US20050043519A1 - Antigen binding domains - Google Patents

Antigen binding domains Download PDF

Info

Publication number
US20050043519A1
US20050043519A1 US10/486,608 US48660804A US2005043519A1 US 20050043519 A1 US20050043519 A1 US 20050043519A1 US 48660804 A US48660804 A US 48660804A US 2005043519 A1 US2005043519 A1 US 2005043519A1
Authority
US
United States
Prior art keywords
antigen binding
binding domain
process according
specific antigen
antigen specific
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US10/486,608
Inventor
Helen Dooley
Andrew Porter
Martin Flajnik
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.)
University of Aberdeen
University of Maryland at Baltimore
Original Assignee
Helen Dooley
Andrew Porter
Martin Flajnik
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from GB0119553A external-priority patent/GB0119553D0/en
Priority claimed from GB0210508A external-priority patent/GB0210508D0/en
Application filed by Helen Dooley, Andrew Porter, Martin Flajnik filed Critical Helen Dooley
Publication of US20050043519A1 publication Critical patent/US20050043519A1/en
Assigned to MARYLAND, UNIVERSITY OF reassignment MARYLAND, UNIVERSITY OF ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: FLAJNIK, MARTIN
Assigned to ABERDEEN UNIVERSITY reassignment ABERDEEN UNIVERSITY ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: DOOLEY, HELEN, PORTER, ANDREW
Assigned to NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF HEALTH AND HUMAN SERVICES (DHHS), U.S. GOVERNMENT reassignment NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF HEALTH AND HUMAN SERVICES (DHHS), U.S. GOVERNMENT CONFIRMATORY LICENSE (SEE DOCUMENT FOR DETAILS). Assignors: UNIVERSITY OF MARYLAND BALT PROF SCHOOL
Priority to US13/280,837 priority Critical patent/US8865431B2/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/005Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies constructed by phage libraries
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/40Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against enzymes
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/565Complementarity determining region [CDR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/569Single domain, e.g. dAb, sdAb, VHH, VNAR or nanobody®

Definitions

  • the present invention relates to the production of antigen specific antigen binding domains (single domain antibodies) from fish, where the term fish encompasses both cartilaginous (subclass Elasmobranchii) and bony fish(class Osteichthyes).
  • antigen specific antigen binding domains we mean the variable region of a Novel Antigen Receptor (NAR).
  • Antibodies are useful in, among other things, molecular diagnostics and therapeutics because of their high affinity binding and specificity.
  • monoclonal antibodies are produced using animal models the production of human monoclonal antibodies remains difficult.
  • the body mounts an immune response because the monoclonal antibody is foreign to the human system.
  • Single domain antibodies are also important as they can penetrate tissues taking with them any linked compounds. In addition, they can bind within cavities on the surface of proteins, for example within enzyme binding sites thus disrupting function.
  • Single domain antibodies produced from Camelidae have been shown to recognize protein cavities and as such have the ability to inhibit enzymes (Lauwereys et al., EMBO 17 pp3512-3520 1998).
  • An object of the present invention is to provide a process for the production of antigen specific antigen binding domains which seeks to alleviate the above problems.
  • a further object of the present invention is to provide a composition comprising antigen specific antigen binding domains for the inhibition of protein activity which seeks to alleviate the above problems.
  • antigen specific antigen binding domains produced from the variable region of NAR found in fish are as stable as single domain antibodies produced from members of the Camelidae family.
  • the immunoglobulin isotype now known as NAR Novel Antigen Receptor
  • NAR Novel Antigen Receptor
  • NAR Novel Antigen Receptor
  • teleost fish which are much more advanced evolutionary than sharks, appear to rely solely upon the production of a low affinity, non-specific IgM type response (Watts et al., Aust Vet J 79 pp570-574 2001).
  • a defining characteristic of teleost IgM is their low affinity and ability to non-specifically bind multiple antigens.
  • IgM neutralisation is through non-specific multiple binding, resulting mainly in agglutination, etc.
  • Neutralisation without complement is usually associated with specific, high affinity binding and had not until this invention been seen in fish species.
  • the antigen specific antigen binding domains of the present invention have been shown to neutralise activity of an enzyme immunogen directly without calling upon other components of the immune system.
  • the NAR variable (V) region conforms to the model of typical Ig superfamily domains with the predicted canonical, intradomain disulphide bond. However, whilst camelid VHH regions have up to 75% sequence identity with other mammalian VH regions, the identity between NAR V and conventional VH domains is as low as 25% (Roux et al., Proceedings of the National Academy of Sciences. USA 95 pp11804-11809 1998).
  • NAR V region sequences Due to this low identity and lack of NAR sequences in the Kabat database, the amino acids of NAR V regions have previously been numbered sequentially (Roux et al., Proceedings of the National Academy of Sciences. USA 95 pp11804-11809 1998). To enable easy comparison of residues in different NAR V molecules, or NAR V region sequences with those of other species during this work, a numbering system was derived for NAR V region based upon that of Kabat et al., (1991) ( Sequences of Proteins of immunological Interest, 5 th Edition. National Institutes of Health, Betheseda, USA ). (Note: this numbering system is used in the Figures attached hereto).
  • NAR V domains were classified into two closely related classes (type I or II), both being constructed from one V, three D and one J segment.
  • Type I regions have non-canonical cys residues in Fr2 (C35) and Fr4 (C103), which likely form a domain-stabilising disulphide bond.
  • C35 Fr2
  • Fr4 C103
  • additional cysteine residue pairs have been observed and almost certainly form disulphide bridges within the CDR, as is found in some cattle VH domains with an unusually long CDR3.
  • Type II regions are very similar in overall structure to type I but instead have non-canonical cysteine residues located in Fr1 (C29) and CDR3, which are proposed to form a constraining disulphide bond like that observed in camelid VHH domains.
  • the presence of cysteines within each NAR type is shown in schematic form in FIG. 1 .
  • NAR type has been identified as the predominant expressed form in nurse shark pups (Type III) but due to its germline joined state displays no junctional diversity.
  • type I and II NAR the DNA encoding the V region is generated by the physical joining of DNA segments which are spatially separate in the genome. This joining process occurs in B-cells and helps generate the diversity of sequence seen for these NAR types. For type III these DNA segments are already physically joined in the DNA of all cells, hence the term germline joined.
  • NAR possesses the cluster type genomic organisation usually observed for Ig receptors in cartilaginous fish, but less than five NAR loci are thought to exist, with only two or three being capable of functional rearrangement and expression.
  • the diversity observed in the primary repertoire is generated through recombination mechanisms and, although extensive (due to the presence of three D segments), is localised to CDR3. On encounter with antigen this repertoire is rapidly expanded by extensive mutation.
  • the pattern of mutation in NAR is unlike that observed in shark IgM, which shows low levels of mutation and poor clustering to CDRs, but rather bears the hallmarks of mammalian-like somatic mutation.
  • NAR V is similar to VH, VL and TCR V but distinct from all three, hence its “unique domain architecture”.
  • the VH name has been used in the past because the constant portion of NAR is a heavy chain but the V region is actually more like VL/TCR V than VH (i.e. groups closer on a phylogenetic tree).
  • NAR V is not like the camel VHH domains which are derived from bona fide heavy chain V regions.
  • the antigen binding domain of the NAR is closer to a VL domain naturally lacking VH rather than the other way round.
  • NAR V and camel VHH clearly shows the huge difference in sequence. If NAR V and camel VHH have the same physical structure (which has been implied but not proven) they achieve this using completely different amino acid sequences, and one would not be able to amplify a NAR V region library using camel VHH library primers. In addition, the ways in which the NAR V and camel VHH gene repertoires are created during VDJ joining are different due to the organisation of the immunoglobulin genes (Schluter et al Immunol Today 18 pp543-549 1997).
  • the transformed host is a prokaryote or a lower eukaryote.
  • the prokaryote host is Escherichia coli.
  • the expressible DNA sequence is in the form of a phagemid vector.
  • Phagemid expression has advantages over phage genome expression in that it results in greater genetic stability and the bacterial transformation efficiency is higher thus enabling the construction of potentially larger and more diverse libraries.
  • the gene encoding the variable region of the antibody can be fused to that of a phage surface protein, usually gene III or VIII.
  • Gene III fusion is favoured due to its limited copy number (3-5 copies) on the tip of each phage, minimising possible avidity effects which are undesirable when trying to isolate binders of high affinity.
  • the antibody fragment genes can be cloned directly into the phage genome or fused to gene segments present within phagemid plasmids.
  • the fish is a member of the Elasmobranchii subclass, for example, a shark or a dogfish.
  • a greater number of smaller members of the Elasmobranchii subclass can be kept in tanks which are smaller in unit area than the grazing area required for the same number of members of the Camelidae family. As the members of the Elasmobranchii subclass are kept in tanks they can easily be caught for extraction of their blood.
  • the shark is a nurse shark, Ginglymostoma cirratum.
  • the antigen specific antigen binding domain has a specific specificity. Accordingly, the antigen specific antigen binding domain can be targeted to a specific antigen(s).
  • the antigen specific antigen binding domain is monoclonal.
  • the antigen specific antigen binding domain is raised to a single antigen.
  • the specificity of the antigen specific antigen binding domain is determined by an antigen which is introduced into the chosen fish.
  • Screening of displayed libraries for specific binding sites involves repeated cycles of selection with the desired antigen in the process of biopanning.
  • the library of phage displayed antigen binding domains is incubated with immobilised antigen, unbound phage are washed out and bound phage eluted.
  • This selected population is expanded by bacterial infection and put through further rounds of selection.
  • Such cyclic panning has thus proven able to enrich for clones of high affinity, much like in vivo antibody selection.
  • step d) the cDNA of the antigen binding domains is generated.
  • restriction enzymes are used to digest the amplified DNA sequences encoding the antigen specific antigen binding domain.
  • the restrictions enzymes can be chosen depending upon, for example, the handle of the primers used in the above process.
  • restriction enzymes are NcoI and NotI.
  • the display vector is any phagemid vector, for example, pHEN2.
  • the expression vector is a soluble expression vector such as pIMS100.
  • vectors are merely examples of the vectors which can be used. It is common general knowledge to those skilled in the art which vectors can be used.
  • an antigen specific antigen binding domain produced by the process as defined above.
  • composition for the inhibition of protein activity comprising antigen specific antigen binding domains derived from a variable region of the immunoglobulin isotype NAR found in fish.
  • NAR V region is 12 kDa which is 20% smaller than any 15 kDa single domain antibody derived from Camelidae, it was still possible to alter protein activity therewith. Size is a significant factor in the therapeutic applications of antigen specific antigen binding domains and other single domain antibodies, with therapeutic benefits of increased tissue penetration, better access to protein clefts for neutralisation via steric hindrance and reduced immunogenicity, resulting from the use of antigen specific antigen binding domains of the present invention.
  • Antigen specific antigen binding domains derived from NAR therefore have a wider target population than single domain antibodies derived from Camelidae by virtue of their smaller size.
  • the potential for immunogenicity is also reduced since in general the smaller the size of a protein the less the immunogenicity.
  • NAR sequences have, in work previous to that of the inventors, been identified at the DNA level, there has been no clue from the DNA evidence that a somatically maturable repertoire, capable of selecting high affinity, specific binders could be a characteristic of the NAR response.
  • a somatically maturable repertoire capable of selecting high affinity, specific binders could be a characteristic of the NAR response.
  • Sequencing of these genes confirms that an atypical (for fish and organisms of this evolutionary lineage) somatically-maturable (showing mutation from the germ line repertoire) response occurs within the NAR repertoire, driven by the immunisation process. This has resulted in the selection of highly specific, high affinity antigen binding domains capable of antigen neutralisation in isolation and not the expected non-specific, low affinity IgM like response typically found in fish and sharks.
  • the inventors have been able to isolate NAR antigen specific antigen binding domains and demonstrate for the first time that the NAR V is able to fold and function in isolation from the rest of the molecule (and in a non-shark environment), that the antigen specific antigen binding domain matures from the germ line genes to become specific for antigen (only possible with a library derived from mRNA and not DNA) and that the antigen specific antigen binding domain is able to bind specifically to the immunising antigen.
  • the inventors have been able to immunize a shark and derive from this immunization a specific, somatically matured antigen specific antigen binding domain that is of high affinity and specific for the immunogen.
  • the antigen specific antigen binding domain is able to neutralise the activity of the immunogen directly, without calling upon other components of the immune system. According to previous understandings, this should not have been possible for a primitive species such as sharks.
  • composition wherein the antigen specific antigen binding domain is a product of the process as defined above.
  • inhibition of protein activity is in a concentration dependent manner.
  • the composition further comprises a pharmaceutical carrier or diluent therefor.
  • Such pharmaceutical carriers are well known in the art.
  • an antigen specific antigen binding domain produced from a variable region of NAR.
  • FIG. 1 shows the presence of cysteine amino acid residues within each NAR type, and human, cattle and camel variable regions for comparison.
  • Canonical cysteines are shown by and non-canonical cysteines are shown by .
  • FIGS. 2A, 2B and 2 C show the amino acid translations of the sequences obtained in the Examples (SEQ ID. 1 to 51).
  • the sequences are aligned against a typical type I and type II clone sequence (top of each Figure with CDR's highlighted in bold) dashes indicate identity to the type I clone and * indicates an in-frame stop codon.
  • FIG. 3 shows NAR type I and II variable region amino acid sequence alignment (SEQ 1 and 2).
  • Germline sequence is given for type I, whilst that given for type II is typical of those observed from somatically mutated cDNA sequences (Roux et al., Proceedings of the National Academy of Sciences. USA 95 pp11804-11809 1998). Sequence identity is indicated by a dash and the CDR's of both sequences are in bold. The numbering above the sequences was generated by comparison of conserved residues (underlined) with those of other species and is used to enable comparison of NAR V region sequences.
  • FIG. 4 shows a variability plot for the 29 immune library sequences identified in the Example (pre-selection and functional). Variability at each position was calculated according to the method of Wu & Kabat (1970) ( Journal of Experimental Medicine 132 pp211-250). The canonical cysteine residues, C22 and 92, are marked by an asterisk.
  • FIGS. 5A and B show polyclonal and monoclonal phage ELISA results for selection on Hen egg white lysozyme (HEL) ( FIG. 5A ) and Chicken ovalbumin (Ova) ( FIG. 5B ). Phage numbers were normalised for each pan prior to polyclonal analysis.
  • HEL Hen egg white lysozyme
  • Ova Chicken ovalbumin
  • FIG. 6 shows the DNA (SEQ ID. 53 & 54) and encoded amino acid sequence (SEQ ID. 52) of the ⁇ -HEL 5A7 clone. CDRs are highlighted in bold.
  • FIG. 7 shows the DNA (SEQ ID. 56 & 57) and encoded amino acid sequence (SEQ ID. 55) of the ⁇ -HEL 4F11 clone. CDRs are highlighted in bold.
  • FIG. 8 shows the amino acid alignment of the two ⁇ -HEL clones, 5A7 (SEQ ID. 52) and 4F11 (SEQ ID. 55), with a typical type I clone (SEQ ID. 1). Sequences are numbered according to FIG. 3 for ease of comparison, differences between the selected clones are highlighted in underlined and CDR's are highlighted in bold, * conserved residues in all sequences, : conserved substitutions, . semi-conserved substitutions.
  • FIG. 9 shows the DNA (SEQ ID. 59 & 60) and encoded amino acid sequence (SEQ ID. 58) of the ⁇ -Ova 4H11 clone. CDRs are highlighted in bold.
  • FIG. 10 shows the DNA (SEQ ID. 62 & 63) and encoded amino acid sequence (SEQ ID. 61) of the ⁇ -Ova 3E4 clone. CDRs are highlighted in bold.
  • FIG. 11 shows amino acid alignment of the two ⁇ -Ova clones, 4H11 (SEQ ID. 58) and 3E4 (SEQ ID. 61), with a typical type I clone (SEQ ID. 1). Sequences are numbered according to FIG. 3 for ease of comparison, differences between the selected clones are underlined and the CDR's are highlighted in bold. * conserved residues in all sequences, : conserved substitutions, . semi-conserved substitutions.
  • FIG. 12 shows binding analysis of ⁇ -HEL clone 5A7. Serial dilutions of crude periplasmic release solution were applied to an ELISA plate coated with each of the test proteins at 10 ⁇ g/ml and blocked with Marvel. Data presented is a mean of triplicate wells and representative of at least three repeat assays.
  • FIG. 13 shows binding analysis of ⁇ -HEL clone 4F11. Serial dilutions of crude periplasmic release solution were applied to an ELISA plate coated with each of the test proteins at 10 ⁇ g/ml and blocked with Marvel. Data presented is a mean of triplicate wells and representative of at least three repeat assays.
  • FIG. 14 shows binding analysis of ⁇ -Ova clone 4H11. Serial dilutions of crude periplasmic release solution were applied to an ELISA plate coated with each of the test proteins at 10 ⁇ g/ml and blocked with Marvel. Data presented is a mean of triplicate wells and representative of at least three repeat assays.
  • FIG. 15 shows a comparison of the stability of the anti-HEL clones 5A7 and 4F11 to irreversible thermal denaturation. Data presented is a mean of triplicate wells and representative of at least three repeat assays.
  • FIG. 16 shows a lysozyme enzymatic inhibition assay.
  • Purified HEL-5A7 NAR V region protein at a final concentration of 2500 nM (filled circle), 250 nM (open triangle) or 25 nM (filled square) were pre-incubated with HEL prior to the introduction of M. lysodeikticus bacterium.
  • the control wells (open diamond) contained buffer in place of HEL-5A7 protein.
  • the data presented is an average of 3 replicates and a typical data set from three repeat experiments.
  • the electroporation-competent strain E. coli XL1-Blue ⁇ recA1 endA1 gyrA96 thi-1 hsdR17 supE44 relA1 lac [F′ proAB lacI q Z ⁇ M15 Tn10 (Tet r )] ⁇ (Stratagene Ltd.) was used to prepare and pan the NAR V region phage display libraries.
  • RNA from bleeds 4 and 5 for each of the three sharks was pooled and stored at ⁇ 80° C. until required for cDNA synthesis.
  • ready-to-go RT-PCR beads 200 ⁇ M each dNTP, 10 mM Tris-HCl buffer, 60 mM KCl, 1.5 mM MgCl 2 , M-MuLV reverse transcriptase, RNAguard7, RNase/DNase free BSA and 2 U Taq DNA polymerase (APB Ltd.) were reconstituted in 45 ⁇ l of DEPC treated H 2 O by incubating on ice for 5 min or until the beads were completely dissolved.
  • To each tube 2 ⁇ l of nurse shark tRNA at 2 ⁇ g/ ⁇ l and 2 ⁇ l of NAR F4 For primer or F4 For2 primer at 25 pM/ ⁇ l were added.
  • Both of these primers are specific for NAR framework region 4 and have a NotI site incorporated in the handle to allow subsequent cloning into the phagemid vector. Tubes were flicked gently to mix contents and incubated on a PCR block pre-warmed to 46° C. for 30 min. Following cDNA synthesis, tubes were incubated at 95° C. for 7 min to inactivate the reverse transcriptase and denature the template.
  • tubes were pre-heated to 95° C. and 1 ⁇ l of Taq DNA polymerase at 1 U/ ⁇ l added to each prior to cycling 32 times at 95° C. for 2 min, 55° C. for 1 min and 72° C. for 1 min 30 s.
  • PAGE-purified PCR product was digested with NcoI and NotI restriction enzymes, at the sites incorporated by the handled primers used for amplification, to allow cloning into the phagemid vector pHEN2. Restricted DNA was purified on a 1.5% agarose gel and the DNA excised and cleaned.
  • Plasmid DNA harvested from an overnight culture of E. coli XL1-Blue and phenol:chloroform treated, was similarly cut with NcoI and NotI restriction enzymes. Double-cut vector was purified on a 0.7% agarose gel and DNA extracted. For library construction digested vector was not treated with calf alkaline phosphatase.
  • Ligated DNA was aliquotted into chilled electroporation cuvettes and to each 40 ⁇ l of freshly thawed electroporation-competent XL1-Blue cells was added. Cells were electroporated and resuspended in 100 ⁇ l ice-cold 2 ⁇ TY media with 1% glucose (w/v) added. Dilutions at 10 ⁇ 2 , 10 ⁇ 4 and 10 ⁇ 6 were performed for each transformation and plated on TYE agar containing 100 ⁇ g/ml ampicillin and 1-2% glucose (w/v). The remaining bacterial suspension was plated straight onto 140 mm petri-dishes containing TYE with ampicillin and glucose (as above). All plates were grown overnight at 37° C.
  • PCR was undertaken as follows; 1 cycle at 95° C. for 3 minutes (to lyse bacteria) and 20 cycles of 95° C. for 1 min, 55° C. for 1 min and 72° C. for 1 min.
  • PCR product was run on a 1.5% agarose gel containing EtBr against molecular weight marker VI (Boehringer Ltd.) to evaluate the percentage of the library carrying NAR V region insert.
  • EtBr against molecular weight marker VI Boehringer Ltd.
  • a further 15 clones carried frameshift mutations leading to the production of non-sense or truncated proteins.
  • the frameshift occurred within CDR3, possibly as a consequence of nucleotide addition or deletion during the recombination process.
  • the frameshift mutation arose within Fr2 (position 41 according to FIG. 3 ) and Fr3 (position 67) respectively and are more likely due to polymerase errors during library construction (the frameshift in clone 14 occurs immediately after a long poly-A tract in the DNA sequence).
  • FIGS. 2 and 4 show good diversity, with each clone having a unique amino acid sequence. Variability is seen to be focussed across CDR3 which, like clones from a similarly constructed na ⁇ ve library, varied greatly in both sequence and length.
  • the immune nature of the library is important as NAR V regions which bound to antigen could not be isolated from a na ⁇ ve library (ie. without prior immunisation).
  • NAR types were represented, with approximately 80% being type I and 20% type II, however a number of clones proved difficult to assign to an NAR type.
  • clone 33 has a type II Fr1 but type I CDR3 and Fr4, whilst clones 06, 40 and 46 have a type I Fr1 and CDR3 but a type II Fr2 and Fr4. This finding suggests the possibility that gene conversion may be occurring between the NAR genes.
  • Clones 24 and 36 are both assigned as type I on the basis of other sequence characteristics but do not carry the pair of cysteine residues normally observed in the type I CDR3.
  • the clones 06, 40, 46 and 48 all encode an uneven number of cysteine residues. As mentioned previously in the case of 06, this may be due to gene conversion. Very few clones bearing an uneven number of cysteines have been observed previously and so it is thought that the V region must be under considerable pressure to maintain an even number of cysteine residues, enabling formation of disulphide bonds. The consequence of unpaired cysteines within the NAR V region is, as yet, unknown but may be detrimental to domain folding. If this is indeed the case then such clones will probably be eliminated from the library during early pans due to their toxicity to the expressing bacteria.
  • Clone 02 encodes 4 cysteine residues in its CDR3, giving this V region a total of 8 cysteine residues and the potential to form 4 disulphide bonds.
  • Such type I domains carrying 4, or occasionally 6 or more, cysteine residues have been previously encountered.
  • the ability to form these additional disulphide bonds, combined with the small size of the NAR V region, may provide an additional source for highly stable antibody fragments.
  • Colonies which were not sequenced, were scraped from the library plates with a sterile spreader into a final volume of 10 ml 2 ⁇ TY medium containing 100 ⁇ g/ml ampicillin and 2% glucose. Cells were combined with sterile glycerol to 20% (v/v), and following thorough mixing aliquotted as 500 ⁇ l shots and flash-frozen prior to storage at ⁇ 80° C.
  • a single aliquot of library stock was added to 200 ml of pre-warmed 2 ⁇ TY medium containing ampicillin at 100 ⁇ g/ml and 1-2% glucose (w/v) and grown at 37° C./250 rpm until log phase (OD 600 of 0.4-0.8) was reached.
  • log phase OD 600 of 0.4-0.8
  • To a 50 ml sample taken from the culture approximately 10 15 of M13K07 helper phage were added and the culture incubated at 37° C. without shaking to allow infection. Following incubation the culture was spun at 3.5K rpm/4° C.
  • the overnight culture was spun at 12K rpm/4° C. for 20 min, 80 ml of supernatant was removed and added to 20 ml of PEG/NaCl, mixed well and incubated on ice for at least 1 h.
  • the precipitated phage was pelleted at 12K rpm/4° C. and re-suspended in 2 ml PBS.
  • the phage suspension was spun at 13K rpm for 10 min to remove any remaining bacterial debris and the phage supernatant stored at 4° C.
  • the phage stock was titrated by performing serial dilutions in PBS and the addition of 900 ⁇ l of a log phase culture to 100 ⁇ l of each dilution.
  • Nunc Maxisorp Immuno test tubes (Gibco BRL, Life technologies Ltd.) were coated with either HEL or Ova in 4 ml of PBS overnight at 4° C. The tube was then washed 3 times with PBS before being blocked with 2% Marvel in PBS (MPBS) for 2 h at room temperature, following which it was washed a further 3 times with PBS. Selection was conducted by incubating the coated immunotube for 1 h at room temperature with 1 ml of phage stock in 3 ml of 2% MPBS on an over-and-under tumbler. A further hour of stationary incubation was allowed before the supernatant containing unbound phage was discarded and bound phage eluted as described below.
  • MPBS 2% Marvel in PBS
  • the immunotube was washed 20 times with PBST, excess liquid drained off and 1 ml of 100 mM triethylamine added. The tube was then rotated for a maximum of 10 min at room temperature to elute bound phage. Following incubation the phage solution was neutralized by mixing with 500 ⁇ l of 1 M Tris-HCl. In this state the phage solution was stored at 4° C. for further use (or long-term at ⁇ 20° C. if glycerol added at 15% v/v).
  • the culture was spun at 3.5 K rpm for 10 min and the resultant cell pellet re-suspended in 2 ⁇ TY containing 100 ⁇ g/ml ampicillin, 50 ⁇ g/ml kanamycin and 0.1-0.25% glucose and incubated overnight at 30° C./250 rpm.
  • the overnight culture was spun at 12K rpm for 10 min and 40 ml of supernatant added to 10 ml of PEG/NaCl, and mixed well prior to incubation on ice for at least 1 h.
  • the phage pellet was again re-suspended in 2 ml of PBS and spun for 10 min at 13K rpm to remove any remaining bacterial debris and the phage stored at 4° C. for the short term.
  • the immune library was subject to five rounds of panning against the protein antigens Hen egg white lysozyme (HEL) and Chicken ovalbumin (Ova), independently, using M13K07 helper phage and triethylamine elution.
  • HEL Hen egg white lysozyme
  • Ova Chicken ovalbumin
  • the antigen coating density was kept constant at 100 ⁇ g/ml for pans 1 and 2 .
  • Following the first round of panning approximately 10 6 phage were eluted from both the HEL and Ova coated immunotubes, increasing 10-fold following pan 2 .
  • the antigen coating density was reduced for each pan in an attempt to select higher affinity binders.
  • the number of phage eluted following HEL selection remained constant at ⁇ 10 6 for both pans that for Ova selection dropped to 10 3 in pan 3 , rising back to 10 6 following pan 4 .
  • the antigen coating concentration was further reduced and selection was accompanied by a significant drop in the number of phage eluted. Due to this reduction in the number of phage eluted polyclonal and monoclonal phage ELISAs were conducted to determine if enrichment of HEL or Ova binders was occurring ( FIG. 5 ).
  • Monoclonal phage ELISAs show an increase in the number of positive phage for both sets of selection over pans 1 to 4 . In the case of HEL selection this increase was from less than 1% to approximately 80% following pan 4 . For Ova selected clones the numbers of positives was slightly lower but regardless increased from less than 1% to approximately 66% after the fourth pan. Following pan 5 the number of HEL-positive clones remained constant at 80% but the number of Ova-positive monoclonals dropped back to the levels observed in earlier pans ( ⁇ 10%).
  • a 96-well Immulon 4 ELISA plate (Dynatech Laboratories Ltd.) was coated with 100 ⁇ l of antigen at 10 ⁇ g/ml for 1 h at 37° C. Following three washes with PBST the wells were blocked with 300 ⁇ l of 2% MPBS (PBS with 2% w/v marvel added) for a further hour at room temperature of overnight at 4° C. Wells were washed 3 times with PBST and to individual wells 10 ⁇ l of PEG precipitated phage from each pan, in 100 ⁇ l of 2% MPBS, was added and the plate incubated for 1 h at room temperature. The phage solution was discarded and the plate washed with PBST 3 times.
  • the overnight plate was spun at 2K rpm for 10 min to give a supernatant containing monoclonal phage supernatant.
  • 50 ⁇ l of this phage supernatant in 50 ⁇ l of MPBS was added per well and the plate incubated at room temperature for 1 h. Following incubation the plate was incubated with anti-M13 HRP conjugated antibody and developed as normal.
  • V region fragments were then ligated into similarly cut, alkaline phosphatase treated and cleaned pIMS100 expression vector. Following overnight incubation at 15° C. the resultant vector, harbouring the NAR V insert fused upstream of the HuCk domain and 6His tail, was transformed into electroporation-competent E. coli XL1-Blue cells. Colonies were picked, grown as overnight cultures in 5 ml TB (containing 2% glucose (v/v), 100 ⁇ g/ml ampicillin, 25 ⁇ g/ml tetracycline) and glycerol stocks and plasmid prepared.
  • Inserts were sequenced from plasmid using the M13 reverse (5′ TTCACACAGGAAACAG 3′) (SEQ ID. 67) and HuCk forward (5′ GAAGATGAAGACAGATGGTGC 3′)(SEQ ID. 68) primer. Once sequence data had been generated the clone was given a unique name to enable identification.
  • the clones 5A7 and 4F11 were chosen to represent the two different amino acid sequences found within the HEL-selected positive clones ( FIGS. 6 and 7 ).
  • the two clones are both conventional NAR type I, and so are illustrated aligned against a typical type I clone in FIG. 8 .
  • the two clones differ from one another at only two positions (43 & 44), both lying within Fr2 and carry identical CDR3 regions.
  • the clones 4H11 and 3E4 were chosen to represent the two different amino acid sequences found within the Ova-selected positive clones ( FIGS. 9 and 10 ). Again these clones were both conventional NAR type I and as such are shown aligned against a typical type I clone in FIG. 11 . These clones differ at 6 amino acids; three within Fr1 (positions 13, 14 & 30), two within Fr2 (positions 46 & 47) and one within CDR3 (position 101).
  • a single colony of transformed E. coli was used to inoculate 5 ml LB containing 1% glucose (v/v), 12.5 ⁇ g/ml tetracycline and 50 ⁇ g/ml ampicillin and grown up at 37° C. /250 rpm overnight.
  • This culture was used to seed 50 ml TB medium containing 1% glucose (v/v), 12.5 ⁇ g/ml tetracycline and 50 ⁇ g/ml ampicillin in 250 ml baffled flasks, at 1% v/v.
  • the 50 ml cultures were grown over a period of 24 hours at 25° C./250 rpm, with one change of media after approximately 10 hours growth. Growth of all the cultures was good with the overnight OD 600 being in the order of 10-20 OD units.
  • the cell pellet resulting from centrifugation was resuspended in 10% of the original culture volume of fractionation buffer (100 ml 200 mM Tris-HCl, 20% sucrose, pH 7.5, 1 ml 100 mM EDTA/L of culture).
  • the suspension was incubated on ice with gentle shaking for 15 min following which an equal volume of ice-cold sterile H 2 O was added and incubation continued for a further 15 min (method modified from French et al., Enzyme & Microbial Technology 19 pp332-338 1996).
  • the suspension was spun at 13K rpm/4° C. for 20 min, the supernatant containing the periplasmic fraction harvested and passed through a 0.22 ⁇ m filter (Sartorius Instruments Ltd.).
  • An Immulon 4 96-well flat bottomed ELISA plate was coated with a suitable concentration of the desired antigen at 100 ⁇ l per well and the plate incubated at 37° C. for 1 h.
  • the plate was washed 3 times with PBST prior to blocking with 200 ⁇ l per well of PBS containing 2% Marvel (w/v) for 1 h at 37° C.
  • Wells were washed a further three times with PBST before addition of samples.
  • a 1 in 5 dilution of crude periplasmic release solution was prepared, added to the top wells of the plate at 200 ⁇ l per well and doubling dilutions in PBS performed. Plates were then incubated at 4° C. for 1 h. Each plate was washed a further 5 times with PBST. Goat anti-HuCk peroxidase conjugate antibody was diluted 1:1000 in PBS and 100 ⁇ l added to wells containing antigen binding domains. Plates were incubated for 1 h at 4° C. and following 6 washes with PBST the ELISA was developed as described previously and the plate read at 450 nm.
  • the HEL-selected clone 5A7 shows good binding to HEL at the top dilution applied and as the sample is serially diluted binding reduces accordingly. Limited binding to the highly related protein turkey egg-white lysozyme (TEL) is observed at the highest dilution but no binding is observed to the proteins Chicken ovalbumin (Ova), Bovine serum albumin (BSA), Keyhole limpet haemocyanin (KLH) or the blocking agent Marvel. An identical pattern of protein binding is also observed for the HEL-selected clone 4F11 ( FIG. 13 ), which is not surprising considering the high degree of amino acid sequence similarity between these two clones (111/113 aa identical).
  • the OD 450 signals obtained for 3F11 are slightly higher than those for 5A7, but this may simply be due to small differences in the amount of protein present in the samples.
  • the Ova-selected clone 4H11 ( FIG. 14 ) showed no binding to any of the proteins tested, including Ova, the antigen it was selected against. To ensure that this was not simply a consequence of there being too little protein present in the assay, a binding assay was performed with undiluted periplasmic release solution. In this instance some binding to all of the proteins was observed for the wells containing the top dilutions of 4H11 protein. This binding was immediately lost once the sample was diluted and so is likely to be non-specific, no doubt resulting from very high concentrations of protein being present.
  • NAR clones capable of binding antigen in a library previously constructed from material from a naive animal and the isolation of HEL-binding, but not Ova-binding clones, from the library constructed from the HEL immunised animals illustrates the highly specific nature of the NAR response following antigen challenge. In other words, antigen specific antigen binding domains with a specific specificity are produced.
  • clones 5A7 and 4F11 were shown to be capable of binding HEL in the antigen binding ELISA it was possible to test the stability of these clones to thermal denaturation.
  • Sub-saturating dilutions of both of the clones, ascertained from the antigen binding curves, were prepared and incubated at a range of temperatures for 3 h prior to their addition to a HEL coated ELISA plate. The samples were then incubated on the ELISA plate for an hour at 4° C. and binding detected with an anti-HuCk HRP conjugated antibody. Stability of the antigen binding domains was plotted as a percentage of that obtained for a control sample which had not been heat treated ( FIG. 15 ).
  • Both clone 5A7 and clone 4F11 show considerable resistance to irreversible denaturation losing 50% functionality at approximately 85° C. and retaining approximately 30% functionality after 3 h at 95° C. This high stability is probably a consequence of the additional, non-canonical cysteine residues found within the NAR V domain. Both clones encode 6 cysteine residues and therefore are capable of forming 3 intradomain disulphide bonds, which (if formed) would contribute greatly to the high stability of these domains. The shape of the stability curves for both of the clones is almost identical and the minor difference in stability between the clones may be simply due to assay variability.
  • HEL-5A7 The ability of HEL-5A7 to inhibit the enzymatic activity of HEL was tested by mixing 12.5 ⁇ l of HEL with 12.5 ⁇ l of purified HEL-5A7 protein in a sterile 96 well tissue culture plate, to give a final HEL concentration of 10 ⁇ g/ml and HEL-5A7 concentrations of 2500 nM, 250 nM and 25 nM.
  • the control well was set up with buffer replacing HEL-5A7.
  • a sample of freeze dried Micrococcus lysodeikticus was reconstituted in 0.1 M phosphate/citrate buffer (pH 5.8) containing 0.09% NaCl, mixed thoroughly and 175 ⁇ l added to the prepared wells. The plate was read over a period of 30 min (at 1 min intervals) at 450 nm. Enzymatic activity was plotted as percentage initial absorbance against time for each sample.
  • HEL-5A7 protein reduced the rate of cell lysis in a concentration dependent manner with respect to the control ( FIG. 16 ).
  • HEL-5A7 protein With HEL-5A7 protein at a final concentration of 2500 nM the rate of cell lysis (9.3 ⁇ 10 ⁇ 3 OD units/min) is almost halved when compared to the control (17 ⁇ 10 ⁇ 3 OD units/min) indicating that the HEL-5A7 region binds within or adjacent to the lysozyme active site cavity.
  • a similarly prepared antigen specific antigen binding domain raised against an unrelated antigen showed no effect upon the rate of cell lysis when introduced to the assay at the same concentrations.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Immunology (AREA)
  • Medicinal Chemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Molecular Biology (AREA)
  • Genetics & Genomics (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • Virology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • General Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Animal Behavior & Ethology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Veterinary Medicine (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Peptides Or Proteins (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Transition And Organic Metals Composition Catalysts For Addition Polymerization (AREA)

Abstract

A process for the production of an antigen specific antigen binding domain using a transformed host containing an expressible DNA sequence encoding the antigen specific antigen binding domain, wherein the antigen specific antigen binding domain is derived from a variable region of the immunoglobulin isotype NAR found in fish.

Description

  • The present invention relates to the production of antigen specific antigen binding domains (single domain antibodies) from fish, where the term fish encompasses both cartilaginous (subclass Elasmobranchii) and bony fish(class Osteichthyes). By antigen specific antigen binding domains we mean the variable region of a Novel Antigen Receptor (NAR).
  • Antibodies, especially monoclonal antibodies, are useful in, among other things, molecular diagnostics and therapeutics because of their high affinity binding and specificity. However, although it is now relatively simple to produce monoclonal antibodies using animal models the production of human monoclonal antibodies remains difficult. As will be appreciated, when monoclonal antibodies from non-human models are introduced into humans, the body mounts an immune response because the monoclonal antibody is foreign to the human system.
  • Recently, it has been appreciated that the activity of the monoclonal antibody can be retained while reducing the rejection thereof in humans by producing single domain antibodies (sda) from the variable chain of the relevant antibody. European patent application number 89311731.7 discloses such single domain antibodies and methods for the production thereof in mice.
  • Single domain antibodies are also important as they can penetrate tissues taking with them any linked compounds. In addition, they can bind within cavities on the surface of proteins, for example within enzyme binding sites thus disrupting function.
  • Single domain antibodies produced from Camelidae have been shown to recognize protein cavities and as such have the ability to inhibit enzymes (Lauwereys et al., EMBO 17 pp3512-3520 1998).
  • Although the small size of single domain antibodies produced from Camelidae has allowed the recognition of protein cavities and inhibition of enzyme activity, the range of possible targets may still be relatively low, since many protein cavities may still be too small to be penetrable by single domain antibodies derived from Camelidae.
  • WO94/25591 and European patent application number 99200439.0 relate to the production of single domain antibodies from Camelidae heavy chain antibodies. Single domain antibodies produced from Camelidae heavy chain antibodies are more stable than mouse single domain antibodies and can be produced in larger quantities. However, as will be appreciated if, even the smaller members of the Camelidae family, for example llama, are to be kept in humane conditions they require significant areas of land to live upon.
  • An object of the present invention is to provide a process for the production of antigen specific antigen binding domains which seeks to alleviate the above problems.
  • A further object of the present invention is to provide a composition comprising antigen specific antigen binding domains for the inhibition of protein activity which seeks to alleviate the above problems.
  • According to an aspect of the present invention there is provided a process for the production of an antigen specific antigen binding domain, using a transformed host containing an expressible DNA sequence encoding the antigen specific antigen binding domain, wherein the antigen specific antigen binding domain is derived from a variable region of NAR found in fish.
  • It has been found that antigen specific antigen binding domains produced from the variable region of NAR found in fish are as stable as single domain antibodies produced from members of the Camelidae family.
  • Further many more fish can be kept per unit area than members of the Camelidae family.
  • The immunoglobulin isotype now known as NAR (Novel Antigen Receptor), was discovered in the serum of the nurse shark (Ginglymostoma cirratum) as a homodimeric heavy chain complex, naturally lacking light chains (Greenberg et al., Nature 374 pp168-173 1995). However, before the present work by the inventors identification of NAR as an antigen binding domain was not fully appreciated neither was its ability to be raised against a specific antigen.
  • Only mammals (humans, mice, rabbits, sheep, camels, llamas, etc.) and some birds (chickens) were believed to be capable of something approaching a secondary immune response such as affinity maturation, antibody class switching etc. as a response to the presence of foreign antigen. For example, teleost fish (bony), which are much more advanced evolutionary than sharks, appear to rely solely upon the production of a low affinity, non-specific IgM type response (Watts et al., Aust Vet J 79 pp570-574 2001). A defining characteristic of teleost IgM is their low affinity and ability to non-specifically bind multiple antigens. IgM neutralisation is through non-specific multiple binding, resulting mainly in agglutination, etc. Neutralisation without complement is usually associated with specific, high affinity binding and had not until this invention been seen in fish species. The antigen specific antigen binding domains of the present invention have been shown to neutralise activity of an enzyme immunogen directly without calling upon other components of the immune system.
  • The NAR variable (V) region conforms to the model of typical Ig superfamily domains with the predicted canonical, intradomain disulphide bond. However, whilst camelid VHH regions have up to 75% sequence identity with other mammalian VH regions, the identity between NAR V and conventional VH domains is as low as 25% (Roux et al., Proceedings of the National Academy of Sciences. USA 95 pp11804-11809 1998).
  • Due to this low identity and lack of NAR sequences in the Kabat database, the amino acids of NAR V regions have previously been numbered sequentially (Roux et al., Proceedings of the National Academy of Sciences. USA 95 pp11804-11809 1998). To enable easy comparison of residues in different NAR V molecules, or NAR V region sequences with those of other species during this work, a numbering system was derived for NAR V region based upon that of Kabat et al., (1991) (Sequences of Proteins of immunological Interest, 5th Edition. National Institutes of Health, Betheseda, USA). (Note: this numbering system is used in the Figures attached hereto).
  • Immediately apparent from the alignment is the deletion of a large portion of CDR2 (residues 54-65) giving the NAR V region its characteristically small size (see, for example, FIG. 2A).
  • Initial sequence analysis allowed the classification of NAR V domains into two closely related classes (type I or II), both being constructed from one V, three D and one J segment. Type I regions have non-canonical cys residues in Fr2 (C35) and Fr4 (C103), which likely form a domain-stabilising disulphide bond. In longer NAR CDR3 loops additional cysteine residue pairs have been observed and almost certainly form disulphide bridges within the CDR, as is found in some cattle VH domains with an unusually long CDR3.
  • Type II regions are very similar in overall structure to type I but instead have non-canonical cysteine residues located in Fr1 (C29) and CDR3, which are proposed to form a constraining disulphide bond like that observed in camelid VHH domains. The presence of cysteines within each NAR type is shown in schematic form in FIG. 1.
  • Recently, an additional NAR type has been identified as the predominant expressed form in nurse shark pups (Type III) but due to its germline joined state displays no junctional diversity.
  • In type I and II NAR the DNA encoding the V region is generated by the physical joining of DNA segments which are spatially separate in the genome. This joining process occurs in B-cells and helps generate the diversity of sequence seen for these NAR types. For type III these DNA segments are already physically joined in the DNA of all cells, hence the term germline joined.
  • NAR possesses the cluster type genomic organisation usually observed for Ig receptors in cartilaginous fish, but less than five NAR loci are thought to exist, with only two or three being capable of functional rearrangement and expression. The diversity observed in the primary repertoire is generated through recombination mechanisms and, although extensive (due to the presence of three D segments), is localised to CDR3. On encounter with antigen this repertoire is rapidly expanded by extensive mutation. The pattern of mutation in NAR is unlike that observed in shark IgM, which shows low levels of mutation and poor clustering to CDRs, but rather bears the hallmarks of mammalian-like somatic mutation.
  • It has recently been found that NAR V is similar to VH, VL and TCR V but distinct from all three, hence its “unique domain architecture”. The VH name has been used in the past because the constant portion of NAR is a heavy chain but the V region is actually more like VL/TCR V than VH (i.e. groups closer on a phylogenetic tree). NAR V is not like the camel VHH domains which are derived from bona fide heavy chain V regions. The antigen binding domain of the NAR is closer to a VL domain naturally lacking VH rather than the other way round.
  • The sequence alignment of NAR V and camel VHH clearly shows the huge difference in sequence. If NAR V and camel VHH have the same physical structure (which has been implied but not proven) they achieve this using completely different amino acid sequences, and one would not be able to amplify a NAR V region library using camel VHH library primers. In addition, the ways in which the NAR V and camel VHH gene repertoires are created during VDJ joining are different due to the organisation of the immunoglobulin genes (Schluter et al Immunol Today 18 pp543-549 1997).
  • Preferably the transformed host is a prokaryote or a lower eukaryote.
  • There are many established prokaryote and lower eukaryote hosts. These hosts are known to correctly express foreign proteins.
  • Conveniently the prokaryote host is Escherichia coli.
  • In preferred embodiments the expressible DNA sequence is in the form of a phagemid vector.
  • Phagemid expression has advantages over phage genome expression in that it results in greater genetic stability and the bacterial transformation efficiency is higher thus enabling the construction of potentially larger and more diverse libraries.
  • To display antibody fragments on phage the gene encoding the variable region of the antibody can be fused to that of a phage surface protein, usually gene III or VIII. Gene III fusion is favoured due to its limited copy number (3-5 copies) on the tip of each phage, minimising possible avidity effects which are undesirable when trying to isolate binders of high affinity. The antibody fragment genes can be cloned directly into the phage genome or fused to gene segments present within phagemid plasmids.
  • Preferably the fish is a member of the Elasmobranchii subclass, for example, a shark or a dogfish.
  • A greater number of smaller members of the Elasmobranchii subclass can be kept in tanks which are smaller in unit area than the grazing area required for the same number of members of the Camelidae family. As the members of the Elasmobranchii subclass are kept in tanks they can easily be caught for extraction of their blood.
  • Conveniently the shark is a nurse shark, Ginglymostoma cirratum.
  • Preferably the antigen specific antigen binding domain has a specific specificity. Accordingly, the antigen specific antigen binding domain can be targeted to a specific antigen(s).
  • Conveniently the antigen specific antigen binding domain is monoclonal. In this connection, the antigen specific antigen binding domain is raised to a single antigen.
  • In preferred embodiments the specificity of the antigen specific antigen binding domain is determined by an antigen which is introduced into the chosen fish.
  • According to a further aspect of the present invention there is provided a process for the production of an antigen specific antigen binding domain comprising the steps of:
    • a) immunising a fish with an antigen;
    • b) isolating lymphocytes from the fish;
    • c) isolating RNA for an antigen specific antigen binding domain from the lymphocytes;
    • d) amplifying DNA sequences encoding the antigen specific antigen binding domain by PCR;
    • e) cloning the amplified DNA into a display vector;
    • f) transforming a host to produce a library;
    • g) selecting the desired clones from the library;
    • h) isolating and purifying the antigen specific antigen binding domain from these clones;
    • i) cloning the DNA sequences encoding the antigen specific antigen binding domain into an expression vector;
    • j) transforming a host to allow expression of the expression vector.
  • Screening of displayed libraries for specific binding sites involves repeated cycles of selection with the desired antigen in the process of biopanning. Generally during selection, the library of phage displayed antigen binding domains is incubated with immobilised antigen, unbound phage are washed out and bound phage eluted. This selected population is expanded by bacterial infection and put through further rounds of selection. As each phage encapsulates the DNA encoding the V region it displays, there is a functional linking of genotype and phenotype, reminiscent of membrane bound immunoglobulin on the surface of B-cells. Such cyclic panning has thus proven able to enrich for clones of high affinity, much like in vivo antibody selection.
  • Preferably before step d) the cDNA of the antigen binding domains is generated.
  • Conveniently restriction enzymes are used to digest the amplified DNA sequences encoding the antigen specific antigen binding domain. The restrictions enzymes can be chosen depending upon, for example, the handle of the primers used in the above process.
  • In preferred embodiments the restriction enzymes are NcoI and NotI.
  • Conveniently the display vector is any phagemid vector, for example, pHEN2.
  • Preferably the expression vector is a soluble expression vector such as pIMS100.
  • The above vectors are merely examples of the vectors which can be used. It is common general knowledge to those skilled in the art which vectors can be used.
  • According to a further aspect of the present invention there is provided an antigen specific antigen binding domain produced by the process as defined above.
  • According to a yet further aspect of the present invention there is provided a composition for the inhibition of protein activity comprising antigen specific antigen binding domains derived from a variable region of the immunoglobulin isotype NAR found in fish.
  • Despite the fact that the NAR V region is 12 kDa which is 20% smaller than any 15 kDa single domain antibody derived from Camelidae, it was still possible to alter protein activity therewith. Size is a significant factor in the therapeutic applications of antigen specific antigen binding domains and other single domain antibodies, with therapeutic benefits of increased tissue penetration, better access to protein clefts for neutralisation via steric hindrance and reduced immunogenicity, resulting from the use of antigen specific antigen binding domains of the present invention.
  • Antigen specific antigen binding domains derived from NAR therefore have a wider target population than single domain antibodies derived from Camelidae by virtue of their smaller size. The potential for immunogenicity is also reduced since in general the smaller the size of a protein the less the immunogenicity.
  • Furthermore, although NAR sequences have, in work previous to that of the inventors, been identified at the DNA level, there has been no clue from the DNA evidence that a somatically maturable repertoire, capable of selecting high affinity, specific binders could be a characteristic of the NAR response. Hence, it is unexpected to be able to generate an NAR library of antigen binding domains derived from sharks and the selection from this of specific and functional antigen specific antigen binding domains and their corresponding receptor genes. Sequencing of these genes confirms that an atypical (for fish and organisms of this evolutionary lineage) somatically-maturable (showing mutation from the germ line repertoire) response occurs within the NAR repertoire, driven by the immunisation process. This has resulted in the selection of highly specific, high affinity antigen binding domains capable of antigen neutralisation in isolation and not the expected non-specific, low affinity IgM like response typically found in fish and sharks.
  • Further still, the inventors have been able to isolate NAR antigen specific antigen binding domains and demonstrate for the first time that the NAR V is able to fold and function in isolation from the rest of the molecule (and in a non-shark environment), that the antigen specific antigen binding domain matures from the germ line genes to become specific for antigen (only possible with a library derived from mRNA and not DNA) and that the antigen specific antigen binding domain is able to bind specifically to the immunising antigen. In summary, as described below, the inventors have been able to immunize a shark and derive from this immunization a specific, somatically matured antigen specific antigen binding domain that is of high affinity and specific for the immunogen. In addition, the antigen specific antigen binding domain is able to neutralise the activity of the immunogen directly, without calling upon other components of the immune system. According to previous understandings, this should not have been possible for a primitive species such as sharks.
  • Conveniently, a composition is provided wherein the antigen specific antigen binding domain is a product of the process as defined above.
  • Preferably inhibition of protein activity is in a concentration dependent manner.
  • Preferably, the composition further comprises a pharmaceutical carrier or diluent therefor.
  • Such pharmaceutical carriers are well known in the art.
  • According to a further aspect of the present invention, there is provided an antigen specific antigen binding domain produced from a variable region of NAR.
  • The invention will now be described, by way of illustrate on only, with reference to the following examples and the accompanying figures.
  • FIG. 1 shows the presence of cysteine amino acid residues within each NAR type, and human, cattle and camel variable regions for comparison. Canonical cysteines are shown by
    Figure US20050043519A1-20050224-P00900
    and non-canonical cysteines are shown by
    Figure US20050043519A1-20050224-P00901
    .
  • FIGS. 2A, 2B and 2C show the amino acid translations of the sequences obtained in the Examples (SEQ ID. 1 to 51). The sequences are aligned against a typical type I and type II clone sequence (top of each Figure with CDR's highlighted in bold) dashes indicate identity to the type I clone and * indicates an in-frame stop codon.
  • FIG. 3 shows NAR type I and II variable region amino acid sequence alignment (SEQ 1 and 2). Germline sequence is given for type I, whilst that given for type II is typical of those observed from somatically mutated cDNA sequences (Roux et al., Proceedings of the National Academy of Sciences. USA 95 pp11804-11809 1998). Sequence identity is indicated by a dash and the CDR's of both sequences are in bold. The numbering above the sequences was generated by comparison of conserved residues (underlined) with those of other species and is used to enable comparison of NAR V region sequences.
  • FIG. 4 shows a variability plot for the 29 immune library sequences identified in the Example (pre-selection and functional). Variability at each position was calculated according to the method of Wu & Kabat (1970) (Journal of Experimental Medicine 132 pp211-250). The canonical cysteine residues, C22 and 92, are marked by an asterisk.
  • FIGS. 5A and B show polyclonal and monoclonal phage ELISA results for selection on Hen egg white lysozyme (HEL) (FIG. 5A) and Chicken ovalbumin (Ova) (FIG. 5B). Phage numbers were normalised for each pan prior to polyclonal analysis.
  • Data presented is a mean of triplicate wells and representative of at least three assays. Monoclonal results are percentages obtained from 96 clones for each pan.
  • FIG. 6 shows the DNA (SEQ ID. 53 & 54) and encoded amino acid sequence (SEQ ID. 52) of the α-HEL 5A7 clone. CDRs are highlighted in bold.
  • FIG. 7 shows the DNA (SEQ ID. 56 & 57) and encoded amino acid sequence (SEQ ID. 55) of the α-HEL 4F11 clone. CDRs are highlighted in bold.
  • FIG. 8 shows the amino acid alignment of the two α-HEL clones, 5A7 (SEQ ID. 52) and 4F11 (SEQ ID. 55), with a typical type I clone (SEQ ID. 1). Sequences are numbered according to FIG. 3 for ease of comparison, differences between the selected clones are highlighted in underlined and CDR's are highlighted in bold, * conserved residues in all sequences, : conserved substitutions, . semi-conserved substitutions.
  • FIG. 9 shows the DNA (SEQ ID. 59 & 60) and encoded amino acid sequence (SEQ ID. 58) of the α-Ova 4H11 clone. CDRs are highlighted in bold.
  • FIG. 10 shows the DNA (SEQ ID. 62 & 63) and encoded amino acid sequence (SEQ ID. 61) of the α-Ova 3E4 clone. CDRs are highlighted in bold.
  • FIG. 11 shows amino acid alignment of the two α-Ova clones, 4H11 (SEQ ID. 58) and 3E4 (SEQ ID. 61), with a typical type I clone (SEQ ID. 1). Sequences are numbered according to FIG. 3 for ease of comparison, differences between the selected clones are underlined and the CDR's are highlighted in bold. * conserved residues in all sequences, : conserved substitutions, . semi-conserved substitutions.
  • FIG. 12 shows binding analysis of α-HEL clone 5A7. Serial dilutions of crude periplasmic release solution were applied to an ELISA plate coated with each of the test proteins at 10 μg/ml and blocked with Marvel. Data presented is a mean of triplicate wells and representative of at least three repeat assays.
  • FIG. 13 shows binding analysis of α-HEL clone 4F11. Serial dilutions of crude periplasmic release solution were applied to an ELISA plate coated with each of the test proteins at 10 μg/ml and blocked with Marvel. Data presented is a mean of triplicate wells and representative of at least three repeat assays.
  • FIG. 14 shows binding analysis of α-Ova clone 4H11. Serial dilutions of crude periplasmic release solution were applied to an ELISA plate coated with each of the test proteins at 10 μg/ml and blocked with Marvel. Data presented is a mean of triplicate wells and representative of at least three repeat assays.
  • FIG. 15 shows a comparison of the stability of the anti-HEL clones 5A7 and 4F11 to irreversible thermal denaturation. Data presented is a mean of triplicate wells and representative of at least three repeat assays.
  • FIG. 16 shows a lysozyme enzymatic inhibition assay. Purified HEL-5A7 NAR V region protein at a final concentration of 2500 nM (filled circle), 250 nM (open triangle) or 25 nM (filled square) were pre-incubated with HEL prior to the introduction of M. lysodeikticus bacterium. The control wells (open diamond) contained buffer in place of HEL-5A7 protein. The data presented is an average of 3 replicates and a typical data set from three repeat experiments.
  • EXAMPLE
  • Bacterial Strains
  • The electroporation-competent strain E. coli XL1-Blue {recA1 endA1 gyrA96 thi-1 hsdR17 supE44 relA1 lac [F′ proAB lacIq ZΔM15 Tn10 (Tetr)]} (Stratagene Ltd.) was used to prepare and pan the NAR V region phage display libraries.
  • PCR Materials
  • All custom oligonucleotides used throughout this work were ordered from Sigma-Genosys Ltd., and were desalted and/or HPLC purified. Library primer sequences were as follows (all 5′ to 3′):
    • NAR F4 For1 ATA ATC AAG CTT GCG GCC GCA TTC ACA GTC ACG ACA GTG CCA CCT C (SEQ ID. 64)
    • NAR F4 For2 ATA ATC AAG CTT GCG GCC GCA TTC ACA GTC ACG GCA GTG CCA TCT C (SEQ ID. 65)
    • NAR F1 Rev ATA ATA AGG AAT TCC ATG GCT CGA GTG GAC CAA ACA CCG (SEQ ID. 66)
  • All PCR reactions were performed on a Hybaid PCR sprint block in Hybaid 0.2 ml thin-walled omnitubes.
  • Construction of NAR V Region Libraries for Phage Display
  • RNA Preparation
  • To enable production of the immune library, three nurse sharks were immunised five times with Hen egg-white lysozyme (HEL) (over a period of approximately 8 months). Blood samples were taken from each shark following each immunisation, peripheral blood lymphocytes isolated, and total RNA prepared for each bleed. The RNA from bleeds 4 and 5 for each of the three sharks was pooled and stored at −80° C. until required for cDNA synthesis.
  • cDNA Synthesis and PCR Amplification
  • For cDNA synthesis, ready-to-go RT-PCR beads (200 μM each dNTP, 10 mM Tris-HCl buffer, 60 mM KCl, 1.5 mM MgCl2, M-MuLV reverse transcriptase, RNAguard7, RNase/DNase free BSA and 2 U Taq DNA polymerase) (APB Ltd.) were reconstituted in 45 μl of DEPC treated H2O by incubating on ice for 5 min or until the beads were completely dissolved. To each tube 2 μl of nurse shark tRNA at 2 μg/μl and 2 μl of NAR F4 For primer or F4 For2 primer at 25 pM/μl were added. Both of these primers are specific for NAR framework region 4 and have a NotI site incorporated in the handle to allow subsequent cloning into the phagemid vector. Tubes were flicked gently to mix contents and incubated on a PCR block pre-warmed to 46° C. for 30 min. Following cDNA synthesis, tubes were incubated at 95° C. for 7 min to inactivate the reverse transcriptase and denature the template.
  • To each tube 2 μl of the common primer NAR F1 Rev at 25 pM/μl, containing a NcoI site in its handle was added, tubes were pre-heated to 95° C. and 1 μl of Taq DNA polymerase at 1 U/μl added to each prior to cycling 32 times at 95° C. for 2 min, 55° C. for 1 min and 72° C. for 1 min 30 s.
  • Following PCR amplification type I and type II, products were PAGE purified on a 1.5% gel a strong band was visualised at approximately 400 bp for both primer sets indicating successful amplification of the NAR V region.
  • Cloning of NAR V Region into the Phagemid Vector pHEN2
  • PAGE-purified PCR product was digested with NcoI and NotI restriction enzymes, at the sites incorporated by the handled primers used for amplification, to allow cloning into the phagemid vector pHEN2. Restricted DNA was purified on a 1.5% agarose gel and the DNA excised and cleaned.
  • Plasmid DNA, harvested from an overnight culture of E. coli XL1-Blue and phenol:chloroform treated, was similarly cut with NcoI and NotI restriction enzymes. Double-cut vector was purified on a 0.7% agarose gel and DNA extracted. For library construction digested vector was not treated with calf alkaline phosphatase.
  • To enable quantification, 2 μl of suitably digested PCR product and pHEN2 vector were run on a 1% agarose gel against 2 μl of DNA marker VI (Boehringer Ltd.) and band intensities evaluated by eye to judge relative amounts of DNA present. Ligations were performed with equal amounts of vector and insert DNA in the presence of 2.5 μl of 10× ligase buffer and 1 μl of T4 ligase. The final volume was made up to 25 μl with H2O and incubated overnight at 15° C. For library construction 30-40 such ligations were performed.
  • Following incubation overnight, ligation products were pooled, phenol:chloroform cleaned and the resultant DNA pellet reconstituted in approximately 100 μl of 1:10 dilution of 10 mM Tris-HCl, pH 8.5. DNA was then ready for transformation into electroporation-competent cells.
  • Transformation of Electroporation-Competent Cells and Evaluation of the Resultant Library
  • Ligated DNA was aliquotted into chilled electroporation cuvettes and to each 40 μl of freshly thawed electroporation-competent XL1-Blue cells was added. Cells were electroporated and resuspended in 100 μl ice-cold 2×TY media with 1% glucose (w/v) added. Dilutions at 10−2, 10−4 and 10−6 were performed for each transformation and plated on TYE agar containing 100 μg/ml ampicillin and 1-2% glucose (w/v). The remaining bacterial suspension was plated straight onto 140 mm petri-dishes containing TYE with ampicillin and glucose (as above). All plates were grown overnight at 37° C.
  • Following incubation overnight, colonies from the dilution plates were counted to give an estimate of the final library size, approximately 5×106 members. Approximately 100 individual colonies were PCR screened using 1 μl each of the primers LMB3 (5′ CAGGAAACAGCTATGAC 3′) (SEQ ID.69) and pHEN seq (5′ CTATGCGGCCCCATTCA 3′) (SEQ ID. 70) at 25 pM/μl, 1 μl of dNTPs at 25 pM each, 2 μl of 50 mM MgCl2, 5 μl of 10×Taq polymerase buffer, 1 μl Taq polymerase (at 1 U/μl) and 39 μl Steripak H2O. PCR was undertaken as follows; 1 cycle at 95° C. for 3 minutes (to lyse bacteria) and 20 cycles of 95° C. for 1 min, 55° C. for 1 min and 72° C. for 1 min. PCR product was run on a 1.5% agarose gel containing EtBr against molecular weight marker VI (Boehringer Ltd.) to evaluate the percentage of the library carrying NAR V region insert. Using this method, 75% of the library was observed to be carrying an insert approximating that expected for the NAR V region, giving a functional library size of 3.75×106 members. Fifty clones, established in this way to be carrying correctly sized inserts, were then sequenced to evaluate library diversity.
  • The encoded amino acid translations of the sequences obtained are shown in FIGS. 2A, B and C.
  • Of the 50 clones sequenced, 6 were found to harbour one or more stop codons encoded by an in-frame TGA codon within CDR3. In the case of clones 13 and 19 the stop codon is probably a consequence of the D3 and D2 segments (respectively) being utilised in a non-preferred reading frame (Roux et al., Proceedings of the National Academy of Sciences. USA 95 pp11804-11809 1998). The reason for the stop codons in the other 5 clones is less distinct but is likely due to somatic hypermutation within this region.
  • A further 15 clones carried frameshift mutations leading to the production of non-sense or truncated proteins. For the majority of these clones the frameshift occurred within CDR3, possibly as a consequence of nucleotide addition or deletion during the recombination process. For clones 14 and 41 the frameshift mutation arose within Fr2 (position 41 according to FIG. 3) and Fr3 (position 67) respectively and are more likely due to polymerase errors during library construction (the frameshift in clone 14 occurs immediately after a long poly-A tract in the DNA sequence).
  • The sequence alignment and the variability plot of the 28 clones encoding functional inserts (FIGS. 2 and 4) show good diversity, with each clone having a unique amino acid sequence. Variability is seen to be focussed across CDR3 which, like clones from a similarly constructed naïve library, varied greatly in both sequence and length. The immune nature of the library is important as NAR V regions which bound to antigen could not be isolated from a naïve library (ie. without prior immunisation).
  • Both NAR types were represented, with approximately 80% being type I and 20% type II, however a number of clones proved difficult to assign to an NAR type. For example, clone 33 has a type II Fr1 but type I CDR3 and Fr4, whilst clones 06, 40 and 46 have a type I Fr1 and CDR3 but a type II Fr2 and Fr4. This finding suggests the possibility that gene conversion may be occurring between the NAR genes.
  • A number of other clones also show some atypical features which were not observed with the naive library pre-selection clones. Clones 24 and 36 are both assigned as type I on the basis of other sequence characteristics but do not carry the pair of cysteine residues normally observed in the type I CDR3. The clones 06, 40, 46 and 48 all encode an uneven number of cysteine residues. As mentioned previously in the case of 06, this may be due to gene conversion. Very few clones bearing an uneven number of cysteines have been observed previously and so it is thought that the V region must be under considerable pressure to maintain an even number of cysteine residues, enabling formation of disulphide bonds. The consequence of unpaired cysteines within the NAR V region is, as yet, unknown but may be detrimental to domain folding. If this is indeed the case then such clones will probably be eliminated from the library during early pans due to their toxicity to the expressing bacteria.
  • Clone 02 encodes 4 cysteine residues in its CDR3, giving this V region a total of 8 cysteine residues and the potential to form 4 disulphide bonds. Such type I domains carrying 4, or occasionally 6 or more, cysteine residues have been previously encountered. The ability to form these additional disulphide bonds, combined with the small size of the NAR V region, may provide an additional source for highly stable antibody fragments.
  • Colonies, which were not sequenced, were scraped from the library plates with a sterile spreader into a final volume of 10 ml 2×TY medium containing 100 μg/ml ampicillin and 2% glucose. Cells were combined with sterile glycerol to 20% (v/v), and following thorough mixing aliquotted as 500 μl shots and flash-frozen prior to storage at −80° C.
  • Panning of NAR V Region Library against Protein Antigens Growth of the Library
  • A single aliquot of library stock was added to 200 ml of pre-warmed 2×TY medium containing ampicillin at 100 μg/ml and 1-2% glucose (w/v) and grown at 37° C./250 rpm until log phase (OD600 of 0.4-0.8) was reached. To a 50 ml sample taken from the culture approximately 1015 of M13K07 helper phage were added and the culture incubated at 37° C. without shaking to allow infection. Following incubation the culture was spun at 3.5K rpm/4° C. for 10 min and the cell pellet re-suspended in 100 ml of 2×TY containing 100 μg/ml ampicillin, 50 μg/ml kanamycin and 0.1-0.25% glucose and incubated overnight at 30° C./250 rpm to allow library expression and rescue.
  • The overnight culture was spun at 12K rpm/4° C. for 20 min, 80 ml of supernatant was removed and added to 20 ml of PEG/NaCl, mixed well and incubated on ice for at least 1 h. The precipitated phage was pelleted at 12K rpm/4° C. and re-suspended in 2 ml PBS. The phage suspension was spun at 13K rpm for 10 min to remove any remaining bacterial debris and the phage supernatant stored at 4° C. The phage stock was titrated by performing serial dilutions in PBS and the addition of 900 μl of a log phase culture to 100 μl of each dilution. Following incubation at 37° C. for 30 min, 100 μl of each dilution was plated on TYE plates containing ampicillin at 100 μg/ml and 1% glucose and incubated overnight at 37° C. The phage titre could be estimated by counting the resulting colonies.
  • Library Selection
  • Nunc Maxisorp Immuno test tubes (Gibco BRL, Life technologies Ltd.) were coated with either HEL or Ova in 4 ml of PBS overnight at 4° C. The tube was then washed 3 times with PBS before being blocked with 2% Marvel in PBS (MPBS) for 2 h at room temperature, following which it was washed a further 3 times with PBS. Selection was conducted by incubating the coated immunotube for 1 h at room temperature with 1 ml of phage stock in 3 ml of 2% MPBS on an over-and-under tumbler. A further hour of stationary incubation was allowed before the supernatant containing unbound phage was discarded and bound phage eluted as described below.
  • Elution and Rescue of Antigen-Bound Phage
  • Triethylamine Elution
  • Binding individuals of the antigen specific antigen binding domain library, displayed on the phage strain M13K07, were eluted using the alkali triethylamine.
  • Following incubation with phage the immunotube was washed 20 times with PBST, excess liquid drained off and 1 ml of 100 mM triethylamine added. The tube was then rotated for a maximum of 10 min at room temperature to elute bound phage. Following incubation the phage solution was neutralized by mixing with 500 μl of 1 M Tris-HCl. In this state the phage solution was stored at 4° C. for further use (or long-term at −20° C. if glycerol added at 15% v/v).
  • To 750 μl of the triethylamine-eluted phage 10 ml of a log phase bacterial culture was added and the culture incubated at 37° C. without shaking for 30 min. Serial dilutions of the culture were prepared in 2×TY and plated on TYE plates containing 100 μg/ml ampicillin and 2% glucose to allow the number of rescued phage to be estimated. The remaining infected culture was spun for 10 min at 13K rpm, re-suspended in 100 μl of 2×TY and plated on a 140 mm petri-dish containing TYE as above. Plates were grown overnight at 37° C.
  • Rescue of Selected Phage
  • After overnight growth, colonies were scraped from the large petri-dishes into 2 ml of 2×TY medium with a sterile scraper and the suspension mixed thoroughly. Following inoculation of 50 ml 2×TY containing 100 μg/ml ampicillin and 1-2% glucose with 50 μl of this suspension, 1 ml of the remaining bacteria was mixed with 15% glycerol (v/v) and stored at −80° C. as a stock. The 50 ml culture was incubated at 37° C./250 rpm until the OD600 reached 0.4, whereupon 15 ml was removed, added to approximately 1010 helper phage and incubated for 30 min at 37° C. Following incubation the culture was spun at 3.5 K rpm for 10 min and the resultant cell pellet re-suspended in 2×TY containing 100 μg/ml ampicillin, 50 μg/ml kanamycin and 0.1-0.25% glucose and incubated overnight at 30° C./250 rpm.
  • The overnight culture was spun at 12K rpm for 10 min and 40 ml of supernatant added to 10 ml of PEG/NaCl, and mixed well prior to incubation on ice for at least 1 h. The phage pellet was again re-suspended in 2 ml of PBS and spun for 10 min at 13K rpm to remove any remaining bacterial debris and the phage stored at 4° C. for the short term.
  • Further rounds of selection were carried out with phage rescued from the previous round of selection, as above, on antigen coated immunotubes.
  • The immune library was subject to five rounds of panning against the protein antigens Hen egg white lysozyme (HEL) and Chicken ovalbumin (Ova), independently, using M13K07 helper phage and triethylamine elution. A summary of the panning results are given in Table 1.
  • In an attempt to minimize loss of clone diversity in early rounds of selection the antigen coating density was kept constant at 100 μg/ml for pans 1 and 2. Following the first round of panning approximately 106 phage were eluted from both the HEL and Ova coated immunotubes, increasing 10-fold following pan 2. For pans 3 and 4 the antigen coating density was reduced for each pan in an attempt to select higher affinity binders. Whilst the number of phage eluted following HEL selection remained constant at ˜106 for both pans that for Ova selection dropped to 103 in pan 3, rising back to 106 following pan 4. For pan 5 the antigen coating concentration was further reduced and selection was accompanied by a significant drop in the number of phage eluted. Due to this reduction in the number of phage eluted polyclonal and monoclonal phage ELISAs were conducted to determine if enrichment of HEL or Ova binders was occurring (FIG. 5).
  • The binding of the HEL-selected polyclonal phage showed a small increase in OD450 over pans 1 and 2, with a significant increase following pan 3. A further small increase in signal followed pan 4, but afterwards pan 5 dropped back to the level observed for earlier pans. A similar pattern was observed for the Ova-selected polyclonal phage with the highest binding being obtained for phage rescued after pan 4, however in this instance the OD450 values remain low (below 0.25) for all pans.
  • Monoclonal phage ELISAs show an increase in the number of positive phage for both sets of selection over pans 1 to 4. In the case of HEL selection this increase was from less than 1% to approximately 80% following pan 4. For Ova selected clones the numbers of positives was slightly lower but regardless increased from less than 1% to approximately 66% after the fourth pan. Following pan 5 the number of HEL-positive clones remained constant at 80% but the number of Ova-positive monoclonals dropped back to the levels observed in earlier pans (˜10%).
  • The drop in the number of clones able to bind Ova after pan 5 indicates that for this pan the protein coating concentration has been reduced such that the selection is too stringent and the majority of clones are no longer able to bind. No such drop is observed for the HEL-selected monoclonal assay, indicating that the affinity of these clones for their antigen is probably higher. This shows that the antigen specific antigen binding domains produced by the sharks are very specific as the sharks were immunised with HEL and only HEL binders could be isolated, Ova data shows no binders. For this reason a selection of clones from pans 3 and 4 were sequenced for Ova but from pans 4 and 5 for HEL.
    TABLE 1
    phage added coating density phage eluted
    Pan (φ/ml) μg/ml (φ/ml)
    Anti-Hel selection
    1 >1012 100 105
    2 >1012 100 106
    3 >1012 50 106
    4 >1012 1 106
    5 >1012 0.1 103
    Anti-Ova selection
    1 >1012 100 105
    2 >1012 100 106
    3 >1012 50 103
    4 >1012 1 105
    5 >1012 0.1 103

    Selection Analysis
    Polyclonal Phage ELISA
  • A 96-well Immulon 4 ELISA plate (Dynatech Laboratories Ltd.) was coated with 100 μl of antigen at 10 μg/ml for 1 h at 37° C. Following three washes with PBST the wells were blocked with 300 μl of 2% MPBS (PBS with 2% w/v marvel added) for a further hour at room temperature of overnight at 4° C. Wells were washed 3 times with PBST and to individual wells 10 μl of PEG precipitated phage from each pan, in 100 μl of 2% MPBS, was added and the plate incubated for 1 h at room temperature. The phage solution was discarded and the plate washed with PBST 3 times. To each well 100 μl of anti-M13 monoclonal HRP conjugate (APB Ltd.), diluted 1 in 5000 in PBS, was added and incubated at room temperature for 1 h. The plate was washed 5 times with PBST and developed with 100 μl per well of TMB substrate, the reaction stopped with 50 μl per well of 1 M H2SO4 and the plate read at 450 nm.
  • Monoclonal Phage ELISA
  • Individual colonies growing on TYE plates were picked into 100 μl 2×TY medium containing 100 μg/ml ampicillin and 1-2% glucose on a sterile 96-well ELISA plate, for each of the pans, and grown overnight at 37° C./250 rpm. Following growth, a 96-well transfer device was used to inoculate a fresh 96-well plate containing 200 μl per well of 2×TY with 100 μg/ml ampicillin and 1-2% glucose. Bacteria were grown for 2 h at 37° C./250 rpm. To the original overnight plate glycerol was added to give a final concentration of 15% and the plates stored at −80° C. as a bacterial stock.
  • After the two hour incubation 25 μl of 2×TY containing 100 μg/ml ampicillin, 1-2% glucose and 1010 helper phage were added to each well. The plate was then incubated for a further hour at 37° C./250 rpm before being spun at 2K rpm for 10 min to pellet the bacteria. Supernatant was aspirated from the plate and the resultant pellet re-suspended in 200 μl 2×TY containing 100 μg/ml ampicillin, 50 μg/ml kanamycin and glucose at 0.25% (w/v). The plate was then incubated overnight at 30° C./250 rpm.
  • The overnight plate was spun at 2K rpm for 10 min to give a supernatant containing monoclonal phage supernatant. To suitably coated and blocked plates, 50 μl of this phage supernatant in 50 μl of MPBS was added per well and the plate incubated at room temperature for 1 h. Following incubation the plate was incubated with anti-M13 HRP conjugated antibody and developed as normal.
  • Subcloning and Sequencing of Positive Monoclonal Phage Clones
  • Following determination of individual clones giving a positive signal for antigen binding, 5 ml of 2×TY containing 2% glucose and 100 μg/ml ampicillin was inoculated from the appropriate clone source. Taking into account the results of the monoclonal phage ELISAs fifteen HEL-positive clones were picked at random from pans 4 and 5, whilst those for Ova were picked from pans 3 and 4. Following overnight incubation of the cultures at 37° C./250 rpm plasmid was prepared as set out above. A 20 μl sample of plasmid was then digested with the restriction enzymes NcoI and NotI and the ˜400 bp fragment corresponding to the NAR V region fragment PAGE purified and recovered. Purified V region fragments were then ligated into similarly cut, alkaline phosphatase treated and cleaned pIMS100 expression vector. Following overnight incubation at 15° C. the resultant vector, harbouring the NAR V insert fused upstream of the HuCk domain and 6His tail, was transformed into electroporation-competent E. coli XL1-Blue cells. Colonies were picked, grown as overnight cultures in 5 ml TB (containing 2% glucose (v/v), 100 μg/ml ampicillin, 25 μg/ml tetracycline) and glycerol stocks and plasmid prepared.
  • Inserts were sequenced from plasmid using the M13 reverse (5′ TTCACACAGGAAACAG 3′) (SEQ ID. 67) and HuCk forward (5′ GAAGATGAAGACAGATGGTGC 3′)(SEQ ID. 68) primer. Once sequence data had been generated the clone was given a unique name to enable identification.
  • On translation only two different sequences were obtained from the 15 HEL-selected clones and two from the 15 Ova-selected clones.
  • The clones 5A7 and 4F11 were chosen to represent the two different amino acid sequences found within the HEL-selected positive clones (FIGS. 6 and 7). The two clones are both conventional NAR type I, and so are illustrated aligned against a typical type I clone in FIG. 8. The two clones differ from one another at only two positions (43 & 44), both lying within Fr2 and carry identical CDR3 regions.
  • The clones 4H11 and 3E4 were chosen to represent the two different amino acid sequences found within the Ova-selected positive clones (FIGS. 9 and 10). Again these clones were both conventional NAR type I and as such are shown aligned against a typical type I clone in FIG. 11. These clones differ at 6 amino acids; three within Fr1 (positions 13, 14 & 30), two within Fr2 (positions 46 & 47) and one within CDR3 (position 101).
  • Expression of Antigen Binding Domains in E. coli
  • Large Scale Expression
  • A single colony of transformed E. coli was used to inoculate 5 ml LB containing 1% glucose (v/v), 12.5 μg/ml tetracycline and 50 μg/ml ampicillin and grown up at 37° C. /250 rpm overnight. This culture was used to seed 50 ml TB medium containing 1% glucose (v/v), 12.5 μg/ml tetracycline and 50 μg/ml ampicillin in 250 ml baffled flasks, at 1% v/v. The 50 ml cultures were grown over a period of 24 hours at 25° C./250 rpm, with one change of media after approximately 10 hours growth. Growth of all the cultures was good with the overnight OD600 being in the order of 10-20 OD units.
  • Overnight cultures were pelleted at 4 K rpm/4° C. for 20 min. Pellets were resuspended in 50 ml fresh TB containing 50 μg/ml ampicillin and given 1 h at 25° C./250 rpm to recover before induction with 1.5 mM IPTG for 3.5-4 h and release of periplasmic contents.
  • Periplasmic Burst Release Method
  • The cell pellet resulting from centrifugation was resuspended in 10% of the original culture volume of fractionation buffer (100 ml 200 mM Tris-HCl, 20% sucrose, pH 7.5, 1 ml 100 mM EDTA/L of culture). The suspension was incubated on ice with gentle shaking for 15 min following which an equal volume of ice-cold sterile H2O was added and incubation continued for a further 15 min (method modified from French et al., Enzyme & Microbial Technology 19 pp332-338 1996). The suspension was spun at 13K rpm/4° C. for 20 min, the supernatant containing the periplasmic fraction harvested and passed through a 0.22 μm filter (Sartorius Instruments Ltd.).
  • None of the cultures showed any sign of bacterial lysis during the 4 h induction period and expression yields in the order of 1 mg crude NAR protein per litre of culture were obtained. In this example the protein expressed from the four selected clones was IMAC purified via the 6His tail.
  • ELISA Analysis of Antigen Binding Domains
  • Antigen Binding ELISA
  • An Immulon 4 96-well flat bottomed ELISA plate was coated with a suitable concentration of the desired antigen at 100 μl per well and the plate incubated at 37° C. for 1 h. The plate was washed 3 times with PBST prior to blocking with 200 μl per well of PBS containing 2% Marvel (w/v) for 1 h at 37° C. Wells were washed a further three times with PBST before addition of samples.
  • A 1 in 5 dilution of crude periplasmic release solution was prepared, added to the top wells of the plate at 200 μl per well and doubling dilutions in PBS performed. Plates were then incubated at 4° C. for 1 h. Each plate was washed a further 5 times with PBST. Goat anti-HuCk peroxidase conjugate antibody was diluted 1:1000 in PBS and 100 μl added to wells containing antigen binding domains. Plates were incubated for 1 h at 4° C. and following 6 washes with PBST the ELISA was developed as described previously and the plate read at 450 nm.
  • The HEL-selected clone 5A7 (FIG. 12) shows good binding to HEL at the top dilution applied and as the sample is serially diluted binding reduces accordingly. Limited binding to the highly related protein turkey egg-white lysozyme (TEL) is observed at the highest dilution but no binding is observed to the proteins Chicken ovalbumin (Ova), Bovine serum albumin (BSA), Keyhole limpet haemocyanin (KLH) or the blocking agent Marvel. An identical pattern of protein binding is also observed for the HEL-selected clone 4F11 (FIG. 13), which is not surprising considering the high degree of amino acid sequence similarity between these two clones (111/113 aa identical). The OD450 signals obtained for 3F11 are slightly higher than those for 5A7, but this may simply be due to small differences in the amount of protein present in the samples. The Ova-selected clone 4H11 (FIG. 14) showed no binding to any of the proteins tested, including Ova, the antigen it was selected against. To ensure that this was not simply a consequence of there being too little protein present in the assay, a binding assay was performed with undiluted periplasmic release solution. In this instance some binding to all of the proteins was observed for the wells containing the top dilutions of 4H11 protein. This binding was immediately lost once the sample was diluted and so is likely to be non-specific, no doubt resulting from very high concentrations of protein being present. This data supported the initial finding that the 4H11 clone does not bind significantly to Ova. The 3E4 clone, like 4H11, does not show binding to the proteins HEL, BSA, KLH, TEL or the blocking agent Marvel, however low level binding is observed for this clone to the selection antigen Ova. The pattern of binding by this clone to Ova is unusual in that binding at the highest protein concentration is low and shows no significant drop on dilution of the sample. When the protein concentration was increased by repeating the assay with undiluted periplasmic solution a similar pattern of binding was observed, thus negating the possibility that the protein concentration was initially too low. The reason for this unusual binding is as yet unknown, but may be due to 3E4 binding only with low affinity to Ova.
  • The distinct lack of NAR clones capable of binding antigen in a library previously constructed from material from a naive animal and the isolation of HEL-binding, but not Ova-binding clones, from the library constructed from the HEL immunised animals illustrates the highly specific nature of the NAR response following antigen challenge. In other words, antigen specific antigen binding domains with a specific specificity are produced.
  • Stability Analysis of Selected Clones
  • As clones 5A7 and 4F11 were shown to be capable of binding HEL in the antigen binding ELISA it was possible to test the stability of these clones to thermal denaturation. Sub-saturating dilutions of both of the clones, ascertained from the antigen binding curves, were prepared and incubated at a range of temperatures for 3 h prior to their addition to a HEL coated ELISA plate. The samples were then incubated on the ELISA plate for an hour at 4° C. and binding detected with an anti-HuCk HRP conjugated antibody. Stability of the antigen binding domains was plotted as a percentage of that obtained for a control sample which had not been heat treated (FIG. 15).
  • Both clone 5A7 and clone 4F11 show considerable resistance to irreversible denaturation losing 50% functionality at approximately 85° C. and retaining approximately 30% functionality after 3 h at 95° C. This high stability is probably a consequence of the additional, non-canonical cysteine residues found within the NAR V domain. Both clones encode 6 cysteine residues and therefore are capable of forming 3 intradomain disulphide bonds, which (if formed) would contribute greatly to the high stability of these domains. The shape of the stability curves for both of the clones is almost identical and the minor difference in stability between the clones may be simply due to assay variability.
  • Repetition of this assay utilising an anti-His HRP conjugated antibody to detect binding generated values which were not significantly different to those obtained with the anti-HuCk secondary antibody, indicating the drop in signal is caused by reduced binding of the NAR V domains, due to denaturation, and not simply reduced detection via the HuCk tag.
  • Inhibition of Protein Activity
  • The ability of HEL-5A7 to inhibit the enzymatic activity of HEL was tested by mixing 12.5 μl of HEL with 12.5 μl of purified HEL-5A7 protein in a sterile 96 well tissue culture plate, to give a final HEL concentration of 10 μg/ml and HEL-5A7 concentrations of 2500 nM, 250 nM and 25 nM. The control well was set up with buffer replacing HEL-5A7. A sample of freeze dried Micrococcus lysodeikticus was reconstituted in 0.1 M phosphate/citrate buffer (pH 5.8) containing 0.09% NaCl, mixed thoroughly and 175 μl added to the prepared wells. The plate was read over a period of 30 min (at 1 min intervals) at 450 nm. Enzymatic activity was plotted as percentage initial absorbance against time for each sample.
  • The introduction of HEL-5A7 protein to the assay reduced the rate of cell lysis in a concentration dependent manner with respect to the control (FIG. 16). With HEL-5A7 protein at a final concentration of 2500 nM the rate of cell lysis (9.3×10−3 OD units/min) is almost halved when compared to the control (17×10−3 OD units/min) indicating that the HEL-5A7 region binds within or adjacent to the lysozyme active site cavity. A similarly prepared antigen specific antigen binding domain raised against an unrelated antigen showed no effect upon the rate of cell lysis when introduced to the assay at the same concentrations.
  • It will be understood that the embodiment illustrated shows one application of the invention only for the purposes of illustration. In practice the invention may be applied to many different configurations, the detailed embodiments being straightforward for those skilled in the art to implement.

Claims (23)

1. A process for the production of an antigen specific antigen binding domain using a transformed host containing an expressible DNA sequence encoding the antigen specific antigen binding domain, wherein the antigen specific antigen binding domain is derived from a variable region of the immunoglobulin isotype NAR found in a species of Elasmobranchii subclass.
2. A process according to claim 1 wherein the transformed host is a prokaryote or a lower eukaryote.
3. A process according to claim 2 wherein the prokaryote host is Escherichia coli.
4. A process according to claim 1 wherein the expressible DNA sequence is in the form of a phagemid vector.
5. A process according to claim 1 wherein the species of Elasmobranchii subclass is a shark or a dog fish.
6. A process according to claim 5 wherein the shark is a nurse shark.
7. A process according to claim 1 wherein the antigen specific antigen binding domain has a specific specificity.
8. A process according to claim 1 wherein the antigen specific antigen binding domain is monoclonal.
9. A process according to claim 7 wherein the specificity of the antigen specific antigen binding domain is determined by an antigen which is introduced into the chosen fish.
10. A process for the production of an antigen specific antigen binding domain comprising the steps of:
a) immunising a member of the Elasmobranchii subclass with an antigen;
b) isolating lymphocytes from the member;
c) isolating RNA from the lymphocytes;
d) amplifying DNA sequences encoding the antigen specific antigen binding domain by PCR;
e) cloning the amplified DNA into a display vector;
f) transforming a host to produce a library;
g) selecting the desired clones from the library;
h) isolating and purifying the antigen specific antigen binding domain from these clones;
i) cloning the DNA sequences encoding the antigen specific antigen binding domain into an expression vector;
j) transforming a host to allow expression of the expression vector.
11. A process according to claim 10 wherein before step d) the cDNA of the antigen specific antigen binding domain is generated.
12. A process according to claim 10 wherein restriction enzymes are used to digest the amplified DNA sequences encoding the antigen specific antigen binding domain.
13. A process according to claim 12 wherein the restriction enzymes are NcoI and NotI.
14. A process according to claim 10 wherein the display vector is any phagemid vector.
15. A process according to claim 14 wherein the display vector is pHEN2.
16. A process according to claim 10 wherein the expression vector is a soluble expression vector.
17. A process according to claim 16 wherein the soluble expression vector is pIMS100.
18. An antigen specific antigen binding domain produced by the process in claim 1.
19. A composition for the inhibition of protein activity comprising antigen specific antigen binding domains derived from a variable region of the immunoglobulin isotype NAR found in a species of Elasmobranchii subclass.
20. A composition according to claim 19, wherein the antigen specific antigen binding domain is produced by the process in claim 1.
21. A composition according to claim 19 whereby inhibition of protein activity is in a concentration dependent manner.
22. A composition according to claim 19, contained in a pharmaceutical carrier or diluent therefor.
23. An antigen specific antigen binding domain produced from a variable region of NAR.
US10/486,608 2001-08-10 2002-08-12 Antigen binding domains Abandoned US20050043519A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US13/280,837 US8865431B2 (en) 2001-08-10 2011-10-25 Antigen binding domains

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
GB0119553A GB0119553D0 (en) 2001-08-10 2001-08-10 Antibodies
GB0119553.6 2001-08-10
GB0210508.8 2002-05-08
GB0210508A GB0210508D0 (en) 2002-05-08 2002-05-08 Antibodies
PCT/GB2002/003714 WO2003014161A2 (en) 2001-08-10 2002-08-12 Antigen binding domains from fish

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB2002/003714 A-371-Of-International WO2003014161A2 (en) 2001-08-10 2002-08-12 Antigen binding domains from fish

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US13/280,837 Continuation US8865431B2 (en) 2001-08-10 2011-10-25 Antigen binding domains

Publications (1)

Publication Number Publication Date
US20050043519A1 true US20050043519A1 (en) 2005-02-24

Family

ID=26246430

Family Applications (2)

Application Number Title Priority Date Filing Date
US10/486,608 Abandoned US20050043519A1 (en) 2001-08-10 2002-08-12 Antigen binding domains
US13/280,837 Expired - Lifetime US8865431B2 (en) 2001-08-10 2011-10-25 Antigen binding domains

Family Applications After (1)

Application Number Title Priority Date Filing Date
US13/280,837 Expired - Lifetime US8865431B2 (en) 2001-08-10 2011-10-25 Antigen binding domains

Country Status (11)

Country Link
US (2) US20050043519A1 (en)
EP (5) EP1419179B1 (en)
JP (1) JP5133494B2 (en)
AT (1) ATE459651T1 (en)
AU (2) AU2002319544B2 (en)
CA (2) CA2457636C (en)
DE (1) DE60235553D1 (en)
DK (1) DK1419179T3 (en)
ES (1) ES2337986T3 (en)
PT (1) PT1419179E (en)
WO (1) WO2003014161A2 (en)

Cited By (43)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060062784A1 (en) * 2004-09-17 2006-03-23 Domantis Limited Compositions monovalent for CD40L binding and methods of use
US20060063921A1 (en) * 2002-06-28 2006-03-23 Domantis Limited Ligand
WO2010097386A1 (en) 2009-02-24 2010-09-02 Glaxo Group Limited Antigen-binding constructs
WO2010097394A1 (en) 2009-02-24 2010-09-02 Glaxo Group Limited Multivalent and/or multispecific rankl-binding constructs
WO2010097385A1 (en) 2009-02-24 2010-09-02 Glaxo Group Limited Antigen-binding constructs
DE112008003232T5 (en) 2007-11-30 2011-02-24 Glaxo Group Limited, Greenford Antigen-binding constructs
WO2012069433A2 (en) 2010-11-23 2012-05-31 Glaxo Group Limited Antigen binding proteins
WO2012069557A1 (en) 2010-11-24 2012-05-31 Glaxo Group Limited Multispecific antigen binding proteins targeting hgf
WO2012135415A1 (en) 2011-03-29 2012-10-04 Glaxosmithkline Llc Buffer system for protein purification
WO2012140627A1 (en) 2011-04-15 2012-10-18 Compugen Ltd. Polypeptides and polynucleotides, and uses thereof for treatment of immune related disorders and cancer
EP2700651A1 (en) 2008-07-18 2014-02-26 Bristol-Myers Squibb Company Compositions monovalent for CD28 binding and methods of use
WO2014120916A1 (en) 2013-02-01 2014-08-07 Bristol-Myers Squibb Company Pegylated domain antibodies monovalent for cd28 binding and methods of use
WO2014141152A2 (en) 2013-03-15 2014-09-18 Glaxosmithkline Intellectual Property (No.2) Limited Low concentration antibody formulations
WO2014141151A1 (en) 2013-03-15 2014-09-18 Glaxosmithkline Intellectual Property (No.2) Limited Use of tricarboxylic acid (tca) intermediates to control ammonia generation in cell culture
WO2014141149A1 (en) 2013-03-15 2014-09-18 Glaxosmithkline Intellectual Property (No.2) Limited Formulations with reduced viscosity
WO2014141150A1 (en) 2013-03-15 2014-09-18 Glaxosmithkline Intellectual Property (No.2) Limited Methods for purifying antibodies
WO2015173782A1 (en) 2014-05-16 2015-11-19 Glaxosmithkline Intellectual Property Management Limited Antibody formulation
WO2016059602A2 (en) 2014-10-16 2016-04-21 Glaxo Group Limited Methods of treating cancer and related compositions
CN105531370A (en) * 2013-04-23 2016-04-27 阿伯丁大学理事会 Isolation of therapeutic target specific VNAR domains to ICOSL
EP3058952A1 (en) 2011-04-07 2016-08-24 Glaxosmithkline LLC Formulations with reduced viscosity
WO2017072716A1 (en) 2015-10-30 2017-05-04 Glaxosmithkline Intellectual Property Development Limited Prognostic method
WO2018047080A1 (en) 2016-09-07 2018-03-15 Glaxosmithkline Intellectual Property Development Limited Methods for purifying antibodies
WO2018050902A2 (en) 2016-09-15 2018-03-22 Quadrucept Bio Limited Multimers, tetramers & octamers
US10370455B2 (en) 2014-12-05 2019-08-06 Immunext, Inc. Identification of VSIG8 as the putative VISTA receptor (V-R) and use thereof to produce VISTA/VSIG8 agonists and antagonists
WO2019171294A1 (en) 2018-03-07 2019-09-12 Glaxosmithkline Intellectual Property Development Limited Methods for purifying antibodies
WO2019171286A1 (en) 2018-03-07 2019-09-12 Glaxosmithkline Intellectual Property Development Limited Methods for purifying recombinant polypeptides
EP3693394A1 (en) 2011-05-27 2020-08-12 Glaxo Group Limited Antigen binding proteins
US10745467B2 (en) 2010-03-26 2020-08-18 The Trustees Of Dartmouth College VISTA-Ig for treatment of autoimmune, allergic and inflammatory disorders
US10781254B2 (en) 2010-03-26 2020-09-22 The Trustees Of Dartmouth College VISTA regulatory T cell mediator protein, VISTA binding agents and use thereof
WO2020187711A1 (en) 2019-03-19 2020-09-24 Quadrucept Bio Limited Multimers
US10899836B2 (en) 2016-02-12 2021-01-26 Janssen Pharmaceutica Nv Method of identifying anti-VISTA antibodies
US10933115B2 (en) 2012-06-22 2021-03-02 The Trustees Of Dartmouth College VISTA antagonist and methods of use
US11009509B2 (en) 2015-06-24 2021-05-18 Janssen Pharmaceutica Nv Anti-VISTA antibodies and fragments
US11014987B2 (en) 2013-12-24 2021-05-25 Janssen Pharmaceutics Nv Anti-vista antibodies and fragments, uses thereof, and methods of identifying same
US11123426B2 (en) 2014-06-11 2021-09-21 The Trustees Of Dartmouth College Use of vista agonists and antagonists to suppress or enhance humoral immunity
WO2021190980A1 (en) 2020-03-22 2021-09-30 Quadrucept Bio Limited Multimers for viral strain evolution
US11180557B2 (en) 2012-06-22 2021-11-23 King's College London Vista modulators for diagnosis and treatment of cancer
US11242392B2 (en) 2013-12-24 2022-02-08 Janssen Pharmaceutica Nv Anti-vista antibodies and fragments
WO2022184659A1 (en) 2021-03-01 2022-09-09 Quadrucept Bio Limited Antibody domains & multimers
US11525000B2 (en) 2016-04-15 2022-12-13 Immunext, Inc. Anti-human VISTA antibodies and use thereof
US11529416B2 (en) 2012-09-07 2022-12-20 Kings College London Vista modulators for diagnosis and treatment of cancer
EP4116427A1 (en) 2012-05-17 2023-01-11 Kymab Limited In vivo guided selection & antibodies
WO2024094527A1 (en) 2022-11-05 2024-05-10 Quadrucept Bio Limited Non-human vertebrates & cells

Families Citing this family (96)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DK2330121T3 (en) 2004-06-02 2014-12-15 Adalta Pty Ltd Binding moieties based on shark IgNar domains
US8329421B2 (en) 2007-07-13 2012-12-11 Ventana Medical Systems, Inc. Methods of predicting response of a neoplasm to an EGFR inhibitor and detecting interactions between EGFR and an EGFR regulatory protein
PE20091163A1 (en) 2007-11-01 2009-08-09 Wyeth Corp ANTIBODIES FOR GDF8
CA2960659C (en) 2007-11-09 2021-07-13 The Salk Institute For Biological Studies Use of tam receptor inhibitors as immunoenhancers and tam activators as immunosuppressors
DK2982695T3 (en) 2008-07-09 2019-05-13 Biogen Ma Inc COMPOSITIONS CONCERNING ANTIBODIES AGAINST LINGO OR FRAGMENTS THEREOF
EP2344180A2 (en) 2008-09-23 2011-07-20 Wyeth LLC Methods for predicting production of activating signals by cross-linked binding proteins
EP4104821A1 (en) 2008-10-29 2022-12-21 Ablynx N.V. Formulations of single domain antigen binding molecules
US10118962B2 (en) 2008-10-29 2018-11-06 Ablynx N.V. Methods for purification of single domain antigen binding molecules
WO2010091182A2 (en) 2009-02-04 2010-08-12 Molecular Innovations Methods for screening candidate agents for modulating prorenin and renin, assays for detecting prorenin, and antibodies used therein
EA201792376A3 (en) 2009-10-23 2018-08-31 Милленниум Фармасьютикалз, Инк. ANTI-GCC MOLECULES ANTIBODIES AND RELATED COMPOSITIONS AND METHODS
WO2011056056A2 (en) 2009-11-04 2011-05-12 Laboratorios Silanes, S.A. De C.V. Anti-cytokine vhnar domains
CN108314733A (en) 2010-07-16 2018-07-24 埃博灵克斯股份有限公司 The single domain antigen binding molecules of modification and its application
WO2012103240A2 (en) 2011-01-25 2012-08-02 Eleven Biotherapeutics, Inc. Receptor binding agents
WO2013033073A1 (en) 2011-08-31 2013-03-07 Ventana Medical Systems, Inc. Expression of ets related gene (erg) and phosphatase and tensin homolog (pten) correlates with prostate cancer capsular penetration
US9156915B2 (en) 2012-04-26 2015-10-13 Thomas Jefferson University Anti-GCC antibody molecules
AU2013257848A1 (en) 2012-05-07 2014-11-27 The University Court Of The University Of Aberdeen Single domain binding molecule
NZ702178A (en) 2012-05-14 2017-01-27 Biogen Ma Inc Lingo-2 antagonists for treatment of conditions involving motor neurons
EP3184121A3 (en) 2012-07-25 2017-09-27 Salk Institute For Biological Studies Lipid membranes with exposed phosphatidylserine as tam ligands, use for treating autoimmune diseases
EP3750560A3 (en) 2012-10-09 2021-03-24 Biogen MA Inc. Combination therapies and uses for treatment of demyelinating disorders
EP2762496A1 (en) 2013-02-05 2014-08-06 EngMab AG Method for the selection of antibodies against BCMA
EP2953972B1 (en) 2013-02-05 2020-07-08 EngMab Sàrl Method for the selection of antibodies against bcma
EP3444278A1 (en) 2013-02-26 2019-02-20 Roche Glycart AG Bispecific t cell activating antigen binding molecules
JP6548630B2 (en) 2013-03-12 2019-07-24 モレキュラー テンプレーツ, インク.Molecular Templates, Inc. Cytotoxic protein comprising a cell targeting binding domain for selective killing of specific cell types and a Shiga toxin A subunit domain
EP3623380A1 (en) 2013-03-15 2020-03-18 Michael C. Milone Targeting cytotoxic cells with chimeric receptors for adoptive immunotherapy
EP2789630A1 (en) 2013-04-09 2014-10-15 EngMab AG Bispecific antibodies against CD3e and ROR1
NL1040254C2 (en) 2013-05-17 2014-11-24 Ablynx Nv Stable formulations of immunoglobulin single variable domains and uses thereof.
RU2714902C2 (en) 2013-12-19 2020-02-20 Новартис Аг Chimeric human mesotheliogen antigen receptors and use thereof
JP6793902B2 (en) 2013-12-20 2020-12-02 ノバルティス アーゲー Adjustable chimeric antigen receptor
WO2015100246A1 (en) 2013-12-24 2015-07-02 Ossianix, Inc. Baff selective binding compounds and related methods
WO2015142661A1 (en) 2014-03-15 2015-09-24 Novartis Ag Regulatable chimeric antigen receptor
EP3593812A3 (en) 2014-03-15 2020-05-27 Novartis AG Treatment of cancer using chimeric antigen receptor
EP3160995A2 (en) 2014-06-26 2017-05-03 Ossianix Inc. Semi-synthetic nurse shark vnar libraries for making and using selective binding compounds
MX2017001011A (en) 2014-07-21 2018-05-28 Novartis Ag Treatment of cancer using humanized anti-bcma chimeric antigen receptor.
US11542488B2 (en) 2014-07-21 2023-01-03 Novartis Ag Sortase synthesized chimeric antigen receptors
JP2017528433A (en) 2014-07-21 2017-09-28 ノバルティス アーゲー Low immunoenhancing dose of mTOR inhibitor and CAR combination
CN107109419B (en) 2014-07-21 2020-12-22 诺华股份有限公司 Treatment of cancer using CD33 chimeric antigen receptor
US20170209492A1 (en) 2014-07-31 2017-07-27 Novartis Ag Subset-optimized chimeric antigen receptor-containing t-cells
SG11201700770PA (en) 2014-08-19 2017-03-30 Novartis Ag Anti-cd123 chimeric antigen receptor (car) for use in cancer treatment
US10577417B2 (en) 2014-09-17 2020-03-03 Novartis Ag Targeting cytotoxic cells with chimeric receptors for adoptive immunotherapy
AU2015329965A1 (en) 2014-10-09 2017-04-27 Engmab Sàrl Bispecific antibodies against CD3epsilon and ROR1
EP3218411B1 (en) 2014-11-14 2022-01-12 Ossianix, Inc. Variable new antigen receptors (vnars) directed against transferrin receptor (tfr) and their use
US20180334490A1 (en) 2014-12-03 2018-11-22 Qilong H. Wu Methods for b cell preconditioning in car therapy
IL303247A (en) 2014-12-29 2023-07-01 Novartis Ag Methods of making chimeric antigen receptor-expressing cells
AU2016205197B2 (en) 2015-01-08 2021-10-21 Biogen Ma Inc. LINGO-1 antagonists and uses for treatment of demyelinating disorders
WO2016126608A1 (en) 2015-02-02 2016-08-11 Novartis Ag Car-expressing cells against multiple tumor antigens and uses thereof
EP3280795B1 (en) 2015-04-07 2021-03-24 Novartis AG Combination of chimeric antigen receptor therapy and amino pyrimidine derivatives
US11896614B2 (en) 2015-04-17 2024-02-13 Novartis Ag Methods for improving the efficacy and expansion of chimeric antigen receptor-expressing cells
EP3286211A1 (en) 2015-04-23 2018-02-28 Novartis AG Treatment of cancer using chimeric antigen receptor and protein kinase a blocker
WO2017015427A1 (en) 2015-07-21 2017-01-26 Novartis Ag Methods for improving the efficacy and expansion of immune cells
PT3331910T (en) 2015-08-03 2020-03-24 Engmab Sarl Monoclonal antibodies against bcma
CN105384825B (en) 2015-08-11 2018-06-01 南京传奇生物科技有限公司 A kind of bispecific chimeric antigen receptor and its application based on single domain antibody
JP6905163B2 (en) 2015-09-03 2021-07-21 ザ トラスティーズ オブ ザ ユニバーシティ オブ ペンシルバニア Biomarkers that predict cytokine release syndrome
CN109069597A (en) 2015-12-22 2018-12-21 诺华股份有限公司 Mesothelin Chimeric antigen receptor (CAR) and the combination of anti-PD-L1 antibody inhibition are in anticancer therapy
US20190269727A1 (en) 2015-12-28 2019-09-05 Novartis Ag Methods of making chimeric antigen receptor-expressing cells
WO2017149515A1 (en) 2016-03-04 2017-09-08 Novartis Ag Cells expressing multiple chimeric antigen receptor (car) molecules and uses therefore
SG11201809041TA (en) 2016-04-15 2018-11-29 Novartis Ag Compositions and methods for selective protein expression
WO2017223419A1 (en) 2016-06-24 2017-12-28 Ig Biosciences Corporation Prebiotic neutraceutical compositions and methods of treatment using the same
AU2017297404A1 (en) 2016-07-13 2019-01-24 Biogen Ma Inc. Dosage regimens of LINGO-1 antagonists and uses for treatment of demyelinating disorders
EP3484455A2 (en) 2016-07-15 2019-05-22 Novartis AG Treatment and prevention of cytokine release syndrome using a chimeric antigen receptor in combination with a kinase inhibitor
WO2018014260A1 (en) 2016-07-20 2018-01-25 Nanjing Legend Biotech Co., Ltd. Multispecific antigen binding proteins and methods of use thereof
CN118021943A (en) 2016-07-28 2024-05-14 诺华股份有限公司 Combination therapy of chimeric antigen receptor and PD-1 inhibitor
EP3490590A2 (en) 2016-08-01 2019-06-05 Novartis AG Treatment of cancer using a chimeric antigen receptor in combination with an inhibitor of a pro-m2 macrophage molecule
CN117866991A (en) 2016-10-07 2024-04-12 诺华股份有限公司 Chimeric antigen receptor for the treatment of cancer
WO2018068201A1 (en) 2016-10-11 2018-04-19 Nanjing Legend Biotech Co., Ltd. Single-domain antibodies and variants thereof against ctla-4
CN110167964B (en) 2016-11-02 2023-12-01 百时美施贵宝公司 Combination of bispecific antibodies and immunopharmaceuticals against BCMA and CD3 for the treatment of multiple myeloma
CN110325547B (en) 2016-11-07 2023-12-26 德格克斯有限公司 Novel amyloid beta oligomer specific binding molecules
US11535662B2 (en) 2017-01-26 2022-12-27 Novartis Ag CD28 compositions and methods for chimeric antigen receptor therapy
EP3589647A1 (en) 2017-02-28 2020-01-08 Novartis AG Shp inhibitor compositions and uses for chimeric antigen receptor therapy
WO2018194441A1 (en) 2017-04-21 2018-10-25 Centro De Investigación Científica Y De Educación Superior De Ensenada, Baja California Vnar having anti-angiogenic activity in solid tumours of pet animals
EP3615055A1 (en) 2017-04-28 2020-03-04 Novartis AG Cells expressing a bcma-targeting chimeric antigen receptor, and combination therapy with a gamma secretase inhibitor
WO2019000223A1 (en) 2017-06-27 2019-01-03 Nanjing Legend Biotech Co., Ltd. Chimeric antibody immune effctor cell engagers and methods of use thereof
CA3075367A1 (en) 2017-09-27 2019-04-04 Elasmogen Ltd Specific binding molecules
KR20200069358A (en) 2017-10-25 2020-06-16 노파르티스 아게 Method for producing chimeric antigen receptor expressing cells
US20210179709A1 (en) 2017-10-31 2021-06-17 Novartis Ag Anti-car compositions and methods
GB201721802D0 (en) 2017-12-22 2018-02-07 Almac Discovery Ltd Ror1-specific antigen binding molecules
AU2018396970A1 (en) 2017-12-28 2020-08-13 Nanjing Legend Biotech Co., Ltd. Single-domain antibodies and variants thereof against TIGIT
EP3740507A4 (en) 2018-01-15 2022-08-24 Nanjing Legend Biotech Co., Ltd. Single-domain antibodies and variants thereof against pd-1
AU2019215031A1 (en) 2018-01-31 2020-08-20 Novartis Ag Combination therapy using a chimeric antigen receptor
EP3774917A4 (en) 2018-03-30 2022-01-19 Nanjing Legend Biotech Co., Ltd. Single-domain antibodies against lag-3 and uses thereof
US20210047405A1 (en) 2018-04-27 2021-02-18 Novartis Ag Car t cell therapies with enhanced efficacy
CN112203725A (en) 2018-06-13 2021-01-08 诺华股份有限公司 BCMA chimeric antigen receptors and uses thereof
WO2019246288A1 (en) * 2018-06-22 2019-12-26 Ossianix, Inc. Anti-cd98hc vnars for crossing the blood brain barrier and type iv vnar libraries
WO2020047452A2 (en) 2018-08-31 2020-03-05 Novartis Ag Methods of making chimeric antigen receptor-expressing cells
EP3844265A2 (en) 2018-08-31 2021-07-07 Novartis AG Methods of making chimeric antigen receptor-expressing cells
WO2020172553A1 (en) 2019-02-22 2020-08-27 Novartis Ag Combination therapies of egfrviii chimeric antigen receptors and pd-1 inhibitors
WO2020210678A1 (en) 2019-04-12 2020-10-15 Novartis Ag Methods of making chimeric antigen receptor-expressing cells
CN114761037A (en) 2019-11-26 2022-07-15 诺华股份有限公司 Chimeric antigen receptor binding to BCMA and CD19 and uses thereof
WO2021163618A1 (en) 2020-02-14 2021-08-19 Novartis Ag Method of predicting response to chimeric antigen receptor therapy
US20230174933A1 (en) 2020-02-27 2023-06-08 Novartis Ag Methods of making chimeric antigen receptor-expressing cells
KR20220146530A (en) 2020-02-27 2022-11-01 노파르티스 아게 Methods of Making Chimeric Antigen Receptor-Expressing Cells
JP2023530919A (en) 2020-06-17 2023-07-20 ヤンセン バイオテツク,インコーポレーテツド Materials and methods for production of pluripotent stem cells
CN112301431B (en) * 2020-08-12 2023-01-31 华东理工大学 Phage library based on shark antibody variable region V-NAR and construction method thereof
GB202020152D0 (en) 2020-12-18 2021-02-03 Elasmogen Ltd Fast-track humanisation of specific binding molecules
US20230174651A1 (en) 2021-06-23 2023-06-08 Janssen Biotech, Inc. Materials and methods for hinge regions in functional exogenous receptors
WO2024018426A1 (en) 2022-07-22 2024-01-25 Janssen Biotech, Inc. Enhanced transfer of genetic instructions to effector immune cells
WO2024056809A1 (en) 2022-09-15 2024-03-21 Novartis Ag Treatment of autoimmune disorders using chimeric antigen receptor therapy

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU634186B2 (en) 1988-11-11 1993-02-18 Medical Research Council Single domain ligands, receptors comprising said ligands, methods for their production, and use of said ligands and receptors
DK0698097T3 (en) 1993-04-29 2001-10-08 Unilever Nv Production of antibodies or (functionalized) fragments thereof derived from Camelidae heavy chain immunoglobulins
EP0954978B1 (en) 1998-03-12 2011-11-30 VHsquared Limited New products comprising inactivated yeasts or moulds provided with active antibodies

Cited By (68)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060063921A1 (en) * 2002-06-28 2006-03-23 Domantis Limited Ligand
US20060062784A1 (en) * 2004-09-17 2006-03-23 Domantis Limited Compositions monovalent for CD40L binding and methods of use
US7927596B2 (en) 2004-09-17 2011-04-19 Domantis Limited Methods of antagonizing binding of CD40 to CD40L with CD40L-specific monovalent polypeptides
US20090297524A1 (en) * 2004-09-17 2009-12-03 Steven Grant Compositions monovalent for CD40L binding and methods of use
US20100092483A1 (en) * 2004-09-17 2010-04-15 Domantis Limited Compositions monovalent for CD40L binding and methods of use
US20100092482A1 (en) * 2004-09-17 2010-04-15 Domantis Limited Compositions monovalent for CD40L binding and methods of use
US8524236B2 (en) 2004-09-17 2013-09-03 Domantis Limited Methods of antagonizing the binding of CD40 to CD40L with CD40L-specific monovalent polypeptides in autoimmune individuals
EP2371862A2 (en) 2004-09-17 2011-10-05 Domantis Limited Compositions monovalent for CD40L binding and methods of use
US20110172400A1 (en) * 2004-09-17 2011-07-14 Domantis Limited Compositions monovalent for cd40l binding and methods of use
US7829096B2 (en) 2004-09-17 2010-11-09 Domantis Ltd. CD40L-specific monovalent polypeptides
US7563443B2 (en) 2004-09-17 2009-07-21 Domantis Limited Monovalent anti-CD40L antibody polypeptides and compositions thereof
DE112008003232T5 (en) 2007-11-30 2011-02-24 Glaxo Group Limited, Greenford Antigen-binding constructs
EP2641919A2 (en) 2007-11-30 2013-09-25 Glaxo Group Limited Antigen-binding constructs
EP2615115A2 (en) 2007-11-30 2013-07-17 Glaxo Group Limited Antigen-binding constructs
EP2700651A1 (en) 2008-07-18 2014-02-26 Bristol-Myers Squibb Company Compositions monovalent for CD28 binding and methods of use
WO2010097385A1 (en) 2009-02-24 2010-09-02 Glaxo Group Limited Antigen-binding constructs
WO2010097394A1 (en) 2009-02-24 2010-09-02 Glaxo Group Limited Multivalent and/or multispecific rankl-binding constructs
WO2010097386A1 (en) 2009-02-24 2010-09-02 Glaxo Group Limited Antigen-binding constructs
US10745467B2 (en) 2010-03-26 2020-08-18 The Trustees Of Dartmouth College VISTA-Ig for treatment of autoimmune, allergic and inflammatory disorders
US10781254B2 (en) 2010-03-26 2020-09-22 The Trustees Of Dartmouth College VISTA regulatory T cell mediator protein, VISTA binding agents and use thereof
WO2012069433A2 (en) 2010-11-23 2012-05-31 Glaxo Group Limited Antigen binding proteins
WO2012069557A1 (en) 2010-11-24 2012-05-31 Glaxo Group Limited Multispecific antigen binding proteins targeting hgf
EP2853542A1 (en) 2010-11-24 2015-04-01 Glaxo Group Limited Multispecific antigen binding proteins targeting HGF
WO2012135415A1 (en) 2011-03-29 2012-10-04 Glaxosmithkline Llc Buffer system for protein purification
EP3058952A1 (en) 2011-04-07 2016-08-24 Glaxosmithkline LLC Formulations with reduced viscosity
WO2012140627A1 (en) 2011-04-15 2012-10-18 Compugen Ltd. Polypeptides and polynucleotides, and uses thereof for treatment of immune related disorders and cancer
EP3693394A1 (en) 2011-05-27 2020-08-12 Glaxo Group Limited Antigen binding proteins
EP4338754A2 (en) 2011-05-27 2024-03-20 Glaxo Group Limited Antigen binding proteins
EP4116427A1 (en) 2012-05-17 2023-01-11 Kymab Limited In vivo guided selection & antibodies
US11180557B2 (en) 2012-06-22 2021-11-23 King's College London Vista modulators for diagnosis and treatment of cancer
US10933115B2 (en) 2012-06-22 2021-03-02 The Trustees Of Dartmouth College VISTA antagonist and methods of use
US11752189B2 (en) 2012-06-22 2023-09-12 The Trustees Of Dartmouth College Vista antagonist and methods of use
US11529416B2 (en) 2012-09-07 2022-12-20 Kings College London Vista modulators for diagnosis and treatment of cancer
WO2014120916A1 (en) 2013-02-01 2014-08-07 Bristol-Myers Squibb Company Pegylated domain antibodies monovalent for cd28 binding and methods of use
WO2014141151A1 (en) 2013-03-15 2014-09-18 Glaxosmithkline Intellectual Property (No.2) Limited Use of tricarboxylic acid (tca) intermediates to control ammonia generation in cell culture
EP3680000A1 (en) 2013-03-15 2020-07-15 GlaxoSmithKline Intellectual Property (No.2) Limited Methods for purifying antibodies
EP3686217A1 (en) 2013-03-15 2020-07-29 GlaxoSmithKline Intellectual Property (No.2) Limited Low concentration antibody formulations
EP4371996A2 (en) 2013-03-15 2024-05-22 GlaxoSmithKline Intellectual Property Development Limited Methods for purifying antibodies
WO2014141150A1 (en) 2013-03-15 2014-09-18 Glaxosmithkline Intellectual Property (No.2) Limited Methods for purifying antibodies
WO2014141149A1 (en) 2013-03-15 2014-09-18 Glaxosmithkline Intellectual Property (No.2) Limited Formulations with reduced viscosity
WO2014141152A2 (en) 2013-03-15 2014-09-18 Glaxosmithkline Intellectual Property (No.2) Limited Low concentration antibody formulations
US11459377B2 (en) 2013-04-23 2022-10-04 Elasmogen Limited Synthetic library of specific binding molecules
CN105531370A (en) * 2013-04-23 2016-04-27 阿伯丁大学理事会 Isolation of therapeutic target specific VNAR domains to ICOSL
US11014987B2 (en) 2013-12-24 2021-05-25 Janssen Pharmaceutics Nv Anti-vista antibodies and fragments, uses thereof, and methods of identifying same
US11242392B2 (en) 2013-12-24 2022-02-08 Janssen Pharmaceutica Nv Anti-vista antibodies and fragments
WO2015173782A1 (en) 2014-05-16 2015-11-19 Glaxosmithkline Intellectual Property Management Limited Antibody formulation
EP3719038A1 (en) 2014-05-16 2020-10-07 GlaxoSmithKline Intellectual Property Management Limited Belimumab formulation
EP3715371A1 (en) 2014-05-16 2020-09-30 GlaxoSmithKline Intellectual Property Management Limited Belimumab formulation
US11123426B2 (en) 2014-06-11 2021-09-21 The Trustees Of Dartmouth College Use of vista agonists and antagonists to suppress or enhance humoral immunity
WO2016059602A2 (en) 2014-10-16 2016-04-21 Glaxo Group Limited Methods of treating cancer and related compositions
US10370455B2 (en) 2014-12-05 2019-08-06 Immunext, Inc. Identification of VSIG8 as the putative VISTA receptor (V-R) and use thereof to produce VISTA/VSIG8 agonists and antagonists
US11009509B2 (en) 2015-06-24 2021-05-18 Janssen Pharmaceutica Nv Anti-VISTA antibodies and fragments
WO2017072716A1 (en) 2015-10-30 2017-05-04 Glaxosmithkline Intellectual Property Development Limited Prognostic method
US10899836B2 (en) 2016-02-12 2021-01-26 Janssen Pharmaceutica Nv Method of identifying anti-VISTA antibodies
US11987630B2 (en) 2016-02-12 2024-05-21 Janssen Pharmaceutica Nv Anti-vista antibodies and fragments, uses thereof, and methods of identifying same
US11603402B2 (en) 2016-04-15 2023-03-14 Immunext, Inc. Anti-human vista antibodies and use thereof
US11525000B2 (en) 2016-04-15 2022-12-13 Immunext, Inc. Anti-human VISTA antibodies and use thereof
US11603403B2 (en) 2016-04-15 2023-03-14 Immunext, Inc. Anti-human vista antibodies and use thereof
US11649283B2 (en) 2016-04-15 2023-05-16 Immunext, Inc. Anti-human vista antibodies and use thereof
WO2018047080A1 (en) 2016-09-07 2018-03-15 Glaxosmithkline Intellectual Property Development Limited Methods for purifying antibodies
US11453726B2 (en) 2016-09-15 2022-09-27 Quadrucept Bio Limited Multimers, tetramers and octamers
WO2018050902A2 (en) 2016-09-15 2018-03-22 Quadrucept Bio Limited Multimers, tetramers & octamers
WO2019171294A1 (en) 2018-03-07 2019-09-12 Glaxosmithkline Intellectual Property Development Limited Methods for purifying antibodies
WO2019171286A1 (en) 2018-03-07 2019-09-12 Glaxosmithkline Intellectual Property Development Limited Methods for purifying recombinant polypeptides
WO2020187711A1 (en) 2019-03-19 2020-09-24 Quadrucept Bio Limited Multimers
WO2021190980A1 (en) 2020-03-22 2021-09-30 Quadrucept Bio Limited Multimers for viral strain evolution
WO2022184659A1 (en) 2021-03-01 2022-09-09 Quadrucept Bio Limited Antibody domains & multimers
WO2024094527A1 (en) 2022-11-05 2024-05-10 Quadrucept Bio Limited Non-human vertebrates & cells

Also Published As

Publication number Publication date
AU2008229871A1 (en) 2008-11-06
CA2763913C (en) 2014-10-28
EP2277914A3 (en) 2012-08-08
WO2003014161A2 (en) 2003-02-20
EP2202243A3 (en) 2012-08-08
AU2008229871B2 (en) 2011-10-06
EP2277913A2 (en) 2011-01-26
JP2005515165A (en) 2005-05-26
ATE459651T1 (en) 2010-03-15
EP2281837A2 (en) 2011-02-09
US20120064074A1 (en) 2012-03-15
US8865431B2 (en) 2014-10-21
EP2202243A2 (en) 2010-06-30
WO2003014161A9 (en) 2004-04-01
AU2002319544B2 (en) 2008-07-10
CA2457636C (en) 2012-01-03
EP1419179A2 (en) 2004-05-19
DK1419179T3 (en) 2010-06-21
EP2281837B1 (en) 2016-10-05
ES2337986T3 (en) 2010-05-03
DE60235553D1 (en) 2010-04-15
EP2277913A3 (en) 2012-08-08
EP1419179B1 (en) 2010-03-03
JP5133494B2 (en) 2013-01-30
WO2003014161A3 (en) 2003-08-28
CA2457636A1 (en) 2003-02-20
EP2277914A2 (en) 2011-01-26
CA2763913A1 (en) 2003-02-20
PT1419179E (en) 2010-03-16
EP2281837A3 (en) 2012-08-08

Similar Documents

Publication Publication Date Title
US8865431B2 (en) Antigen binding domains
AU2002319544A1 (en) Antigen binding domains from fish
JP4323317B2 (en) Methods for cloning variable region sequences
Davies et al. Selection of specific phage-display antibodies using libraries derived from chicken immunoglobulin genes
JP3540315B2 (en) Production of Chimeric Antibodies-Combination Approach
US11685773B2 (en) Method for mass humanization of rabbit antibodies
DE602004012299T2 (en) METHOD FOR CONNECTING INTERESTING SEQUENCES
CA2258518C (en) Recognition molecules interacting specifically with the active site or cleft of a target molecule
JP4213586B2 (en) Camel antibody library production method
Figini et al. In vitro assembly of repertoires of antibody chains on the surface of phage by renaturation
JPH11503918A (en) Variable fragments of immunoglobulins-uses for therapeutic or veterinary purposes
US20100069262A1 (en) Method for Cloning Avian-Derived Antibodies
Romão et al. Construction of high-quality camel immune antibody libraries
US6740747B2 (en) Bovine VDJ cassette, BF1H1, suitable for antigenization
Wang et al. Construction and characterization of phage display library: recognition of mouse serologically detected male (SDM) antigen
Schmidthals Generation and Characterization of heavy chain antibodies derived from Camelids
Jin Characterisation of Protein and DNA Binding Specificity of Human VH Homodimers (homo-VHDs)
Dooley Characterisation of single domain antibody fragments from the nurse shark Ginglymostoma cirratum, using phage display

Legal Events

Date Code Title Description
AS Assignment

Owner name: MARYLAND, UNIVERSITY OF, MARYLAND

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:FLAJNIK, MARTIN;REEL/FRAME:016829/0428

Effective date: 20050316

Owner name: ABERDEEN UNIVERSITY, GREAT BRITAIN

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:DOOLEY, HELEN;PORTER, ANDREW;REEL/FRAME:016825/0750

Effective date: 20050611

AS Assignment

Owner name: NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF

Free format text: CONFIRMATORY LICENSE;ASSIGNOR:UNIVERSITY OF MARYLAND BALT PROF SCHOOL;REEL/FRAME:024513/0956

Effective date: 20100608

STCB Information on status: application discontinuation

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