc. Humanized light chain mice
US20140017228A1 (en) * 2011-12-20 2014-01-16 Regeneron Pharmaceuticals, Inc. Humanized light chain mice
US10561124B2 (en) 2011-12-20 2020-02-18 Regeneron Pharmaceuticals, Inc. Humanized light chain mice
US11617357B2 (en) 2011-12-20 2023-04-04 Regeneron Pharmaceuticals, Inc. Humanized light chain mice
US20130212719A1 (en) * 2012-02-01 2013-08-15 Regeneron Pharmaceuticals, Inc. Humanized Rodents that Express Heavy Chain Containing VL Domains
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
US9801362B2 (en) 2012-03-16 2017-10-31 Regeneron Pharmaceuticals, Inc. Non-human animals expressing pH-sensitive immunoglobulin sequences
US9648856B2 (en) 2012-03-16 2017-05-16 Regeneron 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
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
US11224207B2 (en) 2012-03-16 2022-01-18 Regeneran Pharmaceuticals, Inc. Non-human animals expressing pH-sensitive immunoglobulin sequences
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
US9301510B2 (en) 2012-03-16 2016-04-05 Regeneron Pharmaceuticals, Inc. Mice that produce antigen-binding proteins with pH-dependent binding characteristics
US9896516B2 (en) 2012-03-28 2018-02-20 Kymab Limited Animal models and therapeutic molecules
US11297811B2 (en) 2012-03-28 2022-04-12 Kymab Limited Transgenic non-human vertebrate for the expression of class-switched, fully human, antibodies
US10251377B2 (en) 2012-03-28 2019-04-09 Kymab Limited Transgenic non-human vertebrate for the expression of class-switched, fully human, antibodies
US9253965B2 (en) 2012-03-28 2016-02-09 Kymab Limited Animal models and therapeutic molecules
US9938358B2 (en) 2012-03-28 2018-04-10 Kymab Limited Animal models and therapeutic molecules
US10774155B2 (en) 2012-03-28 2020-09-15 Kymab Limited Animal models and therapeutic molecules
US9445581B2 (en) 2012-03-28 2016-09-20 Kymab Limited Animal models and therapeutic molecules
US9924705B2 (en) 2012-03-28 2018-03-27 Kymab Limited Animal models and therapeutic molecules
US9938357B2 (en) 2012-03-28 2018-04-10 Kymab Limited Animal models and therapeutic molecules
US10667501B2 (en) 2012-05-17 2020-06-02 Kymab Limited Transgenic non-human vertebrate for the in vivo production of dual specificity immunoglobulins or hypermutated heavy chain only immunoglobulins
US10238093B2 (en) 2012-06-12 2019-03-26 Regeneron Pharmaceuticals, Inc. Humanized non-human animals with restricted immunoglobulin heavy chain loci
US10542735B2 (en) 2012-06-12 2020-01-28 Regerneron Pharmaceuticals, Inc. Humanized non-human animals with restricted immunoglobulin heavy chain loci
US11666040B2 (en) 2012-06-12 2023-06-06 Regeneron Pharmaceuticals, Inc. Humanized non-human animals with restricted immunoglobulin heavy chain loci
US11559050B2 (en) 2012-06-12 2023-01-24 Regeneron Pharmaceuticals, Inc. Humanized non-human animals with restricted immunoglobulin heavy chain loci
EP2931030B1 (fr) 2012-12-14 2020-04-29 Open Monoclonal Technology, Inc. Polynucléotides codant pour des anticorps de rongeur ayant des idiotypes humains, et animaux les comprenant
US9930871B2 (en) 2013-02-20 2018-04-03 Regeneron Pharmaceuticals, Inc. Non-human animals with modified immunoglobulin heavy chain sequences
US9788534B2 (en) 2013-03-18 2017-10-17 Kymab Limited Animal models and therapeutic molecules
US10226033B2 (en) 2013-03-18 2019-03-12 Kymab Limited Animal models and therapeutic molecules
US11297810B2 (en) 2013-03-18 2022-04-12 Kymab Limited Animal models and therapeutic molecules
US9783618B2 (en) 2013-05-01 2017-10-10 Kymab Limited Manipulation of immunoglobulin gene diversity and multi-antibody therapeutics
US11707056B2 (en) * 2013-05-02 2023-07-25 Kymab Limited Animals, repertoires and methods
US10730930B2 (en) 2013-05-02 2020-08-04 Kymab Limited Antibodies, variable domains and chains tailored for human use
US11820810B2 (en) 2013-05-02 2023-11-21 Kymab Limited Antibodies, variable domains and chains tailored for human use
US9783593B2 (en) 2013-05-02 2017-10-10 Kymab Limited Antibodies, variable domains and chains tailored for human use
US20150033369A1 (en) * 2013-05-02 2015-01-29 Kymab Limited Animals, repertoires & methods
US11920128B2 (en) 2013-09-18 2024-03-05 Kymab Limited Methods, cells and organisms
EP2877571B1 (fr) 2013-09-18 2018-05-30 Kymab Limited Procédés, cellules et organismes
US10966412B2 (en) 2013-10-01 2021-04-06 Kymab Limited Animal models and therapeutic molecules
EP3794941A1 (fr) 2013-10-01 2021-03-24 Kymab Limited Modèles d'animaux et molécules thérapeutiques
US11399522B2 (en) 2013-10-01 2022-08-02 Kymab Limited Animal models and therapeutic molecules
US10149462B2 (en) 2013-10-01 2018-12-11 Kymab Limited Animal models and therapeutic molecules
US11820997B2 (en) 2013-12-11 2023-11-21 Regeneron Pharmaceuticals, Inc. Methods and compositions for the targeted modification of a genome
US10711280B2 (en) 2013-12-11 2020-07-14 Regeneron Pharmaceuticals, Inc. Methods and compositions for the targeted modification of a mouse ES cell genome
US10881085B2 (en) 2014-03-21 2021-01-05 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
US11111314B2 (en) 2015-03-19 2021-09-07 Regeneron Pharmaceuticals, Inc. Non-human animals that select for light chain variable regions that bind antigen
US10980221B2 (en) 2016-06-03 2021-04-20 Regeneron Pharmaceuticals, Inc. Non-human animals expressing exogenous terminal deoxynucleotidyltransferase
US20210029978A1 (en) * 2016-11-04 2021-02-04 Regeneron Pharmaceuticals, Inc. Non-human animals having an engineered immunoglobulin lambda light chain locus
US12016313B2 (en) 2017-01-19 2024-06-25 Omniab Operations, Inc. Human antibodies from transgenic rodents with multiple heavy chain immunoglobulin loci
CN111031789A (zh) * 2017-04-10 2020-04-17 基因组研究有限公司 动物模型和治疗性分子
WO2018189520A1 (fr) 2017-04-10 2018-10-18 Genome Research Limited Modèles animaux et molécules thérapeutiques
US20210017253A1 (en) * 2018-03-27 2021-01-21 Chongqing Jinmaibo Biotec Co., Ltd Nucleic acid molecules and applications thereof in preparing human single-domain antibody
CN108486125A (zh) * 2018-03-27 2018-09-04 重庆金迈博生物科技有限公司 一种核酸分子及其在制备人源单域抗体中的应用

Also Published As

Publication number Publication date
JP2020150951A (ja) 2020-09-24
US20200337280A1 (en) 2020-10-29
US20140201856A1 (en) 2014-07-17
US20140120582A1 (en) 2014-05-01
HUE055817T2 (hu) 2021-12-28
PL3241435T3 (pl) 2021-12-13
US11564380B2 (en) 2023-01-31
US20130318643A1 (en) 2013-11-28
US20140150126A1 (en) 2014-05-29
SMT201300046B (it) 2013-07-09
EP3622813B1 (fr) 2021-02-17
US20120167237A1 (en) 2012-06-28
EP3622813A1 (fr) 2020-03-18
US20170051045A1 (en) 2017-02-23
CA2767436A1 (fr) 2011-01-13
US20200375158A1 (en) 2020-12-03
EP3241435A1 (fr) 2017-11-08
EP3622814A1 (fr) 2020-03-18
DK2564695T3 (en) 2015-05-26
US20150113669A1 (en) 2015-04-23
EP2792236B2 (fr) 2023-03-22
DK3028564T5 (da) 2024-04-29
JP6612275B2 (ja) 2019-11-27
US20140134156A1 (en) 2014-05-15
EP2604110B1 (fr) 2016-11-30
EP4215043A1 (fr) 2023-07-26
PL2517556T3 (pl) 2016-03-31
PT2564695E (pt) 2015-06-03
DK3241435T3 (da) 2021-08-23
DK3028564T3 (da) 2017-11-13
JP2020156502A (ja) 2020-10-01
NZ597481A (en) 2013-10-25
US20150334998A1 (en) 2015-11-26
HK1180535A1 (en) 2013-10-25
CN102638971A (zh) 2012-08-15
PT2421357E (pt) 2013-04-18
DK2421357T3 (da) 2013-04-15
US20160345552A1 (en) 2016-12-01
ES2884309T3 (es) 2021-12-10
DK2517556T3 (en) 2016-01-11
EP2798950A1 (fr) 2014-11-05
JP2017086084A (ja) 2017-05-25
AU2016244326A1 (en) 2016-11-03
EP2604110A3 (fr) 2013-08-14
US9447177B2 (en) 2016-09-20
DK3622813T3 (da) 2021-05-03
JP2015126736A (ja) 2015-07-09
DK2798950T4 (da) 2023-05-15
HRP20211253T1 (hr) 2021-11-12
EP4014729B1 (fr) 2024-08-07
DK2517557T3 (en) 2016-05-30
ES2537239T3 (es) 2015-06-03
ES2974417T3 (es) 2024-06-27
EP3028564B2 (fr) 2024-01-24
EP2517557B1 (fr) 2016-04-13
SI2421357T1 (sl) 2013-06-28
AU2010269978B2 (en) 2016-11-03
EP3888457B1 (fr) 2024-05-15
EP3241435B1 (fr) 2021-05-26
JP2020028296A (ja) 2020-02-27
AU2021200079A1 (en) 2021-03-18
EP2792236B1 (fr) 2017-11-15
EP2421357B1 (fr) 2013-01-23
EP3888457A1 (fr) 2021-10-06
EP3028564B1 (fr) 2017-09-27
CN105340834B (zh) 2018-11-06
NO2792236T3 (fr) 2018-04-14
HUE061788T2 (hu) 2023-08-28
WO2011004192A1 (fr) 2011-01-13
HRP20130253T1 (hr) 2013-05-31
DK2792236T3 (en) 2018-01-02
KR20130042461A (ko) 2013-04-26
DK2792236T4 (da) 2023-05-15
US20230263143A1 (en) 2023-08-24
US20170099815A1 (en) 2017-04-13
HK1172207A1 (zh) 2013-04-19
EP4014729A1 (fr) 2022-06-22
JP2020156503A (ja) 2020-10-01
EP2792236A3 (fr) 2014-11-12
JP6088562B2 (ja) 2017-03-01
EP2564695B1 (fr) 2015-04-15
ES2403087T3 (es) 2013-05-14
US10165763B2 (en) 2019-01-01
AU2010269978A1 (en) 2012-02-02
EP3028564A1 (fr) 2016-06-08
EP3871497B1 (fr) 2024-02-28
DK3622815T3 (da) 2023-06-26
ES2948572T3 (es) 2023-09-14
US20200352144A1 (en) 2020-11-12
EP2792236A2 (fr) 2014-10-22
EP3028565A1 (fr) 2016-06-08
EP2517557B2 (fr) 2023-08-02
DK2517556T4 (da) 2023-05-15
US9434782B2 (en) 2016-09-06
EP2517556B1 (fr) 2015-11-18
US20140150125A1 (en) 2014-05-29
EP2517556A3 (fr) 2013-07-17
JP6876853B2 (ja) 2021-05-26
EP3888457C0 (fr) 2024-05-15
US20170094956A1 (en) 2017-04-06
US20230270088A1 (en) 2023-08-31
US20170099816A1 (en) 2017-04-13
US11812731B2 (en) 2023-11-14
US20200352145A1 (en) 2020-11-12
US20170099817A1 (en) 2017-04-13
ES2978496T3 (es) 2024-09-13
AU2018206729A1 (en) 2018-08-02
US20140182003A1 (en) 2014-06-26
EP3622813B2 (fr) 2024-06-19
US20150040250A1 (en) 2015-02-05
US11606941B2 (en) 2023-03-21
CN105340834A (zh) 2016-02-24
EP3028565B1 (fr) 2017-09-27
ES2557737T3 (es) 2016-01-28
JP2012532598A (ja) 2012-12-20
SI3241435T1 (sl) 2021-11-30
JP7141427B2 (ja) 2022-09-22
SG177380A1 (en) 2012-02-28
ES2965212T3 (es) 2024-04-11
CY1126083T1 (el) 2023-11-15
EP2604111A2 (fr) 2013-06-19
US20170105396A1 (en) 2017-04-20
EP2421357A1 (fr) 2012-02-29
EP2604111A3 (fr) 2013-12-18
EP2604110A2 (fr) 2013-06-19
US20160345551A1 (en) 2016-12-01
US20170071174A1 (en) 2017-03-16
EP3622815A1 (fr) 2020-03-18
PL2564695T3 (pl) 2015-10-30
CN102638971B (zh) 2015-10-07
EP3871498A1 (fr) 2021-09-01
US20160044900A1 (en) 2016-02-18
LT3241435T (lt) 2021-10-25
EP3871498B1 (fr) 2024-09-25
EP3622814B1 (fr) 2023-11-08
EP3871497C0 (fr) 2024-02-28
EP2564695A1 (fr) 2013-03-06
BR112012000536A2 (pt) 2020-08-11
SG2014010995A (en) 2014-05-29
US20170101482A1 (en) 2017-04-13
DK2604110T3 (en) 2017-02-13
EP2517556B2 (fr) 2023-03-22
US20240057572A1 (en) 2024-02-22
JP5944312B2 (ja) 2016-07-05
US9505827B2 (en) 2016-11-29
EP2517556A2 (fr) 2012-10-31
HK1201026A1 (en) 2015-08-21
DK2517557T4 (da) 2023-09-25
EP2517557A2 (fr) 2012-10-31
US20140201854A1 (en) 2014-07-17
EP3871497A1 (fr) 2021-09-01
DK3028564T4 (da) 2024-04-22
PT3241435T (pt) 2021-08-26
HK1202373A1 (en) 2015-10-02
DK2798950T3 (en) 2017-05-22
AU2021200079B2 (en) 2023-11-09
AU2018206729C1 (en) 2023-08-10
US10064398B2 (en) 2018-09-04
PT2517556E (pt) 2016-01-20
US20130042331A2 (en) 2013-02-14
DK3028565T3 (da) 2017-11-13
JP6930695B2 (ja) 2021-09-01
EP2798950B2 (fr) 2023-03-22
DK3622813T4 (da) 2024-09-16
EP3622814C0 (fr) 2023-11-08
EP2517557A3 (fr) 2013-07-24
KR101875233B1 (ko) 2018-07-05
EP2798950B1 (fr) 2017-04-19
HK1162119A1 (en) 2012-08-24
AU2016244326B2 (en) 2018-04-26
JP6876854B2 (ja) 2021-05-26
AU2018206729B2 (en) 2020-10-08
PL2421357T3 (pl) 2013-07-31
US20160150768A1 (en) 2016-06-02
EP3622815B1 (fr) 2023-04-05
US20230225302A1 (en) 2023-07-20

Similar Documents

Publication Publication Date Title
US20230225302A1 (en) Animal Models and Therapeutic Molecules
US20230159660A1 (en) Animal models and therapeutic molecules
US9445581B2 (en) Animal models and therapeutic molecules
US20140283150A1 (en) Animal Models and Therapeutic Molecules

Legal Events

Date Code Title Description
AS Assignment

Owner name: KYMAB LIMITED, UNITED KINGDOM

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:BRADLEY, ALLAN, MR.;LEE, E-CHIANG, MR.;LIANG, QI;AND OTHERS;SIGNING DATES FROM 20111213 TO 20120116;REEL/FRAME:027718/0485

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

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

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

Free format text: FINAL REJECTION MAILED

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

Free format text: ADVISORY ACTION MAILED

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

Free format text: RESPONSE AFTER FINAL ACTION FORWARDED TO EXAMINER

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

Free format text: NOTICE OF ALLOWANCE MAILED -- APPLICATION RECEIVED IN OFFICE OF PUBLICATIONS

ZAAB Notice of allowance mailed

Free format text: ORIGINAL CODE: MN/=.

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

Free format text: AWAITING TC RESP, ISSUE FEE PAYMENT VERIFIED

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

Free format text: PUBLICATIONS -- ISSUE FEE PAYMENT VERIFIED

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

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

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

Free format text: NON FINAL ACTION MAILED

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

Free format text: FINAL REJECTION MAILED

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

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

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

Free format text: NON FINAL ACTION MAILED

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

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

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

Free format text: FINAL REJECTION MAILED

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

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

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

Free format text: NON FINAL ACTION MAILED

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

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

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

Free format text: FINAL REJECTION MAILED

STCB Information on status: application discontinuation

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