US20090307787A1 - Generation of heavy-chain only antibodies in transgenic animals - Google Patents

Generation of heavy-chain only antibodies in transgenic animals Download PDF

Info

Publication number
US20090307787A1
US20090307787A1 US12/161,981 US16198107A US2009307787A1 US 20090307787 A1 US20090307787 A1 US 20090307787A1 US 16198107 A US16198107 A US 16198107A US 2009307787 A1 US2009307787 A1 US 2009307787A1
Authority
US
United States
Prior art keywords
heavy chain
human
locus
gene segments
antibody
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
US12/161,981
Other languages
English (en)
Inventor
Franklin Gerardus Grosveld
Richard Wilhelm Janssens
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.)
Harbour Antibodies BV
Original Assignee
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from GBGB0601511.9A external-priority patent/GB0601511D0/en
Priority claimed from GBGB0618345.3A external-priority patent/GB0618345D0/en
Application filed by Individual filed Critical Individual
Publication of US20090307787A1 publication Critical patent/US20090307787A1/en
Assigned to ERASMUS UNIVERSITY MEDICAL CENTER reassignment ERASMUS UNIVERSITY MEDICAL CENTER ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: GROSVELD, FRANKLIN GERARDUS, JANSSENS, RICHARD WILHELM
Assigned to HARBOUR ANTIBODIES B.V. reassignment HARBOUR ANTIBODIES B.V. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: ERASMUS UNIVERSITAIR MEDISCH CENTRUM ROTTERDAM
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K67/00Rearing or breeding animals, not otherwise provided for; New breeds of animals
    • A01K67/027New breeds of vertebrates
    • A01K67/0275Genetically modified vertebrates, e.g. transgenic
    • A01K67/0278Humanized animals, e.g. knockin
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/08Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from viruses
    • C07K16/10Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from viruses from RNA viruses
    • C07K16/1009Picornaviridae, e.g. hepatitis A virus
    • 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
    • C07K16/1228Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from bacteria from Gram-negative bacteria from Enterobacteriaceae (F), e.g. Citrobacter, Serratia, Proteus, Providencia, Morganella, Yersinia
    • C07K16/1232Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from bacteria from Gram-negative bacteria from Enterobacteriaceae (F), e.g. Citrobacter, Serratia, Proteus, Providencia, Morganella, Yersinia from Escherichia (G)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/12Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from bacteria
    • C07K16/1267Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from bacteria from Gram-positive bacteria
    • C07K16/1282Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from bacteria from Gram-positive bacteria from Clostridium (G)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/12Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from bacteria
    • C07K16/1267Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from bacteria from Gram-positive bacteria
    • C07K16/1285Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from bacteria from Gram-positive bacteria from Corynebacterium (G)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/24Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against cytokines, lymphokines or interferons
    • C07K16/241Tumor Necrosis Factors
    • CCHEMISTRY; METALLURGY
    • 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
    • C12N15/02Preparation of hybrid cells by fusion of two or more cells, e.g. protoplast fusion
    • 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
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/8509Vectors or expression systems specially adapted for eukaryotic hosts for animal cells for producing genetically modified animals, e.g. transgenic
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2207/00Modified animals
    • A01K2207/15Humanized animals
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • A01K2217/05Animals comprising random inserted nucleic acids (transgenic)
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • A01K2217/07Animals genetically altered by homologous recombination
    • A01K2217/072Animals genetically altered by homologous recombination maintaining or altering function, i.e. knock in
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • A01K2217/07Animals genetically altered by homologous recombination
    • A01K2217/075Animals genetically altered by homologous recombination inducing loss of function, i.e. knock out
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2227/00Animals characterised by species
    • A01K2227/10Mammal
    • A01K2227/105Murine
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2267/00Animals characterised by purpose
    • A01K2267/01Animal expressing industrially exogenous proteins
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins 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
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/569Single domain, e.g. dAb, sdAb, VHH, VNAR or nanobody®
    • 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 to improved methods for the manufacture in transgenic non-human mammals of a diverse repertoire of functional, affinity-matured heavy chain-only antibodies in response to antigen challenge and uses thereof.
  • the invention also relates to the manufacture of a diverse repertoire of class-specific heavy chain-only antibodies from multiple loci.
  • the present invention relates to a method for the generation of human antigen-specific, high affinity, heavy chain-only antibodies of any class or mixture of classes and the isolation and expression of fully functional V H antigen-binding domains.
  • Heavy chain-only antibodies generated using the methods of the present invention are also described.
  • antibodies The structure of antibodies is well known in the art. Most natural antibodies are tetrameric, comprising two heavy chains and two light chains. The heavy chains are joined to each other via disulphide bonds between hinge domains located approximately half way along each heavy chain. A light chain is associated with each heavy chain on the N-terminal side of the hinge domain. Each light chain is normally bound to its respective heavy chain by a disulphide bond close to the hinge domain.
  • each chain fold When an antibody molecule is correctly folded, each chain folds into a number of distinct globular domains joined by more linear polypeptide sequences.
  • V L variable
  • C L constant
  • Heavy chains have a single variable domain V H , a first constant domain (C H 1), a hinge domain and two or three further constant domains.
  • the heavy chain constant domains and the hinge domain together form what is generally known as the constant region of an antibody heavy chain. Interaction of the heavy (V H ) and light (V L ) chain variable domains results in the formation of an antigen binding region (F V ).
  • variable domains of both heavy (V H ) and light (V L ) chains some short polypeptide segments show exceptional variability. These segments are termed hypervariable regions or complementarity determining regions (CDRs). The intervening segments are called framework regions (FRs). In each of the V H and V L domains, there are three CDRs (CDR1-CDR3).
  • IgA In mammals there are five classes of antibody: IgA, IgD, IgE, IgG and IgM, with four IgG and two IgA subtypes present in humans.
  • H Class chain L chain Subunits mg/ml Notes IgG gamma kappa or H 2 L 2 6-13 transferred across lambda placenta IgM mu kappa or (H 2 L 2 ) 5 0.5-3 first antibodies to lambda appear after immunization IgA alpha kappa or (H 2 L 2 ) 2 0.6-3 much higher lambda concentrations in secretions IgD delta kappa or H 2 L 2 ⁇ 0.14 function uncertain lambda IgE epsilon kappa or H 2 L 2 ⁇ 0.0004 binds to basophils and lambda mast cells sensitizing them for certain allergic reactions
  • IgA can be found in areas containing mucus (e.g. in the gut, in the respiratory tract or in the urinogenital tract) and prevents the colonization of mucosal areas by pathogens.
  • IgD functions mainly as an antigen receptor on B cells.
  • IgE binds to allergens and triggers histamine release from mast cells (the underlying mechanism of allergy) and also provides protection against helminths (worms).
  • IgG in its four isotypes provides the majority of antibody-based immunity against invading pathogens.
  • IgM is expressed on the surface of B cells and also in a secreted form with very high affinity for eliminating pathogens in the early stages of B cell mediated immunity (i.e. before there is sufficient IgG to eliminate the pathogens).
  • Normal human B cells contain a single heavy chain locus on chromosome 14 from which the gene encoding a heavy chain is produced by rearrangement. In the mouse the heavy chain locus is located on chromosome 12.
  • a normal heavy chain locus comprises a plurality of V gene segments, a number of D gene segments and a number of J gene segments. Most of a V H domain is encoded by a V gene segment, but the C terminal end of each V H domain is encoded by a D gene segment and a J gene segment.
  • VDJ rearrangement in B-cells, followed by affinity maturation, provides each V H domain with its antigen binding specificity. Sequence analysis of normal H 2 L 2 tetramers demonstrates that diversity results primarily from a combination of VDJ rearrangement and somatic hypermutation [3]. There are over 50 human V gene segments present in the human genome of which only 39 are functional.
  • Fully human antibodies (H 2 L 2 ) can now be derived from transgenic mice in response to antigen challenge.
  • Such transgenic mice comprise a single human heavy chain locus and a separate light chain locus.
  • the comparable mouse heavy and light chain loci are deleted or suppressed so that only human antibodies are produced in the absence of mouse antibodies ([4-10]).
  • heavy chain-only antibody devoid of light chain
  • man Heavy Chain Disease
  • murine model systems Analysis of heavy chain disease at the molecular level showed that mutations and deletions at the level of the genome could result in inappropriate expression of the heavy chain C H 1 domain, giving rise to the expression of heavy chain-only antibody lacking the ability to bind light chain [11, 12].
  • the heavy chain locus in the camelid germline comprises gene segments encoding some or all of the possible heavy chain constant regions.
  • a re-arranged V HH DJ binding domain is spliced onto the 5′ end of the gene segment encoding the hinge domain, to provide a re-arranged gene encoding a heavy chain which lacks a C H 1 domain and is therefore unable to associate with a light chain.
  • Camelid V HH domains contain a number of characteristic amino acids at positions 37, 44, 45 and 47 [49]. These conserved amino acids are thought to be important for conferring solubility on heavy chain-only antibodies. Only certain camelid V H domains are V HH domains with improved solubility characteristics. They are limited to V H subfamily and so respond productively only to a limited range of antigens.
  • Heavy chain-only monoclonal antibodies can be recovered from B-cells of camelid spleen by standard cloning technology or from B-cell mRNA by phage or other display technology [18]. Heavy chain-only antibodies derived from camelids are of high affinity. Sequence analysis of mRNA encoding heavy chain-only antibody demonstrates that diversity results primarily from a combination of V HH DJ rearrangement and somatic hypermutation [49] as is also observed in the production of normal tetrameric antibodies.
  • V H domains An important and common feature of natural camelid and human V H domains is that each domain binds as a monomer with no dependency on dimerisation with a V L domain for optimal solubility and binding affinity.
  • the gene segments encoding each of the constant regions had a deletion of the C H 1 domain to prevent the binding of light chains.
  • the locus contained the antibody LCR at the 3′ end to ensure a high level of expression in cells of the B lineage. This locus was introduced into the mice by microinjection of fertilized eggs.
  • Antibody-based products are usually derived from natural tetrameric antibodies.
  • routes of derivation e.g. from transgenic mice
  • routes of manufacture e.g. from transgenic mice
  • product-specific substances of matter e.g. from transgenic mice
  • Antibody-based products will represent a high proportion of new medicines launched in the 21 st century. Monoclonal antibody therapy is already accepted as a preferred route for the treatment for rheumatoid arthritis and Crohn's disease and there is impressive progress in the treatment of cancer. Antibody-based products are also in development for the treatment of cardiovascular and infectious diseases. Most marketed antibody-based products recognise and bind a single, well-defined epitope on the target ligand (e.g. TNF ⁇ ).
  • TNF ⁇ target ligand
  • V H domains have been: selected from randomised human V H domains in display libraries or derived from heavy chain-only antibody produced naturally from antigen challenge of camelids or derived from V H domain libraries made from camelids. These high affinity V H domains have been incorporated into antibody-based products. These V H domains, also called V HH domains, display a number of differences from classical V H domains, in particular a number of mutations that ensure improved solubility and stability of the heavy chains in the absence of light chains. Most prominent amongst these changes is the presence of a charged amino acid at position 45 [16].
  • Ward et al. [18] demonstrated unambiguously that cloned murine V H regions, when expressed as soluble protein monomers in an E. coli expression system, retain the ability to bind antigen with high affinity.
  • Ward et al [18] describe the isolation and characterisation of V H domains and set out the potential commercial advantages of this approach when compared with classic monoclonal antibody production (see last paragraph). They also recognise that V H domains isolated from heavy chains which normally associate with a light chain lack the solubility of the natural tetrameric antibodies. Hence Ward et al [18] used the term “sticky” to describe these molecules and proposed that this “stickiness” can be addressed through the design of V H domains with improved solubility properties.
  • V H solubility has subsequently been addressed using combinations of randomized and site-directed approaches using phage display.
  • Davies and Riechmann [17] and others see WO92/01047) incorporated some of the features of V H domains from camelid heavy chain-only antibodies in combination with phage display to improve solubility whilst maintaining binding specificity.
  • V H domains may be engineered for improved solubility characteristics [19, 20] or solubility may be acquired by natural selection in vivo [21].
  • V H binding domains have been derived from phage libraries, intrinsic affinities for antigen remain in the low micromolar to high nanomolar range, in spite of the application of affinity improvement strategies involving, for example, affinity hot spot randomisation [22].
  • Human V H or camelid V H domains produced by phage or alternative display technology lack the advantage of improved characteristics as a result of somatic mutations and the additional diversity provided by D and J gene segment recombination in the CDR3 region of the normal antibody binding site.
  • Some camelid V H(VHH) domains whilst showing benefits in solubility relative to human V H , may prove antigenic in man and, moreover, suffer the disadvantage that camelid V H must be generated by immunisation of camelids or by phage display technology.
  • Phage-derived human V H regions are laborious to use since they require many rounds of panning and subsequent mutagenesis in order to achieve high affinity binding characteristics.
  • Camelid V HH domains require the same laborious procedure when isolated from phage or similar display libraries or require the immunization of large animals (llama or camels which also make classical antibodies) not amenable to classic hybridoma technology.
  • camelid binding domains may prove antigenic and require humanization.
  • the present inventors have surprisingly overcome the limitations of the prior art and shown that the repertoire of antibody response can be greatly increased by increasing the number of heavy chain-only loci present in a transgenic non-human mammal used to produce class-specific, heavy chain-only antibodies.
  • the invention relies on the discovery that, where a transgenic non-human mammal possesses multiple heavy chain-only loci, these loci are subject to allelic exclusion. Therefore, only one locus is stochastically chosen and recombined successfully, resulting in the production of a heavy chain-only antibody. Multiple V H heavy chain loci can, therefore, be used in the same transgenic non-human mammal to maximise the antibody repertoire and diversity obtainable from the mammal. When antigenically challenged, the transgenic non-human mammal “selects” the locus comprising the V gene segment which is best suited to respond to the specific antigen challenge to the exclusion of the remaining loci.
  • Heavy chain-only antibodies that can be generated by the methods of the invention show high binding affinity as a result of the transgenic non-human mammal being able to “choose” from a range of loci, from which V, D and J gene segment rearrangements and somatic mutations can occur, generally in the absence of an enlarged CDR3 loop.
  • Essentially normal B-cell maturation is observed with high levels of heavy chain-only antibody present in isolated plasma (provided that the C H 1 domain has been eliminated from all antibody classes present in the recombinant locus).
  • B-cell maturation and the secretion of assembled dimers (e.g. IgG) or multimers (e.g. IgM) has no dependency on the presence or expression of light chain genes.
  • a method for the production of a V H heavy chain-only antibody in a transgenic non-human mammal comprising the step of providing more than one heterologous V H heavy chain locus in that mammal, wherein each V H heavy chain locus comprises one or more V gene segments, one or more D gene segments, one or more J gene segments and a gene segment encoding a heavy chain constant region which, when expressed, does not include a C H 1 domain and expressing a V H heavy chain-only antibody from at least one of said loci.
  • each V H heavy chain locus comprises one or multiple V gene segments, e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 45, 50 or 60 V gene segments, which may be derived from any vertebrate species.
  • each locus may comprise only one V gene segment.
  • each V gene segment is different from all other V gene segments.
  • each V gene segment is identical to all the other V gene segments.
  • the remaining gene segments in each locus may be the same as or may be different from those in all the other loci.
  • the non-human mammal may contain multiple copies of a single V H heavy chain locus. This has the advantage of optimising the chances that a productive re-arrangement in a B cell will take place, thus allowing the production of a useful heavy chain-only antibody.
  • the non-human mammal contains a number of different V H heavy chain loci, this will further optimise the chances of obtaining a heavy chain-only antibody with a desired specificity.
  • each locus comprises multiple V gene segments.
  • the V gene segments in any one locus may all be derived from an organism of the same species, e.g. all V gene segments may be of human origin.
  • the V gene segments in any one locus may be derived from organisms of different species, e.g. some V gene segments from human and others from camelids or from sharks.
  • the V gene segments are of human origin.
  • V gene segment encompasses any naturally occurring V gene segment derived from a vertebrate, including camelids and human.
  • the V gene segment must be capable of recombining with a D gene segment, a J gene segment and a gene segment encoding a heavy chain constant (effector) region (which may comprise several exons but excludes a C H 1 exon) to generate a V H heavy chain-only antibody when the nucleic acid is expressed.
  • a heavy chain constant (effector) region which may comprise several exons but excludes a C H 1 exon
  • a V gene segment also includes within its scope any gene sequence encoding a homologue, derivative or protein fragment which is capable of recombining with a D gene segment, a J gene segment and a gene segment encoding a heavy chain constant region (comprising one or more exons but not a C H 1 exon) to generate a heavy chain-only antibody as defined herein.
  • a V gene segment may for example be derived from a T-cell receptor locus or an immunoglobulin light chain locus.
  • the multiple heavy chain loci of the invention comprise any number or combination of the 39 functional human V gene segments and engineered variants thereof with improved solubility properties distributed across the multiple loci. These may be on any number of loci, e.g. four loci comprising eight V gene segments plus one locus comprising seven V gene segments; seven loci comprising four V gene segments plus one locus comprising three V gene segments; or thirty-nine loci comprising one V gene segment each.
  • Human V genes are classified into seven families, V H 1 to V H 7, and the individual genes within each family numbered. The frequency at which each gene is used is dependent on the varying requirements of the particular immune response. For example, the genes of family V H 3 may be preferentially used in comparison to those of family V H 5 when responding to bacterial antigens. Therefore, in a further preferred embodiment of the invention, groups of V gene segments which have been shown to be useful for generating an antibody response against specific antigens are grouped into separate lines of transgenic non-human mammals. The V gene segments may be grouped according to family or they may be grouped according to individual function.
  • V genes of family V H 3 are shown to be useful for generating an immune response against bacterial antigens, then these may be used to generate a transgenic non-human mammal which is particularly useful for generating heavy chain-only antibodies against bacterial antigens.
  • V H 3 and V H 5 are useful for generating an immune response against bacterial antigens, then these may be grouped together and used to generate a transgenic non-human mammal which is particularly useful for generating heavy chain-only antibodies against bacterial antigens.
  • heterologous means a nucleotide sequence or a locus as herein described which is not endogenous to the mammal in which it is located.
  • V H heavy chain locus in the context of the present invention relates to a minimal micro-locus encoding a V H domain comprising one or more V gene segments, one or more D gene segments and one or more J gene segments, operationally linked to one or more gene segments encoding heavy chain effector regions (each devoid of a C H 1 domain).
  • the primary source of antibody repertoire variability is the CDR3 region formed by the selection of V, D and J gene segments and by the V-D and D-J junctions.
  • the advantage of the present invention is that antibody repertoire and diversity obtained in the rearranged V H gene sequences can be maximised through the use of multiple V H heavy chain loci in the same transgenic non-human mammal.
  • Janssens et al., 2006 [15] have shown that a transgenic locus as described above behaves like a normal immunoglobulin locus in terms of rearrangement and allelic exclusion. This opens up the possibility to have multiple loci in the same animal (on different chromosomes) to maximize the number of possible V H recombinations by exploiting allelic exclusion.
  • Each of the transgenic loci would contain from one to more than forty V H regions.
  • allelic exclusion which randomly chooses one of the loci to start recombination, followed by the next locus if the first recombination was non-productive, etc. until a productive recombination has been produced from one of the loci, would ensure that actually all the V H regions present in the combined loci would be part of the overall recombination process.
  • a number of heavy chain loci will first be introduced separately in a transgenic non-human mammal, generating transgenic non-human mammals with a single heavy chain locus. These animals will then be crossed to generate progeny with multiple heavy chain loci to maximize the number of V H regions, resulting in maximum diversity. Loci can also be added through a new round of transgenesis. New loci would be injected into eggs derived from non-human mammals already comprising one or more heterologous VH heavy chain locus. Stable integration of new heterologous VH heavy loci would result in an increase in available VH regions and hence diversity.
  • ES cells derived from non-human transgenic mammals comprising a heterologous VH heavy chain locus, with additional heterologous VH heavy chain loci, provides an alternative route to increase diversity in non-human mammals (eg mice) where ES cell technology can be used for transgenesis by embryo fusion and blastocyst injection
  • each different heavy chain locus will be present as a single copy in the genome of the transgenic non-human mammal.
  • V, D and J gene segments provides a further increase in the antibody repertoire and diversity obtainable. Subsequent somatic mutation is achieved whilst using a minimal locus (micro-locus) without the need for the V L and L C (light chain) antibody loci.
  • D gene segment and J gene segment also include within their scope derivatives, homologues and fragments thereof as long as the resultant segment can recombine with the remaining components of a heavy chain antibody locus as herein described to generate a heavy chain-only antibody as herein described.
  • D and J gene segments may be derived from naturally occurring sources or they may be synthesised using methods familiar to those skilled in the art and described herein.
  • the V, D and J gene segments are capable of recombination and preferably undergo somatic mutation.
  • the D and J gene segments are preferably derived from a single vertebrate species. This may be any vertebrate species but is preferably a human.
  • each V H heavy chain locus comprises from one to forty (2, 3, 4, 5, 6, 7, 8, 9, 10, 12, 14, 16, 18, 20, 30 or 40) or more D gene segments.
  • the D gene segments may be derived from any vertebrate species but, most preferably, the D gene segments are human D gene segments (normally 25 functional D gene segments).
  • each V H heavy chain locus comprises from one to twenty (2, 3, 4, 5, 6, 7, 8, 9, 10, 12, 14, 16, 18 or 20) or more J gene segments.
  • the J gene segments may be derived from any vertebrate species but, most preferably, the J gene segments are human J gene segments (normally 6 (six) J gene segments).
  • Each V H heavy chain locus may contain the same D and J gene segments. Alternatively, each V H heavy chain locus may contain different combinations of D and J gene segments. For example, where each V H heavy chain locus contains only one V gene segment and this segment is identical in each locus, it is then advantageous to use different combinations of D and J gene segments in each locus to further optimise the chances of obtaining a productive re-arrangement. However, where each V H heavy chain locus contains one or more different V gene segments, it may be advantageous to use the same combination of D and J gene segments in each locus.
  • the V H heavy chain locus comprises one or more V gene segments, twenty-five functional human D gene segments and six human J gene segments.
  • Each gene segment encoding a heavy chain constant region may comprise one or more heavy chain constant region exons of the C ⁇ , C ⁇ 1-4 , C ⁇ , C ⁇ , C ⁇ 1-2 class, with the proviso that the heavy chain constant region gene segments do not express a C H 1 domain.
  • the heavy chain constant region gene segments are selected depending on the preferred class or mixture of antibody classes required.
  • the heterologous heavy chain locus is C ⁇ - and C ⁇ -deficient.
  • Each locus may contain the same constant region-encoding gene segment(s) or each locus may have different or a different combination of constant region-encoding gene segment(s).
  • a heavy chain constant region is encoded by a naturally occurring or engineered gene segment that is capable of recombining with a V gene segment, a D gene segment and a J gene segment in a B cell.
  • the constant region is expressed without a C H 1 domain so that generation of heavy chain-only antibody can occur.
  • the C H 1 exons may be deleted so that the constant region of the V H heavy chain-only antibody as described above does not contain a functional C H 1 domain.
  • class-specific heavy chain constant regions when engineering multivalent binding complexes provides the therapeutic benefits of effector function in vivo dependent on the functionality required, Engineering of individual effector regions can also result in the addition or deletion of functionality [23].
  • IgA constant region functionality would provide improved mucosal function against pathogens [24], whilst the presence of IgG1 constant region functionality provides enhanced serum stability in vivo.
  • the presence of heavy chain C H 2 and C H 3 constant domains provides the basis for stable dimerisation as seen in natural antibodies and provides recognition sites for post-translational glycosylation.
  • the presence of C H 2 and C H 3 also allows for secondary antibody recognition when bispecific and multivalent complexes are used as reagents and diagnostics.
  • IgM antibodies are known to play an important role in the activation of macrophages and of the complement pathway. Due to the close proximity of its binding sites, IgM has a high avidity for pathogens, including viruses. However, IgM is also known to be difficult for use in rapid immunoassay techniques, whereas IgG antibodies can be readily used in these techniques. For such uses, it would be useful to select for the preferred antibody class, i.e. IgG or IgM.
  • heterologous heavy chain C ⁇ locus devoid of C H 1 will produce optionally some or all IgG isotypes, dependent on the IgG1, IgG2, IgG3 and IgG4 isotypes present in the heterologous IgG locus.
  • the heavy chains may comprise C ⁇ genes.
  • the resulting IgE molecule might also be used in therapy.
  • IgA and IgM may be obtained when the heavy chain constant region comprises a C ⁇ and a C ⁇ gene.
  • the heavy chain constant region is of human origin, in particular when the heavy chain-only antibody is to be used for therapeutic applications in humans.
  • the heavy chain constant region is preferably derived from the target organism, vertebrate or mammal in or on which veterinary therapy is to be performed.
  • each heavy chain constant region lacks a C H 1 domain.
  • the C H 1 exon is deleted.
  • C ⁇ and C ⁇ constant regions may be mutated, deleted or substituted. The presence, for example, of IgM with a functional C H 1 domain inhibits B-cell maturation and consequently limits the productive expression of heavy chain-only IgG (devoid of C H 1) within the same locus.
  • C H exon a ‘heavy chain constant region exon’ as herein defined includes the sequences of naturally occurring vertebrate, but especially mammalian, C H exons. This varies in a class specific manner. For example, IgG and IgA are naturally devoid of a C H 4 domain.
  • C H exon also includes within its scope derivatives, homologues and fragments thereof in so far as the C H exon is able to form a functional heavy chain-only antibody as herein defined when it is a component of a heavy chain constant region.
  • Heavy chain effector molecules may be engineered to be free of functional domains, for example the carboxy-terminal C H 4 domains, provided that engineering does not affect secretory mechanisms preventing cell surface assembly and consequently B-cell maturation. Additional features may be engineered into the locus, for example to improve glycosylation or add function.
  • the heterologous heavy chain locus is designed to produce preferred classes or mixtures of heavy chain-only antibodies depending on the antibody class(es) required, with essentially normal B-cell maturation.
  • camelid V, D and J gene segments and camelid effector regions will produce camelid antibodies with features peculiar to camelid antibodies, such as enlarged CDR3 loops.
  • human V, D and J gene segments will produce human heavy chain-only antibodies lacking the enlarged CDR3 loop.
  • V H domain in the context of the present invention refers to an expression product of a V gene segment when recombined with a D gene segment and a J gene segment as defined above.
  • the V H domain has improved ability to bind antigen as a result of VDJ recombination and somatic mutation. There is no dependency on the presence or absence of the enlarged CDR3 loop peculiar to the camelid species.
  • the V H domain is able to bind antigen as a monomer. Any likelihood of combining with a V L domain when expressed as part of a soluble heavy chain-only antibody complex has been eliminated by removal of the C H 1 exon (see [25]).
  • V H domain alone can also be engineered with diverse protein domains to produce fusion proteins for targeted therapeutic and diagnostic purposes, for example with toxins, enzymes and imaging agents.
  • V H domain coding sequences may be derived from a naturally occurring source or they may be synthesised using methods familiar to those skilled in the art.
  • V H domain may be altered or improved by selecting or engineering V, D and/or J gene segments which encode sequences with the required characteristics. As indicated above, some of the 39 functional human V H regions may not be suitable for the production of heavy chain-only antibodies.
  • V H region characteristics Dolk et al., [30] used phage display techniques to generate heavy chain-only antibodies showing improved stability in the harsh conditions associated with anti-dandruff shampoo.
  • the transgenic non-human mammal is preferably a rodent such as a rabbit, guinea pig, rat or mouse. Mice are especially preferred. Alternative mammals such as goats, sheep, cats, dogs or other animals may also be employed. Preferably, the mammal is a mouse.
  • transgenic non-human animals are generated using established oocyte injection technology alone. Where established, ES cell technology or cloning may also be used.
  • antibody heavy and optionally light chain loci endogenous to the mammal are deleted or silenced when a heavy chain-only antibody is expressed according to the methods of the invention.
  • the methods of generating heavy chain-only antibodies as described in the above aspects of the invention may be of particular use in the generation of antibodies for human therapeutic use, as often the administration of antibodies to a species of vertebrate which is of different origin from the source of the antibodies results in the onset of an immune response against those administered antibodies.
  • the antibodies produced according to the invention have the advantage over those of the prior art in that they are of substantially a single or known class and preferably of human origin.
  • Antibodies are of high affinity resulting from a combination of VDJ recombination and affinity maturation in vivo.
  • a further aspect of the invention provides a transgenic non-human mammal comprising more than one heterologous V H heavy chain locus as defined above.
  • the transgenic non-human mammal may be engineered to have a reduced capacity to produce antibodies that include light chains.
  • Antibody-producing cells may be derived from transgenic non-human mammals as defined herein and used, for example, in the preparation of hybridomas for the production of heavy chain-only antibodies as herein defined.
  • nucleic acid sequences may be isolated from these transgenic non-human mammals and used to produce V H domain heavy chain-only chain antibodies or bi-specific/bi-functional complexes thereof, using recombinant DNA techniques which are familiar to those skilled in the art.
  • antigen-specific heavy chain-only antibodies may be generated by immunisation of a transgenic non-human mammal as defined herein.
  • the invention also provides a method for the production of heavy chain-only antibodies by immunising a transgenic non-human mammal as defined above with an antigen.
  • Antibodies and fragments thereof may be may be isolated, characterised and manufactured using well-established methods known to those skilled in the art. These antibodies are of particularly use in the methods described in PCT/GB2005/00292.
  • FIG. 1 Schematic representation of the DNA fragments used to generate the transgenic mice.
  • Two of the llama V HH exons are linked to the human heavy chain diversity (D) and joining (J) gene segments, followed by the C ⁇ , C ⁇ , C ⁇ 2 and C ⁇ 3 human constant region genes and human heavy chain Ig 3′ LCR.
  • Modifications of human C ⁇ 2 and C ⁇ 3 genes were a complete deletion of the CH1 exon from C ⁇ 2 and C ⁇ 3 genes in constructs MG ⁇ and G ⁇ or also from C ⁇ in construct M ⁇ G ⁇ .
  • the presence of two Lox P sites (in red) in the same direction enables the removal of C ⁇ and C ⁇ genes upon Cre mediated recombination.
  • the presence of the Frt site (in green) enables the generation of a single copy transgenic mouse from a multi-copy transgene array by Flp mediated recombination.
  • FIG. 2 Panel A: Table of the flow cytometric analysis of the B lymphocytes expressed as the percentage of B220/CD19 positive cells of total cells in the different organs.
  • Panel B Flow cytometric analysis of B cell populations of wt, ⁇ MT, MG ⁇ / ⁇ MT, M ⁇ G ⁇ / ⁇ MT and G ⁇ / ⁇ MT mice in the BM. Lymphoid cells were gated on the basis of forward and side scatter and surface expression of B220 and human IgM or IgG is plotted. Data are displayed as dot plots.
  • FIG. 3 Panel A-E: DNA FISH of a five copy human G ⁇ locus.
  • Panel A Stretched chromatin fiber from lung cells of G ⁇ line1 transgenic mouse carrying five intact copies (1-5) of the G ⁇ locus, flanked by half a locus containing the LCR (red) and half a locus carrying VHH to J region (green).
  • Panel B Stretched chromatin fiber FISH of a hybridoma (G20) derived from G ⁇ line1 B cells where one copy has rearranged (white arrow).
  • Panel C Non stretched DNA FISH of hybridoma T1 with the LCR probe (red).
  • Panel D Same as C with a probe between VHH and D (green).
  • Panel E overlay of panels C and E.
  • T1 has four rearrangements visible as the loss of 4 green signals compared to no loss of red signals.
  • Panel F Allelic exclusion in G ⁇ transgenic mice is preserved. Flow cytometric analysis of murine surface or intracellular (ic) ⁇ H chain and transgenic human IgG on total BM CD19+ cell fractions from the indicated mice. Data are displayed as dot plots and the percentages of cells within the indicated quadrants are given. Data shown are representative of four mice examined within each group.
  • FIG. 4 Analysis of B cell populations in the spleen of wt, ⁇ MT, G ⁇ , M ⁇ G ⁇ and MG ⁇ mice. Data shown are representative of 4-8 mice examined within each group.
  • Panel A Top, FACS data of spleen cells, stained for mouse IgM, human IgG, human IgM versus B220.
  • Bottom flow cytometric analysis of B cell populations in the spleen. Lymphoid cells were gated on the basis of forward and side scatter and surface expression of B220 and the indicated Ig (upper part) or the CD21/CD23 profile is displayed as dot plots and the percentages of cells within the indicated gates are given.
  • CD21 low CD23 low immature B cells
  • CD21+CD23+ follicular B cells
  • CD21 high CD23 low marginal zone B cells.
  • Panel B Histology of the spleen of wt, ⁇ MT, G ⁇ / ⁇ MT, M ⁇ G ⁇ / ⁇ MT and MG ⁇ / ⁇ MT mice. Immunohistochemical analysis; 5 ⁇ m frozen sections were stained with anti B220 (blue) for B cells and anti-CD11c/N418 (brown) for dendritic cells. Arrows indicate the location of small clusters of B cells in the MG ⁇ spleens.
  • Panel C Sequence alignment of the PCR products obtained from Payer's patches cDNA using VHH1 and VHH2 specific primers in combination with human C ⁇ 2 primer, showing that the transgenic locus undergoes hypermutation in the CDR1 and 2 regions. Sequences are from the transgenic locus G ⁇ with a CH1 deletion.
  • Panel D Top; FACS data of spleen cells, stained with anti-CD19 and anti-B220. Bottom left: Schematic representation of Flp recombination in vivo by breeding to FlpeR transgenic line and FACS data on spleen cells of the single copy recombinant derived from the five copy G ⁇ line 1.
  • FIG. 5 Southern blots showing the absence of the ic light chain rearrangement in G ⁇ / ⁇ MT (panel A) and M ⁇ G ⁇ / ⁇ MT (panel B) transgenic lines.
  • Liver DNA (L) and B cell DNA (B) from a wt mouse and two G ⁇ or four M ⁇ G ⁇ transgenic mice was Hind III digested and probed with the 32 P radiolabeled J ⁇ probe and the carbonic anhydrase II (CAII) probe.
  • the CAII probe which hybridizes to a 4 kb band was used as a loading control.
  • Liver DNA was run to show the ⁇ germline configuration (2.8 Kb band). Only the wt B cells show ic locus rearrangement measured as a decrease in intensity of the 2.8 kb fragment (30% of signal left when compared to liver).
  • FIG. 6 Prot G or concavalin purified serum samples of 6 different G ⁇ lines (A, B), 4 different M ⁇ G ⁇ lines (C) and 2 different MG ⁇ lines (E-G), in the ⁇ MT background run under non-reducing (A) and reducing conditions (B-G).
  • the size of the transgenic human IgG (panels B, F) and IgM (panel C, D) is consistent with the CH1 deletion and the absence of light chains. Mouse ic light chains were normal size (G). Human serum was used as a positive control.
  • Panel D Superose 6 size fractionation of M ⁇ G ⁇ serum aftermixing in a human IgM control under non-reducing conditions. Each fraction was analysed by gel electrophoresis under reducing conditions.
  • Fractions collected from the Superose 6 column are from left (high MW) to right (low MW).
  • the controls are human serum alone (first lane left) and mouse serum before mixing in the human IgM control serum (lane M ⁇ G ⁇ serum). Size markers are indicated.
  • FIG. 7 Panel A: Sequences of monoclonal antibody cDNAs specific for tetanus toxoid; HSP70, rtTA and human TNF ⁇ . The top sequence is the germline VHH2 sequence. The CDR 1, 2 and 3 and hinge regions are indicated above the sequence. Different isotypes and classes are indicated by different colors on the right. The J regions that are used are indicated on the right.
  • Panel B Examples of western blots using the different heavy chain-only antibodies (hybridomas, sera and sdAb). Left panel anti-rtTA serum and hybridoma medium, diluted 1/100 and 1/250 respectively. Middle panel, anti DKTP serum from wt and G ⁇ mice diluted 1/200 and 1/100 respectively.
  • Panels C and D Immunostaining of one of Tet ⁇ on cell lines additionally transfected with a marker plasmid that responds to the presence of rtTA by expressing a marker protein in the cytoplasms 51 .
  • Panel C shows nuclei expressing rtTA (green).
  • Panel D shows doxycycline induced expression of the marker protein in the cytoplasm (red) in response to rtTA and nuclear staining of the cells with DAPI (blue).
  • Panel E Example of BiaCore analysis of the anti rtTA antibody. Affinity is indicated.
  • FIG. 8 Panel A: Schematic drawing of the Ig loci with camelid like splice mutations.
  • the two human IgG constant regions C ⁇ 2 and C ⁇ 3 were first mutated by altering the splice G +1 to A +1 , thought to result in CH1 exon skipping in camelid IgG HCAbs 5 depicted as C ⁇ 2-S and G ⁇ 3-S.
  • the locus contained two llama VHH regions all of the human D and JH regions and human C ⁇ , C ⁇ and C ⁇ 2 and C ⁇ 3 and the LCR (see also main text). These loci were introduced into ⁇ MT transgenic mice and analysed for the expression of the human loci.
  • Panel B Sequencing of bone marrow (BM) human IgG cDNA from the GS mice showed that both VHHs recombined with different human D and J segments and were transcribed with the C ⁇ 2 constant region. However, the CH1 exon was still present, save the last 16 bp, which were spliced out. While in progress, the same cryptic splice site in the CH1 exon was reported in a leukemia patient due to an A to G transition in position 4 of intron 1 [31]. None of the MGS or GS lines rescued B cell development (not shown) in ⁇ MT mice. Although the mouse B cell transcriptional/translational machinery can process rearranged dromedary VHH- ⁇ 2a [34, 60] our data show that in addition to the G to A mutation, other features are important for CH1 exon skipping.
  • BM bone marrow
  • FIG. 9 Schematic representation of the cloning of human V H regions onto the various loci as described in Examples 3 and 4
  • FIGS. 10 and 11 Examples of heavy chain loci containing multiple V H gene segments, the entire D region, the entire JH region, the C ⁇ 2, C ⁇ 3 and Cx regions and the 3′ LCR.
  • FIG. 12 Shows a karyogram with one locus integrated on chromosome 1 and one locus on chromosome 8.
  • a Heavy Chain-Only Antibody Locus is Fully Functional in Mice and Sensitive to Allelic Exclusion
  • Janssens et al. [15] have developed methods for the derivation of heavy chain-only antibodies in transgenic mice. For further details of the methods and experiments described herein, the skilled person should refer to Janssens et al. [15], which is incorporated herein by reference. 1
  • a genomic cosmid library was made from peripheral blood cells of Lama Glama using standard methods. Two different germline VHHs were chosen based on their sequence, an open reading frame without stop codon and the presence of hydrophilic amino acid codons at positions 42, 50 and 52 according to Lefranc numbering [32] and one with and one without a hydrophilic amino acid at position 49. One is identical to IGHV1S1 (acc.num. AF305944) and the other has 94% identity with IGHV1S3 (acc. num.AF305946).
  • clones Two clones were selected from the human genomic Pac library RPCI-11 (BACPAC Resource Center, USA): clone 1065 N8 containing human heavy chain D and J regions, C ⁇ and C ⁇ and clone 1115 N15 containing the C ⁇ 3 gene.
  • Bac clone 11771 from a different human genomic library (Incyte Genomics, Calif., USA) was used as a source of C ⁇ 2 gene and the Ig heavy chain LCR [33].
  • the ⁇ C ⁇ 3 and C ⁇ 2 genes were subcloned separately into pFastBac (Invitrogen). The single point mutation (G to A) [34] or a complete deletion of CH1 exon was achieved by homologous recombination [35].
  • frt and lox P sites were introduced in front of the C ⁇ switch region and a second lox P site was placed in front of the C ⁇ 2 switch region, resulting in MGS or MG ⁇ .
  • the MGS or MG ⁇ vector ( FIG. 1 ), containing two llama VHH genes, followed by human D and J heavy chain regions, C ⁇ , C ⁇ and the modified human C ⁇ 2 and C ⁇ 3 genes and 3′ LCR, was transformed into 16 294 Cre E. coli strain 44 yielding the GS or G ⁇ locus through cre mediated recombination ( FIG. 1 ).
  • M ⁇ G ⁇ was obtained from MG ⁇ by deletion of the C ⁇ CH1 region through homologous recombination.
  • the 220 Kb MGS or MG ⁇ or M ⁇ G ⁇ fragments, 150 Kb GS or G ⁇ fragments ( FIG. 1 ) were purified from vector sequences and injected into pronuclei of fertilized FVB X B16/ ⁇ MT ⁇ / ⁇ eggs at a concentration of 2 ng/ ⁇ l.
  • Transgenic loci were checked for integrity and number of copies by Southern blot analysis of tail DNA using 5′ and 3′ end probes.
  • Transgenic ⁇ MT+/ ⁇ founders were bred as lines in the ⁇ MT ⁇ / ⁇ background. Genotyping was done by PCR (30 cycles with denaturation at 94° C. for 45 s, annealing at 60° C. for 30 s and extension at 72° C.
  • Single cell suspensions were prepared from lymphoid organs in PBS, as described previously 45 . Approximately 1 ⁇ 10 6 cells were incubated with antibodies in PBS/0.5% bovine serum albumin (BSA) in 96 well plates for 30 min at 4° C. Cells were washed twice in PBS/0.5% BSA. For each sample, 3 ⁇ 10 4 events were scored using a FACScan analyzer (Becton Dickinson, Sunnyvale, Calif.). FACS data were analyzed using CellQuest version 1.0 computer software. Four-color analysis was done on a Becton Dickinson FACS Calibur. Most antibodies used have been described [36]; FITC conjugated anti-human IgG and anti human IgM were purchased from Sigma (Zwijndrecht, NL).
  • BSA bovine serum albumin
  • Single cell suspensions were made from spleens and livers of wt mice M ⁇ G ⁇ and G ⁇ transgenic mice.
  • B cells were positively selected using MACS CD45 (B220) MicroBeads (Miltenyi Biotec, Germany) on an Automacs separator according to the manufacturer's instructions to ⁇ 90% purity [37].
  • Genomic DNA was Hind III digested and blotted onto Hybond nylon filters. The filter was hybridized with a 32 P radiolabeled J ⁇ probe (obtained by PCR amplification from genomic DNA over J ⁇ 1 and J ⁇ 5 regions) and 32 P radiolabeled carbonic anhydrase II (CAII) probe. Liver DNA was run to show the signals of the ⁇ germline configuration (2.8 Kb band).
  • the CAII probe which hybridizes to a 4 kb band was used as a loading control. Filters were scanned on a Tyfoon 9200 (Amerscham Biosciences). The intensity of germ line J ⁇ band was quantified using Image Quant 5.2 software, normalized to that of the loading control, and expressed as a percentage of the control liver DNA. (which is 100%).
  • PCR primers used on genomic DNA from hybridomas for amplification of different rearrangement events were as follows:
  • Target DNA Monoclonal hybridoma cells were cultured in DMEM/10% FCS and embedded in agarose as described by Heiskanen [38]. Lungs from a mouse of the G ⁇ transgenic line 1 were collected and a single cell suspension was made. The embedded cells were treated with proteinase K and Rnase H. Mechanically extended DNA was prepared on poly-L-lysine slides (Sigma) using a microwave oven and the edge of another slide.
  • Probes To detect rearranged and non-rearranged copies of the G ⁇ transgene, DNA fragments were purified. A 2.3 kB SpeI and a 3.6 kB SpeI-BssHII fragment for hybridizing the region between VHH and D, and a 5.9 kB BamHI-SpeI fragment or the low copy Bluescript plasmid containing the IgH 3′LCR (16 kB) for LCR detection.
  • the probes were labeled by nick-translation with biotin-11-dUTP (Roche) or digoxigenin-11-dUTP (Roche). Prior to pipetting the probes on the slides, they are denatured by for 5 minutes at 90° C., 5 minutes on ice, and 45 minutes at 37° C.
  • the hybridization mixture contained 50% formamide, 2 ⁇ SSC, salmon sperm DNA (200 ng/ ⁇ l), 5 ⁇ Denhardt's, 1 mM EDTA, and 50 mM sodium phosphate, pH 7.0.
  • Hybridization of the probes is done by pipetting 25 ⁇ l mixture onto the slides, and covering with a 24 ⁇ 32-mm cover slip. To denature the probes and target sequences the slides were put on an 80° C. heating plate for 2 minutes. The probes hybridize overnight at 37° C. in a humidified chamber (humidifier is 50% formamide, 2 ⁇ SSC). Post hybridization washes were performed as described [39].
  • the digoxigenin probe was detected with sheep-anti-digoxigenin (1:500, Sigma), fluorescein-conjugated rabbit-antisheep (1:500, Sigma), and fluorescein-conjugated goat-anti-rabbit (1:500, Sigma).
  • the biotin probe was detected with Texas Red-conjugated avidin (1:500, Sigma) and biotinconjugated goat-anti-avidin (1:500, Boehringer). This step was repeated twice. All incubations and washes were performed as described 48 . After staining the slides were dehydrated in a graded series of ethanol (70, 90, and 100%) 5 minutes each step at room temperature. Cells or DNA was embedded in 25 ⁇ l anti-fading embedding medium Vectashield (Vector Laboratories). Visualization was done with a Leica DMRBE fluorescent microscope using a 100 ⁇ objective.
  • mice 8 weeks old mice were immunized with 5-20 ⁇ g of antigen with Specol adjuvant (IDDLO, Lelystadt, NL) or with preformulated DKTP vaccine s.c. on days 0, 14, 28, 42 and i.p. on day 50. Blood was taken on day 0, 14 and 45. Spleen cells were fused with Sp2-O-Ag14 myeloma cells line (gift from R. Haperen) on day 56 using a ClonalCellTMHY kit (StemCell Technologies, Canada) according to the manufacturer's instructions. DKTP vaccine was obtained from the Netherlands Vaccine Institute (Bilthoven, NL).
  • VHHDJ fragments were amplified by PCR using specific primers: vh1 back Sfi I primer [40-42] in combination with hIgG2hingrev primer (5′-AATCTGGGCAGCGGCCGCCTCGACACAACATTTGCGCTC-3′(SEQ ID NO:15)) or CH2huIgMrev primer (5′-TGGGACGAAGACGGCCGCTTTGGGAGGCAGCTCGGCAAT-3′ (SEQ ID NO:16)).
  • the amplified VHHDJs ( ⁇ 400 bp) were Sfi I/Not I digested, gel purified and cloned into Sfi I/NotI digested phagemid pHEN derived vector.
  • Transformation into TG1 electro-competent cells yielded in a human single domain antibody library. Two rounds of selection were performed using panning on DKTP vaccine antigens adsorbed onto plastic (immunotubes coated with undiluted vaccine) or purified human TNF ⁇ (Biosource International, USA).
  • VHH1 contained all of these VHH hallmark amino acids, but to test the importance of solubility in this proof of principle experiment, the other, VHH2, lacked one of these critical “solubility” amino acids, a Gln (Q) instead of a Glu (E) at position 49.
  • Gln Q
  • Glu E
  • 49 rather than position 50 (Arg, R)
  • LCR Locus control region
  • MG ⁇ mice were unable to rescue B cell development in a ⁇ MT background, whereas the G ⁇ and M ⁇ G ⁇ constructs efficiently rescued B cell development.
  • the rescue of B220/CD19 positive cells was between 30-100% in the different lymphoid compartments independent of copy number ( FIG. 2A ). This is confirmed by flow cytometry of BM using B220 versus human IgM or human IgG staining ( FIG. 2B ).
  • the M ⁇ G ⁇ mice contain human IgM producing cells in the BM absent in wildtype or ⁇ MT mice. Appropriately these cells have not undergone a class switch as they do not contain human IgG.
  • the G ⁇ mice contain only human IgG expressing B cells.
  • the MG ⁇ mice contain very few if any B cells that express human Ig on the cell surface, but interestingly a proportion of the B220 cells express intracellular IgM, but not IgG ( FIG. 2B ). In contrast to the M ⁇ G ⁇ and G ⁇ mice (see below), the MG ⁇ mice express mouse Ig light chains ( FIG. 6G ). These results show that the C ⁇ and C ⁇ genes in the different constructs are expressed and strongly suggest that the absence of CH1 is crucial for cell surface expression of VHH based antibodies.
  • Pro-B cells express high levels of cytoplasmic SLC, IL-7R and CD43, which are downregulated upon pre-BCR expression and absent in mature B cells ( FIG. 2C , compare pro-B cells from ⁇ MT mice and the surface IgM ⁇ pro-B/pre-B cell fraction and the surface IgM+ B cell fraction of wt mice).
  • the human Ig+ B cells from M ⁇ G ⁇ / ⁇ MT or G ⁇ / ⁇ MT mice have low levels of SLC and IL-7R, indicating that the human single chain IgG and IgM receptors functionally replace the murine pre-BCR in the downregulation of SLC and IL-7R.
  • CD43 this appears to be the case only in G ⁇ mice, but the persistence of CD43 expression in M ⁇ G mice could be related to the finding of increased B-1 B cell differentiation in these mice.
  • CD2 and MHC class II expression is induced, as in normal pre-BCR signalling.
  • the levels of the IL-2R/CD25, transiently induced at the pre-B cell stage, are very low on mature M ⁇ G or G ⁇ / ⁇ MT B cells and comparable to those of mature wt B cells ( FIG. 2C ). Furthermore, ic Ig ⁇ expression was not detectable in mature M ⁇ G or G ⁇ / ⁇ MT B cells ( FIG. 2C ) and was also not induced in in vitro BM cultures upon IL-7 withdrawal after 5 days of IL-7+ culture (not shown).
  • mice The human HCAb expressing B cell populations in M ⁇ G or G ⁇ transgenic mice consisted partially of cells that were generated in the BM (HSA high and AA4.1/CD93 high ), and partially of cells that have matured in the periphery and are recirculating (HSA low and CD93 low ), comparable to findings in normal mice.
  • hybridomas were made from the M ⁇ G ⁇ and G ⁇ lines after immunisation, in particular of the five copy G ⁇ line1 (see below). Sequence analysis showed that more than one rearrangement could take place in the multicopy G ⁇ loci.
  • T1 and T3 express two productive mRNA's, that were confirmed by mass spectrometry of the secreted antibodies exactly matching the cDNA (not shown).
  • Control lung cells showed five complete copies plus half a transgene at either end ( FIG. 3A ) in agreement with Southern blot mapping (not shown), while the hybridomas indeed show one and four rearranged copies in G20 and T1 respectively ( FIGS. 3B , C-E).
  • the G ⁇ line1 mice ( FIG. 2A ) contained 5 copies of the G ⁇ locus and hence there was a possibility that the efficient rescue was related to the copy number of the locus.
  • a single copy transgenic line obtained from the 5 copy G ⁇ line1 by Flp recombination through breeding with a FlpeR line 23 rescued B cell development to a similar extent ( FIG. 4D , FIG. 2A ).
  • Murine Ig light chain proteins were not detected in the M ⁇ G ⁇ and G ⁇ mice by Western blots of serum (not shown, but see FIGS. 2C and 6A ) or by FACS, suggesting that the murine light chain genes do not rearrange. This was confirmed by comparing the densities of the Ig ⁇ locus germline signals in DNA from sorted splenic B220+ cell fractions and liver cells by Southern blot analysis ( FIGS. 5A and B), which shows that the mouse light chains do not rearrange and remain in a germline configuration. In contrast light chains are detected in the few human Ig expressing cells in the MG ⁇ / ⁇ MT mice (See FIG. 6G ).
  • HCAb human HCAb in early B cell development in the BM fails to provide the signal leading to light chain rearrangement.
  • the HCAb mimic a BCR rather than a pre-BCR, which is likely related to their failure to bind pseudo-light chains in the absence of CH1 [61].
  • Human IgM was present in M ⁇ G ⁇ serum and human IgG in both M ⁇ G ⁇ (and G ⁇ mouse serum. In non-immunized adult animals, the human IgM ( ⁇ 50 ⁇ g/ml) and IgG (200-1000 ⁇ g/ml) is present at levels comparable to those seen in normal mice or transgenic mice carrying a normal human IgH locus [65].
  • Gel electrophoresis of the serum of all six G ⁇ mice revealed HCAb IgG's with a MW of ⁇ 70 kD under non-reducing and ⁇ 35 kD under reducing conditions, consistent with heavy chain dimers lacking a light chain and each heavy chain lacking the CH1 exon (FIGS. 6 A,B).
  • the serum of M ⁇ G ⁇ mice contained multimeric heavy chain-only human IgM. Under reducing conditions ( FIG. 6C ) all four lines contained IgM chains with the MW as a human control IgM after subtraction of the MW of CH1.
  • the serum was also fractionated ( FIG. 6D horizontal fractions) under non-reducing conditions after which each fraction 10 was analysed by gel electrophoresis under reducing conditions ( FIG. 6D , vertical lanes).
  • FIG. 6D horizontal fractions
  • M ⁇ G ⁇ mice produce HCAb multimeric IgM and dimeric IgG, while G ⁇ produce dimeric IgG in the serum.
  • mice Human IgM and IgGs were below the detection level in a quantitative ELISA assay, but we nevertheless tested whether the MG ⁇ / ⁇ MT mice can respond to immunization. Mice were immunized with human Tumor Necrosis Factor- ⁇ (TNF- ⁇ ) and wt mice developed a strong TNF- ⁇ specific antibody response, while in the two MG ⁇ line 3 mice used, antigen specific human IgGs could not be detected by ELISA or Western blot analysis (not shown).
  • TNF- ⁇ Tumor Necrosis Factor- ⁇
  • mice were immunized with E. Coli hsp70, DKTP ( Diphteria toxoid, whole cell lysate of Bordetella Pertussis , Tetanus toxoid and inactivated poliovirus types 1, 2 and 3) and rtTA [50], while the M ⁇ G ⁇ mice were immunized with human TNF ⁇ . From mice with positive sera by ELISA, individual complete antibodies were isolated using hybridomas or single domain Ab (sdAb) by phage display libraries.
  • sdAb single domain Ab
  • the ⁇ hsp70-, tetanus toxoid- and rtTA-specific monoclonals were sequenced after RTPCR of the antibody RNAs ( FIG. 7A ). This showed that both IgG2 (7 out of 8) and IgG3 (1 out of 8) antibodies were produced (the sdAb were isolated from a IgG2 library). Different D and J regions were used. Although not at high frequency, the VHH from the 11 HCAbs were hypermutated. The three hTNF ⁇ -specific antibodies (one positive IgM hybridoma, FIG. 6 ⁇ -hTNF ⁇ #1 and two sdAb ⁇ -hTNF ⁇ #2 & 3) all had different hypermutations in the CDR2 region.
  • mice expressing a HCD-like human ⁇ protein develop normal CD43-pre-B cells in a SCID background independent of ⁇ 5 [54].
  • the truncated C ⁇ proteins are expressed on the B cell surface without associated L chains and are thought to mimic pre-BCR signaling through self-aggregation [55].
  • BiP chaperones the folding and assembly of antibody molecules by binding to hydrophobic surfaces of the Ig chains that subsequently participate in inter-chain contacts 31 .
  • hydrophilic amino acids in FR2 of VHHs most probably prevents BiP binding to VHH, which needs no (surrogate) light chain to become soluble.
  • CH1 provides the interaction with BiP proposed to hold heavy chains in the ER until assembly (replacement of BiP by a light chain) is complete.
  • the 5 copy G ⁇ line1 rescues B cell development to the same extent as the single copy line integrated at the same position in the genome.
  • one or more rearrangements occur in multicopy transgenic loci ( FIG. 3 ).
  • Two of the hybridomas, originating from two single splenocytes gave two productive HCAb transcripts and proteins. This result confirms that expression of two antibodies in the same B cell is not toxic [63].
  • the prediction 37 that double antibody producing B cells would loose in competition with single antibody producing cells under antigen challenge is not borne out by our result of finding 2 double antibody expressing cells out of 5 hybridomas obtained after antigen challenge.
  • the (multicopy) locus is subject to allelic exclusion in wildtype mice, because BM cells express either mouse or human Ig on the cell surface. There is no significant population of cells expressing both on the cell surface ( FIG. 3F , top panels). Perhaps most interesting is the number of mouse versus human Ig expressing cells. In a wt/5 copy G ⁇ mouse there are three possible alleles available for rearrangement, two mouse alleles with one Ig locus and one allele with five human HCAb loci. If chosen stochastically, a human allele would be chosen only 1 out of 3 times.
  • mice open up completely new possibilities for the production of human HCAbs for clinical or other purposes, particularly in light of the evidence 4 that HCAbs may recognize epitopes that are barely antigenic for conventional antibodies, such as active sites of enzymes.
  • the restricted number of variable regions may explain why not all of the antigens were recognized; the polio and Diphteria proteins gave no response in G ⁇ mice, whereas wt control mice did.
  • all of the antibodies had the llama VHH2 region. This does not include a conserved aminoacid [67] at position 49 in contrast to VHH1 that does have one and should be more soluble.
  • antigen specific high affinity HCAb of potentially any class can be produced in mice.
  • This technology will allow the production of fully human HCAb of any class or fragments thereof in response to antigen challenge for use as therapeutic or diagnostic agents in man.
  • our technology also allows for the production of high affinity matured antibodies from any vertebrate for use as reagents, diagnostics or for the treatment of animals.
  • mice contained the MG ⁇ locus (IgM and IgG locus, Janssens et al., 2006), while the other contained the G ⁇ locus (IgG locus only, Janssens et al., 2006), both mice have the ⁇ MT background.
  • Hybridomas were derived from the B cells from the double transgenic offspring and grown in culture by standard methods. A number of the resulting individual monoclonal cell lines were analysed by PCR and Southern blots, which showed that lines containing a productively rearranged MG ⁇ locus contained a non-rearranged G ⁇ or a non-productively rearranged G ⁇ locus. Conversely cell lines containing a productively rearranged G ⁇ locus contained a non-rearranged or a non-productively rearranged MG ⁇ locus. Thus the sum of the available V H regions is used in the recombination process.
  • all of the number of functional human VH regions is increased by cloning human V H regions (or variants thereof) onto a multiple modified human locus containing the entire D H region, the entire J H region and a combination of the C ⁇ , C ⁇ 2, C ⁇ 3 and C ⁇ regions and the 3′LCR using those methods described in the previous example and known in the art (Janssens et al 2006).
  • This procedure can be carried out using multiple identical V H regions on separate loci or different V H region on separate loci.
  • the different loci can contain identical heavy chain regions or different heavy chain regions.
  • the example 3 is for a locus with identical V H regions on loci that have a different combination of heavy chain regions, example 4 for two loci with identical heavy chain constant regions but different V H regions. Obviously in both examples additional loci could be added.
  • Human V H regions are isolated by PCR amplification of the human genomic DNA using primers that are specific for each selected V H out of the possible 39 functional human V H regions (alternatively human V L regions or TCR V regions or variants of all of these derived by mutagenesis could be used or added).
  • the human V H regions are cloned in sets onto the locus described in the above example ( FIG. 9 ), i.e. comprising the human D H plus J H (or other D and J regions) and C ⁇ , C ⁇ 2, C ⁇ 3 each lacking a CH1 plus 3′ LCR.
  • the C ⁇ region lacking CH1 plus switch regions will be cloned separately in the G ⁇ locus ( FIG. 10 ).
  • This G ⁇ locus is a variant of the original locus in that it does not contain lox sites and the llama V HH regions were removed by standard homologous recombination leaving a unique PI-PspI site ( FIG. 10 ).
  • the functional V H regions may be cloned together, with any multiple on each locus. Initially, 17 functional human V H regions will be cloned together in one set starting with 2 cloned genes per set. To each of these initial constructs, a second set will be added by conventional methodology (e.g. using XhoI-SalI restriction digestion/ligation, ligation of XhoI and SalI compatible sites destroys both). Sets containing 4, 4, 4, 3 and 2 will be linked into one set of 17 genes in a BAC vector. Obviously this procedure could be carried out by combining sets containing other numbers of genes. The above process may be terminated at any point to achieve the desired number of V H regions or extended to achieve a higher number of V H regions.
  • the entire set (e.g. 17 genes) is cloned into the modified G ⁇ locus in the unique PI-PspI site.
  • the C ⁇ region will be cloned into the I-CeuI site of this locus resulting in a A ⁇ G ⁇ locus capable of producing Ig ⁇ and IgG ( FIG. 10 ).
  • other heavy chain regions could be cloned in.
  • mice preferably with a defective mouse IgH locus such as ⁇ MT
  • ⁇ MT defective mouse IgH locus
  • These separate loci are used to generate separate mouse lines, one that would make only IgG and one that would make IgA and/or IgG.
  • These mice are subsequently crossed to bring the total of V H regions available to 34 on two different loci. Crossing these mice to homozygosity for both loci would make 68 V H regions available for recombination. Having multiple copies of an integrated locus would increase this number yet further.
  • hybridomas made from the B cells from these mice would be used to show that when a productively rearranged locus is present, the other loci that were bred in, are either non-rearranged or non-productively rearranged due to allelic exclusion by standard procedures.
  • V H regions similarly isolated to the regions described above (or variants thereof derived by mutagenesis) are cloned onto two separate loci by the same methodology as described above. This would result in two different G ⁇ loci (or variants thereof such as adding C ⁇ ). These loci would be introduced into separate mice resulting in separate transgenic lines (for example 2 loci having 10 VH domains each, FIG. 11 ). These mice would subsequently be crossed to obtain double transgenic mice that would have all of the V H regions used available for the recombination process. Crossing these mice to homozygosity for both loci would double the number of V H regions available for recombination ( FIG.
  • the above process may be terminated at any point to achieve the desired number of V H regions.
  • the D, JH and constant regions will be added to these VH regions.
  • These final loci can then be introduced into separate transgenic mice (preferably with a defective mouse IgH locus) as described in the example above.
  • the position of the lox sites allows the elimination of individual constant regions to generate separate loci that contain or C ⁇ (IgM) alone, or C ⁇ 2 and C ⁇ 3 (IgG2 and IgG3) alone, or C ⁇ alone (IgA) or combinations thereof.
  • These separate loci are used to generate separate mouse lines that would make either human IgM alone, or IgG alone or IgA alone or combinations thereof.
US12/161,981 2006-01-25 2007-01-25 Generation of heavy-chain only antibodies in transgenic animals Abandoned US20090307787A1 (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
GBGB0601511.9A GB0601511D0 (en) 2006-01-25 2006-01-25 Binding Molecules 2
GB0601511.9 2006-01-25
GB0618345.3 2006-09-18
GBGB0618345.3A GB0618345D0 (en) 2006-09-18 2006-09-18 Binding molecules
PCT/IB2007/001491 WO2007096779A2 (fr) 2006-01-25 2007-01-25 Exclusion allelique

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2007/001491 A-371-Of-International WO2007096779A2 (fr) 2006-01-25 2007-01-25 Exclusion allelique

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US14/962,335 Continuation US10638735B2 (en) 2006-01-25 2015-12-08 Generation of heavy-chain only antibodies in transgenic animals

Publications (1)

Publication Number Publication Date
US20090307787A1 true US20090307787A1 (en) 2009-12-10

Family

ID=38437750

Family Applications (3)

Application Number Title Priority Date Filing Date
US12/161,981 Abandoned US20090307787A1 (en) 2006-01-25 2007-01-25 Generation of heavy-chain only antibodies in transgenic animals
US14/962,335 Active 2027-04-27 US10638735B2 (en) 2006-01-25 2015-12-08 Generation of heavy-chain only antibodies in transgenic animals
US16/848,943 Pending US20200267951A1 (en) 2006-01-25 2020-04-15 Generation of heavy-chain only antibodies in transgenic animals

Family Applications After (2)

Application Number Title Priority Date Filing Date
US14/962,335 Active 2027-04-27 US10638735B2 (en) 2006-01-25 2015-12-08 Generation of heavy-chain only antibodies in transgenic animals
US16/848,943 Pending US20200267951A1 (en) 2006-01-25 2020-04-15 Generation of heavy-chain only antibodies in transgenic animals

Country Status (11)

Country Link
US (3) US20090307787A1 (fr)
EP (1) EP1991580A2 (fr)
JP (1) JP5184374B2 (fr)
KR (1) KR101481843B1 (fr)
AU (1) AU2007219159B8 (fr)
BR (1) BRPI0706750A2 (fr)
CA (1) CA2638117A1 (fr)
RU (1) RU2435784C2 (fr)
SG (1) SG169348A1 (fr)
TW (1) TWI404727B (fr)
WO (1) WO2007096779A2 (fr)

Cited By (50)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100122358A1 (en) * 2008-06-06 2010-05-13 Crescendo Biologics Limited H-Chain-only antibodies
US20100197897A1 (en) * 2004-07-22 2010-08-05 Franklin Gerardus Grosveld Binding molecules
US20110004949A1 (en) * 2001-04-24 2011-01-06 Frank Grosveld Immunoglobulin 1
US20110145937A1 (en) * 2009-12-10 2011-06-16 Regeneron Pharmaceuticals, Inc. Mice That Make Heavy Chain Antibodies
US20140201854A1 (en) * 2009-07-08 2014-07-17 Kymab Limited Animal models and therapeutic molecules
WO2014141192A1 (fr) 2013-03-15 2014-09-18 Erasmus University Medical Center Génération d'anticorps à chaînes lourdes uniquement
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
US9896516B2 (en) 2012-03-28 2018-02-20 Kymab Limited Animal models and therapeutic molecules
US9924705B2 (en) 2012-03-28 2018-03-27 Kymab Limited Animal models and therapeutic molecules
US9963716B2 (en) 2011-09-26 2018-05-08 Kymab Limited Chimaeric surrogate light chains (SLC) comprising human VpreB
US10149462B2 (en) 2013-10-01 2018-12-11 Kymab Limited Animal models and therapeutic molecules
US10251377B2 (en) 2012-03-28 2019-04-09 Kymab Limited Transgenic non-human vertebrate for the expression of class-switched, fully human, antibodies
US10266589B2 (en) 2015-07-29 2019-04-23 Allergan, Inc. Heavy chain only antibodies to ANG-2
US10308711B2 (en) 2015-08-14 2019-06-04 Allergan, Inc. Heavy chain only antibodies to PDGF
WO2019129211A1 (fr) 2017-12-28 2019-07-04 Nanjing Legend Biotech Co., Ltd. Anticorps et leurs variants dirigés contre pd-l1
US10358497B2 (en) 2015-09-29 2019-07-23 Amgen Inc. Methods of treating cardiovascular disease with an ASGR inhibitor
US10638735B2 (en) 2006-01-25 2020-05-05 Erasmus University Medical Center Generation of heavy-chain only antibodies in transgenic animals
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
WO2020247854A1 (fr) 2019-06-07 2020-12-10 Amgen Inc. Constructions de liaison bispécifiques à lieurs clivables de manière sélective
US10881085B2 (en) 2014-03-21 2021-01-05 Regeneron Pharmaceuticals, Inc. Non-human animals that make single domain binding proteins
US11051497B2 (en) 2011-09-19 2021-07-06 Kymab Limited Manipulation of immunoglobulin gene diversity and multi-antibody therapeutics
WO2021139777A1 (fr) 2020-01-10 2021-07-15 上海复宏汉霖生物技术股份有限公司 Anticorps anti-tigit et procédé d'utilisation
WO2021170067A1 (fr) 2020-02-28 2021-09-02 上海复宏汉霖生物技术股份有限公司 Construction anti-cd137 et son utilisation
WO2021170071A1 (fr) 2020-02-28 2021-09-02 Shanghai Henlius Biotech, Inc. Constructions anti-cd137, anticorps multispécifique et leurs utilisations
EP3896091A1 (fr) 2015-08-11 2021-10-20 Legend Biotech Ireland Limited Récepteurs d'antigène chimériques ciblant bcma et leurs procédés d'utilisation
WO2021247812A1 (fr) 2020-06-04 2021-12-09 Amgen Inc. Constructions de liaisons bispécifiques
WO2021247769A1 (fr) 2020-06-02 2021-12-09 Dynamicure Biotechnology Llc Constructions anti-cd93 et leurs utilisations
WO2022026763A1 (fr) 2020-07-29 2022-02-03 Dynamicure Biotechnology Llc Constructions anti-cd93 et leurs utilisations
WO2022067262A1 (fr) 2020-09-28 2022-03-31 Dynamicure Biotechnology Llc Constructions anti-cd93 et utilisations associées
WO2022120033A1 (fr) 2020-12-03 2022-06-09 Amgen Inc. Constructions d'immunoglobulines à domaines de liaison multiples
WO2022177394A1 (fr) 2021-02-19 2022-08-25 (주)샤페론 Anticorps bispécifique à domaine unique dirigé contre pd-l1 et cd47 et son utilisation
WO2022177393A1 (fr) 2021-02-19 2022-08-25 (주)샤페론 Anticorps à domaine unique dirigé contre pd-l1 et son utilisation
WO2022177392A1 (fr) 2021-02-19 2022-08-25 (주)샤페론 Anticorps à domaine unique dirigé contre cd47 et son utilisation
WO2022187863A1 (fr) 2021-03-05 2022-09-09 Dynamicure Biotechnology Llc Constructions anti-vista et leurs utilisations
US11447573B2 (en) 2016-07-20 2022-09-20 Nanjing Legend Biotech Co., Ltd. Multispecific antigen binding proteins and methods of use thereof
WO2022204724A1 (fr) 2021-03-25 2022-09-29 Dynamicure Biotechnology Llc Constructions anti-igfbp7 et leurs utilisations
US11472881B2 (en) 2016-10-11 2022-10-18 Nanjing Legend Biotech Co., Ltd. Single-domain antibodies and variants thereof against CTLA-4
US11484027B2 (en) 2020-10-29 2022-11-01 Industrial Polymers and Chemicals, Inc. Air filter with pathogen monitoring and inactivation
WO2022266660A1 (fr) 2021-06-17 2022-12-22 Amberstone Biosciences, Inc. Constructions anti-cd3 et utilisations associées
WO2022269473A1 (fr) 2021-06-23 2022-12-29 Janssen Biotech, Inc. Matériaux et procédés pour régions charnières dans des récepteurs exogènes fonctionnels
US11564380B2 (en) 2009-07-08 2023-01-31 Kymab Limited Animal models and therapeutic molecules
US11707056B2 (en) 2013-05-02 2023-07-25 Kymab Limited Animals, repertoires and methods
US11713353B2 (en) 2018-01-15 2023-08-01 Nanjing Legend Biotech Co., Ltd. Single-domain antibodies and variants thereof against PD-1
US11827713B2 (en) 2017-06-27 2023-11-28 Nanjing Legend Biotech Co., Ltd. Chimeric antibody immune effector cell engagers and methods of use thereof
WO2024018426A1 (fr) 2022-07-22 2024-01-25 Janssen Biotech, Inc. Transfert amélioré d'instructions génétiques à des cellules immunitaires effectrices
US11905327B2 (en) 2017-12-28 2024-02-20 Nanjing Legend Biotech Co., Ltd. Single-domain antibodies and variants thereof against TIGIT
WO2024054929A1 (fr) 2022-09-07 2024-03-14 Dynamicure Biotechnology Llc Constructions anti-vista et leurs utilisations
US11958903B2 (en) 2018-03-30 2024-04-16 Nanjing Legend Biotech Co., Ltd. Single-domain antibodies against LAG-3 and uses thereof
US11981932B2 (en) 2021-06-16 2024-05-14 Janssen Biotech, Inc. Materials and methods for the manufacture of pluripotent stem cells

Families Citing this family (32)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR101886610B1 (ko) 2007-06-01 2018-08-09 오픈 모노클로날 테크놀로지, 인코포레이티드 내생적 면역글로불린 유전자를 억제하고 트랜스제닉 인간 이디오타입 항체를 생산하기 위한 방법 및 조성물
WO2009013620A2 (fr) * 2007-06-11 2009-01-29 Erasmus University Medical Center Rotterdam Recombinaison homologue
ES2436044T3 (es) 2008-05-23 2013-12-26 Ablexis, Llc Procedimiento de generación de anticuerpos de dominio VL individual en animales transgénicos
SG172176A1 (en) 2008-12-18 2011-07-28 Univ Erasmus Medical Ct Non-human transgenic animals expressing humanised antibodies and use therof
GB0905023D0 (en) 2009-03-24 2009-05-06 Univ Erasmus Medical Ct Binding molecules
US9796788B2 (en) 2010-02-08 2017-10-24 Regeneron Pharmaceuticals, Inc. Mice expressing a limited immunoglobulin light chain repertoire
TR201906650T4 (tr) 2010-02-08 2019-05-21 Regeneron Pharma Ortak hafif zincirli fare.
US20130045492A1 (en) 2010-02-08 2013-02-21 Regeneron Pharmaceuticals, Inc. Methods For Making Fully Human Bispecific Antibodies Using A Common Light Chain
NZ707327A (en) 2010-08-02 2017-01-27 Regeneron Pharma Mice that make binding proteins comprising vl domains
ES2872081T3 (es) 2011-08-05 2021-11-02 Regeneron Pharma Ratones con cadena ligera universal humanizada
RU2603090C2 (ru) 2011-10-17 2016-11-20 Регенерон Фармасьютикалс, Инк. Мыши с ограниченной тяжелой цепью иммуноглобулина
MX356429B (es) 2011-12-20 2018-05-29 Regeneron Pharma Ratones con cadena ligera humanizada.
LT3597037T (lt) 2012-06-12 2021-06-10 Regeneron Pharmaceuticals, Inc. Humanizuoti gyvūnai, išskyrus žmones, su apribotu imunoglobulino sunkiosios grandinės lokusu
LT2931030T (lt) 2012-12-14 2020-11-10 Open Monoclonal Technology, Inc. Polinukleotidai, koduojantys graužikų antikūnus su žmogaus idiotipais, ir juos apimantys gyvūnai
RS57582B1 (sr) 2013-02-20 2018-11-30 Regeneron Pharma Nehumane životinje sa modifikovanim sekvencama imunoglobulinskog teškog lanca
WO2014141189A1 (fr) 2013-03-14 2014-09-18 Erasmus University Medical Center Mammifère transgénique non humain destiné à la production d'anticorps
US9783618B2 (en) 2013-05-01 2017-10-10 Kymab Limited Manipulation of immunoglobulin gene diversity and multi-antibody therapeutics
JP2017510273A (ja) 2014-03-21 2017-04-13 リジェネロン・ファーマシューティカルズ・インコーポレイテッドRegeneron Pharmaceuticals, Inc. 異なる結合特性を示すvl抗原結合タンパク質
AU2016232715A1 (en) 2015-03-19 2017-09-28 Regeneron Pharmaceuticals, Inc. Non-human animals that select for light chain variable regions that bind antigen
EP3332637A1 (fr) * 2016-12-09 2018-06-13 Universitätsklinikum Hamburg-Eppendorf Anticorps à chaîne lourde contenant des vhh et sa production
KR20190104400A (ko) 2017-01-19 2019-09-09 오픈 모노클로날 테크놀로지, 인코포레이티드 다중 중쇄 면역글로불린 유전자좌를 갖는 트랜스제닉 설치류 기원의 인간 항체
US20220275066A1 (en) * 2018-03-13 2022-09-01 Smivet B.V. Single domain antibodies binding to tetanus neurotoxin
CN108486125B (zh) * 2018-03-27 2024-01-05 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
WO2020169755A2 (fr) 2019-02-20 2020-08-27 Harbour Antibodies Bv Anticorps
WO2021154993A1 (fr) * 2020-01-28 2021-08-05 University Of Southern California Ingénierie génomique des locus des immunoglobulines humaines pour exprimer des molécules recombinantes à site de liaison
GB202003632D0 (en) 2020-03-12 2020-04-29 Harbour Antibodies Bv SARS-Cov-2 (SARS2, COVID-19) antibodies
TWI806087B (zh) 2020-06-30 2023-06-21 大陸商和鉑醫藥(上海)有限責任公司 免疫細胞銜接多特異性結合蛋白及其製備和應用
JP2023531672A (ja) 2020-06-30 2023-07-25 ハーバー・バイオメド・(シャンハイ)・カンパニー・リミテッド H2L2とHCAb構造を有する結合タンパク質
GB202017555D0 (en) 2020-11-06 2020-12-23 Harbour Antibodies Bv Antibody-conjugated nanoparticles
KR20230150779A (ko) 2020-12-09 2023-10-31 트리아니, 인코포레이티드 중쇄-단독 항체
WO2023036982A1 (fr) 2021-09-10 2023-03-16 Harbour Antibodies Bv Anticorps anti-sars2-s
GB202112935D0 (en) 2021-09-10 2021-10-27 Harbour Antibodies Bv Sars-cov-2 (sars2, covid-19) heavy chain only antibodies

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050287630A1 (en) * 1995-04-27 2005-12-29 Abgenix, Inc. Human antibodies derived from immunized xenomice

Family Cites Families (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4633464A (en) * 1983-08-08 1986-12-30 At&T Bell Laboratories Control signalling arrangement for a digital transmission system
WO1991010741A1 (fr) 1990-01-12 1991-07-25 Cell Genesys, Inc. Generation d'anticorps xenogeniques
GB9015198D0 (en) 1990-07-10 1990-08-29 Brien Caroline J O Binding substance
US7041871B1 (en) 1995-10-10 2006-05-09 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
CA2140638C (fr) 1992-07-24 2010-05-04 Raju Kucherlapati Production d'anticorps xenogeniques
EP0823941A4 (fr) 1995-04-28 2001-09-19 Abgenix Inc Anticorps humains derives de xeno-souris immunisees
EP1500329B1 (fr) 1996-12-03 2012-03-21 Amgen Fremont Inc. Les anticorps humains qui lient en particulier l'alpha de TNF humain
CA2634294A1 (fr) * 2000-08-03 2002-02-14 Therapeutic Human Polyclonals, Inc. Production d'anticorps humanises dans des animaux transgeniques
GB0110029D0 (en) * 2001-04-24 2001-06-13 Grosveld Frank Transgenic animal
GB0228210D0 (en) * 2002-12-03 2003-01-08 Babraham Inst Single chain antibodies
GB0401882D0 (en) 2004-01-28 2004-03-03 Inpharmatica Ltd Protein
SI2311874T1 (sl) * 2004-07-22 2017-12-29 Erasmus University Medical Center Rotterdam Department of Cell Biology and Genetics Vezavne molekule
WO2007096779A2 (fr) 2006-01-25 2007-08-30 Erasmus University Medical Center Rotterdam Exclusion allelique
US20100122358A1 (en) 2008-06-06 2010-05-13 Crescendo Biologics Limited H-Chain-only antibodies
GB0905023D0 (en) * 2009-03-24 2009-05-06 Univ Erasmus Medical Ct Binding molecules

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050287630A1 (en) * 1995-04-27 2005-12-29 Abgenix, Inc. Human antibodies derived from immunized xenomice

Cited By (88)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20110004948A1 (en) * 2001-04-24 2011-01-06 Frank Grosveld Immunoglobulin 2
US8507748B2 (en) * 2001-04-24 2013-08-13 Erasmus Universiteit Rotterdam Method for the production of a single heavy chain antibody
US8502014B2 (en) * 2001-04-24 2013-08-06 Erasmus Universiteit Rotterdam Method for the production of a single heavy chain antibody
US20110004949A1 (en) * 2001-04-24 2011-01-06 Frank Grosveld Immunoglobulin 1
US9353179B2 (en) 2004-07-22 2016-05-31 Erasmus University Medical Centre Binding molecules
US9346877B2 (en) 2004-07-22 2016-05-24 Erasmus University Medical Centre Binding molecules
US10906970B2 (en) 2004-07-22 2021-02-02 Erasmus University Medical Centre Methods of making heavy chain only antibodies using transgenic animals
US20100216974A1 (en) * 2004-07-22 2010-08-26 Franklin Gerardus Grosveld Binding molecules
US20100197897A1 (en) * 2004-07-22 2010-08-05 Franklin Gerardus Grosveld Binding molecules
US20110118444A1 (en) * 2004-07-22 2011-05-19 Franklin Gerardus Grosveld Binding molecules
US8921522B2 (en) * 2004-07-22 2014-12-30 Erasmus University Medical Centre Binding molecules
US8921524B2 (en) * 2004-07-22 2014-12-30 Erasmus University Medical Centre Binding molecules
US10638735B2 (en) 2006-01-25 2020-05-05 Erasmus University Medical Center Generation of heavy-chain only antibodies in transgenic animals
US20100122358A1 (en) * 2008-06-06 2010-05-13 Crescendo Biologics Limited H-Chain-only antibodies
US10064398B2 (en) 2009-07-08 2018-09-04 Kymab Limited Animal models and therapeutic molecules
US20140201854A1 (en) * 2009-07-08 2014-07-17 Kymab Limited Animal models and therapeutic molecules
US11812731B2 (en) 2009-07-08 2023-11-14 Kymab Ltd. Animal models and therapeutic molecules
US10165763B2 (en) 2009-07-08 2019-01-01 Kymab Limited Animal models and therapeutic molecules
US11606941B2 (en) 2009-07-08 2023-03-21 Kymab Limited Animal models and therapeutic molecules
US11564380B2 (en) 2009-07-08 2023-01-31 Kymab Limited Animal models and therapeutic molecules
US20110145937A1 (en) * 2009-12-10 2011-06-16 Regeneron Pharmaceuticals, Inc. Mice That Make Heavy Chain Antibodies
US9888675B2 (en) 2009-12-10 2018-02-13 Regeneron Pharmaceuticals, Inc. Mice that make heavy chain antibodies
US11234419B2 (en) 2009-12-10 2022-02-01 Regeneran Pharmaceuticals, Inc. Mice that make heavy chain antibodies
US8754287B2 (en) 2009-12-10 2014-06-17 Regeneron Pharmaceuticals, Inc. Mice that make heavy chain antibodies
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
US9938358B2 (en) 2012-03-28 2018-04-10 Kymab Limited Animal models and therapeutic molecules
US10251377B2 (en) 2012-03-28 2019-04-09 Kymab Limited Transgenic non-human vertebrate for the expression of class-switched, fully human, antibodies
US9938357B2 (en) 2012-03-28 2018-04-10 Kymab Limited Animal models and therapeutic molecules
US9924705B2 (en) 2012-03-28 2018-03-27 Kymab Limited Animal models and therapeutic molecules
US10774155B2 (en) 2012-03-28 2020-09-15 Kymab Limited Animal models and therapeutic molecules
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
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
WO2014141192A1 (fr) 2013-03-15 2014-09-18 Erasmus University Medical Center Génération d'anticorps à chaînes lourdes uniquement
US10993420B2 (en) 2013-03-15 2021-05-04 Erasmus University Medical Center Production of heavy chain only antibodies in transgenic mammals
US9788534B2 (en) 2013-03-18 2017-10-17 Kymab Limited Animal models and therapeutic molecules
US11297810B2 (en) 2013-03-18 2022-04-12 Kymab Limited Animal models and therapeutic molecules
US10226033B2 (en) 2013-03-18 2019-03-12 Kymab Limited Animal models and therapeutic molecules
US10730930B2 (en) 2013-05-02 2020-08-04 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
US9783593B2 (en) 2013-05-02 2017-10-10 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
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
US10881085B2 (en) 2014-03-21 2021-01-05 Regeneron Pharmaceuticals, Inc. Non-human animals that make single domain binding proteins
US11046756B2 (en) 2015-07-29 2021-06-29 Allergan, Inc. Heavy chain only antibodies to ANG-2
US10266589B2 (en) 2015-07-29 2019-04-23 Allergan, Inc. Heavy chain only antibodies to ANG-2
EP3792279A2 (fr) 2015-07-29 2021-03-17 Allergan, Inc. Anticorps contenant seulement une chaîne lourde contre ang-2
EP4282878A2 (fr) 2015-08-11 2023-11-29 Legend Biotech Ireland Limited Récepteurs d'antigène chimériques ciblant bcma et leurs procédés d'utilisation
EP4282877A2 (fr) 2015-08-11 2023-11-29 Legend Biotech Ireland Limited Récepteurs d'antigène chimériques ciblant bcma et leurs procédés d'utilisation
EP3896091A1 (fr) 2015-08-11 2021-10-20 Legend Biotech Ireland Limited Récepteurs d'antigène chimériques ciblant bcma et leurs procédés d'utilisation
EP4063397A1 (fr) 2015-08-11 2022-09-28 Legend Biotech Ireland Limited Récepteurs d'antigènes chimériques basés sur des anticorps à domaine unique et leurs méthodes d'utilisation
US10308711B2 (en) 2015-08-14 2019-06-04 Allergan, Inc. Heavy chain only antibodies to PDGF
US11028163B2 (en) 2015-08-14 2021-06-08 Allergan, Inc. Heavy chain only antibodies to PDGF
US10358497B2 (en) 2015-09-29 2019-07-23 Amgen Inc. Methods of treating cardiovascular disease with an ASGR inhibitor
US11066472B2 (en) 2015-09-29 2021-07-20 Amgen Inc. Methods of treating cardiovascular disease with an anti-ASGR antibody or binding fragments thereof
US11447573B2 (en) 2016-07-20 2022-09-20 Nanjing Legend Biotech Co., Ltd. Multispecific antigen binding proteins and methods of use thereof
US11472881B2 (en) 2016-10-11 2022-10-18 Nanjing Legend Biotech Co., Ltd. Single-domain antibodies and variants thereof against CTLA-4
US11827713B2 (en) 2017-06-27 2023-11-28 Nanjing Legend Biotech Co., Ltd. Chimeric antibody immune effector cell engagers and methods of use thereof
US11905327B2 (en) 2017-12-28 2024-02-20 Nanjing Legend Biotech Co., Ltd. Single-domain antibodies and variants thereof against TIGIT
WO2019129211A1 (fr) 2017-12-28 2019-07-04 Nanjing Legend Biotech Co., Ltd. Anticorps et leurs variants dirigés contre pd-l1
US11713353B2 (en) 2018-01-15 2023-08-01 Nanjing Legend Biotech Co., Ltd. Single-domain antibodies and variants thereof against PD-1
US11958903B2 (en) 2018-03-30 2024-04-16 Nanjing Legend Biotech Co., Ltd. Single-domain antibodies against LAG-3 and uses thereof
WO2020247854A1 (fr) 2019-06-07 2020-12-10 Amgen Inc. Constructions de liaison bispécifiques à lieurs clivables de manière sélective
WO2020247852A1 (fr) 2019-06-07 2020-12-10 Amgen Inc. Constructions de liaison bispécifiques
WO2021139777A1 (fr) 2020-01-10 2021-07-15 上海复宏汉霖生物技术股份有限公司 Anticorps anti-tigit et procédé d'utilisation
WO2021170067A1 (fr) 2020-02-28 2021-09-02 上海复宏汉霖生物技术股份有限公司 Construction anti-cd137 et son utilisation
WO2021170071A1 (fr) 2020-02-28 2021-09-02 Shanghai Henlius Biotech, Inc. Constructions anti-cd137, anticorps multispécifique et leurs utilisations
WO2021247769A1 (fr) 2020-06-02 2021-12-09 Dynamicure Biotechnology Llc Constructions anti-cd93 et leurs utilisations
WO2021247812A1 (fr) 2020-06-04 2021-12-09 Amgen Inc. Constructions de liaisons bispécifiques
WO2022026763A1 (fr) 2020-07-29 2022-02-03 Dynamicure Biotechnology Llc Constructions anti-cd93 et leurs utilisations
WO2022067262A1 (fr) 2020-09-28 2022-03-31 Dynamicure Biotechnology Llc Constructions anti-cd93 et utilisations associées
US11684066B1 (en) 2020-10-29 2023-06-27 Industrial Polymers and Chemicals, Inc. Air filter with pathogen monitoring and inactivation
US11793194B2 (en) 2020-10-29 2023-10-24 Industrial Polymers and Chemicals, Inc. Air filter with pathogen monitoring and inactivation
US11484027B2 (en) 2020-10-29 2022-11-01 Industrial Polymers and Chemicals, Inc. Air filter with pathogen monitoring and inactivation
US11766040B2 (en) 2020-10-29 2023-09-26 Industrial Polymers and Chemicals, Inc. Air filter with pathogen monitoring and inactivation
WO2022120033A1 (fr) 2020-12-03 2022-06-09 Amgen Inc. Constructions d'immunoglobulines à domaines de liaison multiples
WO2022177392A1 (fr) 2021-02-19 2022-08-25 (주)샤페론 Anticorps à domaine unique dirigé contre cd47 et son utilisation
WO2022177393A1 (fr) 2021-02-19 2022-08-25 (주)샤페론 Anticorps à domaine unique dirigé contre pd-l1 et son utilisation
WO2022177394A1 (fr) 2021-02-19 2022-08-25 (주)샤페론 Anticorps bispécifique à domaine unique dirigé contre pd-l1 et cd47 et son utilisation
WO2022187863A1 (fr) 2021-03-05 2022-09-09 Dynamicure Biotechnology Llc Constructions anti-vista et leurs utilisations
WO2022204724A1 (fr) 2021-03-25 2022-09-29 Dynamicure Biotechnology Llc Constructions anti-igfbp7 et leurs utilisations
US11981932B2 (en) 2021-06-16 2024-05-14 Janssen Biotech, Inc. Materials and methods for the manufacture of pluripotent stem cells
WO2022266660A1 (fr) 2021-06-17 2022-12-22 Amberstone Biosciences, Inc. Constructions anti-cd3 et utilisations associées
WO2022269473A1 (fr) 2021-06-23 2022-12-29 Janssen Biotech, Inc. Matériaux et procédés pour régions charnières dans des récepteurs exogènes fonctionnels
WO2024018426A1 (fr) 2022-07-22 2024-01-25 Janssen Biotech, Inc. Transfert amélioré d'instructions génétiques à des cellules immunitaires effectrices
WO2024054929A1 (fr) 2022-09-07 2024-03-14 Dynamicure Biotechnology Llc Constructions anti-vista et leurs utilisations

Also Published As

Publication number Publication date
US10638735B2 (en) 2020-05-05
RU2008134517A (ru) 2010-02-27
EP1991580A2 (fr) 2008-11-19
US20200267951A1 (en) 2020-08-27
CA2638117A1 (fr) 2007-08-30
TWI404727B (zh) 2013-08-11
AU2007219159A1 (en) 2007-08-30
RU2435784C2 (ru) 2011-12-10
US20160295843A1 (en) 2016-10-13
WO2007096779A3 (fr) 2008-06-26
KR20090013748A (ko) 2009-02-05
SG169348A1 (en) 2011-03-30
AU2007219159B2 (en) 2012-06-14
BRPI0706750A2 (pt) 2011-04-05
AU2007219159B8 (en) 2012-06-28
TW200808825A (en) 2008-02-16
WO2007096779A2 (fr) 2007-08-30
JP2009524641A (ja) 2009-07-02
KR101481843B1 (ko) 2015-01-12
JP5184374B2 (ja) 2013-04-17

Similar Documents

Publication Publication Date Title
US20200267951A1 (en) Generation of heavy-chain only antibodies in transgenic animals
US20230133028A1 (en) Transgenic mice
ES2635497T3 (es) Moléculas de unión
US5545806A (en) Ransgenic non-human animals for producing heterologous antibodies
WO2009013620A2 (fr) Recombinaison homologue
US20090271880A1 (en) Binding molecules
US20040231012A1 (en) Murine expression of human IgA Lambda locus
WO1992003918A1 (fr) Animaux non humains transgeniques capables de produire des anticorps heterologues
JP2001527386A (ja) 異種抗体を産生し得るトランスジェニック非ヒト動物
KR20110089846A (ko) 키메라 항체의 제조를 위한 인간 이외의 포유동물
EP2288623A2 (fr) Procédé de génération d anticorps à domaine vl unique dans des animaux transgéniques
CN101410412B (zh) 在转基因动物中产生仅有重链之抗体
Brüggemann et al. Selection strategies III: transgenic mice
MX2008009685A (es) Generacion de anticuerpos solo de cadena pesada en animales transgenicos
US20230270086A1 (en) Transgenic animals expressing heavy chain antibodies
EP1288229A2 (fr) Animaux transgéniques non humains, capables de fabriquer des anticorps hétérologues
NZ617158B2 (en) Non-human animals expressing antibodies having a common light chain

Legal Events

Date Code Title Description
AS Assignment

Owner name: ERASMUS UNIVERSITY MEDICAL CENTER, NETHERLANDS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:GROSVELD, FRANKLIN GERARDUS;JANSSENS, RICHARD WILHELM;REEL/FRAME:027562/0661

Effective date: 20111010

STCB Information on status: application discontinuation

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

AS Assignment

Owner name: HARBOUR ANTIBODIES B.V., NETHERLANDS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:ERASMUS UNIVERSITAIR MEDISCH CENTRUM ROTTERDAM;REEL/FRAME:065157/0018

Effective date: 20231005