GB2495083A - Human VpreB and chimaeric surrogate light chains in transgenic non-human vertebrates - Google Patents

Human VpreB and chimaeric surrogate light chains in transgenic non-human vertebrates Download PDF

Info

Publication number
GB2495083A
GB2495083A GB1116495.1A GB201116495A GB2495083A GB 2495083 A GB2495083 A GB 2495083A GB 201116495 A GB201116495 A GB 201116495A GB 2495083 A GB2495083 A GB 2495083A
Authority
GB
United Kingdom
Prior art keywords
human
vertebrate
gene
text
cell
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
GB1116495.1A
Other versions
GB201116495D0 (en
Inventor
Allan Bradley
E-Chiang Lee
Qi Liang
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Kymab Ltd
Original Assignee
Kymab Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Kymab Ltd filed Critical Kymab Ltd
Priority to GB1116495.1A priority Critical patent/GB2495083A/en
Publication of GB201116495D0 publication Critical patent/GB201116495D0/en
Priority to PCT/GB2012/052380 priority patent/WO2013045916A1/en
Priority to EP12766714.5A priority patent/EP2761008A1/en
Publication of GB2495083A publication Critical patent/GB2495083A/en
Priority to US14/226,698 priority patent/US9963716B2/en
Priority to US15/973,376 priority patent/US20180282761A1/en
Withdrawn legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/46Hybrid immunoglobulins
    • C07K16/461Igs containing Ig-regions, -domains or -residues form different species
    • C07K16/462Igs containing a variable region (Fv) from one specie and a constant region (Fc) from another
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • 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
    • 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
    • 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
    • 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
    • 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

Abstract

Non-human vertebrates (e.g. a mouse or rat) or cells are disclosed comprising an antibody heavy chain transgene, wherein the transgene comprises one or more human VH , D or JH gene segments operably connected upstream of a constant region gene so that the transgene can undergo VDJ recombination in vivo to produce an antibody gene comprising a rearranged VDJC encoding an antibody heavy chain having a human variable region and a constant region, or wherein the transgene comprises a rearranged VDJ encoding a human variable region operably connected upstream of a constant region gene, the genome further comprising a human VpreB gene capable of expressing human a VpreB. Embodiments also provide for species-matching of the entire surrogate light chain for efficient pairing with chimaeric heavy chains during B-cell development in vivo in a non-human transgenic vertebrate. The invention is directed towards improving the production of chimaeric antibodies.

Description

Human VoreB & Chimperic Surrogate Light Chains in Transgenic Non-Human Vertebrates The present invention relates inter a/ia to improvements in the production of chimaeric antibodies in non-human transgenic vertebrates such as mice and rats bearing one or more chimaeric antibody transgenes. In particular, the invention provides for improved non-human vertebrates and cells in which VpreB has been species-matched with the variable region of the chimaeric antibodies. Also, embodiments also provide for species-matching of the entire surrogate light chain for efficient pairing with chimaeric heavy chains during 8-cell development in viva in a non-human transgenic vertebrate setting.
Furthermore, the invention relates to a method of using the vertebrates to isolate antibodies or nucleotide sequences encoding antibodies. Antibodies, nucleotide sequences, pharmaceutical compositions and uses are also provided by the invention.
BACKGROUND
Advancements in the construction of transgenic mice bearing human antibody gene loci have led researchers to move away from the provision of entirely human transgenic antibody loci (bearing human variable and constant region gene segments) to chimaeric transgenic antibody loci. The transgenic loci comprise human V(D)J segments operably connected upstream of non-human constant regions. The use of constant regions that are endogenous to the transgenic non-human vertebrate (eg, endogenous mouse or rat constant regions) are desirable to harness the endogenous control of antibody generation and maturation following immunisation of the vertebrate with antigen.
B-cell development is characterized by the ordered rearrangement of immunoglobulin variable region genes. Alter the V0J rearrangement of the H chain gene segments, a precursor (pre)-B-cell is generated. After the functional rearrangement of a light chain gene, the cells develop into surface 1gM-bearing mature B-cells. Fully assembled IgH and L chains are transported onto the surface of B-cells, while free H chains are retained in the endoplasmic reticulum(ER) in association with BiP.
A critical step in B-cell differentiation is the selective expansion of cells with a functional i heavy chain resulting from productive IgH rearrangement. This is achieved by the association of the p heavy chain with the surrogate light chain (SLC) proteins KS and VpreB and the signal transducing heterodimer Iga and to form the pre-B-cell receptor (pre-BCR). The expression and formation of a S pre-BCR dramatically improves the efficiency of pre-B and B-cell production, by signalling proliferative expansion of pre-B-cells. The major function of the pre-BCR is the selection and expansion of cells that have undergone a productive VDJ rearrangement. The expression of membrane-bound.t chains is essential for the clonal expansion, and initiation of L chain gene rearrangement.
The association of SLC with p chain works as a checkpoint to determine whether the cell has successfully completed VDJ combination and expresses a functional kH chain. More than half of all p chains cannot assemble with the SLC, accounting for most of the changes in the V heavy repertoire during B-cell development.
VpreB is homologous to variable regions, AS is homologous to light chain constant region. The protein encoded by the two genes can form a tightly, but noncovalently bound heterodimer with the general structure of an L chain. KS protein can be covalently disulphide-bonded to p H chains in pre-B-cells. Whereas VpreB alone can associate with lgp, K 5 alone cannot. Only when it is non-covalently associated with VpreB does A 5 form a disulphide bridge with the first constant region (Cl-Il) of an lgp chain to form the pre-BCR.
The AS and Vpreb, which together form the SLC, are early markers of B-cell commitment. They are expressed at the pro-and pre-B-cell stages and silenced in immature and mature B-cells. In pre-B-cells, following rearrangement of the heavy chain locus, the surrogate light chain (SLC) acts as a chaperone, mediating transport of the newly synthesised heavy chain mu to the cell surface and together with C mu forms part of the pre-BCR (Immunol Today. 1993 Feb;14(2):60-8; The surrogate light chain in B-cell development; Melchers F, Karasuyama H, Haasner D, Bauer S, Kudo A, Sakaguchi N, Jameson B, Rolink A). The pre-BCR mediates signalling, leading to proliferation of pre-B-cells that have a productive heavy chain rearrangement. Mice that lack a functional KS show a drastic reduction in the number of B-cells. Using VpreB knock-out mice, it has been shown that VpreB is also required for efficient B-cell development, particularly for the transition to pre-BCR bearing cells (pre-BlI stage) (see, Int Immunol. 1999 Mar;11(3):453-60; Partial block in B lymphocyte development at the transition into the pre-B cell receptor stage in Vpre-B1-deficient mice; Màrtensson A, Argon Y, Melchers F, Dul JL, Mrtensson IL; and Sabbattini & Dillon 2005 infra). As the degree of proliferation has been proposed to be dependent upon the stability of the association of the mu heavy chain with the components of the SLC, the pre-BCR is thought to influence the choice, and thus the extent and diversity of, the heavy chain variable region repertoire.
Besides its role in signalling proliferation, the pre-BCR is also thought to mediate down regulation of the RAG genes, thereby preventing the rearrangement of other IgH loci (allelic exclusion) and the occurrence of double-strand breaks in dividing pre-B-cells. The pre-BCR has also been proposed to exert a negative feedback on KS and VpreB expression so that, after a phase of clonal expansion, the large pre-BIl cells become depleted of pre-BCR, exit the cell cycle and differentiate into resting small pre-BlI cells. At this stage, the SLC is repressed while the IgL loci start to be rearranged. The product of a successfully rearranged lgL gene will then pair with the mu heavy chain to form a BCR with antigen-binding capability on the surface of immature B-cells. These cells migrate to the peripheral blood and secondary lymphoid organs, and develop into mature B-cells ready for subsequent encounter with antigen (Sabbattini & Dillon 2005 infra).
SUMMARY OF THE INVENTION
The inventors were aware of the desirability to use endogenous (eg, mouse or rat) constant regions to harness the endogenous control of antibody generation and maturation following immunisation.
They have realised, however, that these considerations do not address earlier B-cell stages in vivo (prior to antigen stimulation) when the animal's B-cell repertoire is maturing. Such maturation includes transition from immature pro-B-cells to pie-B-cells, including the pre-BlI stage when B-cells bear surface B-cell receptors (BCR) in which the heavy chain variable region is paired with the VpreB.
CH1 regions of heavy chains are paired with KS in the pre-BCRs. The inventors realised that the provision of genes encoding chimaeric antibodies is not tailored to the pre-BlI stage, and they realised that it is important to address this since this stage features huge clonal expansion required to provide a pool of B-cells for functional heavy chain selection during subsequent B-cell development To this end, the present invention provides for species-matching of human V regions in chimaeric heavy chains with human VpreR. In embodiments, the invention also provides for species-matching of the mu constant region with A5, so that heavy chains in pre-BCRs are completely species-matched with chimaeric surrogate light chain (SLC).
Thus, a first configuration of the present invention provides, A non-human vertebrate (eg, a mouse or rat) or cell (eg, a mouse cell or rat cell) whose genome comprises an antibody heavy chain transgene, the transgene comprising (a) one or more human VE-] gene segments, one or more human D gene segments and one or more human JH gene segments operably connected upstream of a constant region gene so that the transgene is capable of undergoing VDJ recombination in vivo to produce an antibody gene comprising a rearranged VDJC encoding an antibody heavy chain having a human variable region and a constant region; or (b) a rearranged VDJ encoding a human variable region operably connected upstream of a constant region gene so that the transgene encodes (optionally following VDJ combination with the constant region) a chimaeric antibody heavy chain having a human variable region and a constant region; the genome further comprising a human VpreB gene capable of expressing a human VpreB.
The present invention is applicable to the production of 4-chain antibodies in transgenic animals, where the antibodies each contain 2 heavy chains and 2 light chains. Alternatively, the invention can be applied to the production of H2 antibodies (heavy chain antibodies) which are devoid of Cl-Il and light chains.
In a second embodiment, the invention provides A method of constructing a transgenic non-human vertebrate cell (eg, an ES cell, eg, a mouse or rat ES cell), the method comprising (i) introducing into the genome of a non-human vertebrate cell (or an ancestor thereof) one or more human VH gene segments, one or more human D gene segments and one or more human JH gene segments so that said gene segments are operably connected upstream of a constant region gene (optionally an endogenous non-human vertebrate constant region gene) to form a heavy chain transgene, wherein in said cell or a progeny thereof the transgene is capable of undergoing VDJ recombination in vivo to produce an antibody gene comprising a rearranged VDJC encoding an antibody heavy chain having a human variable region and a constant region; or (ii) introducing into the genome of a non-human vertebrate cell (or an ancestor thereof) a rearranged VDJ encoding a human variable region so that said VDJ is operably connected upstream of a constant region gene (optionally an endogenous non-human vertebrate constant region gene) to form a heavy chain transgene, wherein in said cell or a progeny thereof the transgene encodes (optionally following VDJ combination with the constant region) an antibody heavy chain having a human variable region and a constant region; and wherein the method further comprises introducing into the cell a human VpreB gene capable of expressing a human VpreB.
In a third embodiment, the invention provides A method of promoting B-cell development in a non-human vertebrate (eg, a mouse or rat), the method comprising (a) providing in a non-human vertebrate embryonic stem (ES) cell genome an immunoglobulin transgene capable of expressing an antibody mu heavy chain) wherein the antibody heavy chain comprises a human variable region and a mu constant region (optionally an endogenous non-human vertebrate mu constant region); and creating a first non-human vertebrate from said ES cell or a progeny thereof; (b) providing in a second ES cell genome a second transgene comprising a human VpreB gene capable of expressing a human VpreB; and creating a second non-human vertebrate from said ES cell or a progeny thereof; and (c) creating by breeding a third non-human vertebrate capable of co-expressing the mu antibody heavy chain and human vpreB wherein a pre-B-cell receptor can form to promote B-cell development of cells bearing a mu heavy chain in said third vertebrate; the third vertebrate being made by crossing said first and second vertebrates or progeny thereof by breeding to create said third vertebrate, the third vertebrate comprising the first and second transgenes, and wherein endogenous heavy chain expression has been inactivated in said third vertebrate.
In a fourth embodiment, the invention provides A method of promoting B-cell development in a non-human vertebrate (eg, a mouse or rat), the method comprising (i) inactivating endogenous heavy chain expression in said vertebrate; (ii) providing in the genome of said vertebrate an immunoglobulin transgene capable of expressing an antibody mu heavy chain, wherein the antibody heavy chain comprises a human variable region and a non-human vertebrate mu constant region (optionally an endogenous non-human vertebrate mu constant region); and (ii) providing in the genome of said vertebrate a second transgene capable of expressing a human VpreB wherein a pre-B-cell receptor can form to promote B-cell development of cells bearing a mu heavy chain in said vertebrate; Optionally wherein the genome is homozygous for said first transgene.
An aspect of the invention further provides for the possibility of species-matching in viva the variable and constant regions of the antibody heavy chain with a chimaeric SLC. This is useful for promoting B-cell development in the transgenic non-human vertebrate (eg, a mouse or rat).
To this end, the invention provides a fifth embodiment: A non-human vertebrate (eg, a mouse or rat) or cell (eg, a mouse cell or rat cell) whose genome comprises an antibody heavy chain transgene, the transgene comprising (a) one or more human VH gene segments, one or more human D gene segments and one or more human JH gene segments operably connected upstream of a constant region gene of a non-human vertebrate (eg, mouse or rat) species so that the transgene is capable of undergoing VDJ recombination in vivo to produce an antibody gene comprising a rearranged VDJC encoding a chimaeric antibody heavy chain having a human variable region and a non-human vertebrate constant region; or (b) a rearranged VDJ encoding a human variable region operably connected upstream of a constant region gene of a non-human vertebrate (eg, mouse or rat) species so that the transgene encodes (optionally following VDJ combination with the constant region) a chimaeric antibody heavy chain having a human variable region and a non-human vertebrate constant region; the genome further comprising one or more genes together encoding a chimaeric surrogate light chain, the surrogate light chain comprising a human VpreB and 1.5 of said non-human vertebrate (eg, mouse or rat) species.
A sixth embodiment of the invention provides A method of constructing a transgenic non-human vertebrate cell (eg, an ES cell, eg, a mouse or rat ES cell), the method comprising (i) introducing into a non-human vertebrate cell (or an ancestor thereof) one or more human VH gene segments, one or more human D gene segments and one or more human.IH gene segments so that said gene segments are operably connected upstream of an endogenous non-human vertebrate constant region gene, wherein in said cell or a progeny thereof the transgene is capable of undergoing VDJ recombination in vivo to produce an antibody gene comprising a rearranged VDJC encoding a chimaeric antibody heavy chain having a human variable region and a non-human vertebrate constant region; or (ii) introducing into a non-human vertebrate cell (or an ancestor thereof) a rearranged VDJ encoding a human variable region so that said VDJ is operably connected upstream of an endogenous non-human vertebrate constant region gene, wherein in said cell or a progeny thereof the transgene encodes (optionally following VDJ combination with the constant region) a chimaeric antibody heavy chain having a human variable region and a non-human vertebrate constant region; and wherein the method further comprises introducing into the cell a human VpreB gene such that the cell or a progeny thereof is capable of expressing a chimaeric surrogate light chain as well as said chimaeric antibody heavy chain, wherein the surrogate light chain comprises a human VpreB and an endogenous non-human vertebrate AS.
S
A seventh embodiment of the invention further provides A method of promoting B-cell development in a non-human vertebrate (eg, a mouse or rat), the method comprising (a) providing in a non-human vertebrate embryonic stem (Es) cell an immunoglobulin transgene capable of expressing a chimaeric antibody mu heavy chain, wherein the chimaeric antibody heavy chain comprises a human variable region and an endogenous non-human vertebrate mu constant region; and creating a first non-human vertebrate from said ES cell or a progeny thereof; (b) providing in a second ES cell a second transgene capable of expressing a human VpreB so that the human VpreB and an endogenous X5 form a chimaeric surrogate light chain; and creating a second non-human vertebrate from said ES cell or a progeny thereof; and (c) creating by breeding a third non-human vertebrate capable of co-expressing the chimaeric mu antibody heavy chain and chimaeric surrogate light chain wherein a pre-BCR can form to promote B-cell development in said third vertebrate; the third vertebrate being made by crossing said first and second vertebrates or progeny thereof by breeding to create said third vertebrate, the third vertebrate comprising the first and second transgenes, and wherein endogenous heavy chain expression has been inactivated in said third vertebrate.
A transgenic mouse according to the vertebrate of this method, or a progeny thereof, is provided.
An eighth embodiment of the invention provides A method of promoting B-cell development in a non-human vertebrate (eg, a mouse or rat), the method comprising (i) inactivating endogenous heavy chain expression in said vertebrate; (ii) providing in the genome of said vertebrate an immunoglobulin transgene capable of expressing a chimaeric antibody mu heavy chain, wherein the chimaeric antibody heavy chain comprises a human variable region and an endogenous non-human vertebrate mu constant region; and (ii) providing in the genome of said vertebrate a second transgene capable of expressing a human VpreB so that the human VpreB and an endogenous AS form a chimaeric surrogate light chain; so that the vertebrate is capable of co-expressing the chimaeric mu antibody heavy chain and chimaeric surrogate light chain wherein a pre-BCR can form to promote B-cell development in said S vertebrate; Optionally wherein the genome is homozygous for said first transgene.
A transgenic mouse according to the vertebrate of this method, or a progeny thereof, is provided.
DETAILED DESCRIPTION OF THE INVENTION
The invention is useful for promoting the appropriate human IGH repertoire on the mature B-cells in the vertebrate (or progeny thereof) or in transgenic vertebrates derived from non-human vertebrate cells. At the transition of pre-BI to pre-BIl cells, VH to DH-JH rearrangements are initiated (see, eg, Màrtensson & Ceredig, Immunology 2000, 101, 435-441; Role of the surrogate light chain and the pre-B-cell receptor in mouse B-cell development). Whenever they occur in-frame, a mu chain can be produced and presented on the cell surface only it associates with the surrogate light chain to form the pre-B-cell receptor (pre-BCR). The pre-BCR on the cell surface is required for clonal expansion of pre-BlI cells before light chain rearrangement. Compromised presentation of pre-BCR on the cell surface due to the impairment of association of a mu chain carrying a certain variable region with a surrogate light chain will result in no clonal expansion of such a clone and reduce the representation of this variable region in the mature B cells. Human VpreBl only shares a low identity with rodent VpreBl/2, suggesting that species specificity for the pre-BCR assembly exists. To this end, the inventors realised the importance for appropriate VpreB/variable region pairing on generation of effective antibody diversity in the context of an antibody heavy chain transgene encoding human ZS variable regions, when this transgene is harboured in a non-human environment such as a transgenic mouse or rat.
The CH1 constant region pairs with the AS protein. Together, the VpreB and AS make up the surrogate light chain (SLC) that pairs with mu heavy chains to form the pre-BCR. For H2 antibodies pairing with the CH1 is not possible (since no CH1 is present), but the desirability to pair human VH regions with human VpreB according to the invention still applies.
Pairing Human Variable Regions of Pre-BCRs with Human VøreB The extent of homology between human VpreB with mouse and rat VpreB is relatively low (about 73% amino acid identity). As discussed above, the inventors have realised the advantage of matching human antibody heavy chain variable regions with human VpreB in receptors of pre-B cells for promoting B-cell development in transgenic non-human vertebrates bearing human antibody variable region genes.
In a first configuration, the invention thus provides:-Anon-human vertebrate (eg, a mouse or rat) or cell (eg, a mouse cell or rat cell) whose genome comprises an antibody heavy chain transgene, the transgene comprising (a) one or more human VH gene segments, one or more human U gene segments and one or more human JH gene segments operably connected upstream of a constant region gene so that the transgene is capable of undergoing VDJ recombination in vivo to produce an antibody gene comprising a rearranged VDJC encoding an antibody heavy chain having a human variable region and a constant region; or (b) a rearranged VDJ encoding a human variable region operably connected upstream of a constant region gene so that the transgene encodes (optionally following VDJ combination with the constant region) a chimaeric antibody heavy chain having a human variable region and a constant region; the genome further comprising a human VpreB gene capable of expressing a human VpreB.
The sequences of VpreB's and X5's are given below:-SEQ ID NO: 1 Human VpreBl nucleotide sequence (GenBank Accession No = NG_029387.1) SEQ ID NO: 2 Human VpreRl amino acid sequence (GenBank Accession No = NP_009059.1) SEQ ID NO: 3 Human VpreB3 nucleotide sequence (GenBank Accession No = NC_000022.10) SEQIDNO:4 Human VpreB3 amino acid sequence (GenBank Accession No = NP_037510) SEQ ID NO: 5 Mouse VpreBl nucleotide sequence (GenBank Accession No = NC_000082.5) SEQ ID NO: 6 Mouse VpreB1 amino acid sequence (GenBank Accession No = NP_058678) SEQ ID NO: 7 Mouse VpreB2 nucleotide sequence (GenBank Accession No = NC_000082) SEQ ID NO: 8 Mouse VpreB2 amino acid sequence (GenBank Accession No = NP_058679.1) SEQIDNO:9 Rat VpreBl nucleotide sequence (GenBank Accession No = NM_001108845.1) SEQ ID NO: 10 Rat VpreBl amino acid sequence (GenBank Accession No = NP_0011023151) SEQ ID NO: 11 Rat VpreB2 nucleotide sequence (GenBank Accession No = NC_005110) SEQ ID NO: 12 Rat VpreB2 amino acid sequence (GenBank Accession No = NP_001128260) SEQ ID NO: 13 Human X5 nucleotide sequence (GenBank Accession No = NG 009791) SEQ ID NO: 14 Human Xsamino acid sequence (GenBank Accession No = NP_064455) SEQ ID NO: 15 Mouse Xsnucleotide sequence (GenBank Accession No = AC_000038) SEQ ID NO: 16 Mouse Asamino acid sequence (GenBank Accession No = NP_001177254) SEQ ID NO: 17 Rat A5nucleotide sequence (GenBank Accession No = NC_005110) SEQIDNO:18 Rat A5amino acid sequence (GenBank Accession No = NP_001177270) All sequences listed under these GenBank Accession numbers are incorporated herein by reference as though explicitly written herein and for possible use in the present invention and possible inclusion of any claim below.
In one embodiment of any configuration of the invention, the human VpreB is human VpreBl. In another embodiment, the human Vpreb is human VpreB3.
Table 1: Sequence identity: VpreB: Human vs Mouse vs Rat IIl't)!: c.: 2S 72.t. il 2 Li:;'
Ll I..
Table 2' Sequence identity: Lambda 5: Human vs Mouse vs Rat Wi n:.t;cr. S7.? VI.::ir:. -q.) 4 r t; All nucleotide co-ordinates for the mouse are those corresponding to NCBI m37 for the mouse CS7BL/6J strain, e.g. April 2007 ENSEMBL Release 55.37h. e.g. NCBI37 July 2007 (NCBI build 37) (e.g. UCSC version mrn9 see www.genorne.ucsc.edu and http://genome.ucsc.eclu/FAOjFAQreleases.html) unless otherwise specified. Human nucleotides coordinates are those corresponding to GRCh37 (e.g. UCSC version hg 19, http://genome.ucsc.edu/FA0jFAQxeleases.html), Feb 2009 ENSEMBL Release 55.or are those corresponding to NCBI36. Ensemble release 54 unless otherwise specified. Rat nucleotides are those corresponding to RGSC 3.4 Dec 2004 ENSEMBL release 55.34w, or Baylor College of Medicine HGSC v3.4 Nov 2004(e.g., UCSC rn4, see www.genome.ucsc.edu and http://genome.ucsc.edu/FAOjfAQreleases.htnil) unless otherwise specified.
In one example of the vertebrate or cell, the genome is homozygous for said transgene and endogenous non-human vertebrate antibody heavy chain expression has been inactivated.
Alternatively or additionally, endogenous non-human vertebrate antibody light chain expression has been inactivated. Details on possible methods of inactivation are explained in more detail below.
The present invention is applicable to the production of 4-chain antibodies in transgenic animals, where the antibodies each contain 2 heavy chains and 2 light chains. Alternatively, the invention can be applied to the production of H2 antibodies (heavy chain antibodies) which are devoid of CH1 and light chains (see, eg, Nature. 1993 Jun 3;363(6428):446-8; Naturally occurring antibodies devoid of light chains; Hamers-Casterman C, Atarhouch T, Muyldermans 5, Robinson G, Hamers C, Songa ER, Bendahman N, Hamers R). These antibodies function to specifically bind antigen, such antibodies being found in the blood of Camel/doe (eg, llamas, camels, alpacas). Such antibodies with VH pairs can also be synthetically produced to provide therapeutic and prophylactic medicaments (eg, see W01994004678, W02004041862, W02004041863). Transgenic mice also can produce such heavy chain antibodies and the in vivo production of the antibodies allows the mouse's immune system to select for human VF-I-VH pairings, sometimes selecting for such pairings in which mutations have been introduced in vivo by the mouse to accommodate the pairing (W02010109165A2). Thus, in an embodiment of the present invention, the heavy chain transgene is devoid of a CH1 gene segment and the genome comprises no functional antibody light chain locus.
Throughout this text, and with application to any configuration, aspect, embodiment or example of the invention, the term "endogenous" (eg, endogenous constant region gene or endogenous AS) in relation to a non-human vertebrate or cell indicates that the constant region, AS etc is a type of constant region or AS that is normally found in the vertebrate or cell (as opposed to an exogenous constant region or AS whose sequence is not normally found in such a vertebrate or cell). For example, the endogenous constant region and AS can be those encoded by the wild-type genome of the non-human vertebrate/cell. So, in an example wherein the vertebrate cell is a mouse ES cell, the endogenous constant region and AS would be mouse constant region and AS. Going further, the endogenous regions are, in an example, strain-matched to the vertebrate/cell. So, in one embodiment, the vertebrate cell is a mouse 129 ES cell, the endogenous constant region and A5 would be mouse 129 constant region and AS. In another embodiment, the vertebrate cell is a mouse JM8 ES cell. the endo2enous constant region and AS would be mouse JMS constant region and AS. In another embodiment, the vertebrate is a Black 6 mouse) the endogenous constant region and AS would be mouse Black 6 constant region and A5.
In any configuration, aspect, embodiment or example of the invention, the constant region of the heavy chain transgene is a non-human vertebrate constant region (eg, mouse or rat constant region). Optionally, the constant region is endogenous to said non-human vertebrate or cell.
Alternatively, the constant region of the heavy chain transgene is human constant region. For example, the constant region is human and devoid of a CH1. This is useful for producing human H2 antibodies (especially when the vertebrate or cell is not capable of expressing light chains).
In one example of the vertebrate or cell of the invention, the constant region is a Cmu, eg, a mouse or rat Cmu. For example, where the vertebrate is a mouse (or cell is a mouse cell), the Cmu is an endogenous mouse Cmu. The transgene, in an example, comprises a Smu switch 5' of the Cmu and a Cgamma 3' of the Cmu, with as gamma switch between the Cmu and Cgamma. In an embodiment, the Cmu, Cgamma and switches are endogenous mouse C regions and switches. For example, the C regions and switches are mouse 129 C regions and switches; or the C regions and switches are mouse Black 6 C regions and switches. In another embodiment, the S gamma and C regions are mouse S gamma and C regions, and the Smu is a rat Smu.
In an example of any configuration, aspect, embodiment or example of the invention, the human VpreB gene has a nucleotide sequence that is at least 80, 85, 90, 91, 92, 93, 94, 95, 96, 97, 98 or 99 % identical to SEQ ID NO: 1. The VpreB gene encodes a human VpreB that pairs with the heavy chain human variable region. In an example, the human VpreB gene has a nucleotide sequence that is identical to SEQ ID NO:1.
In an example of any configuration, aspect, embodiment or example of the invention, the human VpreB gene encodes a human VpreB comprising an amino acid sequence that is at least 80, 85, 90, 91, 92, 93, 94, 95, 96, 97, 98 or 99% identical to SEQ ID NO: 2. The VpreB gene encodes a human VpreB that pairs with the heavy chain human variable region. In an example, the human VpreB gene encodes a human VpreB comprising an amino acid sequence that is identical to SEQ ID NO:2; or ootionallv with from one to 15 amino acid changes (eg, 1,2,3,4,5,6, 7, 8, 9, 10, 11, 12, 13, 14 or 15 changes) compared with SEQ ID NO: 2.
In any configuration, aspect, embodiment or example of the invention, the human VpreB gene encodes a human VpreB that pairs with the heavy chain human variable region, the human VpreB gene having from one to 25 nucleotide changes (eg, 1, 2, 3,4, 5, 6, 7,8,9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24 or 25 changes) compared to human VpreB (SEQ ID NO: 1). The VpreB gene can be manipulated in vitro by creation of a targeting vector comprising the gene sequence and homology arms flanking the sequence. This can be introduced into the genome of, for example an ES cell, using the standard technique of homologous recombination as will be apparent to the skilled person. Alternatively) recombinase mediated cassette exchange (RMCE) -another standard technique -can be used to effect precise insertion of the VpreB gene into the genome.
Homologous recombination or RMCE are generally-useful techniques for the specific, targeted, introduction of sequences into a genome. One or both of these techniques are readily applicable for introducing into a vertebrate or cell genome a human VpreB sequence (wild-type or mutant); V, D, J and C regions; switches; enhancers and other immunoglobulin control elements; and As genes.
These and other techniques for target insertion are known to the skilled addressee.
In an example of any configuration, aspect, embodiment or example of the invention, the genome does not comprise a non-human vertebrate (eg, mouse or rat) species VpreBl and/or VpreB2 gene.
In this embodiment, the possibility of non-human VpreB competing with the human VpreB for binding to the human variable regions is eliminated. Thus, pairing of human V regions with human VpreB on pre-B cells is not hampered by such competition, which is advantageous for promoting B-cell development.
In an example of any configuration, aspect, embodiment or example of the invention, the genome further comprises a As gene (eg, a human, mouse or rat AS); optionally wherein the AS gene is a As of a non-human vertebrate (eg, mouse or rat) species and said constant region gene is a constant region gene of the same non-human vertebrate (eg, mouse or rat) species as the AS gene. For example, the constant region and AS genes are mouse constant region and AS genes, eg, mouse 129 constant region and As genes; or mouse JM8 constant region and AS genes). For example, the constant region and AS genes are rat constant region and AS genes. In an example) constant region gene and AS gene are endogenous genes of said vertebrate or cell. Thus) the invention contemplates close matching of not only the V-regionJVpreB part of pre-BCR5 but also close matching of the Cmu/ A5 part. While not wishing to be bound to any theory, the inventors believe that this is useful for stabilising the pre-BCRs to promote B-cell development in vivo, thus improving the B-cell pool (and potentially diversity) from which to select antigen-specific antibodies following immunisation of vertebrates according to the inventions.
In another embodiment of any configuration, aspect, embodiment or example of the invention, the As is a chimaeric As comprising a non-human vertebrate AS (eg, endogenous mouse or rat A5) in which amino acid sequences have been replaced by corresponding amino acids from a human AS.
For example) the region of AS that interfaces with VpreB to form the SLC is replaced with the corresponding part from a human AS to form a chimaeric AS with a human part (ie, species-matched with the human VpreB) and a non-human vertebrate (eg, mouse or rat) part that is species matched with the CH1 of the non-human vertebrate Cmu of the heavy chain transgene. In the alternative embodiment where the As is a human As, the As is advantageously species-matched to interface with the human X5.
In one aspect the non-human vertebrate is able to generate a diversity of at least lx io different functional chimaeric antibody sequence combinations.
In a second configuration, the method provides A method of constructing a transgenic non-human vertebrate cell (eg, an ES cell, eg, a mouse or rat ES cell)) the method comprising (i) introducing into the genome of a non-human vertebrate cell (or an ancestor thereof) one or more human VH gene segments, one or more human D gene segments and one or more human JH gene segments so that said gene segments are operably connected upstream of a constant region gene (optionally an endogenous non-human vertebrate constant region gene) to form a heavy chain transgene, wherein in said cell or a progeny thereof the transgene is capable of undergoing VDJ recombination in vivo to produce an antibody gene comprising a rearranged VDJC encoding an antibody heavy chain having a human variable region and a constant region; or (ii) introducing into the genome of a non-human vertebrate cell (or an ancestor thereof) a rearranged VDJ encoding a human variable region so that said VDJ is operably connected upstream of a constant region gene (optionally an endogenous non-human vertebrate constant region gene) to form a heavy chain transgene, wherein in said cell or a progeny thereof the transgene encodes (optionally following VDJ combination with the constant region) an antibody heavy chain having a human variable region and a constant region; and wherein the method further comprises introducing into the cell (before, after or concomitantly with step (i) or (ii)) a human VpreB gene capable of expressing a human VpreR.
Targeting of the genome of an ES cell to produce a transgenic cell and subsequently a transgenic vertebrate (eg, a mouse) may be carried out using a protocol as explained by reference to the figures 1-18 and description in W02011004129, the disclosure of which is incorporated herein by reference. A suitable mouse ES cell is selected from AB2.1 cells, AR2.2 cells and JM8 cells.
In any configuration, aspect, embodiment or example of the invention, the cell is an ES cell is capable of developing into a non-human mammal able to produce a repertoire of antibodies which are chimaeric, said chimaeric antibodies having a non-human mammal constant region and a human variable region. Optionally the genome of the cell is modified to prevent expression of fully host-species specific antibodies.
In one aspect the cell is an induced pluripotent stem cell (iPS cell).
In one aspect cells are isolated non -human mammalian cells.
In one aspect a cell as disclosed herein is preferably a non -human mammalian cell.
In one aspect the cell is a cell from a mouse strain selected from C57BL/6, M129 such as 129/SV, BALB/c, and any hybrid of C57BL/6, M129 such as 129/SV, or BALB/c.
Maintaining the performance of the ES cell clones through multiple rounds of manipulation without the need to test the germ line potential of the ES cell line at every step may be important in the method of constructing a transgenic locus. The cell lines currently in use for the KOMP and EUCOMM global knockout projects have been modified twice prior to their use for this project and their germ line transmission rates are unchanged from the parental cells (these lines are publicly available, see www.komp.org and www.eucomm.org). This cell line, called JMS, can generate 100% ES cell-derived mice under published culture conditions (Pettitt, S.J., Liang, 0., Rairdan, X.Y., Moran, J.L., Prosser, ElM., Beier, D.R., Lloyd, K.C., Bradley, A., and Skarnes, W.C. (2009). Agouti C57BL/6N embryonic stem cells for mouse genetic resources. Nature Methods.). These cells have demonstrated ability to reproducibly contribute to somatic and germ line tissue of chimaeric animals using standard mouse ES cell culture conditions. This capability can be found with cells cultured on a standard feeder cell line (SNL) and even feeder-free, grown only on gelatine-coated tissue culture plates. One particular sub-line, JM8A3, maintained the ability to populate the germ line of chimeras after several serial rounds of sub-cloning. Extensive genetic manipulation via, for example, homologous recombination -as could be the case in the present method -cannot compromise the pluripotency of the cells. The ability to generate chimeras with such high percentage of ES cell-derived tissue has other advantages. First, high levels of chimaerism correlates with germ line transmission potential and provide a surrogate assay for germ line transmission while only taking 5 to 6 weeks. Second, since these mice are 100% ES cell derived the engineered loci can be directly tested, removing the delay caused by breeding. Testing the integrity of the new Ig loci is possible in the chimera since the host embryo will be derived from animals that are mutant for the RAG-i gene as described in the next section.
Another cell line that may be used is an I-IPRT-negative cell line, such as AB2.1, as disclosed in "Chromosome engineering in mice, RamIrez-Solis R, Liu P and Bradley A, Nature 1995;378;6558;720-4.
RAG-i complementation While many clones will generate 100% ES derived mice some will not. Thus, at every step mice can be generated in a RAG-i-deficient background. This provides mice with 100% ES-derived B-and T-cells which can be used directly for immunization and antibody production. Cells having a RAG-2 deficient background, or a combined RAG-1/RAG-2 deficient background may be used, or equivalent mutations in which mice produce only Es cell-derived B cells and/or T cells.
In order that only the human-mouse IgH (and optionally also light chain) loci are active in these mice, the human-mouse Igi-) (and optionally light chain loci) can be engineered in a cell line in which one allele of the IgH (and optionally light chain) locus has already been inactivated. Alternatively the inactivation of the host Ig locus/loci can be carried out after insertion.
Mouse strains that have the RAG-i gene mutated are immunodeficient as they have no mature B-or T-lymphocytes (us 5,859,307). 1-and B-lymphocytes only differentiate if proper V(D)J recombination occurs. Since RAG-i is an enzyme that is crucial for this recombination, mice lacking RAG-i are immunodeficient. If host embryos are genetically RAG-i homozygous mutant, a chimera produced by injecting such an embryo will not be able to produce antibodies if the animal's lymphoid tissues are derived from the host embryo. However, JM8 cells and AB2.1 cells, for example, generally contribute in excess of 80% of the somatic tissues of the chimaeric animal and would therefore usually populate the lymphoid tissue. JM8 cells have wild-type RAG-i activity and therefore antibodies produced in the chimaeric animal would be encoded by the engineered JM8 ES cell genome only. Therefore, the chimaeric animal can be challenged with an antigen by immunization and subsequently produce antibodies to that antigen. This allows one skilled in the art to test the performance of the engineered human/mouse Ig loci as described in the present invention.
One skilled in the art could use the chimaeric animal as described to determine the extent of antibody diversity (see e.g. Harlow, E. & Lane, D. 1998, 5th edition, Antibodies: A Laboratory Manual, Cold Spring Harbor Lab. Press, Plainview, NY). For example, the existence in the chimaeric animal's serum of certain antibody epitopes could be ascertained by binding to specific anti-idiotype antiserum, for example, in an ELISA assay. One skilled in the art could also sequence the genomes of B-cell clones derived from the chimaeric animal and compare said sequence to wild-type sequence (ie, minus human VpreB) to assess the B-cell repertoire and ascertain the level of hypermutation, such hypermutation being indicative of normal antibody maturation.
One skilled in the art would also use said chimaeric animal to examine antibody function wherein said antibodies are encoded from the engineered Ig loci (see e.g. Harlow, E. & Lane, D. 1998, 5th edition, Antibodies: A Laboratory Manual, Cold Spring Harbor Lab. Press, Plainview, NY). For example, antisera could be tested for binding an antigen, said antigen used to immunize the chirnaeric animal. Such a measurement could be made by an ELISA assay. Alternatively, one skilled in the art could test for neutralization of the antigen by addition of the antisera collected from the appropriately immunized chimaeric animal.
It is well known to those skilled in the art that positive outcomes for any of these tests demonstrate the ability of the engineered Ig loci in the presence of VpreB during B-cell development according to the instant invention, to encode antibodies with human variable regions and non-human vertebrate constant regions, said antibodies capable of functioning in the manner of wild-type antibodies.
The introduction of human gene segment DNA into the genome can be carried out using vectors carrying the DNA, as more fully explained below. In one aspect such vectors are BACs (bacterial artificial chromosomes). It will be appreciated that other cloning vectors may be used in the invention, and therefore reference to BACs herein may be taken to refer generally to any suitable vector. As described in W02011004129, in one aspect the inserted DNA is built up in the genome of a cell, such as an ES cell, in a stepwise manner using 2, 3,4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20 or more separate insertions for the heavy chain gene segments. Fragments are suitably inserted at the same or substantially the same cell locus, e.g. ES cell locus, one after another, to form the complete VDJ region.
In a further aspect the method comprises the insertion of human heavy chain gene segments upstream of the non-human vertebrate constant region by step-wise insertion of multiple fragments by homologous recombination, preferably using an iterative process. Suitably fragments of approximately 100KB from the human VDJ locus are inserted) suitably to form a VDJ region after the final iteration of the insertion process, as described in W02011004129.
In one aspect the insertion process commences at a site where an initiation cassette has been inserted into the genome of a cell, such as an ES cell, providing a unique targeting region. In one aspect the initiation cassette is inserted in the non-human vertebrate heavy chain locus, for use in insertion of human heavy chain DNA. The initiation cassette suitably comprises a vector backbone sequence with which a vector having a human DNA fragment in the same backbone sequence can recombine to insert the human DNA into the cell (e.g. Es) cell genome, and suitably a selection marker, such as a negative selection marker. Suitably the vector backbone sequence is that of a BAC library, to allow BACs to be used in the construction of the ES cells and mammals. The vector backbone sequence may however be any sequence which serves as a target site into which a homologous sequence can insert, for example by homologous recombination and,for example RMCE, and is optionally not DNA encoding any of the VDJ or constant region.
Suitable BACs are available from the Sanger centre, see "A genome-wide, end-sequenced 1295v BAC library resource for targeting vector construction". Adams Di, Quail MA, Cox T, van der Weyden L, Gorick BD, Su Q, Chan WI, Davies R, Bonfield JK, Law F, Humphray 5, Plumb B, Liu P, Rogers J, Bradley A. Genomics. 2005 Dec; 86(6):753-8. Epub 2005 Oct 27. The Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire CB1O iSA, UK. BACs containing human DNA are also available from, for example, Invitrogen. A suitable library (source of human heavy chain gene segments) is described in Osoegawa K et al, Genome Research 2001. 11: 483-496 and obtainable from the Roswell Park In any configuration, aspect, embodiment or example of the invention, non-human vertebrates, such as mice, are generated in a RAG-i or RAG-2 -deficient background, or other suitable genetic background which prevents the production of mature host B and T lymphocytes.
In any configuration, aspect, embodiment or example of the invention, the non-human vertebrate is a rodent, suitably a mouse, and cells of the invention, are rodent cells or ES cells, suitably mouse ES cells.
The ES cells of the present invention can be used to generate animals using techniques well known in the art, which comprise injection of the ES cell into a blastocyst followed by implantation of chimaeric blastocystys into females to produce offspring which can be bred and selected for homozygous recombinants having the required insertion. In one aspect the invention relates to a chimeric non-human vertebrate comprised of ES cell-derived tissue and host embryo derived tissue, wherein the ES cell is according to the invention. In one aspect the invention relates to genetically-altered subsequent generation non-human vertebrates, which include vertebrates homozygous for the human VDJ region.
In one any configuration, aspect, example or embodiment, the genome of the cell or vertebrate comprises: one or more human Ig light chain kappa V regions and one or more human Ig light chain kappa J regions upstream of all or part of a kappa constant region.
In one any configuration, aspect, example or embodiment, the genome of the cell or vertebrate comprises: one or more human Ig light chain lambda V regions and one or more human Ig light chain lambda J regions upstream of all or part of a lambda constant region.
Suitably the light chain Vi and C regions are able to form antibody light chains in vivo capable of forming an antibody with one or more of the chimaeric heavy chains to form an antibody that binds a predetermined antigen.
In such aspects a human kappa and/or lambda region is inserted into the genome, in combination with insertion of the heavy chain VW region or part thereof, upstream of the host heavy chain constant region as disclosed herein.
Suitably the insertion of the human VJC light chain DNA, or part thereof as disclosed above, is made at the equivalent mouse locus. In one aspect the human light chain kappa VJC DNA, or part thereof, is inserted immediately unstream or downstream of the mouse kappa VJC region. In one aspect, the human light chain lambda VJC region or part thereof is inserted immediately upstream or downstream of the mouse lambda VJC region. In one aspect only the human kappa VIC locus is inserted and not the human lambda VJC locus. In one aspect only the human lambda VJC locus is inserted and not the human kappa VJC locus. Insertions may be made using the techniques disclosed herein, and suitably do not remove the host sequences from the genome. In one aspect the non -human mammal host VJC sequences may be inactivated in some way, by mutation, or inversion, or by insertion of the human variable region DNA, or by any other means. In one aspect the cell or non -human mammal of the invention may comprise an insertion of the complete VJC human region.
The human kappa variable region DNA might be inserted into the genome in functional arrangement with a lambda constant region, for example inserted upstream of a lambda constant region.
Alternatively human lambda region variable DNA might be inserted in functional arrangement with a kappa constant region, for example inserted upstream of a kappa constant region.
In one any configuration, aspect, example or embodiment, the genome of the cell or vertebrate comprises: one or more non-human vertebrate control sequences such as the enhancer sequence(s) is maintained upstream of the nonhuman vertebrate Mu constant region, suitably in its native position with respect to the distance from the constant region.
In one any configuration, aspect, example or embodiment, one or more non-human mammal control sequences such as an enhancer sequence(s) are maintained downstream of the nonhuman vertebrate Mu constant region, suitably in its native position with respect to the distance from the constant region.
In one any configuration, aspect, example or embodiment, a non-human mammal switch sequence, suitably the endogenous switch sequence, is maintained upstream of the non-human vertebrate Mu constant region, suitably in its native position with respect to distance from the constant region.
In such location the host enhancer or switch sequences are operative in vivo with the host constant region sequence(s).
In one aspect a switch sequence is neither human, nor native in the non-human mammal, for example in one aspect a non -human mammal switch sequence is not a mouse or human switch sequence. The switch sequence may be, for example, a rodent or primate sequence, or a synthetic sequence. In particular the switch sequence maybe a rat sequence where the non -human mammal is a mouse. By way of example, a mouse or human constant mu sequence maybe placed under the control of a switch sequence from a rat, or chimp, or other switch sequence, suitably capable of allowing isotype switching to occur in vivo.
One combination envisaged is a rat switch with mouse enhancer sequences and mouse constant regions in a mouse cell.
In one aspect the human promoter and/cr other control elements that are associated with the different human V, D on regions are maintained in after insertion of the human VDJ into the mouse genome.
The functional replacement of human promoter or other control regions by non-human mammal promoter or control regions may be carried out by use of recombineering, or other recombinant DNA technologies, to insert a part of the human Ig region (such as a human V region) into a vector (such as a BAC) containing a non-human Ig region. The recombineering/recombinant technique suitably replaces a portion of the non-human (e.g. mouse) DNA with the human Ig region, and thus places the human Ig region under control of the non-human mammal promoter or other control region. Suitably the human coding region for a human V region replaces a mouse V region coding sequence. Suitably the human coding region for a human D region replaces a mouse D region coding sequence. Suitably the human coding region for a human J region replaces a mouse J region coding sequence. In this way human V. D or J regions may be placed under the control of a non-human mammal promoter, such as a mouse promoter.
In one any configuration, aspect, example or embodiment, the human DNA inserted into the genome of the non-human vertebrate or cell are placed under control of the host regulatory sequences or other (non-human, non-host) sequences, In one aspect reference to human coding regions includes both human introns and exons, or in another aspect simply exons and no introns, which may be in the form of cDNA.
It is also possible to use recombineering, or other recombinant DNA technologies, to insert a non -human-mammal (e.g. mouse) promoter or other control region, such as a promoter for a V region or VpreB, into a BAC containing a human Ig region. The A recombineering step then places a portion of human DNA under control of the mouse promoter or other control region.
Generally, insertion of human variable region DNA at or close to the equivalent endogenous locus in the recipient genome is preferred, for example within 1,2,3,4,5,6,7,8,9, or 10kb of the boundary (upstream or downstream) of a host immunoglobulin locus.
In one embodiment of the method of the second configuration of the invention, said genome is homozygous for said transgene and endogenous non-human vertebrate antibody heavy chain expression is inactivated.
In one embodiment of the method of the second configuration of the invention, endogenous non-human vertebrate antibody light chain expression is inactivated.
In one embodiment of the method of the second configuration of the invention, the heavy chain transgene is devoid of a CH1 gene segment and the genome comprises no functional antibody light chain locus.
In one embodiment of the method of the second configuration of the invention, said constant region is a Cmu. For example, the Cmu is a non-human vertebrate Cmu or an endogenous Cmu of said vertebrate or cell.
In one embodiment of the method of the second configuration of the invention) said genome does not comprise a non-human vertebrate (eg, mouse or rat) species VpreBl and/or VpreB2 gene.
In one embodiment of the method of the second configuration of the invention, the genome further comprises a X5 gene; optionally wherein the A5 gene is a AS of a non-human vertebrate (eg, mouse or rat) species and said constant region gene is a constant region gene of the same non-human vertebrate (eg, mouse or rat) species as the X5 gene.
In one embodiment of the method of the second configuration of the invention) said constant region gene and AS gene are endogenous genes of said vertebrate or cell.
In one embodiment of the method of the second configuration of the invention, the method comprises making a progeny of the cell made according to the method, wherein the progeny is homozygous for said heavy chain transgene and endogenous non-human vertebrate antibody expression has been inactivated.
A third configuration of the invention provides A method of promoting B-cell development in a non-human vertebrate (eg, a mouse or rat), the method comprising (a) providing in a non-human vertebrate embryonic stem (Es) cell genome an immunoglobulin transgene capable of expressing an antibody mu heavy chain, wherein the antibody heavy chain comprises a human variable region and a mu constant region (optionally an endogenous non-human vertebrate mu constant region); and creating a first non-human vertebrate from said ES cell or a progeny thereof; (b) providing in a second ES cell genonie a second transgene comprising a human VpreB gene capable of expressing a human VpreB; and creating a second non-human vertebrate from said ES cell or a progeny thereof; and (c) creating by breeding a third non-human vertebrate capable of co-expressing the mu antibody heavy chain and human vpreB wherein a pre-B-ceII receptor can form to promote B-cell development of cells bearing a mu heavy chain in said third vertebrate; the third vertebrate being made by crossine said first and second vertebrates or progeny thereof by breeding to create said third vertebrate, the third vertebrate comprising the first and second transgenes, and wherein endogenous heavy chain expression has been inactivated in said third vertebrate.
In one embodiment of the method of the third configuration of the invention, in (a) the heavy chain transgene is constructed to be devoid of a CH1 gene segment and the genome of the third non-human vertebrate comprises no functional antibody light chain locus.
In one embodiment of the method of the third configuration of the invention, the genome of the third vertebrate does not comprise a non-human vertebrate (eg, mouse or rat) species VpreBl and/or VpreB2 gene.
In one embodiment of the method of the third configuration of the invention, the third non-human vertebrate expresses a AS of a non-human vertebrate (eg, mouse or rat) species and said constant region is a constant region of the same non-human vertebrate (eg, mouse or rat) species as the AS gene; optionally wherein the AS and constant region are an endogenous AS and constant region of mouse or rat.
The invention further provides a transgenic mouse according to the third vertebrate, or a progeny thereof.
In a fourth aspect, the invention provides A method of promoting B-cell development in a non-human vertebrate (eg, a mouse or rat), the method comprising (i) inactivating endogenous heavy chain expression in said vertebrate; (ii) providing in the genome of said vertebrate an immunoglobulin transgene capable of expressing an antibody mu heavy chain, wherein the antibody heavy chain comprises a human variable region and a non-human vertebrate mu constant region (optionally an endogenous non-human vertebrate mu constant region); and (ii) providing in the genome of said vertebrate a second transgene capable of expressing a human VpreB wherein a pre-B-ceIl receptor can form to promote B-cell development of cells bearing a mu heavy chain in said vertebrate; Optionally wherein the genome is homozygous for said first transgene.
In one embodiment of the method of the fourth configuration of the invention, the heavy chain transgene is devoid of a CH1 gene segment and the genome of the vertebrate comprises no functional antibody light chain locus.
In one em bodiment of the method of the fourth configuration of the invention, the genome of the vertebrate does not comprise a non-human vertebrate (eg, mouse or rat) species VpreBl and/or VpreB2 gene.
In one embodiment of the method of the fourth configuration of the invention, the non-human vertebrate expresses a X5 of a non-human vertebrate (eg, mouse or rat) species and said constant region is a constant region of the same non-human vertebrate (eg, mouse or rat) species as the A5 gene; optionally wherein the A5 and constant region are an endogenous X5 and constant region of mouse or rat.
The invention also provides a transgenic mouse according to the fourth configuration, or a progeny thereof.
Chimaeric Surrogate Light Chains The extent of homology between human X5 with mouse and rat XS is relatively low (about 56% amino acid identity). The inventors realised the advantage of species-matching the X5 in the SLC in a transgenic non-human vertebrate with the species of mu constant region in the pre-BCRs in such transgenic animals. Thus, the invention further provides aspects that provide for the possibility of species-matching in vivo the variable and constant regions of the antibody heavy chain with both components (VpreB and X5) of a chimaeric SLC. This is useful for promoting B-cell development in the transgenic non-human vertebrate (eg, a mouse or rat).
Thus, in an embodiment of the vertebrates, cells and methods of the invention, the genome further comDrises a X5 gene: ootionally wherein the AS gene is a AS of a non-human vertebrate (eg, mouse or rat) species and said constant region gene is a constant region gene of the same non-human vertebrate (eg, mouse or rat) species as the X5 gene. For example, the vertebrate is a mouse or rat (or cell is a mouse or rat cell) and the A5 is aX5 that is endogenous to the mouse or rat (or mouse or rat cell). In this example, the constant region of the heavy chain transgene is an constant region (eg, Cmu) endogenous to the mouse or rat. In this way, the A5 is matched for pairing to the constant region at the cell surface in the pre-BlI stage, and the human VpreB is matched for pairing with the human variable region encoded by the transgene.
To this end, the invention provides in a fifth configuration A non-human vertebrate (eg, a mouse or rat) or cell (eg, a mouse cell or rat cell) whose genome comprises an antibody heavy chain transgene, the transgene comprising (a) one or more human VH gene segments, one or more human D gene segments and one or more human JH gene segments operably connected upstream of a constant region gene of a non-human vertebrate (eg, mouse or rat) species so that the transgene is capable of undergoing VDJ recombination in vivo to produce an antibody gene comprising a rearranged VDJC encoding a chimaeric antibody heavy chain having a human variable region and a non-human vertebrate constant region; or (b) a rearranged VDJ encoding a human variable region operably connected upstream of a constant region gene of a non-human vertebrate (eg, mouse or rat) species so that the transgene encodes (optionally following VDJ combination with the constant region) a chimaeric antibody heavy chain having a human variable region and a non-human vertebrate constant region; the genome further comprising one or more genes together encoding a chimaeric surrogate light chain, the surrogate light chain comprising a human VpreB and AS of said non-human vertebrate (eg, mouse or rat) species.
In one embodiment of the method of the fifth configuration of the invention, said genome is homozygous for said transgene and endogenous non-human vertebrate antibody heavy chain expression has been inactivated.
In one embodiment of the method of the fifth configuration of the invention, said constant region gene is a Cmu gene.
In one embodiment of the method of the fifth configuration of the invention, said constant region gene and AS gene are endogenous genes of said vertebrate or cell.
In one embodiment of the method of the fifth configuration of the invention, said genome does not comprise a non-human vertebrate (eg, mouse or rat) species VpreBl and/or VpreB2 gene.
In a sixth configuration, the invention further provides A method of constructing a transgenic non-human vertebrate cell (eg, an ES cell, eg, a mouse or rat ES cell), the method comprising (i) introducing into a non-human vertebrate cell (or an ancestor thereof) one or more human VH gene segments, one or more human D gene segments and one or more human IH gene segments so that said gene segments are operably connected upstream of an endogenous non-human vertebrate constant region gene, wherein in said cell or a progeny thereof the transgene is capable of undergoing VDJ recombination in vivo to produce an antibody gene comprising a rearranged VDJC encoding a chimaeric antibody heavy chain having a human variable region and a non-human vertebrate constant region; or (U) introducing into a non-human vertebrate cell (or an ancestor thereof) a rearranged VDJ encoding a human variable region so that said VDJ is operably connected upstream of an endogenous non-human vertebrate constant region gene, wherein in said cell or a progeny thereof the transgene encodes (optionally following VDJ combination with the constant region) a chimaeric antibody heavy chain having a human variable region and a non-human vertebrate constant region; and wherein the method further comprises introducing into the cell a human VpreB gene such that the cell or a progeny thereof is capable of expressing a chimaeric surrogate light chain as well as said chimaeric antibody heavy chain, wherein the surrogate light chain comprises a human VpreB and an endoeenous non-human vertebrate AS.
In one embodiment of the method of the sixth configuration of the invention, said constant region gene is a Cmu gene.
In one embodiment of the method of the sixth configuration of the invention, each endogenous non-human vertebrate VpreB gene is inactivated or deleted from said genome.
In one embodiment, the method of the sixth configuration of the invention comprises making a progeny of the cell made according to the method, wherein the progeny is homozygous for said heavy chain transgene and endogenous non-human vertebrate antibody heavy chain expression has been inactivated.
In a seventh configuration, the invention provides A method of promoting B-cell development in a non-human vertebrate (eg, a mouse or rat), the method comprising (a) providing in a non-human vertebrate embryonic stem (Es) cell an immunoglobulin transgene capable of expressing a chimaeric antibody mu heavy chain, wherein the chimaeric antibody heavy chain comprises a human variable region and an endogenous non-human vertebrate mu constant region; and creating a first non-human vertebrate from said ES cell or a progeny thereof; (b) providing in a second ES cell a second transgene capable of expressing a human VpreB so that the human VpreB and an endogenous ItS form a chimaeric surrogate light chain; and creating a second non-human vertebrate from said ES cell or a progeny thereof; and (c) creating by breeding a third non-human vertebrate capable of co-expressing the chiniaeric mu antibody heavy chain and chimaeric surrogate light chain wherein a pre-BCR can form to promote B-cell development in said third vertebrate; the third vertebrate being made by crossing said first and second vertebrates or progeny thereof by breeding to create said third vertebrate, the third vertebrate comprising the first and second transgenes, and wherein endogenous heavy chain expression has been inactivated in said third vertebrate.
The invention provides a transgenic mouse according to the vertebrate of this method, or a progeny thereof.
In an eighth configuration, the invention provides A method of promoting B-cell development in a non-human vertebrate (eg, a mouse or rat), the method comprising (i) inactivating endogenous heavy chain expression in said vertebrate; (ii) providing in the genome of said vertebrate an immunoglobulin transgene capable of expressing a chimaeric antibody mu heavy chain, wherein the chimaeric antibody heavy chain comprises a human variable region and an endogenous non-human vertebrate mu constant region; and (U) providing in the genome of said vertebrate a second transgene capable of expressing a human VpreB so that the human VpreB and an endogenous X5 form a chimaeric surrogate light chain; so that the vertebrate is capable of co-expressing the chimaeric mu antibody heavy chain and chimaeric surrogate light chain wherein a pre-BCR can form to promote B-cell development in said vertebrate; Optionally wherein the genome is homozygous for said first transgene.
A transgenic mouse according to the vertebrate of this method, or a progeny thereof, is provided.
In any configuration, aspect, embodiment or example of the invention, the inserted human genes may be derived from the same individual or different individuals, or be synthetic or represent human consensus sequences.
Techniques for constructing non-human vertebrates and vertebrate cells whose genomes comprise a transgene containing human V, J and D regions are well known in the art. For example, reference is made to WO2011004192, U57501552, U56673986, US6130364, W02009/076464 and U56586251, the disclosures of which are incorporated herein by reference in their entirety.
In one embodiment in any configuration of the invention, the vertebrate is a non-human mammal and the vertebrate cell is a non-human mammalian cell. In one embodiment in any configuration of the invention, the vertebrate is a mouse, rat, rabbit, Camelid (eg, a llama, alpaca or camel) or shark; or the vertebrate cell is a mouse, rat, rabbit, Camelid (eg, a llama, alpaca or camel) or shark cell.
In one aspect the human heavy chain gene segments are inserted into the genome so that they are placed under control of the host regulatory sequences (eg, enhancers, promoters and/or switches) or other (non-human, non-host) sequences. In one aspect reference to human coding regions includes both human introns and exons, or in another aspect simply exoris and no introns, which may be in the form of cDNA.
Alternatively it is possible to use recombineering, or other recombinant DNA technologies, to insert a non human-vertebrate (e.g. mouse) promoter or other control region, such as a promoter for a V region, into a BAC containing a human Ig region. The recombineering step then places a portion of human DNA under control of the mouse promoter or other control region.
The invention also relates to a cell line which is grown from or otherwise derived from cells as described herein, including an immortalised cell line. The cell line may comprise inserted human V, D on genes as described herein, either in germline configuration or after rearrangement following in vivo maturation. The cell may be immortalised by fusion to a tumour cell to provide an antibody producing cell and cell line, or be made by direct cellular immortalisation.
In one aspect the non-human vertebrate of any configuration of the invention is able to generate a diversity of at least 1 X io6 different functional chimaeric immunoglobulin sequence combinations.
Optionally in any configuration of the invention the constant region is endogenous to the vertebrate n endogenous switch. In one embodiment, the constant region comprises a Cgamma (CI) region and/or a Smu (Si4 switch. Switch sequences are known in the art, for example, see Nikaido et al, Nature 292: 845-848 (1981) and also W02011004192, US7501552, U56673986, US6130364, W02009/076464 and U56586251, eg, SEQ ID NOs: 9-24 disclosed in U57501552. Optionally the constant region comprises an endogenous S gamma switch and/or an endogenous Smu switch. One or more endogenous switch regions can be provided, in one embodiment, by constructing a transgenic immunoglobulin locus in the vertebrate or cell genome in which at least one human V region, at least one human J region, and optionally at least one human D region, or a rearranged VDJ or Vi region, are inserted into the genome in operable connection with a constant region that is endogenous to the vertebrate or cell. For example, the human V(D)J regions or rearranged VDJ or Vi can be inserted in a cis orientation onto the same chromosome as the endogenous constant region. A trans orientation is also possible, in which the human V(D)J regions or rearranged VDJ or Vi are inserted into one chromosome of a pair (eg, the chromosome 6 pair in a mouse or the chromosome 4 in a rat) and the endogenous constant region is on the other chromosome of the pair, such that trans-switching takes place in which the human V(D)i regions or rearranged VDJ or Vi are spliced inoperable linkage to the endogenous constant region. In this way, the vertebrate can express antibodies having a chain that comprises a variable region encoded all or in part by human V(D)J or a rearranged VDJ or Vi, together with a constant region (eg, a Cgamma or Cmu) that is endogenous to the vertebrate.
Human variable regions are suitably inserted upstream of non-human vertebrate constant region, the latter comprising all of the DNA required to encode the full constant region or a sufficient portion of the constant region to allow the formation of an effective chimaeric antibody capable of specifically recognising an antigen.
In one aspect the chimaeric antibodies or antibody chains have a part of a host constant region sufficient to provide one or more effector functions seen in antibodies occurring naturally in a host vertebrate, for example that they are able interact with Ec receptors, and/or bind to complement.
Reference to a chimaeric antibody or antibody chain having a non-human vertebrate constant region herein therefore is not limited to the complete constant region but also includes chimaeric antibodies or chains which have all of the host constant region) or a part thereof sufficient to provide one or more effector functions. This also applies to non-human vertebrate mammals and cells and methods of the invention in which human variable region DNA may be inserted into the host genome such that it forms a chimaeric antibody chain with all or part of a host constant region.
In one aspect the whole of a host non-human vertebrate constant region is operably linked to human variable region DNA.
The host non-human vertebrate constant region herein is optionally the endogenous host wild-type constant region located at the wild type locus, as appropriate for the heavy or light chain. For example, the human heavy chain DNA is suitably inserted on mouse chromosome 12, suitably adjacent the mouse heavy chain constant region, where the vertebrate is a mouse.
In one optional aspect where the vertebrate is a mouse, the insertion of the human DNA, such as the human VDJ region is targeted to the region between the 14 exon and the C1t locus in the mouse genome IgH locus, and in one aspect is inserted between coordinates 114,667,090 and 114,665,190, suitably at coordinate 114,667,091, after 114,667,090. In one aspect the insertion of the human DNA, such as the human light chain kappa Vi is targeted into mouse chromosome 6 between coordinates 70,673,899 and 70,675,515, suitably at position 70,674,734, or an equivalent position in the lambda mouse locus on chromosome 16.
In one aspect the host non-human vertebrate constant region for forming the chimaeric antibody may be at a different (non endogenous) chromosomal locus. In this case the inserted human DNA, such as the human variable VDJ or Vi region(s) may then be inserted into the non-human genonie at a site which is distinct from that of the naturally occurring heavy or light constant region. The native constant region may be inserted into the genome, or duplicated within the genome, at a different chromosomal locus to the native position, such that it is in a functional arrangement with the human variable region such that chimaeric antibodies of the invention can still be produced.
In one aspect the human DNA is inserted at the endogenous host wild-type constant region located at the wild type locus between the host constant region and the host VDJ region.
Reference to location of the variable region upstream of the non-human vertebrate constant region means that there is a suitable relative location of the two antibody portions, variable and constant, to allow the variable and constant regions to form a chimaeric antibody or antibody chain in vivo in the vertebrate. Thus, the inserted human DNA and host constant region are in operable connection with one another for antibody or antibody chain production.
In one aspect the inserted human DNA is capable of being expressed with different host constant regions through isotype switching. In one aspect isotype switching does not require or involve trans switching. Insertion of the human variable region DNA on the same chromosome as the relevant host constant region means that there is no need for trans-switching to produce isotype switching.
In the present invention, optionally host non-human vertebrate constant regions are maintained and it is preferred that at least one non-human vertebrate enhancer or other control sequence, such as a switch region, is maintained in functional arrangement with the non-human vertebrate constant region, such that the effect of the enhancer or other control sequence, as seen in the host vertebrate, is exerted in whole or in part in the transgenic animal. This approach is designed to allow the full diversity of the human locus to be sampled, to allow the same high expression levels that would be achieved by non-human vertebrate control sequences such as enhancers, and is such that signalling in the B-cell, for example isotype switching using switch recombination sites, would still use non-human vertebrate sequences.
A non-human vertebrate having such a genome would produce chimaeric antibodies with human variable and non-human vertebrate constant regions, but these are readily humanized, for example in a cloning step. Moreover the in vivo efficacy of these chimaeric antibodies could be assessed in these same animals.
In one aspect the inserted human IgH VDJ region comprises, in germline configuration, all of the V, D and I regions and intervening sequences from a human. Optionally, non-functional V and/or D and/or 1 gene segments are omitted. For example, VH which are inverted or are pseudogenes may be omitted.
In one aspect 800-1000kb of the human IgH VDJ region is inserted into the non-human vertebrate lgF-l locus, and in one aspect a 940, 950 or 960kb fragment is inserted, Suitably this includes bases 105,400,051 to 106,368,585 from human chromosome 14.
In one aspect the inserted IgH human fragment consists of bases 105,400,051 to 106,368,585 from chromosome 14. In one aspect the inserted human heavy chain DNA, such as DNA consisting of bases 105,400,051 to 106,368,585 from chromosome 14, is inserted into mouse chromosome 12 between the end of the mouse 14 region and the Ep region, suitably between co-ordinates 114,667,090 and 114,665,190, or at co-ordinate 114,667,091, after 114,667,090. In one aspect the insertion is between co-ordinates 114,667,089 and 114,667,090 (co-ordinates refer to NCBI m37, for the mouse C57RL/6J strain), or at equivalent position in another non-human vertebrate genome.
A cell or non-human vertebrate of the invention, in one embodiment, comprises an insertion of human heavy chain variable region DNA between co-ordinates 114,665, 183 and 114, 566, 725, such as between 114 666 283 and 114 666 625, optionally between co-ordinates 114,666,335 and 114,666,536, optionally between 114,666,385 and 114,666,486, or between 114,666,425 and 114,666,446, or between 114,666,435 and 114,666,436 of mouse chromosome 12 with reference to NCBIM37 for the mouse genome, relating to mouse strain C57BL/6J or an equivalent position of mouse chromosome 12 from a different mouse strain or an equivalent position in the genome of another non-human vertebrate, e.g., a rat. The insertion between co-ordinates 114,666,435 and 114,666,436 relating to mouse strain CS7BL/6J is equivalent to an insertion between co-ordinates 1207826 and 1207827 on chromosome 12 with reference to the 129/SvJ genomic sequence of the geneBank access number NT114985.2. An insertion may be made at equivalent position in another genonie, such as another mouse genome. In an example of this embodiment, the cell or mammal of the invention comprises a human IgH VDJ region which comprises or consists of nucleotides 106,328,851-107,268,544, such as nucleotides 106,328,901-107,268,494, such as nucleotides 106,328,941-107,268,454, such as nucleotides 106,328,951-107,268,444 of human Chromosome 14, with reference to the GRCH37/hgl9 sequence database, or insertion of equivalent nucleotides relating to chromosome 14 from a different human sequence or database. The human insertion may be made between the regions indicated above.
In one aspect there is inserted into the genome the human kappa Vi region which comprises, in germline configuration, all of the V and J regions and intervening sequences from a human.
Optionally, non-functional V and/or i gene segments are omitted.
Suitably this includes bases 88,940,356 to 89,857,000 from human chromosome 2, suitably approximately 917kb. In a further aspect the light chain Vi insert may comprise only the proximal clusters of V segments and I segments. Such an insert would be of approximately 473 kb.
In one aspect the human light chain kappa DNA, such as the human IgK fragment of bases 88,940,356 to 89,857,000 from human chromosome 2, is suitably inserted into mouse chromosome 6 between coordinates 70,673,899 and 70,675,515, suitably at position 70,674,734.
In one aspect the human lambda V (or Vi) region is inserted into the genome, which comprises, in germline configuration, all of the V (and optionally i) regions and intervening sequences from a human (which will thus include the human VpreB gene and a promoter). Suitably this includes analogous bases to those selected for the kappa fragment, from human chromosome 2. optionally, non-functionai V and/or I gene segments are omitted.
All specific human fragments described above may vary in length, and may for example be longer or shorter than defined as above, such as 500 bases, 1KB, 2K, 3K, 4K, 5KB, 10KB, 20KB, 30KB, 40KB or 50KB or more, which suitably comprise all or part of the human V(D)J region, whilst preferably retaining the requirement for the final insert to comprise human genetic material encoding the complete heavy chain region and light chain region, as appropriate, as described above.
In one aspect the 3' end of the last inserted human sequence, generally the last human J sequence, is inserted less than 2kb, preferably less than 1KB from the human/non-human vertebrate (eg, human/mouse or human/rat) join region.
Optionally, the genome is homozygous at at least the IgH, or a second, or all three immunoglobulin loci (IgH, lgX and lgK).
In another aspect the genome may be heterozygous at one or more of the loci, such as heterozygous for DNA encoding a chimaeric antibody chain and native (host cell) antibody chain. In one aspect the genome may be heterozygous for DNA capable of encoding 2 different antibody heavy chains encoded by immunoglobulin transgenes of the invention, for example, comprising 2 different chimaeric heavy chains and a human lambda light chain.
In one aspect the invention relates to a non-human vertebrate or cell, and methods for producing said vertebrate or cell, as described herein, wherein the inserted human DNA, such as the human IgH VDJ region and/or light chain V, J regions are found on only one allele and not both alleles in the mammal or cell. In this aspect a mammal or cell has the potential to express both an endogenous host antibody heavy or light chain and a chimaeric heavy or light chain.
In one embodiment in any configuration of the invention, the genome has been modified to prevent or reduce the expression of fully-endogenous antibody. Examples of suitable techniques for doing this can be found in WO2011004192, US7501552, US6673986, U56130364, W02009/076464, EP1399559 and U56586251, the disclosures of which are incorporated herein by reference. In one embodiment, the non-human vertebrate VDJ region of the endogenous heavy chain immunoglobulin locus, and optionally Vi region of the endogenous light chain immunoglobulin loci (lambda and/or kappa loci), have been inactivated. For example, all or part of the non-human vertebrate VDJ region is inactivated by inversion in the endogenous heavy chain immunoglobulin locus of the mammal, optionally with the inverted region being moved upstream or downstream of the endogenous Ig locus. For example, all or part of the non-human vertebrate Vi region is inactivated by inversion in the endogenous kappa chain immunoglobulin locus of the mammal, optionally with the inverted region being moved upstream or downstream of the endogenous Ig locus. For example, all or part of the non-human vertebrate Vi region is inactivated by inversion in the endogenous lambda chain immunoglobulin locus of the mammal, optionally with the inverted region being moved upstream or downstream of the endogenous Ig locus. In one embodiment the endogenous heavy chain locus is inactivated in this way as is one or both of the endogenous kappa and lambda loci.
Additionally or alternatively, the vertebrate has been generated in a genetic background which prevents the production of mature host B and T lymphocytes, optionally a RAG-i-deficient and/or RAG-2 deficient background. See US5859301 for techniques of generating RAG-i deficient animals.
In one embodiment in any configuration of the invention, the human V,i and optional D regions are provided by all or part of the human IgH locus; optionally wherein said all or part of the IgH locus includes substantially the full human repertoire of IgH V, D and J regions and intervening sequences.
A suitable part of the human IgH locus is disclosed in W02011004192. In one embodiment, the human IgH part includes (or optionally consists of) bases 105,400,051 to 106,368,585 from human chromosome 14 (coordinates from NCBI36). Additionally or alternatively, optionally wherein the vertebrate is a mouse or the cell is a mouse cell, the human V, J and optional D regions are inserted into mouse chromosome 12 at a position corresponding to a position between coordinates 114,667,091 and 114,665,190, optionally at coordinate 114,667,091 (coordinates from NCBIM37, relating to mouse strain C57BL/6J).
In one embodiment of any configuration of the vertebrate or vertebrate cell of the invention when the vertebrate is a mouse, (i) the constant region comprises a mouse or rat switch and optionally a mouse CF.t region. For example the constant region is provided by the constant region endogenous to the mouse, eg, by inserting human V(D)i region sequences into operable linkage with the endogenous constant region of a mouse genome or mouse cell genome.
In one embodiment of any configuration of the vertebrate or vertebrate cell of the invention when the vertebrate is a rat, (i) the constant region comprises a mouse or rat Sp switch and optionally a rat Cji region. For example the constant region is provided by the constant region endogenous to the rat, eg, by inserting human V(D)J region sequences into operable linkage with the endogenous constant region of a rat genome or rat cell genome.
In one embodiment of any configuration of the vertebrate or vertebrate cell of the invention the genonie comprises an antibody light chain transgene which comprises all or part of the human lgX locus including at least one human JA region and at least one human CA region, optionally Cx6 and/or C7. Optionally, the transgene comprises a plurality of human JA regions,optionally two or more of i1, i2, J6 and 1x7, optionally all of J4, Jx2, J6 and i7. The human lambda immunoglobulin locus comprises a unique gene architecture composed of serial i-c clusters. In order to take advantage of this feature, the invention in optional aspects employs one or more such human i-C clusters inoperable linkage with the constant region in the transgene, eg, where the constant region is endogenous to the non-human vertebrate or non-human vertebrate cell. Thus, optionally the transgene comprises at least one human i-C cluster, optionally at least J7-c7. The construction of such transgenes is facilitated by being able to use all or part of the human lambda locus such that the transgene comprises one or more i-C clusters in germline configuration, advantageously also including intervening sequences between clusters and/or between adjacent I and C regions in the human locus. This preserves any regulatory elements within the intervening sequences which may be involved in Vi and/or ic recombination and which may be recognised by AID (activation-induced deaminase) or AID homologues.
Where endogenous regulatory elements are involved in cSR (class-switch recombination) in the non-human vertebrate, these can be preserved by including in the transgene a constant region that is endogenous to the non-human vertebrate. In the invention, one can match this by using an AID or AID homologue that is endogenous to the vertebrate or a functional mutant thereof. Such design elements are advantageous for maximising the enzymatic spectrum for SHM (somatic hypermutation) and/or CSR and thus for maximising the potential for antibody diversity.
Optionally) the lambda transgene comprises a human EX enhancer. Optionally, the kappa transgene comprises a human EK enhancer. Optionally) the heavy chain transgene comprises a heavy chain human enhancer.
In one embodiment of any configuration of the invention the constant region is endogenous to the non-human vertebrate or derived from such a constant region. For example, the vertebrate is a mouse or the cell is a mouse cell and the constant region is endogenous to the mouse. For example, the vertebrate is a rat or the cell is a rat cell and the constant region is endogenous to the rat.
In one embodiment of any configuration of the invention the heavy chain transgene comprises a plurality human IgH V regions, a plurality of human D regions and a plurality of human J regions, optionally substantially the full human repertoire of IgH V, D and J regions.
In one embodiment of any configuration of the invention, the vertebrate or cell comprises a heavy chain further transgene, the further transgene comprising at least one human IgH V region, at least one human D region and at least one human J region, optionally substantially the full human repertoire of IgH V, D and J regions.
In one embodiment of any configuration of the invention, (i) the heavy chain transgene comprises at least one human IgH V region) at least one human J region, and optionally at least one human D region; and (ii) the vertebrate or cell comprises a kappa transgene, the kappa transgene comprising at least one human lgK V region and at least one human J region.
In one embodiment of any configuration of the invention, (i) the heavy chain transgene comprises at least one human IgH V region, at least one human J region, and optionally at least one human D region; and (ii) the vertebrate or cell comprises a lambda transgene, the lambda transgene comprising at least one human lgX V region and at least one human J region.
In one embodiment of any configuration of the invention, (i) the heavy chain transgene comprises substantially the full human repertoire of IgH V, D and J regions; and (ii) the vertebrate or cell comprises substantially the full human repertoire of lgK V and J regions and/or substantially the full human repertoire of lgX V and J regions.
An aspect provides a B-cell, hybridoma or a stem cell, optionally an embryonic stem cell or haematopoietic stem cell, according to any configuration of the invention. In one embodiment, the cell is a BALB/c, JM8 or AB2.1 or AB2.2 embryonic stem cell (see discussion of suitable cells, and in particular JMS and AB2.1 cells, in W02011004192, which disclosure is incorporated herein by reference). In one aspect the ES cell is derived from the mouse BALB/c, C57BL/6N, C57BL/6J 129S5 or 129Sv strain.
An aspect provides a method of isolating an antibody or nucleotide sequence encoding said antibody, the method comprising (a) immunising (see e.g. Harlow, E. & Lane, D. 1998, 5th edition, Antibodies: A Laboratory Manual, Cold Spring Harbor Lab. Press, Plainview, NY; and Pasqualini and Arap, Proceedings of the National Academy of Sciences (2004) 101:257-259) a vertebrate according to any configuration or aspect of the invention with an antigen such that the vertebrate produces antibodies; and (b) isolating from the vertebrate an antibody that specifically binds to said antigen and/or a nucleotide sequence encoding at least the heavy and/or the light chain variable regions of said antibody; optionally wherein the variable regions of said antibody are subsequently joined to a human constant region. Such joining can be effected by techniques readily available in the art, such as using conventional recombinant DNA and RNA technology as will be apparent to the skilled person. See e.g. Sambrook, J and Russell, D. (2001, 3'd edition) Molecular Cloning: A Laboratory Manual (Cold Spring Harbor Lab. Press, Plainview, NY).
Suitably an immunogenic amount of the antigen is delivered. The invention also relates to a method for detecting a target antigen comprising detecting an antibody produced as above with a secondary detection agent which recognises a portion of that antibody.
Isolation of the antibody in step (b) can be carried out using conventional antibody selection techniques, eg, panning for antibodies against antigen that has been immobilised on a solid support, optionally with iterative rounds at increasing stringency, as will be readily apparent to the skilled person.
As a further optional step, after step (b) the amino acid sequence of the heavy and/or the light chain variable regions of the antibody are mutated to improve affinity for binding to said antigen.
Mutation can be generated by conventional techniques as will be readily apparent to the skilled person, eg, by error-prone PCR. Affinity can be determined by conventional techniques as will be readily apparent to the skilled person, eg, by surface plasmon resonance, eg, using BiacoreTM.
Additionally or alternatively, as a further optional step, after step (b) the amino acid sequence of the heavy and/or the light chain variable regions of the antibody are mutated to improve one or more biophysical characteristics of the antibody, eg, one or more of melting temperature, solution state (monomer or dimer), stability and expression (eg, in CR0 orE co/i).
An aspect provides an antibody produced by the method of the invention, optionally for use in medicine, eg, for treating and/or preventing a medical condition or disease in a patient, eg, a human.
An aspect provides a nucleotide sequence encoding the antibody of the invention, optionally wherein the nucleotide sequence is part of a vector. Suitable vectors will be readily apparent to the skilled person, eg, a conventional antibody expression vector comprising the nucleotide sequence together in operable linkage with one or more expression control elements.
An aspect provides a pharmaceutical composition comprising the antibody of the invention and a diluent, excipient or carrier, optionally wherein the composition is contained in an IV container (eg, and IV bag) or a container connected to an IV syringe.
An aspect provides the use of the antibody of the invention in the manufacture of a medicament for the treatment and/or prophylaxis of a disease or condition in a patient, eg a human.
In a further aspect the invention relates to a method for producing an antibody specific to a desired antigen the method comprising immunizing a transgenic non-human vertebrate as above with a predetermined antigen and recovering a chimaeric antibody (see e.g. Harlow, E. & Lane, D. 1998, 5th edition, Antibodies: A Laboratory Manual, Cold Spring Harbor Lab. Press, Plainview, NY; and Pasqualini and Arap, Proceedings of the National Academy of Sciences (2004) 101:257-259). Suitably an immunogenic amount of the antigen is delivered. The invention also relates to a method for detecting a target antigen comprising detecting an antibody produced as above with a secondary detection agent which recognises a portion of that antibody.
In a further aspect the invention relates to a method for producing a fully humanised antibody comprising immunizing a transgenic non-human vertebrate as above with a predetermined antigen, recovering a chimaeric antibody or cells expressing the antibody, and then replacing the non-human vertebrate constant region with a human constant region. This can be done by standard cloning techniques at the DNA level to replace the non-human vertebrate constant region with an appropriate human constant region DNA sequence -see e.g. Sambrook, J and Russell, D. (2001, 3'd edition) Molecular Cloning: A Laboratory Manual (Cold Spring Harbor Lab. Press, Plainview, NY).
In a further aspect the invention relates to humanised antibodies and antibody chains produced according to the present invention, both in chimaeric and fully humanised form, and use of said antibodies in medicine. The invention also relates to a pharmaceutical composition comprising such an antibody and a pharmaceutically acceptable carrier or other excipient.
Antibody chains containing human sequences, such as chimaeric human-non-human antibody chains, are considered humanised herein by virtue of the presence of the human protein coding regions region. Fully humanised antibodies may be produced starting from DNA encoding a chimaeric antibody chain of the invention using standard techniques.
Methods for the generation of both monoclonal and polyclonal antibodies are well known in the art, and the present invention relates to both polyclonal and monoclonal antibodies of chimaeric or fully humanised antibodies produced in response to antigen challenge in non-human vertebrates of the present invention.
In a yet further aspect, chimaeric antibodies or antibody chains generated in the present invention may be manipulated, suitably at the DNA level, to generate molecules with antibody-like properties or structure, such as a human variable region from a heavy chain absent a constant region, for example a domain antibody; or a human variable region with any constant region from either heavy or light chain from the same or different species; or a human variable region with a non-naturally occurring constant region; or human variable region together with any other fusion partner. The invention relates to all such chimaeric antibody derivatives derived from chimaeric antibodies identified according to the present invention.
In an example, the genome of the cell or non-human vertebrate of the invention encodes an antibody comprising an antibody chain having a human heavy chain variable region upstream of a mouse light chain constant region in combination with one of: a fully human antibody light chain; a non-human vertebrate (e.g., mouse or rat) antibody light chain; a chimaeric non-human vertebrate (e.g., mouse or rat) -human antibody chain; or an antibody chain having a human heavy chain variable region upstream of a non-human vertebrate (e.g., mouse or rat) light chain constant region; In a further aspect, the invention relates to use of non-human vertebrates of the present invention in the analysis of the likely effects of drugs and vaccines in the context of a quasi-human antibody repertoire.
The invention also relates to a method for identification or validation of a drug or vaccine, the method comprising delivering the vaccine or drug to a vertebrate of the invention and monitoring one or more of: the immune response, the safety profile; the effect on disease.
The invention also relates to a kit comprising an antibody or antibody derivative as disclosed herein and either instructions for use of such antibody or a suitable laboratory reagent, such as a buffer, antibody detection reagent.
The invention also relates to a method for making an antibody, or part thereof, the method comprising providing: (i) a nucleic acid encoding an antibody) or a part thereof, obtained according to the present invention; or (ii) sequence information from which a nucleic acid encoding an antibody obtained according to the present invention, or part thereof, can be expressed to allow an antibody to be produced.
In an embodiment, the invention provides The non-human vertebrate, mouse, rat, cell or method of any preceding configuration, wherein the genome comprises (a) said antibody heavy chain transgene; and (b) an antibody kappa light chain transgene and/or an antibody lambda chain transgene; wherein all of the V. D and J in said transgenes are human V, D and J; wherein endogenous antibody heavy and light chain expression has been inactivated; and optionally wherein said genome is homozygous for said heavy and light chain transgenes.
In an embodiment, the the kappa and lambda chain transgenes comprise constant regions of said non-human vertebrate species capable of pairing with the constant region of the heavy chain.
In an embodiment, the heavy chain transgene comprises a substantially complete human functional VH, D and JH repertoire.
In an embodiment, the kappa chain transgene comprises a substantially complete human functional VK and JK repertoire; and the lambda chain transgene comprises a substantially complete human functional VA and iN repertoire.
In a ninth embodiment, the invention provides A transgenic mouse or rat, or a transgenic mouse or rat cell (eg, an ES cell), whose genome comprises (a) an antibody heavy chain transgene, the transgene comprising a substantially complete human functional VH, D and JH repertoire operably connected upstream of an endogenous (mouse or rat) mu constant region gene so that the transgene is capable of undergoing VDJ recombination in vivo to produce an antibody gene comprising a rearranged VDJC encoding an antibody heavy chain having a human variable region and an endogenous mu constant region; (b) an antibody kappa light chain transgene and/or an antibody lambda chain transgene; -wherein all of the V, D and J in said transgenes are human V, D and I; -wherein the kappa chain transgene comprises a substantially complete human functional VK and JK repertoire; and the lambda chain transgene comprises a substantially complete human functional VA and iN repertoire; -wherein the kappa and/or lambda chain transgenes comprise endogenous constant regions capable of pairing with the constant region of the heavy chain; (c) a human VpreB gene capable of expressing a human VpreB; -optionally wherein the human VpreB gene is operably linked to an endogenous VpreB promoter; and (d) an endogenous As gene capable of expressing an endogenous As; -wherein endogenous antibody heavy and light chain expression has been inactivated; and -optionally wherein said genome is homozygous for said heavy and light chain transgenes.
In a tenth embodiment, the invention provides A transgenic mouse or rat, or a transgenic mouse or rat cell (eg, an ES cell), whose genome comprises (a) an antibody heavy chain transgene, the transgene comprising a substantially complete human functional VH, D and JH repertoire operably connected upstream of an endogenous (mouse or rat) mu constant region gene so that the transgene is capable of undergoing VDJ recombination in vivo to produce an antibody gene comprising a rearranged VDJC encoding an antibody heavy chain having a human variable region and an endogenous mu constant region; -wherein all of the V, D and J in said transgene are human V. D and J; -wherein the heavy chain transgene is devoid of a CH1 gene segment and the genome of the vertebrate comprises no functional antibody light chain locus; -wherein endogenous antibody heavy chain expression has been inactivated; and -optionally wherein said genome is homozygous for said heavy chain transgene; (c) a human VpreB gene capable of expressing a human VpreB; -optionally wherein the human VpreB gene is operably linked to an endogenous VpreB promoter; and (d) an endogenous AS gene capable of expressing an endogenous AS.
Insertion of Human VrreB Gene As described above, precise insertion of exogenous DNA (such as human VpreB or antibody gene segment DNA) can be effected by homolgous recombination, RMCE or other techniques that will be apparent to the skilled person. Vector manipulation can be effected using recombineering as is well known.
The human VpreB gene is found in human cells within the human antibody VA gene segment cluster (in the nucleotide region between human IgA V gene segments lV-53 and 5-52 (the nucleotide region being oositions 22599200 to 22599926 on human chromosome 22)). Thus, in one embodiment, a Si nucleotide sequence found between human IgA V gene segments IV-53 and 5-52, eg, positions 22599200 to 22599926 on human chromosome 22, is inserted into the genome of the cell or vertebrate, wherein the nucleotide sequence comprises a human VpreB gene and optionally the associated human promoter. For example, the nucleotide sequence is inserted in the endogenous lambda antibody locus upstream of the endogenous lambda constant region. In one embodiment, a region of the human VA gene segment cluster comprising at least one (or substantially all) human VA gene segments and the human VpreB and promoter is inserted upstream of the endogenous (eg, mouse or rat) CA. In one example, the insertion is between the most 3) endogenous JA and the endogenous CA. In another embodiment, the insertion is in the endogenous VA cluster, optionally replacing the VA in whole or in part. In one embodiment, all of the human lambda VA gene segment cluster (including introns) is inserted into the genome so that it replaces the enogenous VA gene segment cluster. In one embodiment, the cell or vertebrate is a mouse or rat, so "endogenous" refers to the regions found in the genome of a mouse or rat.
Expression of the AS and VpreB genes is B-cell lineage restricted and is also subject to stage-specific regulation during B-cell development. Binding sites for the transcription factors EBF, E47, Pax-5 and Ikaros are present in the the AS and VpreB promoters. The mouse VpreBl and AS genes are both located in the AS -VpreBl locus and 4kb away from each other. A locus control region (LCR) located within the AS -VpreBl locus provides the regulatory elements for these genes are. Hence, the region around the two genes are important for the transcription of the VpreBl and AS genes.
Furthermore, these two genes are subject to tight stage-specific control during B-cell development.
For activation at the pro-B cell stage, early B cell factor (EBF) initiates the activation of the AS -VpreBl locus. It has been observed that the EBF with some other factors (E2A5) bind the promoters of the two genes. The transcription of these two genes has to be silenced during the transition from the pre-B to the immature B cell stage, in order for light chain rearrangement and binding with heavy chain. Ikaros gene acts as a repressor by competing with EBF for binding to the same sites in the promoters of the VpreBl and AS genes. In summary, the AS -VpreBl locus seems to be important for the temporally-controlled activation and inactivation of the surrogate light chain during B-cell development. See:- * Mol Cell Biol. 1999 Jan;19(1):671-9; Analysis of mice with single and multiple copies of transgenes reveals a novel arrangement for the lambdaS-VpreBl locus control region; Sabbattini P, Georgiou A, Sinclair C, Dillon N. * Seminars in Immunology, 2005 Apr; 17(2):121-7; The lambdas-VpreBl locus--a model system for studying gene regulation during early B cell development; Sabbattini P, Dillon N. Thus, in one embodiment the invention provides a non-human vertebrate, mouse, rat, cell or method according to any configuration, aspect, embodiment or example, wherein the expression of the human VpreB gene is under endogenous control. For example, the vertebrate is a mouse or rat or the cell is a mouse or rat cell (eg, an ES cell) and the human VpreB is under the control of the gene expression control elements of the mouse or rat (cell). This can be achieved, in one example, by replacing an endogenous VpreB (eg, VpreBl) gene with a human VpreB gene at the location where the former usually resides in the genome. Precise gene replacement in genomes can be carried out by homolgous recombination or RMCE as herein described.
Mouse VpreBl and VpreB2 are found at the following position in a mouse genome:-Mouse VpreBl: chromosome 16: 16868494 to 16869348 Mouse VpreB2: chromosome 16: 17980658 to 17981173 Thus, the invention provides a non-human vertebrate, mouse, rat, cell or method according to any configuration, aspect, embodiment or example, wherein the human VpreB gene is operably linked to an endogenous promoter, optionally an endogenous VpreB promoter (eg, a VpreBl or VpreB2 promoter). Thus, where the vertebrate (or cell) is a mouse or rat (cell), the human VpreB is operably linked to a VpreBl or VpreB2 promoter of said mouse or rat (cell). Operable linkage enables the promoter to control expression of the human gene. It is desirable to use endogenous gene expression control (eg, using an endogenous promoter) for human VpreB expression since this will harness the endogenous (eg, mouse or rat) temporal expression control for turning on and off the human VpreB gene (and coordinating this with AS expression) during the various stages of B-cell development, particularly up to the point where light chain pairing with heavy chains occurs in immature B-cells. Thereafter, the usual switching off of SLC-component gene expression can be controlled properly by the endogenous control mechanisms when the pre-BCR is no longer oarticinatinp in B-cell maturation.
Thus, the invention provides a non-human vertebrate, mouse, rat, cell or method according to any configuration, aspect, embodiment or example, wherein the human VpreB gene is operably linked to an endogenous VpreB-X5 locus control region (LCR). Thus, where the vertebrate (or cell) is a mouse or rat (cell), the human VpreB is operably linked to a VpreB-A5 LCR of said mouse or rat (cell). LCRs are dominant activating sequences that are able to activate gene expression at any location in the genome. Optionally, the LCR is present at the wild-type position in the genome of the vertebrate or cell. In another example, the LCR is not at the wild-type position in the genome of the vertebrate or cell. In this case, therefore, the human VpreB is positioned away from the endogenous VpreB gene in the genome, which may be advantageous for controlling the human VpreB expression independently of the endogenous As.
In an embodiment, the invention provides a non-human vertebrate, mouse, rat, cell or method according to any configuration, aspect, embodiment or example, wherein the human VpreB gene is operably linked to an endogenous VpreB locus control region (LCR). Thus, where the vertebrate (or cell) is a mouse or rat (cell), the human VpreB is operably linked to a VpreB LCR of said mouse or rat (cell). Optionally, the LCR is present at the wild-type position in the genome of the vertebrate or cell. In another example, the LCR is not at the wild-type position in the genome of the vertebrate or cell. In this case, therefore, the human VpreB is positioned away from the endogenous VpreB gene in the genome, which may be advantageous for controlling the human VpreR expression independently of the endogenous AS.
Optionally one or more DNase I hypersensitive sites (HS) is operably linked in the genome to the human VpreB gene. For example, the HS forms part of the LCR or is 3' of the LCR or 3' of the LCR-human VpreB (see, Sabbattini eta! 1999, supra).
Optionally in any configuration, aspect, embodiment or example of the inventions, the human VpreB gene is present in multiple copies in the genome, eg, the genome comprises 2,3,4,5, 6, 7,8, 9 or 10 copies of the human VpreB gene. For example, 2, 3,4 or 5 copies are operably linked to the control element (eg, promoter and/or LCR) mentioned above.
Assessing the development of the B-Cell Repertoire Some assays can be used to investigate if the invention leads to promotion of B-cell development and repertoire. Although described in the context of transgenic mice below, these comments apply equally to other transgenic non-human vertebrates of the invention.
1. In chimaeric mouse, after introducing the human DNA fragment covering several human Vs, all Ds and is between mouse is and mouse Cs, the usage of the inserted human Vs can be tested by sequencing the mRNA products of the IgH locus in chimaeric mouse. In this case, human Vs will compete with endogenous mouse Vs for formation of the heavy chain. The usage of human Vs vs mouse Vs will be compared in mice with and without introduction of human VpreB gene into the chimaeric mouse with/without the endogenous mouse VpreB gene. More detail is provided in the non-limiting example below.
2. The use of human VpreB or mouse VpreB protein for pre-BCR assembly can be analysed in pre-B cell populations from mice with or without the human VpreB. Surrogate light chain can only be detected in the transitional pre-BI and large pre-BlI stages in bone marrow. To enrich the cell populations in bone marrow for investigating pre-BCR, the deregulation of surrogate light chain can be blocked by inactivation of SLP6S gene (Nature Immunol. 20034, 38-43; The adaptor protein SLP-acts as a tumor suppressor that limits pre-B cell expansion. Flemming, A., Brummer, T., Reth, M. & Jumaa, H.) or both IRF4 (interferon-regulatory factor 4) and IRF8 genes (Genes Dev. 2003 17, 1703-1708; lRF-4,8 orchestrate the pre-B-to-B transition in lymphocyte development. Lu, R., Medina, K. L., Lancki, D. W. & Singh, H). It has been reported that pre-BlI cells from mice that are deficient in these factors continue to express surrogate-light-chain genes and therefore retain pre-BCR expression. The unlimited supply of surrogate light chains lead to a hyperplastic pre-BlI-cell phenotype in vivo and provide large number of pre-B cells for us to analyze.
3. In instances (eg, as described in part 1 above), where mouse Vs are retained following insertion of human gene segments, the mouse endogenous heavy chain VD1 is inactivated, and this may be done while leaving mouse Vs intact. Thus, for the formation of chimaeric heavy chain genes, the heavy chain V regions should only be transcribed from human heavy VDis. To assess the relative use of human v mouse Vs for heavy chain gene formation, the B-cell repertoire and diversity will be compared between mice having human VpreB gene v mice lacking the human VpreB (eg, mice with mouse VpreB only). Pairing of human Vs with human VpreB will be preferable to pairing of mouse Vs with human VpreB, and thus antibody heavy chains comprising human variable regions will be selected during B-cell development.
As cells transit from the pre-BI to pre-BlI stage, the pre-BCR induces a signal for proliferative expansion. Pre-BCR controls the number of developing pre-B cells by controlling proliferation, which represents clonal expansion. Pre-BlI cells enter two to seven rounds of cell division when lgj.x chain assembles into pre-BCRs. This ultimately results in a more diverse antibody repertoire, as later in development, each cell will recombine and express a unique L chain. In this process, the association of surrogate light chain with chain monitors the cells to determine whether it has successfully completed VDJH recombination and expresses a functional pH chain which will form the pre-PCR for downstream signalling. In this case, the strength of the pairing between the pH and SL chains may determine the amplitude of the transmitted signal, and thereby, the extent of proliferation (J.
Immunol. 2006, 177 pp. 2242-2249; Pre-B cell receptor assesses the quality of IgH chains and tunes the Pre-B cell repertoire by delivering differential signals. Y. Kawano, S. Yoshikawa, V. Minegishi and H. Karasuyama). Therefore, in the mouse with chiniaeric heavy chain (human VDJs and mouse Cs),, by introducing the human VpreB gene to provide complete species-matching with the chimaeric heavy chain, It is expected that the repertoire and so the diversity of chiniaeric antibodies will be improved by using human VpreB instead of mouse VpreB for association with the heavy chains. The improvement for B cell repertoire and diversity can be showed by increased usage of human heavy VDJs and/or a shift in VH gene family usage in mouse expressing human VpreB (see the examples below for possible techniques).
It will be understood that particular embodiments described herein are shown by way of illustration and not as limitations of the invention. The principal features of this invention can be employed in various embodiments without departing from the scope of the invention. Those skilled in the art will recognize, or be able to ascertain using no more than routine study, numerous equivalents to the specific procedures described herein. Such equivalents are considered to be within the scope of this invention and are covered by the claims. All publications and patent applications mentioned in the specification are indicative of the level of skill of those skilled in the art to which this invention pertains. All publications and patent applications are herein incorporated by reference to the same extent as if each individual publication or patent application was specifically and individually indicated to be incorporated by reference. The use of the word "a" or "an" when used in conjunction with the term "comprising" in the claims and/or the specification may mean "one," but it is also consistent with the meaning of "one or more," "at least one," and "one or more than one." The use "_ -" L-ms is used to mean "and/or" unless explicitly indicated to refer to alternatives only or the alternatives are mutually exclusive, although the disclosure supports a definition that refers to only alternatives and "and/or." Throughout this application, the term "about" is used to indicate that a value includes the inherent variation of error for the device, the method being employed to determine the value, or the variation that exists among the study subjects.
As used in this specification and claim(s), the words "comprising" (and any form of comprising, such as "comprise" and "comprises"), "having" (and any form of having, such as "have" and "has"), "including" (and any form of including) such as "includes" and "include") or "containing" (and any form of containing, such as "contains" and "contain") are inclusive or open-ended and do not exclude additional, unrecited elements or method steps The term "or combinations thereof" as used herein refers to all permutations and combinations of the listed items preceding the term. For example, "A, B, C, or combinations thereof is intended to include at least one of: A) B, C, AB, AC, BC, or ABC) and if order is important in a particular context, also BA, CA, CB, CBA, BCA, ACB, BAC, or CAB. Continuing with this example, expressly included are combinations that contain repeats of one or more item or term, such as RB, MA, MB, BBC, AAABCCCC, CBBAAA, CABABB, and so forth. The skilled artisan will understand that typically there is no limit on the number of items or terms in any combination, unless otherwise apparent from the context.
Any part of this disclosure may be read in combination with any other part of the disclosure, unless otherwise apparent from the context.
All of the compositions and/or methods disclosed and claimed herein can be made and executed without undue experimentation in light of the present disclosure. While the compositions and methods of this invention have been described in terms of preferred embodiments, it will be apparent to those of skill in the art that variations may be applied to the compositions and/or mnthndc nnrl in thø ctønc nr in the sequence of steps of the method described herein without departing from the concept, spirit and scope of the invention. All such similar substitutes and modifications apparent to those skilled in the art are deemed to be within the spirit) scope and concept of the invention as defined by the appended claims.
The present invention is described in more detail in the following non limiting prophetic exemplification.
EXAMPLES
EXAMPLE 1: Introduction of human DNA gene segments into ES cells By way of illustration, the following example is provided on the introduction of human antibody gene segment DNA into Cell culture of C57BL/6N-derived cell lines, such as the JM8 male ES cells.
This will follow standard techniques. The JM8 ES cells have been shown to be competent in extensively contributing to somatic tissues and to the germline, and are being used for large mouse mutagenesis programs at the Sanger Institute such as EUCOMM and KOMP (Pettitt, Si., Liang, 0., Rairdan, XV., Moran, iL., Prosser, H.M., Beier, D.R., Lloyd, K.C., Bradley, A., and Skarnes, W.C.
(2009). Agouti CS7BL/6N embryonic stem cells for mouse genetic resources. Nature Methods.). 1M8 ES cells (1.0X107) will be electroporated (5001.LF, 230V; BioRad) with 10ig 1-Scel linearized human BAC DNA. The transfectants will be selected with either Puromycin (3ig/ml) or G418 (150ig/ml).
The selection will begin either 24 hours (with G418) or 48 hours (with Puromycin) post electroporation and proceed for 5 days. lOjig linearized human BAC DNA can yield up to 500 Puromycin or G418 resistant ES cell colonies. The antibiotic resistant ES cell colonies will be picked into 96-well cell culture plates for genotyping to identify the targeted clones.
Once targeted mouse ES cell clones are identified, they will be analyzed by array Comparative Genomic Hybridization (CGH) for total genome integrity (Chung, VJ., Jonkers, J., Kitson, H., Fiegler, H., F-lumphray, S., Scott, C., Hunt, S., Vu, V., Nishijima, I., Velds, A., et a!. (2004). A whole-genome mouse BAC microarray with 1-Mb resolution for analysis of DNA copy number changes by array comparative genomic hybridization. Genome research 14, 188-196.and Liang, 0., Conte, N., Skarnes, W.C., and Bradley, A. (2008). Extensive genomic copy number variation in embryonic stem cells.
Proceedings of the National Academy of Sciences of the United States of America 105, 17453- 17456.). ES cells that have abnormal genomes do not contribute to the germline of the chimaeric mice efficiently. BAC integrity will be examined by PCR-amplifying each known functional V gene in the BAC. For example, in one approach the first human BAC chosen for the IgH locus has 6 functional VH gene segments and all human D and JH gene segments. To confirm the integrity of this BAC for the presence of these IgH genes, pairs of PCR primers will be designed and used to PCR-amplify genomic DNA from the targeted ES cells. The human wild-type size and sequence of these fragments will ensure that the inserted BAC has not been rearranged. More detailed CGH will also confirm the integrity of the inserted BACs. For example, one skilled in the art could use an oligo aCGH platform, which is developed by Agilent Technologies, Inc. This platform not only enables one to study genome-wide DNA copy number variation at high resolution (Barrett, M.T., Scheffer, A., Ben-Dor, A., Sampas, N., Lipson, D., Kincaid, R., Tsang, P., Curry, B., Baird, K., Meltzer, P.S., et a!. (2004).
Comparative genomic hybridization using oligonucleotide microarrays and total genomic DNA.
Proceedings of the National Academy of Sciences of the United States of America 101, 17765- 17770.), but permit examination of a specific genome region using custom designed arrays.
Comparing the traditional aCGH techniques which rely on cDNA probes or whole BAC probes, the 60-mer oligonucleotides probes can ensure specific hybridization and high sensitivity and precision that is needed in order to detect the engineered chromosome alterations that we have made. For example, oligos designed to hybridize at regular intervals along the entire length of the inserted BAC would detect even quite short deletions, insertions or other rearrangements. Also, this platform provides the greatest flexibility for customized microarray designs. The targeted ES cell genomic DNA and normal human individual genomic DNA will be labelled separately with dyes and hybridized to the array. Arrays slides will be scanned using an Aglient Technologies DNA microarray scanner.
Reciprocal fluorescence intensities of dye Cy5 and dye Cy3 on each array image and the log2 ratio values will be extracted by using Bluefuse software (Bluegnorne). Spots with inconsistent fluorescence patterns ("confidence" <0.29 or "quality" = 0) will be excluded before normalizing all log2 ratio values. Within an experiment, Log2 ratio between -0.29 and +0.29 for the signal from any oligo probe are regarded as no copy number change. The log2 ratio threshold for "Duplication" is usually >0.29999, and for deletion is <0.29999.
EXAMPLE 2: Introducing the human VpreB gene into the mouse lambda locus by inserting the human lambda V gene segment cluster The human Vpreb gene is inserted into a mouse genome by introducing human lambda gene segment DNA, wherein the nucleotide sequence of the DNA comprises a human VpreB gene and optionally the associated human promoter. Human VpreBl and VpreB3 genes are both located in chromosome 22. The human VpreBl gene is between human lgX V gene segments lV-53 and 5-52 (Chromosome 22: 22599200 to 22599926). Therefore, the human lambda gene segment which covers the human VpreBl gene and contains multiple Vs flanking this gene is inserted in the endogenous lambda antibody locus upstream of the endogenous lambda constant region between the most 3' endogenous Pt and the endogenous CA following the approach in Example 1 and as -"""11004129.
EXAMPLE 3: Introducing the human VpreB gene within the mouse VpreB-X5 locus and targeting into the mouse heavy or lambda locus The following example illustrates the insertion of the human VpreBl gene within the mouse VpreB-S AS locus to bring the human gene under the endogenous LCR influence.
The mouse VpreBl and AS genes are closely associated, both within a 19-kb fragment called the AS-VpreBl locus. Apart from these two genes, this locus contains a locus control region (LCR) required for correct levels of expression and tissue-specificity of AS and VpreBl. Locus control regions (LCRs) are sequences that mediate reorganisation of chromatin and activation of transcription by sequence-specific transcription factors. The characteristic of an LCR is the ability to drive gene expression in transgenic mice at any site of integration at levels that are substantially equivalent to those of the gene in its natural location. It has been shown that the LCR in the A5-VpreBl locus is able to promote efficient and stage-specific expression of both genes in transgenic mice at all sites of integration tested.
To make sure that the expression of the inserted human VpreBl gene is properly controlled in mouse, the mouse A5-VpreBl locus is retrieved and cloned into a vector (pBlueScript II SK(+)). The mouse VpreBl nucleotide sequence is replaced by the human VpreBl sequence by cloning. The construct containing the fragment of mouse AS-human VpreBl locus is then cloned into a BAC containing a human heavy chain locus fragment. Then it is targeted into the genome of a mouse ES cell following the introduction of the human BAC into the mouse heavy chain locus upstream of the endogenous heavy chain mu constant region. In this case, the human VpreB function can be directly tested in the Fl mouse generated from the ES cells. Manipulation of constructs and BACs can be effected by standard recombineering.
Insertion of the mouse AS-human VpreBl can be carried out using homologous recombination or RMCE. Homologous combination can employ homology arms corresponding to the endogenous sequences flanking the mouse AS-mouse VpreBl in the mouse ES genome, so that insertion of the mouse A5-human VpreBl replaces (and thus deletes) the endogenous mouse A5-mouse VpreBl and places the inserted construct under the control of the endogenous LCR.
RMCE can be used to insert the mouse X5-human VpreBl construct (optionally with the endogenous mouse IS-mouse VpreBl LCR) anywhere in the genome. RMCE can also be used to precisely delete the endogenous mouse X5-mouse VpreBl so that the inserted mouse its-human VpreBl construct is the only source of a VpreBl gene.
In the alternative) the method can be modified to insert the mouse lambda 5-human VpreBl construct into the endogenous lambdas-VpreB locus on the mouse lambda locus) for example to bring the human VpreBl gene under endogenous control (eg, control of the lambda5-VpreB LCR).
To do this, the cloned mouse lambda 5-human VpreBl construct (produced by standard recombineering) is inserted into a homologous recombination vector using in vitro recombineering to add homology arms flanking the construct. Using homologous recombination in the presence of a marker carried by the vector, the construct is inserted precisely to replace the endogenous mouse lambda 5-mouse VpreBl in the genome of a mouse ES cell. Alternatively, instead of homologous recombination, standard RMCE can be used to effect precise insertion into the genome.
EXAMPLE 4: Assessing the B-cell development and repertoire in chimaeric mouse containing human VpreB gene The following example is provided on assessing the advantage of having the human VpreB gene in the chimaeric mouse.
The mouse embryonic stem cells (eg, AB2.1 cells; Baylor College of Medicine) containing the targeted human VpreBl gene are grown on a feeder layer of SNL7 fibroblasts (Baylor College of Medicine) in embryonic stem cell medium containing 15% serum. Then the cells are microinjected into blastocysts which are transferred to the uteri of pseudopregnant F1 female mice. To test for germline transmission) male chimeras are bred to C57BL/6-Tyr8 albino female mice (Baylor College of Medicine). After obtaining mice with both chimaeric heavy chain gene (human V region and mouse C region) and human VpreBl gene, the following assays are performed to show the advantage of human VpreB v mouse VpreB.
1. Pre-B cell populations containing pre-BCR from human VpreB v mouse VpreB In the pre-B cells, p heavy chain associated with surrogate light chain will deposit on the cell surface to signal the cell survival and proliferation. The pre-B cell populations with the pre-BCR from human VpreB protein v mouse VpreB protein are analyzed by flow cytometry.
Cells from bone marrow are surface stained with fluorescence-labelled mAb and then analysied with MACSQuant (Miltenyi Biotech) on B220 cells. Pre-B cell population can be detected using pre-BCR-specific mAR and/or mouse X5cells. The cells with pre-BCR of human VpreB v mouse VpreB are distinguished by using monoclonal antibody specifically to human or mouse Vpreb.
2. Usage of human Vs in the mouse with chimaeric antibody In the mouse with the insertion of human VHDHJHS between endogenous mouse VHDHJHS and CHS, the usage of human Vs v endogenous mouse Vs are compared by sequencing the heavy chain products in naive mice.
Approach: a. Total RNA extraction (use TRIzol® Reagent) from mouse spleen b. By a standard 5' RACE (Rapid amplification of 5' complementary DNA ends) method, using a mouse Cp-specific primer, PCR fragments from mouse synthesized heavy chain mRNAs can be amplified.
c. The PCR products are cloned into a vector and then sequenced.
d. The sequences map to heavy V Di sequences which contain either mouse V or human V. Then we compare the usage of human Vs v mouse Vs.
e. The usage of human Vs v mouse Vs is analysed in the mice with/without a human VpreBl gene. We expect by introducing human VpreBl gene, the human V usage will increase in the mouse with chimaeric lgF-I gene.
3. Improvement of repertoire and diversity of chimaeric antibody In this embodiment, in mice with the chimaeric heavy locus, endogenous mouse V genes are inactivated by deletion or inversion of the mouse V locus. Therefore, all the heavy chain products are transcribed from human Vs.
Using the same methods in Example 3, the heavy chain products from naïve and/or immunized mice are sequenced and map to human VH DRJH5 with different CDR sequences.
The repertoire of heavy V Di selected by surrogate light chain from human VpreB and mouse VpreB can be compared from several aspects. First is the improvement of usage of human heavy V D is and hypervariabilities for mice having human VpreB. Second is the shift of usage in terms of human heavy V gene families caused by human VpreB v mouse VpreB.
SEQUENCE LISTING
SEQ ID NO: 1 Human VpreBl nucleotide sequence (GenBank Accession No = NG_029387.1) gagtcagagctctgcatgtctgcaccatgtcctgggctcctgtcctgctcatgctgtttgtctactgcacaggt gaggga acccccagatcccaaagactcctgccccttccttcatcctgccctgcccccacggcccacatgcatctgtgtca ccaggt tgtggtcctcagccggtgctgcatcagccgccggccatgtcctcggcccttggaaccacaatccgcctcacctg caccct gaggaacgaccatgacatcggtgtgtacagcgtctactggtaccagcagaggccgggccaccctcccaggttcc tgctga gatatttctcacaatcagacaagagccagggcccccaggtcccccctcgcttctctggatccaaagatgtggcc aggaac agggggtatttgagcatctctgagctgcagcctgaggacgaggctatgtattactgtgctatgggggcccgcag ctcgga gaaggaggagagggagagggagtgggaggaagaaatggaacccactgcagccaggacacgtgtcccttgaactg aagaca gcagaggcacgcatccccttggagaga ctgtcatgga agagggtggagccgccgcccgaagcgccgaggaggctgagcca ctcagcatctcctggtcctgcagtgttgctgtaaatccccattggagactgcattagggaattaaagctgcttg tcactt tttgctg SEQ ID NO: 2 Human VpreBl amino acid sequence (GenBank Accession No = NP_009059.1) rnswapvllmlfvyctgcgpqpvlhqpparnssalgttirltctlrndhdigvysvywyqqrpghpprfllryf sqsdksqg pqvpprfsgskdvarnrgylsiselqpedeamyycamgarssekeerereweeemeptaartrvp SEQIDNO:3 Human VpreB3 nucleotide sequence (GenBank Accession No = NC_000022.1O) cttcccagccctgtgccccaaagcacctggagcatatagccttgcagaacttctacttgcctgcctccctgcct ctggcc atggcctgccggtgcctcagcttccttctgatggggaccttcctgtcaggtgaatctttcctgggcctcagtca cctggg tgtggggtggggaacagtactggccccagaaggcccctgcaaggaggcaaatcgatggggacagtagggcaggt cctggg aggggtattttttttttttttgagacggagttttgctcttgttgcccaggctggagtgcaacggtgcaatctcg gctcac tgcacctctgcctcccaggttcaagcgattcttctgcctcagcgtcctgagtagctgggattacaggcatgcgc caccac gcctggctaattttgtatttttagtagagacaaggtttcttcatgttggtcaggctggtctcaaactcctgacc tcaggt gatccgcccacctcagcttcccaaagtgctaggattacaggcttgagccactgcacctggctgggaggcgtatt caggta gaggggtgacctggcttcagacagctctgcccttgacctggggcaaatcacttccctgtgtgggccttggtttt ctcatt cgtgaatgaccagatcactagagccctggactctgactttgggtctccttcttagtttttcacaggggaagcta ttgtgg gttggtccctaccccacagggcctgaggcaatctaacctccctgagagggtccctgcagccagttgcctgaggc tgagtt gatgtgtgggacagcccagg2ggttcctgggggtggttagtctgtattcagggtttggaagagctgaagtgaag tgggcg gggaagtgggggagaggggtgcagttctgcagagaaacgtgggtgggtagcagagggacctagaggctgctagt ccaacc ttctgagctctgggcctttaactgaacacagatccttgaggatatggcactaatggagatttgggggctaactc caaacc cctcactcacaaagggagatggaggtccagatagggcaccctaagtcacacagaaccaggcctcctgcaccccg ttcatt gctaatcccatagcactgggctatgagccctttgggactgggagtctccatggagtccaaccaagccttcacag ggcagg ggtgggagggaaggggctcaggctgagtgggtttgtgtctcgcagtttcccagacagtcctggcccagctggat gcactg ctggtcttcccaggccaagtggctcaactctcctgcacgctcagcccccagcacgtcaccatcagggactacgg tgtgtc ctggtaccagcagcgggcaggcagtgcccctcgatatctcctctactaccgctcggaggaggatcaccaccggc ctgctg acatccccgatcgattctcggcagccaaggatgaggcccacaatgcctgtgtcctcaccattagtcccgtgcag cctgaa gacgacgcggattactactgctctgttggctacggctttagtccctaggggtggggtgtgagatgggtgcctcc cctctg cctcccatttctgcccctgaccttgggtcccttttaaactttctctgagccttgcttcccctctgtaaaatggg ttaata atattcaacatgtcaacaaca SEQ ID NO: 4 Human VpreB3 amino acid sequence (GenBank Accession No = NP_037510) macrclsfllmgtfisvsqtvlaqldallvfpgqvaqlsctlspqhvtirdygvswyqqragsapryllyyrse edhhrp adipdrfsaakdeahnacvltispvqpeddadyycsvgygfsp SEQIDNO:5 Mouse VpreBl nucleotide sequence (GenBank Accession No = NC_000082.5) agagcccagaaagcctgggagggtggtgagcaggaaccaggggtgcagtgaccctctccccaaagcagggagga gagtgc ttcccagctggtcagggcccaggagcagtggctgtagggggcagggtgctgcaggtctggagccatggcctgga cgtctg tcctgctcatgctgctggcctatctcacaggtaaggaaactcttggggcccagggcttctttgctcctcctatg gccttg ctctgccccagtgatggacatgcttctatcttctcaggttgtggccctcagcccatggtgcatcagccaccatt agcatc ttcttcccttggagccaccatccgcctctcctgtaccctgagcaacgaccataacattggcatttacagcattt actggt accagcagaggccgggccaccctcccaggttcctgctgagatacttctcacactcagacaagcaccagggtccc gatatc ccacctcgcttctccgggtccaaagatacgaccaggaacctggggtatctgagcatctctgaactgcagcctga ggacga ggctgtgtattactgtgccgtggggctccggagccaggaaaagaagaggatggagagggagtgggaaggagaaa agtcgt atacagatttgggatcttaggctctggagacattcagaccctgaactgaagacagagtttgctttgctcggcta gtctgg tatgggaaggaggggtagaacgtgaggttttgcagagcctagaagatggaattatgcagcttttccttgttctg cggtgt tgctatgagcccccattggaggctggattgtagaattaaagctgtttttactgaa SEQ ID NO: 6 Mouse VpreBl amino acid sequence (GenBank Accession No = NP_058678) mawtsvllmllayltgcgpqpmvhqpplassslgatirlsctlsndhnigiysiywyqqrpghpprfllryfsh sdkhqg pdipprfsgskdttrnlgylsiselqpedeavyycavglrsqekkrrnerewegeksytdlgs SEQ ID NO: 7 Mouse VpreBZ nucleotide sequence (GenBank Accession No = NC_000082) atggcctggacgtctgtcctgctcatgctgctggcccacctcacaggtaagggaactcttggggtccagggctt ccttgc tcctcctgtggccttgctctgccccagtgatggacatgcttctatcttctcaggttgtggccctcagcccatgg tgcatc agcca ccatcagcatcttcttcccttggagcca cca tccgcctctcctgtaccctgagcaacgaccata acattggcatt tacagcatttactggtaccagcagaggccgggccaccctcccaggttcctgctgagatacttctcacactcaga caagca ccagggtcccgatatcccacctcgcttctccgggtccaaagatacggccaggaacctggggtatctgagcatct ctgaac tgcagcctgaggacgaggctgtgtattactgcgctgtggggctccggagccacgaaaagaagagaatggagaga gagtgg gaaggagaaaagtcgtatacagatttgggatcttag SEQ ID NO: 8 Mouse VpreBZ amino acid sequence (GenBank Accession No = NP_058679.1) mawtsvllmllahltgcgpqpmvhqppsassslgatirlsctlsndhnigiysiywyqqrpghpprfllryfsh sdkhqg pdipprfsgskdtarnlgylsiselqpecleavyycavglrshekkrmerewegeksytdlgs SEQ ID NO: 9 Rat VpreBl nucleotide sequence (GenBank Accession No = NM_001108845.1) ccagaaagcctgggagggtggtgagcaggaaccagtggtgcaaagcaggggcgagactgcttcctagctggtca gggcac cggagcagtggctgtaggggtcagggtgctgcaggtctggaaccatggcctggacgtctgccctgctcatactg ctggcc catctcacaggtacgggaactcttggggcccagagcctccttgctcctcctcttgccttgctctgccgcagtga tgggca cgcttctattttctcaggttgtggccctcagcccgtgctgcatcagccaccatcggcctcttccttccttggaa cctcca tccgcctcacctgtgccctgagcagcaaccataacattggcatttacagcatttactggtaccagcagaggccg ggccac cctcccacgttcctgctgagattcttctcacactcagacaagctccagggtcccaagatcccccctcgcttctc cggatc caaagatacagccaggaacctggggtacctgagcatctctgacctgcagccagaggacgaggctgtgtattact gcgccg tggggcttcggagctgggaaaaggagaagaggatggagagggagtgggaagaagaaaagtagcggacagattcg ggatct taggctctggagacattcagacctagaaccgaagacggagtttgctttgctcggctaggctggtttggggagga ggggta gaacaccgggcttcgcagagccaggaaggtggagccagccgcttttccttgtattgcagtgttgctatgcgccc catcgg aggctggattgtagaattaaagctgttttttttttttttgttttgtttttttgttttttgtttcttttttctta actg SEQIDNO:1O Rat VpreBl amino acid sequence (GenBank Accession No = NP_001102315.1) mawtsal Iii a hi tgcgpqpvlh q ppsassfigtsirltcaissn h ii igiys iywyqq rpgh p ptfI I rffshsd klqg pkipprfsgskdtarnlgylsisdlqpedeavyycavglrswekekrrnereweeek SEQ ID NO: 11 Rat VpreB2 nucleotide sequence (GenBank Accession No = NC_005110) atggcctggacgtctgccctgctcatactgctggcccatctcacaggtacgggaactcttggggcccagagcct ccttgc tcctcctcttgccttgctctgccgcagtgatgggcacgcttctattttctcaggttgtggccctcagcccgtgc tgcatc agccaccatcggcctcttccttccttggaacctccatccgcctcacctgtgccctgagcagcaaccataacatt ggcatt tacagcatttactggtaccagcagaggccgggccaccctcccacgttcctgctgagattcttctcacactcaga caagct ccagggtcccaagatcccccctcgcttctccggatccaaagatatagccaggaacctggggtacctgagcatct ctgacc tgcagccagaggacgaggctgtgtattactgcgccgtggggcttcggagctgggaaaaggagaagaggatggag agggag tgggaagaagaaaagtagcggacagattcgggatcttaggctctggagacattcagacctagaaccgaagacgg agtttg ctttgctcggctaggctggtttggggaggaggggtagaacaccgggcttcgcagagccaggaaggtggagccag ccgctt ttccttgtattgcagtgttgctatgcgccccatcggaggctggattgtagaattaaagctgttttttactg SEQ ID NO: 12 Rat VpreBZ amino acid sequence (GenBank Accession No = NP_001128260) mawtsallillahltgcgpqpvlhqppsassfigtsirltcalssnhnigiysiywyqqrpghpptfllrffsh sdklqg pkipprfsgskdiarnlgylsisdlqpedeavyycavglrswekekrmereweeek SEQIDNO: 13 Human 1'5 nucleotide sequence (GenBank Accession No = NG_009791) ggccacatggactggggtgcaatgggacagctgctgccagcgagagggaccagggcaccactctctagggagcc cacact gcaagtcaggccacaaggacctctgaccctgagggccgatgaggccagggacaggccaggggggccttgaggcc cctggt gagccaggccccaacctcaggcagcgctggcccctgctgctgctgggtctggccgtggtaacccatggcctgct gcgccc aacagctgcatcgcagagcagggccctgggccctggagcccctggaggaagcagccggtccagcctgaggagcc ggtggg gcaggtaaggggtgagagattccaggggatgtgggggtctgggtggcagaggcgggaaaggatgaccaagggga gacgag ccagaggggtgaggaggaaggttaatccctggaggggagccacagacactgactttaactaaagtgtcaagatt ttgtcc atctttgaattaatttttattgcttaatgtcatattaaaatattatttatcttgattcctgagatttcttcccc cactta catttggcaccaaggccaatgtctccctcacctccccctagtccttggggtagggcaggactggaggcaggggc aggacg tccacaggagtggtggccgctatccctgaaggatgcccaggcctctccctcctcctcctcccactcctcctccc ccccct cctcttttccccttggcctatgtcacctgtccactcccaccctcactgggcaggggccactccctggagctcca gctaag gtgtgaggggcctttcctggagtccctgggtcactagacctcagccagcatcgcctcctgaaaccagcccctag gagaca caagcttatccagggtgcaagtgcctccaaagaagaagcccaggagaggctctagggaggccacaactccctgt gtgacc tcagccattgccaccactctgcgtgtggtgggaggtccccagacaaagcaccaagcatcggggtccatttatga gcattt gggacacaacagcctgttcactggtgcatgttatacccacaggcgataatcatttcaggggcagaacctccctc tcggtg gccccataggcaggtcctgctgtgatcccctttgtgcagacggggatagagcccggagaggtgaggtgacccgt ccgagt cactcagctcatgggcacagattctaaggcccaagctatcccctctagctctcccctgtcccatcctctaagct gatcga gcggacacgtgcatctctgggacctgagtttccctttttctctcttttttttctttttcaaataaagtttcaca gagttt cactcttgtcgcccagtctggagtgcaatggcgagatcttggctcattgaaacctccgtctcccaggttcaaga cattct cctgcctcagcttccggagtagctgggattacaggcatctgccaccacacctggctaattttgtgtattttttt ttagta aagacagagtttcaccatgttggccaggctggtctcaaactcctgacctcaggtgatccacccacctcagcctc ccaaag tgctgggattacaggcatgagacaccacaccgggcctgagtttccccttctgcaatctgaggggccctgactgg tgaggg ccttcagcgtcccacccacccagaggatgctggggtggctgtggtgagagctccagcagtggcagccgacctga cccaca ccaggagcccggccatggaggcggggtcagcatggtggcaggccgggaccgggtgtcagtgtcctgcacggact tctgag caaggagtccccatcagggtcaggctctgtgctggggctgaggtcccagaggatctagatttgccccaattcaa gtccac aaggagcgggggccgggtgaggagacagccacatgcagggtgatgcctacagaacagagactgggatggggaag gcccga ggggtctccacaagggacgggtgacaggtggagggagacacagataataaaaaatggtattatgttggggggct attaat gtaagtttttatattagaatctttagaaatcttatagaaatactatgggccgggtgctgtgtctcatgcctgta atccca gcactttaggacgccaagatgggcagatcacgaggtcaggagattgagaccatcctggccaacatggcaaaact ccgtct cta ctaa a a ataca a aa a ctagctgggcgtggtggcgcgagcctatagtcccagctactcgggaggggaggcaggaga at cgcttgaatcagggaggtggaggttgcagtgagctgagattgcaccgctgcactccagcctggacgacagaacg agactc cca ctcaaa a a a a a a a a a aggtattatgctgggggggatatgaatatgagtttttata atctttagaa atactatgggcc aggtgcagtggctcatgcctgtaatcccacaactttgggaggctgaggcgggattgtttgagcccaggagtttg agatca gcctgagcaacatagcaagaccccatttctacaaaacatataaaaactagctagtcatggtggcacttgcctgt ggtccc agctacttaggaggctggtgagaggattgcttaagcctcggaggttgaggatgcagtaagctgagatcccacca ctgcac tccagcctgggtga cagaaggagaccctgtctca aa a a a a a aaaaaaaaaaaaaa ga ctgattattcctgtaga attctg gtaaatatctcctatcaaataaatgacttttctattcatagcttattaaaagatattttcattgtttttaagaa ataggt tgtgtatcactttttatatttagttgtaaatttatttgttttatttatttttttagagatgggggtctcactat gttgcc tgggctggcctccaactgctgggctcaagcaatatccctgcctcagcgtccccagtagctgggactacaagcat gcgcta ccacaccggcataattttttgtagagatgaggtttcgccatgttgcctgggctggtcttgaacccctgggctca agccat ccacccgcctcggcctcctaaagtgctgagattacaga cgtgagccaccctacctagcctgtactatttttaataggtct taataggttttgaatgttaattatttttaaattaatttcaaaaatcttctacaacatggatgaaacctgaagac attata cttagtga a ata agccaga cacaaaaggaca a atgtcatttga atcca cttctatgaggtacctagaataggca aattca cttggacaaaaagtagatttgaggttagcagggtgagggggagggaagaatgggggactgtagttaacgggttt agagtt tctgtttgggaagatgaaagagttctggagatggatggtggtgaaggttgcccaacggtgtgaatgtacttagt gccacg gagctgta cgtttaaaaatagttaaagtggaaatattgatgctatgtata a a a atggagcggggtgtggtggctca Ca cc tataatcccagcactttgagaggccaaggtgggcagatcacctgaggtcgggagttcgagaccagcctgacaaa catgga ga a a caccgtctcta ctga a a ata ca aaaaatta gccaagtgtgctggcacatgcctgtaatcccagctactcgggaggc tgaagcaggagaatcgcttgaacccaggatgtggaggttgcggtgagccaaggtggcaccattgcactccagcc tgggcg a caagggtga a attccatttca aaaa a ataaaaggaaatggga ctgtacatagcaggaga gagagggagagatca atatg acacttctttttttttttttttttttttgggacagtcttgctctgttgccaggctgcactccagcctgggcaac agagcg agactctgtctcaaaaaaaaaaagaatgtctagatgcagtagctaagttcgcagaagccatttcagtgtgggag gcgagg tgactgagtccagggactccaggtttctggctgaggggttgagaccgcccatggtcattgtgatgagatgaaga cagaaa atgagctgggctgggggaaggtggtcatctcctctggacgtgattagtttgaggctcctgttgggtagcccact gggcaa ggtcagtaggcaattgaggagctgagagggtttggagctgggatagactccagcctcaccatgtgggcaatagt gggggt ca cagagtgtga 22 atggctaagaaagaggtctgggtggggtctggggagtcagcagccaggcatgggccatggaga a a c agatgccaggggaagaggaaaggggagtctcagaccccaaggggaaaagagtcactggaaagaggggccagccc tgtgtc gcatccagcggagacaccaggtacagcagaaggaccttggacatgaccatgaggagggccttgttgaccatggc ctggga gagatgggggtgagagcctgggggaccacaccatgtccccagcacacagtgcctggtagacaggatggatttat ggatgg acggacaggtagatggatgga cgaatggacagatgatagatggatgcaaaga cagatga atagatgga cagatgcataga tggacagatgga cagatggatggacggacggatggaatga atgatcagaaaaggcttcatgaacaaagtgagactgagct gcatctccatgggtagatataaaagcagaggactctcctcttgagtcaggaatgacccaatgtcctggtccagg gaggaa gtcagcctccttgactggggacacttgtggcagatttcagaggcccttaaaatgaggccaagtgaggtggacag gtccga gccagctgaggactcctcagccacacggcacagctgcctgaggggatgtgtcactcagggagttgctgggacct actggg cccagcgttgccatcagcaccaacagtttcagagagggggacacacgctggggcagcacctgcctcagagaagg gacagg cacagagacactactgggggacactactgggacactggccacccccctaccctgtgcctgggtcacagcctaca cactgc agccctgtgcccctcactcccagcaggttcctgctccagcgcggctcctggactggccccaggtgctggccccg ggggtt tcaatccaagcataactcagtgacgcatgtgtttggcagcgggacccagctcaccgttttaagtaagtggctct aacttc ccaggctgtcccaccctctcctgtctctggaaaatgtgttttctctctctggggcttcttcccctctgccctcc cagcct taagcactgacccctacctttgtccatggggcctggaggagatgtgttagtctcagggtaatggcaggaagggc ccccac agtgggagcagccgccttcaggttccaacagcaggacacagcctggtcccagggcctgggctgggattgggcgg ggteag ggctcctcccctctcccagggcagatgtctgagtgagggacagaggctggttctgatgaggggccctgcagtgg ccttag agacagtccctgggaecccaggttctaggctgagggetggatgcccatccagcctgggagggccacacgggggc ctgggg acacaggggtcacccccaggggagaccaatggagggcacagagagggctctgggtctaggctgcagctctgtgg cctctg ctgggtcttcagggcatggggacacagaggaacggatgaggtcccagagcccagccctcccaggacagtcacca gaaagg agagggtctcttagtgcagagatgtgcctgtccctggagccctgtcatctctggggcctggtgtctctctgttc atgggt cgacctcccaccttcatttgaggaagggcaccttagactcagaaggtgactagcggggagtaaacgggagtgca gagaac tccatggctgccaggtgaagtccaggggcatcagaggctgctggggtgggcatgggggctgcggtgccccaaag tctggg ggagcagccccaagaacccagccgatgtgaagggtcctgtggtcgggctggtggggacaggggcgacggcagag ccccag ggtgtgtctgggtggagcccacgcttcaccaggagagctgagtgggccaggctggggcacagcctggtgcccca ggggat gggaagctccaggccatgccaggcttgggtctccccacatcctgccagtatagttttgtgtgctgtgagggaga ccccta gattccaa a ctcaga ctccaga a a ccagga aggagggagca cagcctgccctgggtgca cacggggaaaccgaggctgca gaggaaagggctgggccaggacacctgggaaaggtga cttgggaagggctcctagga aggcacagggctgtctgctctcc agagggctccagtggaaaggagggaatgaggagggaaggagaggccctgggtggaccaggcggccacaccatga accctc ccagagactttagacagagagaggcgctccacaacaccccacactccctctgccatctctcaccccctcctctg tccaca caggtcagcccaaggccaccccctcggtcactctgttcccgccgtcctctgaggagctccaagccaacaaggct acactg gtgtgtctcatgaatgacttttatccgggaatcttgacggtgacctggaaggcagatggtacccccatcaccca gggcgt ggagatgaccacgccctccaaacagagcaacaacaagtacgcggccagcagctacctgagcctgacgcccgagc agtgga ggtcccgcagaagctacagctgccaggtcatgcacgaagggagcaccgtggagaagacggtggcccctgcagaa tgttca taggttcccagccccgaccccacccaaaggggcctggagctgcaggatcccaggggaagggtctctctctgcat cccaag ccatcca gcccttctccctgtacccagta a a ccctaaataaataccctctttgtca a ccagaaa SEQ ID NO: 14 Human Asamino acid sequence (GenBank Accession No = NP_064455) mrpgtgqggleapgepgpnlrqrwpllllglavvthgllrptaasqsralgpgapggssrsslrsrwgrfllqr gswtgp rcwprgfqskhnsvthvfgsgtqltvlsqpkatpsvtifppsseelqankatlvclmndfypgiltvtwkadgt pitqgv emttpskqsnn kyaassyl sit peqwrsrrsyscqvrn hegstvektva paecs SEQ1DNO:15 Mouse Asnucleotide sequence (GenBank Accession No = AC_000038) ctggaatagcttttggecaccagaggaggaacaatcettttgccgggagatctacactgcaagtgaggctagag ttgact ttggacttgagggtcaatgaagctcagagtaggacagactctgggcactatccccaggcagtgtgaagttctcc tcctgc tgctgctgttgggtctagtggatggtgtccaccacata ctttccccaagctcagcagaaaggagcagagctgtgggccct ggagcttcagtgggaagcaacaggcctagcctatgggcccttcccggcaggtaagagacttgctttttggggaa ggtacg cgtgtaggtccacggactagaggctagaatgagtgactgggaaggaaggtggctattgaggccatgggtgtgag aaggaa ggatctgcctaagaggagggggctgtgcaaatgctagcttaaacttggtacctcgatcattgcaaggccagtgt tctacc aatgaaccacatccatagcccacctccattttccatttattttgagaagaggtcttgctaggtttcccaggctg atctta ctctctgtagcctatgcaagatttgaacgtcctatagctttcctagatccttcctccttcctaagtcctcttta gtagct agaattaagactgtggtggcgcgtttgtttttgttttttgttttttgtttttttgtgtgtgtgtgtgtgtgtgt catggt cctagttcatttagaattaaatgtgtgtgtttatcgtagtcaggtagttgcagttctagaagtttattcttagg taatta ttaaaacatccattaaatattgtgagggtgtcttgggttgggaggaggagacaccatggccatggagatcaccc agtcaa ga agaccttgcctgtaaagcatgaggactta attcagactctcagca accattcatcta ctatca agatgtttttactca gataaatagaaaaaaaaaaaaaaa aggtttggtgtggtggca caccttttatatcccagcagttgggaggcagaggcagg cagatctttgtgagtttgaggccagcctggtctacatagtagtctcatgataga a ata a a a a ca a a aga atccttgcaat atagtgtgcatgggtcctgggaggaagcatagggccagtaggaactgatgcacacacgcatgtatgcacacacg cacaag cacatatgca ca cacaaatattcaccagagattcctggttcca ccaca cccaccagagaagatataccacccaggaattt ctgagaatctgctgggcctgatattgccatcaacactgaaaaaattcagagatggaagtaagagatggatagat ggactt gtcgggatactggctgctcatctattttctgccaaggacatagtttatttcctgaagttctgtgtctgactcac ccaaca ggctcctgttccagatcatcccacggggagcaggtcccaggtgctcgccccataggcttccatctaagccccag ttttgg tatgtctttggtggtgggacccagctcacaatcctaggtaagtggttctcatggtctcatgatccagctggctc agggaa gtccatttttgctctggggaattcttactatctgcctttcctagccttgcagtctgaactgtaaaggcagtagt aattct a aggtaaatca cagggaaaggcccta a cagcttcatcta ctcttctgaggcagatgccca a a a agagatcaagga atgag atttttcaggccatagagaccactacacctcagcatctaaaccgaggcccagatgcccatccagactaagaaga ccacat agtaggtgcagggacatggctggtgccatgaaccccaccttcaaagattcaggcca ccctagataga a a a ccacagaggc tgagagagaaatttggttcaatttaatcttctgctggcccatgaggtcacagagacacacaaaaggctcagagt gatcag gtta ctagaaccaggtcctcccaggatgatta ctaga a a aga a atgtagactgtctattgttcctgagggctggagcctg ttgtctaacttgtccatcttcctcaaatatccttagacttagatgaggaatgaaggagcaaatggggccaagtg aaatca ggagtaaccagagacttcctggatggtcaggggtttgctatccttcctagtctgggagatggagcctcaggaac acagcc agtataggtcttgtgattactgtggacaaagcagtaggtccttggaggagttggaggatttttctggatggaat ctaatc ctcggtgagataactaaatggaatctggagcacaggccccgaagcccttatacagcaggcacacctcaagacca actctc caggaccagtcttgcagaataaatgagcagcaattctcagaggagtctctgagcactggagaagcaattcaggt tgggga gctgccctctgcctcaccagaaggccagggtcagatcccaattcactaccaggaggcctgggttagatcccaaa tcaggt tccagcttcaaggggctagagaattcagctggtcttagtctcagcgggggaactgagattgcaagggtctgggt ctgggt cattttatctggaagaggaacatgttctaatgggatgctaggctgtctgctctccaggggactcaagtggtcag aggaga agaaggaagcatccctggatggaagactgatgctgtagtgaatggccacagagctcctgataagagaaggacgc ttcctt atcacgtgggctctcctatgctaactcttatctccttctctatctgcgcaggtcagcccaagtctgaccccttg gtcact ctgttcctgccttccttaaagaatcttcaggccaacaaggccacactagtgtgtttggtgagcgaattctaccc aggtac tttggtggtggactggaaggtagatggggtccctgtcactcagggtgtagagacaacccaaccctccaaacaga ccaaca acaaatacatggtcagcagctacctgacactgatatctgaccagtggatgcctcacagtagatacagctgccgg gtcact catgaaggaaacactgtggagaagagtgtgtcacctgctgagtgttcttagaccacaatcctccctgaagcctc aggggc ctggatctga agtgccaga a a a agttgttttttgttttgttttttgttttttttcccatta a ccatctcactgtctttcc tgtgcctaata ctcaata a atatcttaccaccaac SEQ ID NO: 16 Mouse Asamino acid sequence (GenBank Accession No = NP_001177254) mklrvgqtlgtiprqcevlllllllglvdgvhhilspssaersravgpgasvgsnrpslwalpgrllfqiiprg agprcs phrlpskpqfwyvfgggtqltilgqpksdplvtlflpslknlqankatlvclvsefypgtlvvdwkvdgvpvtq gvettq pskqtnnkyrnvssyltlisdqwmphsryscrvthegntveksvspaecs SEQ ID NO: 17 Rat Asnucleotide sequence (GenBank Accession No = NC_OOS11O) atgaagctcagggcaggacagacactgggcactatccccaggcaatgtgaaattctccttctgttgctgctgtt gggcct ggtggatggtgtccaccatatactttccccaagctcagcacaaaggggcagagctgtgggccctggagcctcag tgggaa gcagcaggtctagcctgtggacccttccaggcaggtaagagactttcttataggggaatgtatgtgtgtgggtc catgga ctggaggctgaaatgggtgactgggaaggaaggtaaccattgaggccataggtgtgagaaggaaggatctgcct aagagg agggggctgggcaaatgctagcttaaacttagtacctaactcatgcaaggccagtattctatcaatgagccata tccata gcccacctcccttttccatttatttagagaagagggcttgcctgattgcctaggctgatcttactttctgtagc ctatgc aagatttgacctcctagatccttcctccttcctaagtcctctttagtagctggaattaaggctggtaccaggtt atttta gtgtgtcatggtcctagttcattcagaattgtgtgtgctcatcttagtcaaatagttgcagttttagaagttta ttctta ggcaattatcaaaacatccattaaatattctaagagtgtcttgggctgggaagagtagacactggggacatgga gaccac ccagtgaagaagactttga ctgtcaagcatgagga cctagttcaga ctctcagca ctcatgcatctgctatca agataca atcacatgtttttttactcagataaatagaaaaaaattaaggtttggtgtggtggcacatcccttatatcccag cagttg gcaggcagaggcagatagacctttgtgagtttgaggccagtctggtttacatagtggcctcatgacagaaataa aacaaa agaattctcacaaaatata gtgtgcatgggtcctgggatgaagcacagga ccaggagga ata ca caca cacacacaca ca cacacacacaca Ca ca cacacacacacaca a ata ca catcagagattcctggttccaccaca ctcaccagaga agatata ccacccagaacttttctgagaatcagttgggcctggtgttgccatcaactggatggaaataggagatggataga tggact tattttctgccaaggacacagttcatttcctgaagtccggtgcctacctcacccaacaggttcctgttccagat catccc acggggagcaggtcccaggtgctggccccataggcttccatccaagtcccagttatggtacgtctttggtagtg ggaccc agctcacaatcctaggtaagcgattcccatggtctcatgatccagctgtcttagggaagtccctttttcctctg gggagt tcttctcacctgcctttcctagtcttgtagtctgaatggtacctttttctgtgagtgaggggaaggcaagtagt tctaag gtaaaccacaggaaaggtcccaatagcttcagcta ctcttccgaggcagatgtcca a a a agggatcaggggctgaggttt tcaggctgtagagaccactgcacttcagcatcta a a ctgaggcccagatgcccatccagatta aga aggccacatagtag gtgcagggatatgactggtgccatgattcctgcctttaaagattcaggtaaccctagatagaaaaccacagagg ctgaga gaagaatctggcccagtttaatcttctgctggccaatgaggtcatagagacacagaaaaggcttagagtgacca ggtaac tagaaccagctcctcccaggatgatta ctaga a a a a a a atgtagactgtctattgttcctggggtttctcaggcctggag cctgttgtctaacttgtccatctccctcaaatgtccttagagttagatgaggaatgagggagcaaatggcgcca agtgaa atcaggagtcaccagagacttcctgggtggtcaggggtttgctatccttcctagtctgggagacggagccccag gagcac agtcagagtaggtcttgtgattactgtggacaaagcatcaggcccttggaggagttcctaggacttttcaggat ggaatc ta atccttggtgagataacta a ataga a tctggagca caggcccgggagccctta tacagcaggca ca cctcaagaccaa ctccccaggaccagtcttgcagaataaatgagcagtaattctcagaggaggctctgacactggagaagcaatgg gggttg gggagctgctccctgaactcccccacccccataggaggccagcgtcagatcccaattcagattccagctcctag tggcta gagagtacagatggccttggtcttagtggggaaactgagattgcaaggggcagggtgtgggtcatttcacctgg aagagg aacacggtctaatggggcaccaggctgtctgctctccaggggactcaggtgggcagaggaaaagaaggaagcat ccttga tggaacactctgagctgtagtgaatggctacagggctcctgataagaggaggatgcttccctgtcatgtgggct ctccta tgccaactcttatccccttctctatctgcacagggcagcccaagtctgaccccttggtcactctgttcctgcct tcctta aagaatctccaggttaagaaggcgacactagtgtgtctggtgagcgaattctacccaggtactttggtggtgga ctggaa ggta gatgggatccctgtcactcagggtgtggagacaacccaaccctccaaacagacca a ca a caagtacgtggccagca gctacctgacactgatgtctgaccaatggatgcctcacagtagatacacctgccaggtcactcatgaaggaaac actgtg gagaagagtgtgtcacctgctgaatgttcttag SEQ ID NO: 18 Rat A5amino acid sequence (GenBank Accession No = NP_001177270) mklragqtlgtiprqceilllllllglvdgvhhilspssaqrgravgpgasvgssrsslwtlpgrflfqiiprg agprcw phrlpsksqlwyvfgsgtqltilgqpksdplvtlflpslknlqvkkatlvclvsefypgtlvvdwkvdgipvtq gvettq pskqtnnkyvassyltlrnsdqwmphsrytcqvthegntveksvspaecs

Claims (1)

  1. <claim-text>CLAIMS: 1. A non-human vertebrate (eg, a mouse or rat) or cell (eg, a mouse cell or rat cell) whose genome comprises an antibody heavy chain transgene, the transgene comprising (a) one or more human Vt-i gene segments, one or more human D gene segments and one or more human il-I gene segments operably connected upstream of a constant region gene so that the transgene is capable of undergoing VDJ recombination in vivo to produce an antibody gene comprising a rearranged VDJC encoding an antibody heavy chain having a human variable region and a constant region; or (b) a rearranged VDJ encoding a human variable region operably connected upstream of a constant region gene so that the transgene encodes (optionally following VDJ combination with the constant region) a chimaeric antibody heavy chain having a human variable region and a constant region; the genome further comprising a human VpreB gene capable of expressing a human VpreB.</claim-text> <claim-text>2. The vertebrate or cell of claim 1, wherein said genome is homozygous for said transgene and endogenous non-human vertebrate antibody heavy chain expression has been inactivated.</claim-text> <claim-text>3. The vertebrate or cell of claim 2, wherein endogenous non-human vertebrate antibody light chain expression has been inactivated.</claim-text> <claim-text>4. The vertebrate or cell of claim 3, wherein the heavy chain transgene is devoid of a CR1 gene segment and the genome comprises no functional antibody light chain locus.</claim-text> <claim-text>5. The vertebrate or cell of any preceding claim, wherein said constant region is a Cmu, optionally endogenous Cmu.</claim-text> <claim-text>6. The vertebrate or cell of any preceding claim, wherein the human VpreB gene has a nucleotide sequence that is at least 85% identical to SEQ ID NO: 1.</claim-text> <claim-text>7. The vertebrate or cell of any preceding claim, wherein said genome does not comprise a non-human vertebrate (eg, mouse or rat) species VpreBl and/or VpreB2 gene.</claim-text> <claim-text>8. The vertebrate or cell of any preceding claim, wherein the genome further comprises a X5 gene; optionally wherein the itS gene is a itS of a non-human vertebrate (eg, mouse or rat) species and said constant region gene is a constant region gene of the same non-human vertebrate (eg, mouse or rat) species as the itS gene.</claim-text> <claim-text>9. The vertebrate or cell of claim 8, wherein said constant region gene and AS gene are endogenous genes of said vertebrate or cell.</claim-text> <claim-text>10. A method of constructing a transgenic non-human vertebrate cell (eg, an ES cell, eg, a mouse or rat ES cell), the method comprising (i) introducing into the genome of a non-human vertebrate cell (or an ancestor thereof) one or more human VH gene segments, one or more human D gene segments and one or more human JH gene segments so that said gene segments are operably connected upstream of a constant region gene (optionally an endogenous non-human vertebrate constant region gene) to form a heavy chain transgene, wherein in said cell or a progeny thereof the transgene is capable of undergoing VDJ recombination in vivo to produce an antibody gene comprising a rearranged VDJC encoding an antibody heavy chain having a human variable region and a constant region; or (ii) introducing into the genome of a non-human vertebrate cell (or an ancestor thereof) a rearranged VDJ encoding a human variable region so that said VDJ is operably connected upstream of a constant region gene (optionally an endogenous non-human vertebrate constant region gene) to form a heavy chain transgene, wherein in said cell or a progeny thereof the transgene encodes (optionally following VDJ combination with the constant region) an antibody heavy chain having a human variable region and a constant region; and wherein the method further comprises introducing into the cell a human VpreB gene capable of expressing a human VpreB.</claim-text> <claim-text>11. The method of claim 10, wherein said genome is homozygous for said transgene and endogenous non-human vertebrate antibody heavy chain expression is inactivated.</claim-text> <claim-text>12. The method of claim 10 or 11, wherein endogenous non-human vertebrate antibody light chain expression is inactivated.</claim-text> <claim-text>13. The method of claim 12, wherein the heavy chain transgene is devoid of a Cl-Il gene segment and the genome comprises no functional antibody light chain locus.</claim-text> <claim-text>14. The method of any one of claims 10 or 13, wherein said constant region is a Cmu.</claim-text> <claim-text>15. The method of any one of claims 10 to 14, wherein the human VpreB gene has a nucleotide sequence that is at least 85% identical to SEQ ID NO: 1.</claim-text> <claim-text>16. The method of any one of claims 10 to 15, wherein said genome does not comprise a non-human vertebrate (eg, mouse or rat) species VpreBl and/or VpreB2 gene.</claim-text> <claim-text>17. The method of any one of claims 10 to 16, wherein the genome further comprises a AS gene; optionally wherein the AS gene is a AS of a non-human vertebrate (eg, mouse or rat) species and said constant region gene is a constant region gene of the same non-human vertebrate (eg, mouse or rat) species as the AS gene.</claim-text> <claim-text>18. The method of claim 17, wherein said constant region gene and AS gene are endogenous genes of said vertebrate or cell.</claim-text> <claim-text>19. The method of any one of claims 10 to 18, comprising making a progeny of the cell made according to any one of claims 10 to 18, wherein the progeny is homozygous for said heavy chain transgene and endogenous non-human vertebrate antibody expression has been inactivated.</claim-text> <claim-text>20. A method of promoting B-cell development in a non-human vertebrate (eg, a mouse or rat), the method comprising (a) providing in a non-human vertebrate embryonic stem (Es) cell genome an immunoglobulin transgene capable of expressing an antibody mu heavy chain, wherein the antibody heavy chain comprises a human variable region and a mu constant region (optionally an endogenous non-human vertebrate mu constant region); and creating a first non-human vertebrate from said ES cell or a progeny thereof; (b) providing in a second ES cell genome a second transgene comprising a human VpreB gene capable of expressing a human VpreB; and creating a second non-human vertebrate from said ES cell or a progeny thereof; and (c) creating by breeding a third non-human vertebrate capable of co-expressing the mu antibody heavy chain and human vpreB wherein a pre-R-cell receptor can form to promote B-cell development of cells bearing a mu heavy chain in said third vertebrate; the third vertebrate being made by crossing said first and second vertebrates or progeny thereof by breeding to create said third vertebrate, the third vertebrate comprising the first and second transgenes, and wherein endogenous heavy chain expression has been inactivated in said third vertebrate.</claim-text> <claim-text>21. The method of claim 20, wherein in (a) the heavy chain transgene is constructed to be devoid of a CF-Il gene segment and the genome of the third non-human vertebrate comprises no functional antibody light chain locus.</claim-text> <claim-text>22. The method of claim 20 or 21, wherein the human VpreB gene has a nucleotide sequence that is at least 85% identical to SEQ ID NO: 1.</claim-text> <claim-text>23. The method of any one of claims 20 to 22, wherein the genome of the third vertebrate does not comprise a non-human vertebrate (eg, mouse or rat) species VpreBl and/or VpreBZ gene.</claim-text> <claim-text>24. The method of any one of claims 20 to 23, wherein the third non-human vertebrate expresses a X5 of a non-human vertebrate (eg, mouse or rat) species and said constant region is a constant region of the same non-human vertebrate (eg, mouse or rat) species as the X5 gene; optionally wherein the X5 and constant region are an endogenous X5 and constant region of mouse or rat.</claim-text> <claim-text>25. A transgenic mouse or rat according to the third vertebrate of any one of claims 20 to 24, or a progeny thereof.</claim-text> <claim-text>26. A method of promoting B-cell development in a non-human vertebrate (eg, a mouse or rat), the method comprising (i) inactivating endogenous heavy chain expression in said vertebrate;S(ii) providing in the genome of said vertebrate an immunoglobulin tiansgene capable of expressing an antibody mu heavy chain, wherein the antibody heavy chain comprises a human variable region and a non-human vertebrate mu constant region (optionally an endogenous non-human vertebrate mu constant region); and (ii) providing in the genome of said vertebrate a second transgene capable of expressing a human VpreB wherein a pre-B-cell receptor can form to promote B-cell development of cells bearing a mu heavy chain in said vertebrate; optionally wherein the genome is homozygous for said first transgene.</claim-text> <claim-text>27. The method of claim 26, wherein in the heavy chain transgene is devoid of a CH1 gene segment and the genome of the vertebrate comprises no functional antibody light chain locus.</claim-text> <claim-text>28. The method of claim 26 or 27, wherein the human VpreB gene has a nucleotide sequence that is at least 85% identical to SEQ ID NO: 1.</claim-text> <claim-text>29. The method of any one of claims 26 to 28, wherein the genome of the vertebrate does not comprise a non-human vertebrate (eg, mouse or rat) species VpreBl and/or VpreBZ gene.</claim-text> <claim-text>30. The method of any one of claims 26 to 29, wherein the non-human vertebrate expresses a AS of a non-human vertebrate (eg, mouse or rat) species and said constant region is a constant region of the same non-human vertebrate (eg, mouse or rat) species as the AS gene; optionally wherein the AS and constant region are an endogenous AS and constant region of mouse or rat.</claim-text> <claim-text>31. A transgenic mouse or rat according to the vertebrate of any one of claims 26 to 30, or a progeny thereof.</claim-text> <claim-text>32. A non-human vertebrate, mouse, rat, cell or method according to any preceding claim, wherein the expression of the human VpreB gene is under edogenous control.</claim-text> <claim-text>33. The non-human vertebrate, mouse, rat, cell or method according to claim 32, wherein the human VpreB gene is operably linked to an endogenous promoter, optionally an endogenous VpreB promoter (eg, a VpreBl promoter).</claim-text> <claim-text>34. The non-human vertebrate, mouse, rat, cell or method of any preceding claim, wherein the genome comprises (a) said antibody heavy chain transgene; and (b) an antibody kappa light chain transgene and/or an antibody lambda chain transgene; wherein all of the V1 D and J in said transgenes are human V, D and J; wherein endogenous antibody heavy and light chain expression has been inactivated; and optionally wherein said genome is homozygous for said heavy and light chain transgenes.</claim-text> <claim-text>35. The non-human vertebrate, mouse, rat, cell or method of claim 34, wherein the kappa and lambda chain transgenes comprise constant regions of said non-human vertebrate species capable of pairing with the constant region of the heavy chain.</claim-text> <claim-text>36. The non-human vertebrate, mouse, rat, cell or method of claim 340135, wherein the heavy chain transgene comprises a substantially complete human functional Vh, D and JH repertoire.</claim-text> <claim-text>37. The non-human vertebrate, mouse, rat, cell or method of claim 34,35 or 36, wherein the kappa chain transgene comprises a substantially complete human functional VK and JK repertoire; and the lambda chain transgene comprises a substantially complete human functional Vk and JA repertoire.</claim-text> <claim-text>38. A transgenic mouse or rat, or a transgenic mouse or rat cell (eg, an ES cell), whose genome comprises (a) an antibody heavy chain transgene, the transgene comprising a substantially complete human functional VH, D and JH repertoire operably connected upstream of an endogenous (mouse or rat) mu constant region gene so that the transgene is capable of undergoing VDJ recombination in vivo to produce an antibody gene comprising a rearranged VDJC encoding an antibody heavy chain having a human variable region and an endogenous mu constant region; (b) an antibody kappa light chain transgene and/or an antibody lambda chain transgene; -wherein all of the V, D and J in said transgenes are human V, D and J; -wherein the kappa chain transgene comprises a substantially complete human functional VK and Ji< repertoire; and the lambda chain transgene comprises a substantially complete human functional VA and JA repertoire; -wherein the kappa and/or lambda chain transgenes comprise endogenous constant regions capable of pairing with the constant region of the heavy chain; (c) a human VpreB gene capable of expressing a human VpreB; -optionally wherein the human VpreB gene is operably linked to an endogenous VpreB promoter; and (d) an endogenous AS gene capable of expressing an endogenous AS; -wherein endogenous antibody heavy and light chain expression has been inactivated; and -optionally wherein said genorne is homozygous for said heavy and light chain transgenes.</claim-text> <claim-text>39. A transgenic mouse or rat, or a transgenic mouse or rat cell (eg, an ES cell), whose genome comprises (a) an antibody heavy chain transgene, the transgene comprising a substantially complete human functional VH, D and JH repertoire operably connected upstream of an endogenous (mouse or rat) mu constant region gene so that the transgene is capable of undergoing VDJ recombination in vivo to produce an antibody gene comprising a rearranged VDJC encoding an antibody heavy chain having a human variable region and an endogenous mu constant region; -wherein all of the V, D and J in said transgene are human V, D and J; -wherein the heavy chain transgene is devoid of a CF-Il gene segment and the genome of the vertebrate comprises no functional antibody light chain locus; -wherein endogenous antibody heavy chain expression has been inactivated; and -optionally wherein said genome is homozygous for said heavy chain transgene; (c) a human VpreB gene capable of expressing a human VpreB; -optionally wherein the human VpreB gene is operably linked to an endogenous VpreB promoter; and (d) an endogenous X5 gene capable of expressing an endogenous A5.</claim-text>
GB1116495.1A 2011-09-26 2011-09-26 Human VpreB and chimaeric surrogate light chains in transgenic non-human vertebrates Withdrawn GB2495083A (en)

Priority Applications (5)

Application Number Priority Date Filing Date Title
GB1116495.1A GB2495083A (en) 2011-09-26 2011-09-26 Human VpreB and chimaeric surrogate light chains in transgenic non-human vertebrates
PCT/GB2012/052380 WO2013045916A1 (en) 2011-09-26 2012-09-26 Chimaeric surrogate light chains (slc) comprising human vpreb
EP12766714.5A EP2761008A1 (en) 2011-09-26 2012-09-26 Chimaeric surrogate light chains (slc) comprising human vpreb
US14/226,698 US9963716B2 (en) 2011-09-26 2014-03-26 Chimaeric surrogate light chains (SLC) comprising human VpreB
US15/973,376 US20180282761A1 (en) 2011-09-26 2018-05-07 Chimaeric Surrogate Light Chains (SLC) Comprising Human VpreB

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
GB1116495.1A GB2495083A (en) 2011-09-26 2011-09-26 Human VpreB and chimaeric surrogate light chains in transgenic non-human vertebrates

Publications (2)

Publication Number Publication Date
GB201116495D0 GB201116495D0 (en) 2011-11-09
GB2495083A true GB2495083A (en) 2013-04-03

Family

ID=44993319

Family Applications (1)

Application Number Title Priority Date Filing Date
GB1116495.1A Withdrawn GB2495083A (en) 2011-09-26 2011-09-26 Human VpreB and chimaeric surrogate light chains in transgenic non-human vertebrates

Country Status (1)

Country Link
GB (1) GB2495083A (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016062990A1 (en) * 2014-10-22 2016-04-28 Crescendo Biologics Limited Transgenic mice
GB2561352A (en) * 2017-04-10 2018-10-17 Genome Res Ltd Animal models and therapeutic molecules
GB2584219A (en) * 2014-10-22 2020-11-25 Crescendo Biologics Ltd Transgenic mice

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008118970A2 (en) * 2007-03-27 2008-10-02 Sea Lane Biotechnologies, Llc Constructs and libraries comprising antibody surrogate light chain sequences
WO2011004192A1 (en) * 2009-07-08 2011-01-13 Genome Research Limited Animal models and therapeutic molecules
WO2011062206A1 (en) * 2009-11-17 2011-05-26 協和発酵キリン株式会社 Human artificial chromosome vector
WO2011097603A1 (en) * 2010-02-08 2011-08-11 Regeneron Pharmaceuticals, Inc. Common light chain mouse

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008118970A2 (en) * 2007-03-27 2008-10-02 Sea Lane Biotechnologies, Llc Constructs and libraries comprising antibody surrogate light chain sequences
WO2011004192A1 (en) * 2009-07-08 2011-01-13 Genome Research Limited Animal models and therapeutic molecules
WO2011062206A1 (en) * 2009-11-17 2011-05-26 協和発酵キリン株式会社 Human artificial chromosome vector
WO2011097603A1 (en) * 2010-02-08 2011-08-11 Regeneron Pharmaceuticals, Inc. Common light chain mouse

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016062990A1 (en) * 2014-10-22 2016-04-28 Crescendo Biologics Limited Transgenic mice
CN107002092A (en) * 2014-10-22 2017-08-01 克雷森多生物制剂有限公司 Transgenic mice
GB2547587A (en) * 2014-10-22 2017-08-23 Crescendo Biologics Ltd Transgenic mice
EP3461848A1 (en) * 2014-10-22 2019-04-03 Crescendo Biologics Limited Transgenic mice
GB2584219A (en) * 2014-10-22 2020-11-25 Crescendo Biologics Ltd Transgenic mice
GB2547587B (en) * 2014-10-22 2020-12-09 Crescendo Biologics Ltd Transgenic mice
GB2584219B (en) * 2014-10-22 2021-03-24 Crescendo Biologics Ltd Transgenic Mice
US11547099B2 (en) 2014-10-22 2023-01-10 Crescendo Biologies Limited Transgenic mice
GB2561352A (en) * 2017-04-10 2018-10-17 Genome Res Ltd Animal models and therapeutic molecules
GB2561352B (en) * 2017-04-10 2023-01-18 Genome Res Ltd Animal models and therapeutic molecules

Also Published As

Publication number Publication date
GB201116495D0 (en) 2011-11-09

Similar Documents

Publication Publication Date Title
AU2021200079B2 (en) Animal models and therapeutic molecules
US20180282761A1 (en) Chimaeric Surrogate Light Chains (SLC) Comprising Human VpreB
US20150033372A1 (en) Human VpreB &amp; Chimaeric Surrogate Light Chains in Transgenic Non-Human Vertebrates
TR201906650T4 (en) Common light chain mouse.
GB2495083A (en) Human VpreB and chimaeric surrogate light chains in transgenic non-human vertebrates

Legal Events

Date Code Title Description
WAP Application withdrawn, taken to be withdrawn or refused ** after publication under section 16(1)