WO2004003019A2 - Immunoglobin single variant antigen-binding domains and dual-specific constructs - Google Patents

Immunoglobin single variant antigen-binding domains and dual-specific constructs Download PDF

Info

Publication number
WO2004003019A2
WO2004003019A2 PCT/GB2003/002804 GB0302804W WO2004003019A2 WO 2004003019 A2 WO2004003019 A2 WO 2004003019A2 GB 0302804 W GB0302804 W GB 0302804W WO 2004003019 A2 WO2004003019 A2 WO 2004003019A2
Authority
WO
WIPO (PCT)
Prior art keywords
ligand
binding
specific
domains
epitope
Prior art date
Application number
PCT/GB2003/002804
Other languages
French (fr)
Other versions
WO2004003019A9 (en
WO2004003019A3 (en
Inventor
Greg Winter
Ian Tomlinson
Olga Ignatovich
Lucy Holt
Elena De Angelis
Phillip Jones
Original Assignee
Domantis Limited
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=30001956&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=WO2004003019(A2) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority claimed from PCT/GB2002/003014 external-priority patent/WO2003002609A2/en
Priority claimed from GB0230202A external-priority patent/GB0230202D0/en
Priority to SI200330414T priority Critical patent/SI1517921T1/en
Priority to AU2003244817A priority patent/AU2003244817B2/en
Priority to DE60305919T priority patent/DE60305919T2/en
Priority to JP2004516979A priority patent/JP2006512895A/en
Priority to EP03738294A priority patent/EP1517921B1/en
Priority to CA002492092A priority patent/CA2492092A1/en
Application filed by Domantis Limited filed Critical Domantis Limited
Priority to EP03782689A priority patent/EP1578801A2/en
Priority to JP2005509717A priority patent/JP2006523090A/en
Priority to CA002511910A priority patent/CA2511910A1/en
Priority to AU2003290330A priority patent/AU2003290330A1/en
Priority to PCT/GB2003/005646 priority patent/WO2004058821A2/en
Publication of WO2004003019A2 publication Critical patent/WO2004003019A2/en
Priority to EP10177693A priority patent/EP2267028A3/en
Priority to AU2004220325A priority patent/AU2004220325B2/en
Priority to JP2006518331A priority patent/JP5087274B2/en
Priority to AT04737302T priority patent/ATE414106T1/en
Priority to PL04737302T priority patent/PL1639011T3/en
Priority to CN2004800249396A priority patent/CN1845938B/en
Priority to PCT/GB2004/002829 priority patent/WO2004081026A2/en
Priority to CA002529819A priority patent/CA2529819A1/en
Priority to PT04737302T priority patent/PT1639011E/en
Priority to DK04737302T priority patent/DK1639011T3/en
Priority to DE602004017726T priority patent/DE602004017726D1/en
Priority to SI200431021T priority patent/SI1639011T1/en
Priority to EP08166692A priority patent/EP2221317A3/en
Priority to EP04737302A priority patent/EP1639011B1/en
Priority to ES04737302T priority patent/ES2315664T3/en
Priority to US10/925,366 priority patent/US20050271663A1/en
Publication of WO2004003019A3 publication Critical patent/WO2004003019A3/en
Priority to US10/985,847 priority patent/US20060002935A1/en
Priority to US11/023,959 priority patent/US20060106203A1/en
Priority to HK05103582A priority patent/HK1070081A1/en
Priority to GBGB0501752.0A priority patent/GB0501752D0/en
Priority to US11/098,758 priority patent/US20060073141A1/en
Priority to US11/141,661 priority patent/US20060257406A1/en
Priority to US11/217,919 priority patent/US20060063921A1/en
Priority to US11/664,542 priority patent/US20080008713A1/en
Priority to US11/667,393 priority patent/US20070298041A1/en
Priority to US11/315,848 priority patent/US20060286066A1/en
Priority to US11/338,863 priority patent/US20070104710A1/en
Publication of WO2004003019A9 publication Critical patent/WO2004003019A9/en
Priority to US11/501,522 priority patent/US20070093651A1/en
Priority to US11/501,546 priority patent/US20100234570A1/en
Priority to CY20061101247T priority patent/CY1105191T1/en
Priority to ZA200610474A priority patent/ZA200610474B/en
Priority to US11/704,832 priority patent/US9321832B2/en
Priority to IL183027A priority patent/IL183027A0/en
Priority to US11/981,821 priority patent/US20100081792A1/en
Priority to US12/006,933 priority patent/US20080241166A1/en
Priority to US12/150,567 priority patent/US20080233130A1/en
Priority to US12/150,487 priority patent/US20080233129A1/en
Priority to CY20091100035T priority patent/CY1108691T1/en
Priority to US12/409,617 priority patent/US20090258012A1/en
Priority to AU2011200930A priority patent/AU2011200930B2/en
Priority to US13/181,834 priority patent/US9028822B2/en
Priority to US13/733,675 priority patent/US20130216538A1/en
Priority to US14/173,204 priority patent/US20150087813A1/en
Priority to US14/924,971 priority patent/US20170145080A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/24Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against cytokines, lymphokines or interferons
    • C07K16/241Tumor Necrosis Factors
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2875Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the NGF/TNF superfamily, e.g. CD70, CD95L, CD153, CD154
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2878Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the NGF-receptor/TNF-receptor superfamily, e.g. CD27, CD30, CD40, CD95
    • 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
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/42Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against immunoglobulins
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/21Immunoglobulins specific features characterized by taxonomic origin from primates, e.g. man
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/31Immunoglobulins specific features characterized by aspects of specificity or valency multispecific
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/34Identification of a linear epitope shorter than 20 amino acid residues or of a conformational epitope defined by amino acid residues
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/52Constant or Fc region; Isotype
    • C07K2317/53Hinge
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/55Fab or Fab'
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/565Complementarity determining region [CDR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/567Framework region [FR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/569Single domain, e.g. dAb, sdAb, VHH, VNAR or nanobody®
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • C07K2317/62Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising only variable region components
    • C07K2317/622Single chain antibody (scFv)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • C07K2317/62Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising only variable region components
    • C07K2317/626Diabody or triabody
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2318/00Antibody mimetics or scaffolds
    • C07K2318/20Antigen-binding scaffold molecules wherein the scaffold is not an immunoglobulin variable region or antibody mimetics

Definitions

  • the present invention relates to dual specific ligands.
  • the invention provides a method for the preparation of dual-specific ligands comprising a first immunoglobulin single variable domain binding to a first antigen or epitope, and a second immunoglobulin single variable domain binding to a second antigen or epitope. More particularly, the invention relates to dual-specific ligands wherein binding to at least one of the first and second antigens or epitopes acts to increase the half-life of the ligand in vivo. Open and closed conformation ligands comprising more than one binding specificity are described.
  • the antigen binding domain of an antibody comprises two separate regions: a heavy chain variable domain (v H ) ⁇ d a light chain variable domain (V : which can be either K or V ⁇ ).
  • the antigen binding site itself is formed by six polypeptide loops: three from V H domain (HI, H2 and H3) and three from v L domain (LI, L2 and L3).
  • V H domain HI, H2 and H3
  • v L domain LI, L2 and L3
  • a diverse primary repertoire of V genes that encode the V H an d V L domains is produced by the combinatorial rearrangement of gene segments.
  • the V H S ene * s produced by the recombination of three gene segments, V H> D an d JH- T ⁇ .
  • V H segments there are approximately 51 functional V H segments (Cook and Tomlinson (1995) Immunol Today, 16: 237), 25 functional D segments (Corbett et al. (1997) J. Mol. Biol, 268: 69) and 6 functional JJJ segments (Ravetch et al. (1981) Cell, 27: 583), depending on the haplotype.
  • the V H segment encodes the region of the polypeptide chain which forms the first and second antigen binding loops of the v H domain (HI and H2), whilst the VH > D an JH segments combine to form the third antigen binding loop of the VH domain (H3).
  • the A S ene is produced by the recombination of only two gene segments, V an ⁇ L- In humans, there are approximately 40 functional V ⁇ segments (Schable and Zachau (1993) Biol. Chem. Hoppe-Seyler, 374: 1001), 31 functional V ⁇ segments (Williams et al (1996) J. Mol. Biol, 264: 220; Kawasaki et al. (1997) Genome Res., 7: 250), 5 functional J ⁇ segments (Hieter et al. (1982) J Biol. Chem., 257: 1516) and 4 functional j segments (Vasicek and Leder (1990) J Exp. Med., 172: 609), depending on the haplotype.
  • the ⁇ L segment encodes the region of the polypeptide chain which forms the first and second antigen binding loops of the y L domain (LI and L2), whilst the VL an d JL segments combine to form the third antigen binding loop of the V domain (L3).
  • Antibodies selected from this primary repertoire are believed to be sufficiently diverse to bind almost all antigens with at least moderate affinity. High affinity antibodies are produced by "affinity maturation" of the rearranged genes, in which point mutations are generated and selected by the immune system on the basis of improved binding.
  • the main-chain conformations are determined by (i) the length of the antigen binding loop, and (ii) particular residues, or types of residue, at certain key position in the antigen binding loop and the antibody framework.
  • H3 region is much more diverse in terms of sequence, length and structure (due to the use of D segments), it also forms a limited number of main-chain conformations for short loop lengths which depend on the length and the presence of particular residues, or types of residue, at key positions in the loop and the antibody framework (Martin et al. (1996) J. Mol. Biol, 263: 800; Shirai et al. (1996) FEBS Letters, 399: 1.
  • Bispecific antibodies comprising complementary pairs of V H an N L regions are known in the art. These bispecific antibodies must comprise two pairs of VH an d V L s, eac V H ⁇ V L pair binding to a single antigen or epitope. Methods described involve hybrid hybridomas (Milstein & Cuello AC, Nature 305:537-40), minibodies (Hu et al, (1996) Cancer Res 56:3055-3061;), diabodies (Holliger et al, (1993) Proc. Natl. Acad. Sci. USA 90, 6444- 6448; WO 94/13804), chelating recombinant antibodies (CRAbs; (Neri et al, (1995) j. Mol. Biol.
  • each antibody species comprises two antigen-binding sites, each fashioned by a complementary pair of V H an d V domains. Each antibody is thereby able to bind to two different antigens or epitopes at the same time, with the binding to EACH antigen or epitope mediated by a V H an it complementary V L domain.
  • inactive VH ⁇ V L P arrs can greatly reduce the fraction of bispecific IgG.
  • WO 02/02773 (Abbott Laboratories) describes antibody molecules with "dual specificity".
  • the antibody molecules referred to are antibodies raised or selected against multiple antigens, such that their specificity spans more than a single antigen.
  • Each complementary V H /V L pair in the antibodies of WO 02/02773 specifies a single binding specificity for two or more structurally related antigens; the V H and V L domains in such complementary pairs do not each possess a separate specificity.
  • the antibodies thus have a broad single specificity which encompasses two antigens, which are structurally related.
  • natural auto antibodies have been described that are polyreactive (Casali & Notkins, Ann. Rev. Immunol.
  • Protein engineering methods have been suggested that may have a bearing on this.
  • a catalytic antibody could be created with a binding activity to a metal ion through one variable domain, and to a hapten (substrate) through contacts with the metal ion and a complementary variable domain (Barbas et al., 1993 Proc. Natl. Acad. Sci USA 90, 6385-6389).
  • the binding and catalysis of the substrate is proposed to require the binding of the metal ion (second antigen).
  • the binding to the V H ⁇ V L pairing relates to a single but multi- component antigen.
  • Single heavy chain variable domains have also been described, derived from natural antibodies which are normally associated with light chains (from monoclonal antibodies or from repertoires of domains; see EP-A-0368684). These heavy chain variable domains have been shown to interact specifically with one or more related antigens but have not been combined with other heavy or light chain variable domains to create a ligand with a specificity for two or more different antigens . Furthermore, these single domains have been shown to have a very short in vivo half-life. Therefore such domains are of limited therapeutic value.
  • the inventors have described, in their copending international patent application WO 03/002609 as well as copending unpublished UK patent application 0230203.2, dual specific immunoglobulin ligands which comprise immunoglobulin single variable domains which each have different specificities.
  • the domains may act in competition with each other or independently to bind antigens or epitopes on target molecules.
  • the present invention provides a further improvement in dual specific ligands as developed by the present inventors, in which one specificity of the ligand is directed towards a protein or polypeptide present in vivo in an organism which can act to increase the half-life of the ligand by binding to it.
  • a dual-specific ligand comprising a first immunoglobulin single variable domain having a binding specificity to a first antigen or epitope and a second complementary immunoglobulin single variable domain having a binding activity to a second antigen or epitope, wherein one or both of said antigens or epitopes acts to increase the half-life of the ligand in vivo and wherein said first and second domains lack mutually complementary domains which share the same specificity, provided that said dual specific ligand does not consist of an anti-HSA V H domain and an anti- ⁇ galactosidase V ⁇ domain.
  • HSA human serum albumin
  • Antigens or epitopes which increase the half-life of a ligand as described herein are advantageously present on proteins or polypeptides found in an organism in vivo. Examples include extracellular matrix proteins, blood proteins, and proteins present in various tissues in the organism. The proteins act to reduce the rate of ligand clearance from the blood, for example by acting as bulking agents, or by anchoring the ligand to a desired site of action. Examples of antigens/epitopes which increase half-life in vivo are given in Annex 1 below.
  • Increased half-life is useful in in vivo applications of immunoglobulins, especially antibodies and most especially antibody fragments of small size.
  • Such fragments (Fvs, disulphide bonded Fvs, Fabs, scFvs, dAbs) suffer from rapid clearance from the body; thus, whilst they are able to reach most parts of the body rapidly, and are quick to produce and easier to handle, their in vivo applications have been limited by their only brief persistence in vivo.
  • the invention solves this problem by providing increased half-life of the ligands in vivo and consequently longer persistence times in the body of the functional activity of the ligand.
  • Half lives (i l A alpha and X l A beta) and AUC can be determined from a curve of serum concentration of ligand against time.
  • the WinNonlin analysis package (available from Pharsight Corp., Mountain View, CA94040, USA) can be used, for example, to model the curve.
  • a first phase the alpha phase
  • a second phase (beta phase) is the terminal phase when the ligand has been distributed and the serum concentration is decreasing as the ligand is cleared from the patient.
  • the t alpha half life is the half life of the first phase and the t beta half life is the half life of the second phase.
  • the present invention provides a ligand or a composition comprising a ligand according to the invention having a t ⁇ half-life in the range of 15 minutes or more.
  • the lower end of the range is 30 minutes, 45 minutes, 1 hour, 2 hours, 3 hours, 4 hours, 5 hours, 6 hours, 7 hours, 10 hours, 11 hours or 12 hours.
  • a ligand or composition according to the invention will have a t ⁇ half life in the range of up to and including 12 hours.
  • the upper end of the range is 11, 10, 9, 8, 7, 6 or 5 hours.
  • An example of a suitable range is 1 to 6 hours, 2 to 5 hours or 3 to 4 hours.
  • the present invention provides a ligand or a composition comprising a ligand according to the invention having a t ⁇ half-life in the range of 2.5 hours or more.
  • the lower end of the range is 3 hours, 4 hours, 5 hours, 6 hours, 7 hours, 10 hours , 11 hours, or 12 hours.
  • a ligand or composition according to the invention has a t ⁇ half-life in the range of up to and including 21 days.
  • the upper end of the range is 12 hours, 24 hours, 2 days, 3 days, 5 days, 10 days, 15 days or 20 days.
  • a ligand or composition according to the invention will have a t ⁇ half life in the range 12 to 60 hours. In a further embodiment, it will be in the range 12 to 48 hours. In a further embodiment still, it will be in the range 12 to 26 hours.
  • the present invention provides a ligand or a composition comprising a ligand according to the invention having an AUC value (area under the curve) in the range of 1 mg.min/ml or more.
  • the lower end of the range is 5, 10, 15, 20, 30, 100, 200 or 300mg.min ml.
  • a ligand or composition according to the invention has an AUC in the range of up to 600 mg.min/ml.
  • the upper end of the range is 500, 400, 300, 200, 150, 100, 75 or 50 mg.min ml.
  • a ligand according to the invention will have a AUC in the range selected from the group consisting of the following: 15 to 150mg.min/ml, 15 to 100 mg.min/ml, 15 to 75 mg.min/ml, and 15 to 50mg.min/ml.
  • the dual specific ligand comprises two complementary variable domains, i.e. two variable domains that, in their natural environment, are capable of operating together as a cognate pair or group even if in the context of the present invention they bind separately to their cognate epitopes.
  • the complementary variable domains may be immunoglobulin heavy chain and light chain variable domains
  • V H and V L domains are advantageously provided by scFv or Fab antibody fragments.
  • Variable domains may be linked together to form multivalent ligands by, for example: provision of a hinge region at the C-terminus of each V domain and disulphide bonding between cysteines in the hinge regions; or provision of dAbs each with a cysteine at the C-terminus of the domain, the cysteines being disulphide bonded together; or production of V-CH & V-CL to produce a Fab format; or use of peptide linkers (for example Gly 4 Ser linkers discussed hereinbelow) to produce dimers, trimers and further multimers.
  • peptide linkers for example Gly 4 Ser linkers discussed hereinbelow
  • the inventors have found that the use of complementary variable domains allows the two domain surfaces to pack together and be sequestered from the solvent. Furthermore the complementary domains are able to stabilise each other. In addition, it allows the creation of dual-specific IgG antibodies without the disadvantages of hybrid hybridomas as used in the prior art, or the need to engineer heavy or light chains at the sub-unit interfaces.
  • the dual-specific ligands of the first aspect of the present invention have at least one V H /V L pair.
  • a bispecific IgG according to this invention will therefore comprise two such pairs, one pair on each arm of the Y-shaped molecule.
  • bi-specific molecules can be created in two different ways. Firstly, they can be created by association of two existing V H /V pairings that each bind to a different antigen or epitope (for example, in a bi-specific IgG). In this case the V H /VL pairings must come all together in a 1:1 ratio in order to create a population of molecules all of which are bi-specific. This never occurs (even when complementary CH domain is enhanced by "knobs into holes” engineering) leading to a mixture of bi-specific molecules and molecules that are only able to bind to one antigen or epitope but not the other.
  • the second way of creating a bispecific antibody is by the simultaneous association of two different V H chain with two different V L chains (for example in a bi-specific diabody).
  • Bi-specific antibodies constructed according to the dual-specific ligand approach according to the first aspect of the present invention overcome all of these problems because the binding to antigen or epitope 1 resides within the V H or V L domain and the binding to antigen or epitope 2 resides with the complementary V L or V H domain, respectively. Since VH and V L domains pair on a 1:1 basis all V H /V L pairings will be bispecific and thus all formats constructed using these VH/V L pairings (Fv, scFvs, Fabs, minibodies, IgGs etc) will have 100% bi-specific activity.
  • first and second "epitopes” are understood to be epitopes which are not the same and are not bound by a single monospecific ligand.
  • they are advantageously on different antigens, one of which acts to increase the half-life of the ligand in vivo.
  • the first and second antigens are advantageously not the same.
  • the dual specific ligands of the invention do not include ligands as described in WO 02/02773.
  • the ligands of the present invention do not comprise complementary V H /V L pairs which bind any one or more antigens or epitopes co-operatively.
  • the ligands according to the first aspect of the invention comprise a VH VL complementary pair, wherein the V domains have different specificities.
  • the ligands according to the first aspect of the invention comprise V H /V L complementary pairs having different specificities for non-structurally related epitopes or antigens.
  • Structurally related epitopes or antigens are epitopes or antigens which possess sufficient structural similarity to be bound by a conventional V H V L complementary pair which acts in a co-operative manner to bind an antigen or epitope; in the case of structurally related epitopes, the epitopes are sufficiently similar in structure that they "fit" into the same binding pocket formed at the antigen binding site of the V H /V L dimer.
  • the present invention provides a ligand comprising a first immunoglobulin variable domain having a first antigen or epitope binding specificity and a second immunoglobulin variable domain having a second antigen or epitope binding specificity wherein one or both of said first and second variable domains bind to an antigen which increases the half-life of the ligand in vivo, and the variable domains are not complementary to one another.
  • binding to one variable domain modulates the binding of the ligand to the second variable domain.
  • variable domains may be, for example, pairs of V H domains or pairs of V L domains.
  • Binding of antigen at the first site may modulate, such as enhance or inhibit, binding of an antigen at the second site.
  • binding at the first site at least partially inhibits binding of an antigen at a second site.
  • the ligand may for example be maintained in the body of a subject organism in vivo through binding to a protein which increases the half-life of the ligand until such a time as it becomes bound to the second target antigen and dissociates from the half-life increasing protein. Modulation of binding in the above context is achieved as a consequence of the structural proximity of the antigen binding sites relative to one another.
  • Such structural proximity can be achieved by the nature of the structural components linking the two or more antigen binding sites, eg by the provision of a ligand with a relatively rigid structure that holds the antigen binding sites in close proximity.
  • the two or more antigen binding sites are in physically close proximity to one another such that one site modulates the binding of antigen at another site by a process which involves steric hindrance and/or conformational changes within the immunoglobulin molecule.
  • the first and the second antigen binding domains may be associated either covalently or non-covalently.
  • Ligands according to the invention may be combined into non-immunoglobulin multi- ligand structures to form multivalent complexes, which bind target molecules with the same antigen, thereby providing superior avidity, while at least one variable domain binds an antigen to increase the half life of the multimer.
  • natural bacterial receptors such as SpA have been used as scaffolds for the grafting of CDRs to generate ligands which bind specifically to one or more epitopes. Details of this procedure are described in US 5,831,012.
  • Other suitable scaffolds include those based on fibronectin and affibodies. Details of suitable procedures are described in WO 98/58965.
  • Suitable scaffolds include lipocallin and CTLA4, as described in van den Beuken et al, J. Mol. Biol. (2001) 310, 591-601, and scaffolds such as those described in WO0069907 (Medical Research Council), which are based for example on the ring structure of bacterial GroEL or other chaperone polypeptides.
  • Protein scaffolds may be combined; for example, CDRs may be grafted on to a CTLA4 scaffold and used together with immunoglobulin VH or V L domains to form a ligand. Likewise, fibronectin, lipocallin and other scaffolds may be combined.
  • variable domains are selected from V-gene repertoires selected for instance using phage display technology as herein described, then these variable domains can comprise a universal framework region, such that is they may be recognised by a specific generic ligand as herein defined.
  • the use of universal frameworks, generic ligands and the like is described in WO99/20749.
  • reference to phage display includes the use of both phage and/or phagemids.
  • variable domains variation in polypeptide sequence is preferably located within the structural loops of the variable domains.
  • the polypeptide sequences of either variable domain may be altered by DNA shuffling or by mutation in order to enhance the interaction of each variable domain with its complementary pair.
  • the 'dual-specific ligand' is a single chain Fv fragment.
  • the 'dual-specific ligand' consists of a Fab region of an antibody.
  • the term "Fab region" includes a Fab-like region where two VH or two VL domains are used.
  • variable regions may be derived from antibodies directed against target antigens or epitopes. Alternatively they may be derived from a repertoire of single antibody domains such as those expressed on the surface of filamentous bacteriophage. Selection may be performed as described below.
  • the invention provides a method for producing a ligand comprising a first immunoglobulin single variable domain having a first binding specificity and a second single immunoglobulin single variable domain having a second (different) binding specificity, one or both of the binding specificities being specific for an antigen which increases the half-life of the ligand in vivo, the method comprising the steps of:
  • variable domains (b) selecting a second variable region by its ability to bind to a second epitope, (c) combining the variable domains;
  • step (d) selecting the ligand by its ability to bind to said first epitope and to said second epitope.
  • the ligand can bind to the first and second epitopes either simultaneously or, where there is competition between the binding domains for epitope binding, the binding of one domain may preclude the binding of another domain to its cognate epitope. h one embodiment, therefore, step (d) above requires simultaneous binding to both first and second (and possibly further) epitopes; in another embodiment, the binding to the first and second epitoes is not simultaneous.
  • the epitopes are preferably on separate antigens.
  • Ligands advantageously comprise V H /V L combinations, or V H /VH or V L /V L combinations of immunoglobulin variable domains, as described above.
  • the ligands may moreover comprise camelid V HH domains, provided that the V HH domain which is specific for an antigen which increases the half-life of the ligand in vivo does not bind Hen egg white lysozyme (HEL), porcine pancreatic alpha-amylase or NmC-A; hcg, BSA-linked RR6 azo dye or S.
  • HEL Hen egg white lysozyme
  • NmC-A hcg
  • BSA-linked RR6 azo dye or S Hen egg white lysozyme
  • said first variable domain is selected for binding to said first epitope in absence of a complementary variable domain.
  • said first variable domain is selected for binding to said first epitope/antigen in the presence of a third variable domain in which said third variable domain is different from said second variable domain and is complementary to the first domain.
  • the second domain may be selected in the absence or presence of a complementary variable domain.
  • the antigens or epitopes targeted by the ligands of the invention may be any antigen or epitope but advantageously is an antigen or epitope that is targeted with therapeutic benefit.
  • the invention provides ligands, including open conformation, closed conformation and isolated dAb monomer ligands, specific for any such target, particularly those targets further identified herein. Such targets may be, or be part of, polypeptides, proteins or nucleic acids, which may be naturally occurring or synthetic.
  • the ligand of the invention may bind the epiotpe or antigen and act as an antagonist or agonist (eg, EPO receptor agonist).
  • EPO receptor agonist eg, EPO receptor agonist
  • cytokines and growth factors include but are not limited to: ApoE, Apo-SAA, BDNF, Cardiotrophin-1, EGF, EGF receptor, ENA-78, Eotaxin, Eotaxin-2, Exodus-2, EpoR, FGF-acidic, FGF-basic, fibroblast growth factor- 10, FLT3 ligand, Fractalkine (CX3C), GDNF, G-CSF, GM-CSF, GF- ⁇ l, insulin, IFN- ⁇ , IGF-I, IGF-II, IL-l ⁇ , IL-l ⁇ , IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8 (72 a.a.), IL-8 (77 a.a.), IL-9, IL-10, IL-11, IL-12, IL-13
  • variable domains are derived from a respective antibody directed against the antigen or epitope. In a preferred embodiment the variable domains are derived from a repertoire of single variable antibody domains.
  • the present invention provides one or more nucleic acid molecules encoding at least a dual-specific ligand as herein defined.
  • the dual specific ligand may be encoded on a single nucleic acid molecule; alternatively, each domain may be encoded by a separate nucleic acid molecule.
  • the domains may be expressed as a fusion polypeptide, in the manner of a scFv molecule, or may be separately expressed and subsequently linked together, for example using chemical linking agents.
  • Ligands expressed from separate nucleic acids will be linked together by appropriate means.
  • the nucleic acid may further encode a signal sequence for export of the polypeptides from a host cell upon expression and may be fused with a surface component of a filamentous bacteriophage particle (or other component of a selection display system) upon expression.
  • the present invention provides a vector comprising nucleic acid encoding a dual specific ligand according to the present invention.
  • the present invention provides a host cell transfected with a vector encoding a dual specific ligand according to the present invention.
  • Expression from such a vector may be configured to produce, for example on the surface of a bacteriophage particle, variable domains for selection. This allows selection of displayed variable regions and thus selection of 'dual-specific ligands' using the method of the present invention.
  • the present invention further provides a kit comprising at least a dual-specific ligand according to the present invention.
  • Dual-Specific ligands preferably comprise combinations of heavy and light chain domains.
  • the dual specific ligand may comprise a V H domain and a V L domain, which may be linked together in the form of an scFv.
  • the ligands may comprise one or more C H or CL domains.
  • the ligands may comprise a CHI domain, C H 2 or C H 3 domain, and/or a CL domain, C ⁇ l, C ⁇ 2, C ⁇ 3 or C ⁇ 4 domains, or any combination thereof.
  • a hinge region domain may also be included.
  • Such combinations of domains may, for example, mimic natural antibodies, such as IgG or IgM, or fragments thereof, such as Fv, scFv, Fab or F(ab') 2 molecules.
  • Other structures such as a single arm of an IgG molecule comprising VH, V L , C H I and C L domains, are envisaged.
  • variable regions are selected from single domain V gene repertoires.
  • the repertoire of single antibody domains is displayed on the surface of filamentous bacteriophage.
  • each single antibody domain is selected by binding of a phage repertoire to antigen.
  • each single variable domain may be selected for binding to its target antigen or epitope in the absence of a complementary variable region.
  • the single variable domains may be selected for binding to its target antigen or epitope in the presence of a complementary variable region.
  • the first single variable domain may be selected in the presence of a third complementary variable domain
  • the second variable domain may be selected in the presence of a fourth complementary variable domain.
  • the complementary third or fourth variable domain may be the natural cognate variable domain having the same specificity as the single domain being tested, or a non-cognate complementary domain - such as a "dummy" variable domain.
  • the dual specific ligand of the invention comprises only two variable domains although several such ligands may be incorporated together into the same protein, for example two such ligands can be incorporated into an IgG or a multimeric immunoglobulin, such as IgM.
  • a plurality of dual specific ligands are combined to form a multimer.
  • two different dual specific ligands are combined to create a tetra-specific molecule.
  • variable regions of a dual-specific ligand produced according to the method of the present invention may be on the same polypeptide chain, or alternatively, on different polypeptide chains.
  • variable regions are on different polypeptide chains, then they may be linked via a linker, generally a flexible linker (such as a polypeptide chain), a chemical linking group, or any other method known in the art.
  • the present invention provides a composition comprising a dual- specific ligand, obtainable by a method of the present invention, and a pharmaceutically acceptable carrier, diluent or excipient. Moreover, the present invention provides a method for the treatment and/or prevention of disease using a 'dual-specific ligand' or a composition according to the present invention.
  • the present invention provides multispecific ligands which comprise at least two non-complementary variable domains.
  • the ligands may comprise a pair of V H domains or a pair of VL domains.
  • the domains are of non-camelid origin; preferably they are human domains or comprise human framework regions (FWs) and one or more heterologous CDRs.
  • CDRs and framework regions are those regions of an immunoglobulin variable domain as defined in the Kabat database of Sequences of Proteins of rmmunological Interest.
  • Preferred human framework regions are those encoded by germline gene segments DP47 and DPK9.
  • FWl, FW2 and FW3 of a V H or V L domain have the sequence of FWl, FW2 or FW3 from DP47 or DPK9.
  • the human frameworks may optionally contain mutations, for example up to about 5 amino acid changes or up to about 10 amino acid changes collectively in the human frameworks used in the ligands of the invention.
  • variable domains in the multispecific ligands according to the second configuration of the invention may be arranged in an open or a closed conformation; that is, they may be arranged such that the variable domains can bind their cognate ligands independently and simultaneously, or such that only one of the variable domains may bind its cognate ligand at any one time.
  • non-complementary variable domains for example two light chain variable domains or two heavy chain variable domains
  • a ligand such that binding of a first epitope to a first variable domain inhibits the binding of a second epitope to a second variable domain, even though such non-complementary domains do not operate together as a cognate pair.
  • the ligand comprises two or more pairs of variable domains; that is, it comprises at least four variable domains.
  • the four variable domains comprise frameworks of human origin.
  • the human frameworks are identical to those of human germline sequences.
  • the present invention provides a method for producing a multispecific ligand comprising the steps of: a) selecting a first epitope binding domain by its ability to bind to a first epitope, b) selecting a second epitope binding domain by its ability to bind to a second epitope, c) combining the epitope binding domains; and d) selecting the closed conformation multispecific ligand by its ability to bind to said first second epitope and said second epitope.
  • the invention provides method for preparing a closed conformation multi-specific ligand comprising a first epitope binding domain having a first epitope binding specificity and a non-complementary second epitope binding domain having a second epitope binding specificity, wherein the first and second binding specificities compete for epitope binding such that the closed conformation multi-specific ligand may not bind both epitopes simultaneously, said method comprising the steps of:
  • a) selecting a first epitope binding domain by its ability to bind to a first epitope b) selecting a second epitope binding domain by its ability to bind to a second epitope, c) combining the epitope binding domains such that the domains are in a closed conformation; and d) selecting the closed conformation multispecific ligand by its ability to bind to said first second epitope and said second epitope, but not to both said first and second epitopes simultaneously.
  • the invention provides a closed conformation multi-specific ligand comprising a first epitope binding domain having a first epitope binding specificity and a non- complementary second epitope binding domain having a second epitope binding specificity, wherein the first and second binding specificities compete for epitope binding such that the closed conformation multi-specific ligand may not bind both epitopes simultaneously.
  • An alternative embodiment of the above aspect of the of the second configuration of the invention optionally comprises a further step (bl) comprising selecting a third or further epitope binding domain.
  • the multi-specific ligand produced whether of open or closed conformation, comprises more than two epitope binding specificities.
  • the multi-specific ligand comprises more than two epitope binding domains
  • at least two of said domains are in a closed conformation and compete for binding; other domains may compete for binding or may be free to associate independently with their cognate epitope(s).
  • the term 'multi-specific ligand' refers to a ligand which possesses more than one epitope binding specificity as herein defined.
  • the term 'closed conformation' means that the epitope binding domains of the ligand are attached to or associated with each other, optionally by means of a protein skeleton, such that epitope binding by one epitope binding domain competes with epitope binding by another epitope binding domain. That is, cognate epitopes may be bound by each epitope binding domain individually but not simultaneosuly.
  • the closed conformation of the ligand can be achieved using methods herein described.
  • Open conformation means that the epitope binding domains of the ligand are attached to or associated with each other, optionally by means of a protein skeleton, such that epitope binding by one epitope binding domain does not compete with epitope binding by another epitope binding domain.
  • the term 'competes' means that the binding of a first epitope to its cognate epitope binding domain is inhibited when a second epitope is bound to its cognate epitope binding domain.
  • binding may be inhibited sterically, for example by physical blocking of a binding domain or by alteration of the structure or environment of a binding domain such that its affinity or avidity for an epitope is reduced.
  • the epitopes may displace each other on binding.
  • a first epitope may be present on an antigen which, on binding to its cognate first binding domain, causes steric hindrance of a second binding domain, or a coformational change therein, which displaces the epitope bound to the second binding domain.
  • binding is reduced by 25% or more, advantageously 40%, 50%, 60%, 70%, 80%, 90% or more, and preferably up to 100% or nearly so, such that binding is completely inhibited.
  • Binding of epitopes can be measured by conventional antigen binding assays, such as ELISA, by fluorescence based techniques, including FRET, or by techniques such as suface plasmon resonance which measure the mass of molecules.
  • each epitope binding domain is of a different epitope binding specificity.
  • first and second “epitopes” are understood to be epitopes which are not the same and are not bound by a single monospecific ligand. They may be on different antigens or on the same antigen, but separated by a sufficient distance that they do not form a single entity that could be bound by a single mono-specific V H /V L binding pair of a conventional antibody.
  • domain antibodies or dAbs are separately competed by a monospecific VH/V L ligand against two epitopes then those two epitopes are not sufficiently far apart to be considered separate epitopes according to the present invention.
  • the closed conformation multispecific ligands of the invention do not include ligands as described in WO 02/02773.
  • the ligands of the present invention do not comprise complementary VH ⁇ V L P a i rs which bind any one or more antigens or epitopes cooperatively.
  • the ligands according to the invention preferably comprise non- complementary V H 'VH or VL"VL pairs.
  • each V H or V L domain in each V H "V H or V L -V L P arr h s a different epitope binding specificity, and the epitope binding sites are so arranged that the binding of an epitope at one site competes with the binding of an epitope at another site.
  • each epitope binding domain comprises an immunoglobulin variable domain.
  • each immunoglobulin variable domain will be either a variable light chain domain (V ) or a variable heavy chain domain V H - * 1 the second configuration of the present invention, the immunoglobulin domains when present on a ligand according to the present invention are non-complementary, that is they do not associate to form a V H V L antigen binding site.
  • multi-specific ligands as defined in the second configuration of the invention comprise immunoglobulin domains of the same sub-type, that is either variable light chain domains (y-) or variable heavy chain domains (v H )- Moreover, where the ligand according to the invention is in the closed conformation, the immunoglobulin domains may be of the camelid V HH type.
  • the ligand(s) according to the invention do not comprise a camelid V HH domain. More particularly, the ligand(s) of the invention do not comprise one or more amino acid residues that are specific to camelid V HH domains as compared to human V H domains.
  • variable domains are derived from antibodies selected for binding activity against different antigens or epitopes.
  • the variable domains may be isolated at least in part by human immunisation. Alternative methods are known in the art, including isolation from human antibody libraries and synthesis of artificial antibody genes.
  • the variable domains advantageously bind superantigens, such as protein A or protein L. Binding to superantigens is a property of correctly folded antibody variable domains, and allows such domains to be isolated from, for example, libraries of recombinant or mutant domains.
  • Epitope binding domains according to the present invention comprise a protein scaffold and epitope interaction sites (which are advantageously on the surface of the protein scaffold).
  • Epitope binding domains may also be based on protein scaffolds or skeletons other than immunoglobulin domains.
  • natural bacterial receptors such as SpA have been used as scaffolds for the grafting of CDRs to generate ligands which bind specifically to one or more epitopes. Details of this procedure are described in US 5,831,012.
  • Other suitable scaffolds include those based on fibronectin and affibodies. Details of suitable procedures are described in WO 98/58965.
  • Other suitable scaffolds include lipocallin and CTLA4, as described in van den Beuken et al, J. Mol. Biol. (2001) 310, 591-601, and scaffolds such as those described in WO0069907 (Medical Research Council), which are based for example on the ring structure of bacterial GroEL or other chaperone polypeptides.
  • Protein scaffolds may be combined; for example, CDRs may be grafted on to a CTLA4 scaffold and used together with immunoglobulin V H or V L domains to form a multivalent ligand. Likewise, fibronectin, lipocallin and other scaffolds maybe combined.
  • the epitope binding domains of a closed conformation multispecific ligand produced according to the method of the present invention may be on the same polypeptide chain, or alternatively, on different polypeptide chains.
  • the variable regions are on different polypeptide chains, then they may be linked via a linker, advantageously a flexible linker (such as a polypeptide chain), a chemical linking group, or any other method known in the art.
  • the first and the second epitope binding domains may be associated either covalently or non-covalently. In the case that the domains are covalently associated, then the association may be mediated for example by disulphide bonds.
  • the first and the second epitopes are preferably different. They may be, or be part of, polypeptides, proteins or nucleic acids, which may be naturally occurring or synthetic.
  • the ligand of the invention may bind an epiotpe or antigen and act as an antagonist or agonist (eg, EPO receptor agonist).
  • the epitope binding domains of the ligand in one embodiment have the same epitope specificity, and may for example simultaneously bind their epitope when multiple copies of the epitope are present on the same antigen.
  • these epitopes are provided on different antigens such that the ligand can bind the epitopes and bridge the antigens.
  • epitopes and antigens may be for instance human or animal proteins, cytokines, cytokine receptors, enzymes co-factors for enzymes or DNA binding proteins.
  • Suitable cytokines and growth factors include but are not limited to: ApoE, Apo-SAA, BDNF, Cardiotrophin-1, EGF, EGF receptor, ENA-78, Eotaxin, Eotaxin-2, Exodus-2, EpoR, FGF-acidic, FGF-basic, fibroblast growth factor- 10, FLT3 ligand, Fractalkine (CX3C), GDNF, G-CSF, GM-CSF, GF- ⁇ l, insulin, IFN- ⁇ , IGF-L IGF-IL IL-l ⁇ , IL-l ⁇ , IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8 (72 a.a.), IL-8 (77 a.a).
  • IL-10R IL-18R
  • dual specific ligands may be used to target cytokines and other molecules which cooperate synergistically in therapeutic situations in the body of an organism.
  • the invention therefore provides a method for synergising the activity of two or more cytokines, comprising administering a dual specific ligand capable of binding to said two or more cytokines.
  • the dual specific ligand may be any dual specific ligand, including a ligand composed of complementary and/or non- complementary domains, a ligand in an open conformation, and a ligand in a closed conformation.
  • this aspect of the invention relates to combinations of V H domains and V L domains, V H domains only and V L domains only.
  • Target combinations may be therapeutically active only if both targets are targeted by the ligand, whereas targeting one target alone is not therapeutically effective, hi another embodiment, one target alone may provide some low or minimal therapeutic effect, but together with a second target the combination provides a synergistic increase in therapeutic effect.
  • the cytokines bound by the dual specific ligands ofthis aspect of the invention are sleeted from the list shown in Annex 2.
  • dual specific ligands may be used in oncology applications, where one specificity targets CD89, which is expressed by cytotoxic cells, and the other is tumour specific.
  • examples of tumour antigens which may be targetted are given in Annex 3.
  • variable domains are derived from an antibody directed against the first and/or second antigen or epitope.
  • variable domains are derived from a repertoire of single variable antibody domains.
  • the repertoire is a repertoire that is not created in an animal or a synthetic repertoire.
  • the single variable domains are not isolated (at least in part) by animal immunisation. Thus, the single domains can be isolated from a naive library.
  • the second configuration of the invention in another aspect, provides a multi-specific ligand comprising a first epitope binding domain having a first epitope binding specificity and a non-complementary second epitope binding domain having a second epitope binding specificity.
  • the first and second binding specificities may be the same or different.
  • the present invention provides a closed conformation multi-specific ligand comprising a first epitope binding domain having a first epitope binding specificity and a non-complementary second epitope binding domain having a second epitope binding specificity wherein the first and second binding specificities are capable of competing for epitope binding such that the closed conformation multi-specific ligand cannot bind both epitopes simultaneously.
  • the invention provides open conformation ligands comprising non-complementary binding domains, wherein the deomains are specific for a different epitope on the same target. Such ligands bind to targets with increased avidity.
  • the invention provides multivalent ligands comprising non-complementary binding domains specific for the same epitope and directed to targets which comprise multiple copies of said epitope, such as IL-5, PDGF-AA, PDGF-BB, TGF beta, TGF beta2, TGF beta3 and TNF ⁇ , for eample human TNF Receptor 1 and human TNF ⁇ .
  • ligands according to the invention can be configured to bind individual epitopes with low affinity, such that binding to individual epitopes is not therapeutically significant; but the increased avidity resulting from binding to two epitopes provides a theapeutic benefit.
  • epitopes may be targetted which are present individually on normal cell types, but present together only on abnormal or diseased cells, such as tumour cells. In such a situaton, only the abnormal or diseased cells are effectively targetted by the bispecific ligands according to the invention.
  • Ligand specific for multiple copies of the same epitope, or adjacent epitopes, on the same target may also be trimeric or polymeric (tertrameric or more) ligands comprising three, four or more non-complementary binding domains.
  • ligands may be constructed comprising three or four V H domains or V L domains.
  • ligands are provided which bind to multisubunit targets, wherein each binding domain is specific for a subunit of said target.
  • the ligand may be dimeric, trimeric or polymeric.
  • the multi-specific ligands according to the above aspects of the invention are obtainable by the method of the first aspect of the invention.
  • the first epitope binding domain and the second epitope binding domains are non-complementary immunoglobulin variable domains, as herein defined. That is either V H "V H or V L "V L variable domains.
  • Chelating dAbs in particular may be prepared according to a preferred aspect of the invention, namely the use of anchor dAbs, in which a library of dimeric, trimeric or multimeric dAbs is constructed using a vector which comprises a constant dAb upstream or downstream of a linker sequence, with a repertoire of second, third and further dAbs being inserted on the other side of the linker.
  • the anchor or guiding dAb maybe TAR1-5 (VK), TARl-27(V ⁇ ), TAR2h-5(VH) or TAR2h-6(V ⁇ ).
  • the invention accordingly provides a method for preparing a chelating multimeric ligand comprising the steps of: (a) providing a vector comprising a nucleic acid sequence encoding a single binding domain specific for a first epitope on a target;
  • the first and second epitopes are adjacent such that a multimeric ligand is capable of binding to both epitopes simultaneously. This provides the ligand with the advantages of increased avidity if binding. Where the epitopes are the same, the increased avidity is obtained by the presence of multiple copies of the epitope on the target, allowing at least two copies to be simultaneously bound in order to obtain the increased avidity effect.
  • the binding domains may be associated by several methods, as well as the use of linkers.
  • the binding domains may comprise cys residues, avidin and streptavidin groups or other means for non-covalent attachment post-synthesis; those combinations which bind to the target efficiently will be isolated.
  • a linker may be present between the first and second binding domains, which are expressed as a single polypeptide from a single vector, which comprises the first binding domain, the linker and a repertoire of second binding domains, for instance as described above.
  • the first and second binding domains associate naturally when bound to antigen; for example, VH and V ⁇ domains, when bound to adjacent epitopes, will naturally associate in a three-way interaction to form a stable dimer.
  • Such associated proteins can be isolated in a target binding assay. An advantage of this procedure is that only binding domains which bind to closely adjacent epitopes, in the correct conformation, will associate and thus be isolated as a result of their increased avidity for the target.
  • At least one epitope binding domain comprises a non-immunoglobulin 'protein scaffold' or 'protein skeleton' as herein defined.
  • Suitable non-immunoglobulin protein scaffolds include but are not limited to any of those selected from the group consisting of: SpA, fibronectin, GroEL and other chaperones, lipocallin, CCTLA4 and affibodies, as set forth above.
  • a protein skeleton according to the invention is an immunoglobulin skeleton.
  • the term 'immunoglobulin skeleton' refers to a protein which comprises at least one immunoglobulin fold and which acts as a nucleus for one or more epitope binding domains, as defined herein.
  • Preferred immunoglobulin skeletons as herein defined includes any one or more of those selected from the following: an immunoglobulin molecule comprising at least (i) the CL (kappa or lambda subclass) domain of an antibody; or (ii) the CHI domain of an antibody heavy chain; an immunoglobulin molecule comprising the CHI and CH2 domains of an antibody heavy chain; an immunoglobulin molecule comprising the CHI, CH2 and CH3 domains of an antibody heavy chain; or any of the subset (ii) in conjunction with the CL (kappa or lambda subclass) domain of an antibody.
  • a hinge region domain may also be included.
  • Such combinations of domains may, for example, mimic natural antibodies, such as IgG or IgM, or fragments thereof, such as Fv, scFv, Fab or F(ab') 2 molecules. Those skilled in the art will be aware that this list is not intended to be exhaustive.
  • Linking of the skeleton to the epitope binding domains may be achieved at the polypeptide level, that is after expression of the nucleic acid encoding the skeleton and/or the epitope binding domains. Alternatively, the linking step may be performed at the nucleic acid level.
  • Methods of linking a protein skeleton according to the present invention, to the one or more epitope binding domains include the use of protein chemistry and/or molecular biology techniques which will be familiar to those skilled in the art and are described herein.
  • the closed conformation multispecific ligand may comprise a first domain capable of binding a target molecule, and a second domain capable of binding a molecule or group which extends the half-life of the ligand.
  • the molecule or group may be a bulky agent, such as HSA or a cell matrix protein.
  • the phrase "molecule or group which extends the half-life of a ligand" refers to a molecule or chemical group which, when bound by a dual-specific ligand as described herein increases the in vivo half-life of such dual specific ligand when admimstered to an animal, relative to a ligand that does not bind that molecule or group. Examples of molecules or groups that extend the half-life of a ligand are described hereinbelow.
  • the closed conformation multispecific ligand may be capable of binding the target molecule only on displacement of the half-life enhancing molecule or group.
  • a closed conformation multispecific ligand is maintained in circulation in the bloodstream of a subject by a bulky molecule such as HSA.
  • a target molecule When a target molecule is encountered, competition between the binding domains of the closed conformation multispecific ligand results in displacement of the HSA and binding of the target.
  • Ligands according to any aspect of the present invention, as well as dAb monomers useful in constructing such ligands, may advantageously dissociate from their cognate target(s) with a K d of 300nM to 5pM (ie, 3 x 10 "7 to 5 x 10 "12 M), preferably 50nM to20 ⁇ M, or 5nM to 200pM or InM to lOOpM, 1 x 10 "7 M or less, 1 x 10 "8 M or less, 1 x 10 "9 M or less, 1 x 10 "10 M or less, 1 x 10 "11 M or less; and/or a K o ff rate constant of 5 x 10 "1 to 1 x 10 "7 S “1 , preferably 1 x 10 "2 to 1 x 10 "6 S “1 , or 5 x 10 "3 to 1 x 10 "5 S '1 , or 5 x 10 "1 S “1 or less, or 1 x 10 "2 S “1 or less, or
  • the invention provides an anti-TNF ⁇ dAb monomer (or dual specific ligand comprising such a dAb), homodimer, heterodimer or homotrimer ligand, wherein each dAb binds TNF ⁇ .
  • the ligand binds to TNF ⁇ with a K d of 300nM to 5 ⁇ M (ie, 3 x 10 -7 to 5 x 10 "12 M), preferably 50nM to 20pM, more preferably 5nM to 200pM and most preferably InM to lOOpM; expressed in an alternative manner, the Kd is 1 x 10 "7 M or less, preferably 1 x 10 "8 M or less, more preferably 1 x 10 "9 M or less, advantageously 1 x 10 "10 M or less and most preferably 1 x 10 "11 M or less; and/or a Koff rate constant of 5 x
  • the ligand neutralises TNF ⁇ in a standard L929 assay with an ND50 of 500nM to 50pM, preferably or lOOnM to 50pM, advantageously lOnM to lOOpM, more preferably InM to lOOpM; for example 50nM or less, preferably 5nM or less, advantageously 500pM or less, more preferably 200pM or less and most preferably lOOpM or less.
  • an ND50 500nM to 50pM, preferably or lOOnM to 50pM, advantageously lOnM to lOOpM, more preferably InM to lOOpM; for example 50nM or less, preferably 5nM or less, advantageously 500pM or less, more preferably 200pM or less and most preferably lOOpM or less.
  • the ligand inhibits binding of TNF alpha to TNF alpha Receptor I (p55 receptor) with an IC50 of 500nM to 50pM, preferably lOOnM to 50pM, more preferably lOnM to lOOpM, advantageously InM to lOOpM; for example 50nM or less, preferably 5nM or less, more preferably 500pM or less, advantageously 200pM or less, and most preferably lOOpM or less.
  • the TNF ⁇ is Human TNF ⁇ .
  • the invention provides a an anti-TNF Receptor I dAb monomer, or dual specific ligand comprising such a dAb, that binds to TNF Receptor I with a Kd of 300nM to 5pM (ie, 3 x 10 "7 to 5 x 10 "12 M), preferably 50nM to20pM, more preferably 5nM to 200pM and most preferably InM to lOOpM, for example 1 x 10 "7 M or less, preferably 1 x 10 "8 M or less, more preferably 1 x 10 "9 M or less, advantageously 1 x 10 "10 M or less and most preferably 1 x 10 "11 M or less; and/or a K o f rate constant of 5 x 10 "1 to 1 x 10 "7 S “ ⁇ preferably 1 x 10 "2 to 1 x 10 "6 S “1 , more preferably 5 x 10 "3 to 1 x 10 ⁇ 5 S “1 , for example 5 x 10 "1
  • the dAb monomeror ligand neutralises TNF ⁇ in a standard assay (eg, the L929 or HeLa assays described herein) with an ND50 of 500nM to 50pM, preferably lOOnM to 50pM, more preferably lOnM to lOOpM, advantageously InM to lOOpM; for example 50nM or less, preferably 5nM or less, more preferably 500pM or less, advantageously 200pM or less, and most preferably lOOpM or less.
  • a standard assay eg, the L929 or HeLa assays described herein
  • an ND50 500nM to 50pM, preferably lOOnM to 50pM, more preferably lOnM to lOOpM, advantageously InM to lOOpM; for example 50nM or less, preferably 5nM or less, more preferably 500pM or less, advantageously 200pM or less, and most preferably lOOpM or
  • the dAb monomer or ligand inhibits binding of TNF alpha to TNF alpha Receptor I (p55 receptor) with an IC50 of 500nM to 50pM, preferably lOOnM to 50pM, more preferably lOnM to lOOpM, advantageously InM to lOOpM; for example 50nM or less, preferably 5nM or less, more preferably 500pM or less, advantageously 200pM or less, and most preferably lOOpM or less.
  • the TNF Receptor I target is Human TNF ⁇ .
  • the invention provides a dAb monomer(or dual specific ligand comprising such a dAb) that binds to serum albumin (SA) with a K d of InM to 50 ⁇ M (ie, x 10 "9 to 5 x 10 "4 ), preferably lOOnM to lO ⁇ M.
  • SA serum albumin
  • the affinity (eg K d and/or K off as measured by surface plasmon resonance, eg using BiaCore) of the second dAb for its target is from 1 to 100000 times (preferably 100 to 100000, more preferably 1000 to 100000, or 10000 to 100000 times) the affinity of the first dAb for SA.
  • the first dAb binds SA with an affinity of approximately lO ⁇ M
  • the second dAb binds its target with an affinity of lOOpM.
  • the serum albumin is human serum albumin (HSA).
  • the first dAb (or a dAb monomer) binds SA (eg, HSA) with a K of approximately 50, preferably 70, and more preferably 100, 150 or 200 nM.
  • SA eg, HSA
  • the invention moreover provides dimers, trimers and polymers of the aforementioned dAb monomers, in accordance with the foregoing aspect of the present invention.
  • Ligands according to the invention can be linked to an antibody Fc region, comprising one or both of CH2 and C H 3 domains, and optionally a hinge region.
  • vectors encoding ligands linked as a single nucleotide sequence to an Fc region may be used to prepare such polypeptides.
  • the present invention provides one or more nucleic acid molecules encoding at least a multispecific ligand as herein defined.
  • the ligand is a closed conformation ligand. In another embodiment, it is an open conformation ligand.
  • the multispecific ligand may be encoded on a single nucleic acid molecule; alternatively, each epitope binding domain may be encoded by a separate nucleic acid molecule. Where the ligand is encoded by a single nucleic acid molecule, the domains may be expressed as a fusion polypeptide, or may be separately expressed and subsequently linked together, for example using chemical linking agents. Ligands expressed from separate nucleic acids will be linked together by appropriate means.
  • the nucleic acid may further encode a signal sequence for export of the polypeptides from a host cell upon expression and may be fused with a surface component of a filamentous bacteriophage particle (or other component of a selection display system) upon expression.
  • Leader sequences which may be used in bacterial expresion and/or phage or phagemid display, include pelB, stll, ompA, phoA, bla and pelA.
  • the present invention provides a vector comprising nucleic acid according to the present invention.
  • the present invention provides a host cell transfected with a vector according to the present invention.
  • Expression from such a vector may be configured to produce, for example on the surface of a bacteriophage particle, epitope binding domains for selection. This allows selection of displayed domains and thus selection of 'multispecific ligands' using the method of the present invention.
  • the epitope binding domains are immunoglobulin variable regions and are selected from single domain V gene repertoires. Generally the repertoire of single antibody domains is displayed on the surface of filamentous bacteriophage. In a preferred embodiment each single antibody domain is selected by binding of a phage repertoire to antigen.
  • the present invention further provides a kit comprising at least a multispecific ligand according to the present invention, which may be an open conformation or closed conformation ligand. Kits according to the invention may be, for example, diagnostic kits, therapeutic kits, kits for the detection of chemical or biological species, and the like.
  • the present invention provides a homogenous immunoassay using a ligand according to the present invention.
  • the present invention provides a composition comprising a closed conformation multispecific ligand, obtainable by a method of the present invention, and a pharmaceutically acceptable carrier, diluent or excipient.
  • the present invention provides a method for the treatment of disease using a 'closed conformation multispecific ligand' or a composition according to the present invention.
  • the disease is cancer or an inflammatory disease, eg rheumatoid arthritis, asthma or Crohn's disease.
  • the present invention provides a method for the diagnosis, including diagnosis of disease using a closed conformation multispecific ligand, or a composition according to the present invention.
  • a closed conformation multispecific ligand may be exploited to displace an agent, which leads to the generation of a signal on displacement.
  • binding of analyte (second antigen) could displace an enzyme (first antigen) bound to the antibody providing the basis for an immunoassay, especially if the enzyme were held to the antibody through its active site.
  • the present invention provides a method for detecting the presence of a target molecule, comprising: (a) providing a closed conformation multispecific ligand bound to an agent, said ligand being specific for the target molecule and the agent, wherein the agent which is bound by the ligand leads to the generation of a detectable signal on displacement from the ligand; (b) exposing the closed conformation multispecific ligand to the target molecule; and (c) detecting the signal generated as a result of the displacement of the agent.
  • the agent is an enzyme, which is inactive when bound by the closed conformation multi-specific ligand.
  • the agent may be any one or more selected from the group consisting of the following: the substrate for an enzyme, and a fluorescent, luminescent or chromogenic molecule which is inactive or quenched when bound by the ligand.
  • sequences similar or homologous are also part of the invention.
  • the sequence identity at the amino acid level can be about 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or higher.
  • the sequence identity can be about 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%) or higher.
  • substantial identity exists when the nucleic acid segments will hybridize under selective hybridization conditions (e.g., very high stringency hybridization conditions), to the complement of the strand.
  • the nucleic acids may be present in whole cells, in a cell lysate, or in a partially purified or substantially pure form.
  • sequence identity or “sequence identity” or “similarity” between two sequences (the terms are used interchangeably herein) are performed as follows.
  • the sequences are aligned for optimal comparison purposes (e.g., gaps can be introduced in one or both of a first and a second amino acid or nucleic acid sequence for optimal alignment and non-homologous sequences can be disregarded for comparison purposes).
  • the length of a reference sequence aligned for comparison purposes is at least 30%, preferably at least 40%, more preferably at least 50%, even more preferably at least 60%, and even more preferably at least 70%, 80%, 90%, 100% of the length of the reference sequence.
  • amino acid residues or nucleotides at corresponding amino acid positions or nucleotide positions are then compared.
  • a position in the first sequence is occupied by the same amino acid residue or nucleotide as the corresponding position in the second sequence, then the molecules are identical at that position (as used herein amino acid or nucleic acid "homology” is equivalent to amino acid or nucleic acid “identity”).
  • the percent identity between the two sequences is a function of the number of identical positions shared by the sequences, taking into account the number of gaps, and the length of each gap, which need to be introduced for optimal alignment of the two sequences.
  • the BLAST algorithm (version 2.0) is employed for sequence alignment, with parameters set to default values.
  • the BLAST algorithm is described in detail at the world wide web site ("www") of the National Center for Biotechnology Information (“.ncbi”) of the National Institutes of Health (“nih”) of the U.S. government (“.gov”), in the "/Blast/” directory, in the "blast_help.html” file.
  • the search parameters are defined as follows, and are advantageously set to the defined default parameters.
  • BLAST Basic Local Alignment Search Tool
  • blastp, blastn, blastx, tblastn, and tblastx these programs ascribe significance to their findings using the statistical methods of Karlin and Altschul, 1990, Proc. Natl. Acad. Sci. USA 87(6):2264-8 (see the "blast_help.html” file, as described above) with a few enhancements.
  • the BLAST programs were tailored for sequence similarity searching, for example to identify homologues to a query sequence. The programs are not generally useful for motif-style searching. For a discussion of basic issues in similarity searching of sequence databases, see Altschul et al. (1994).
  • blastp compares an amino acid query sequence against a protein sequence database
  • blastn compares a nucleotide query sequence against a nucleotide sequence database
  • blastx compares the six-frame conceptual translation products of a nucleotide query sequence (both strands) against a protein sequence database
  • tblastn compares a protein query sequence against a nucleotide sequence database dynamically translated in all six reading frames (both strands).
  • tblastx compares the six-frame translations of a nucleotide query sequence against the six-frame translations of a nucleotide sequence database.
  • BLAST uses the following search parameters:
  • HISTOGRAM Display a histogram of scores for each search; default is yes. (See parameter H in the BLAST Manual).
  • DESCRIPTIONS Restricts the number of short descriptions of matching sequences reported to the number specified; default limit is 100 descriptions. (See parameter V in the manual page). See also EXPECT and CUTOFF. ALIGNMENTS Restricts database sequences to the number specified for which high- scoring segment pairs (HSPs) are reported; the default limit is 50. If more database sequences than this happen to satisfy the statistical significance threshold for reporting (see EXPECT and CUTOFF below), only the matches ascribed the greatest statistical significance are reported. (See parameter B in the BLAST Manual). EXPECT The statistical significance threshold for reporting matches against database sequences; the default value is 10, such that 10 matches are expected to be found merely by chance, according to the stochastic model of Karlin and Altschul (1990).
  • CUTOFF Cutoff score for reporting high-scoring segment pairs.
  • the default value is calculated from the EXPECT value (see above).
  • HSPs are reported for a database sequence only if the statistical significance ascribed to them is at least as high as would be ascribed to a lone HSP having a score equal to the CUTOFF value. Higher CUTOFF values are more stringent, leading to fewer chance matches being reported. (See parameter S in the BLAST Manual). Typically, significance thresholds can be more intuitively managed using EXPECT.
  • MATRIX Specify an alternate scoring matrix for BLASTP, BLASTX, TBLASTN and TBLASTX.
  • the default matrix is BLOSUM62 (Henikoff & Henikoff, 1992, Proc. Natl. Aacad. Sci. USA 89(22):10915-9).
  • the valid alternative choices include: PAM40, PAM120, PAM250 and IDENTITY.
  • No alternate scoring matrices are available for BLASTN; specifying the MATRIX directive in BLASTN requests returns an error response.
  • STRAND Restrict a TBLASTN search to just the top or bottom strand of the database sequences; or restrict a BLASTN, BLASTX or TBLASTX search to just reading frames on the top or bottom strand of the query sequence.
  • FILTER Mask off segments of the query sequence that have low compositional complexity, as determined by the SEG program of Wootton & Federhen (1993) Computers and Chemistry 17:149-163, or segments consisting of short-periodicity internal repeats, as determined by the XNU program of Claverie & States, 1993, Computers and Chemistry 17:191-201, or, for BLASTN, by the DUST program of Tarusov and Lipman (see the world wide web site of the NCBI). Filtering can eliminate statistically significant but biologically uninteresting reports from the blast output (e.g., hits against common acidic-, basic- or proline-rich regions), leaving the more biologically interesting regions of the query sequence available for specific matching against database sequences.
  • Filtering is only applied to the query sequence (or its translation products), not to database sequences. Default filtering is DUST for BLASTN, SEG for other programs.
  • NCBI-gi Causes NCBI gi identifiers to be shown in the output, in addition to the accession and/or locus name.
  • sequence comparisons are conducted using the simple BLAST search algorithm provided at the NCBI world wide web site described above, in the "/BLAST" directory.
  • Figure 1 shows the diversification of V H /HSA at positions H50, H52, H52a, H53,
  • V K H55, H56, H58, H95, H96, H97, H98 (DVT or NNK encoded respectively) which are in the antigen binding site of VH HSA.
  • the sequence of V K. is diversified at positions L50, L53.
  • Figure 2 shows Library 1 : Germline V ⁇ /DVT V H ,
  • Figure 3 shows an alignment of V H chains and V ⁇ chains.
  • Figure 4 shows the characterisation of the binding properties of the K8 antibody, the binding properties of the K8 antibody characterised by monoclonal phage ELISA, the dual specific K8 antibody was found to bind HSA and /3-gal and displayed on the surface of the phage with absorbant signals greater than 1.0. No cross reactivity with other proteins was detected.
  • Figure 5 shows soluble scFv ELISA performed using known concentrations of the
  • K8 antibody fragment A 96-well plate was coated with lOO ⁇ g of HSA, BSA and /3-gal at lO ⁇ g/ml and lOO ⁇ g/ml of Protein A at 1 ⁇ g/ml concentration. 50 ⁇ g of the serial dilutions of the K8 scFv was applied and the bound antibody fragments were detected with Protein L-HRP. ELISA results confirm the dual specific nature of the K8 antibody.
  • Figure 6 shows the binding characteristics of the clone K8V ⁇ /dummy VH analysed using soluble scFv ELISA. Production of the soluble scFv fragments was induced by IPTG as described by Harrison et al, Methods Enzymol. 1996;267:83-109 and the supernatant containing scFv assayed directly.
  • Soluble scFv ELISA is performed as described in example 1 and the bound scFvs were detected with Protein L-HRP.
  • the ELISA results revealed that this clone was still able to bind /3-gal, whereas binding BSA was abolished.
  • Figure 7 shows the sequence of variable domain vectors 1 and 2.
  • Figure 8 is a map of the C H vector used to construct a V H 1/V H 2 multipsecific ligand.
  • Figure 9 is a map of the V ⁇ vector used to construct a V ⁇ l/V ⁇ 2 multispecific ligand.
  • FIG. 10 TNF receptor assay comparing TAR1-5 dimer 4, TAR1-5-19 dimer 4 and TAR1-5-19 monomer.
  • FIG. 11 TNF receptor assay comparing TAR1-5 dimers 1-6. All dimers have been FPLC purified and the results for the optimal dimeric species are shown.
  • FIG. 12 TNF receptor assay of TAR1-5 19 homodimers in different formats: dAb- linker-dAb format with 3U, 5U or 7U linker, Fab format and cysteine hinge linker format.
  • Figure 13 Dummy VH sequence for library 1.
  • Figure 14 Dummy VH sequence for library 2.
  • Figure 15 Dummy VK sequence for library 3.
  • Figure 16 Nucleotide and amino acid sequence of anti MSA dAbs MSA 16 and MSA 26.
  • FIG. 17 Inhibition biacore of MSA 16 and 26.
  • Purified dAbs MSA16 and MSA26 were analysed by inhibition biacore to determine K d . Briefly, the dAbs were tested to determine the concentration of dAb required to achieve 200RUs of response on a biacore CM5 chip coated with a high density of
  • MSA MSA antigen at a range of concentrations around the expected K d was premixed with the dAb and incubated overnight. Binding to the MSA coated biacore chip of dAb in each of the premixes was then measured at a high flow-rate of 30 ⁇ l/minute.
  • FIG. 18 Serum levels of MSA16 following injection. Serum half life of the dAb MSA16 was determined in mouse. MSA16 was dosed as single i.v. injections at approx 1.5mg/kg into CD1 mice. Modelling with a 2 compartment model showed MSA16 had a tl/2 ⁇ of 0.98hr, a tl/2/3 of
  • MSA16 had a considerably lengthened half life compared with HEL4 (an anti-hen egg white lysozyme dAb) which had a tl/2 ⁇ of 0.06hr and a tl/2/3 of 0.34hr.
  • FIG. 19 ELISA (a) and TNF receptor assay (c) showing inhibition of TNF binding with a Fab-like fragment comprising MSA26Ck and TAR1-5-19CH. Addition of MSA with the Fab-like fragment reduces the level of inhibition.
  • the signal in the dual specific well was reduced by more than 50% but the signal in the dAb well was not reduced at all (see figure 19a).
  • the same dual specific protein was also put into the receptor assay with and without MSA and competition by MSA was also shown (see figure 19c). This demonstrates that binding of MSA to the dual specific is competitive with binding to TNF ⁇ !.
  • FIG. 20 TNF receptor assay showing inhibiton of TNF binding with a disulphide bonded heterodimer of TAR1-5-19 dAb and MSA16 dAb.
  • Addition of MSA with the dimer reduces the level of inhibiton in a dose dependant manner.
  • the TNF receptor assay (figure 19 (b)) was conducted in the presence of a constant concentration of heterodimer (18nM) and a dilution series of MSA and HSA.
  • the presence of HSA at a range of concentrations did not cause a reduction in the ability of the dimer to inhibit TNF ⁇ ; .
  • the addition of MSA caused a dose dependant reduction in the ability of the dimer to inhibit TNFo; (figure
  • Complementary Two immunoglobulin domains are "complementary" where they belong to families of structures which form cognate pairs or groups or are derived from such families and retain this feature. For example, a H domain and a V L domain of an antibody are complementary; two V H domains are not complementary, and two domains are not complementary. Complementary domains may be found in other members of the immunoglobulin superfamily, such as the V ⁇ and V ⁇ (or ⁇ and ⁇ ) domains of the T-cell receptor. In the context of the second configuration of the present invention, non-complementary domains do not bind a target molecule cooperatively, but act independently on different target epitopes which may be on the same or different molecules.
  • Domains which are artificial such as domains based on protein scaffolds which do not bind epitopes unless engineered to do so, are non-complementary.
  • two domains based on (for example) an immunoglobulin domain and a fibronectin domain are not complementary.
  • Immunoglobulin This refers to a family of polypeptides which retain the immunoglobulin fold characteristic of antibody molecules, which contains two ⁇ sheets and, usually, a conserved disulphide bond.
  • Members of the immunoglobulin superfamily are involved in many aspects of cellular and non-cellular interactions in vivo, including widespread roles in the immune system (for example, antibodies, T-cell receptor molecules and the like), involvement in cell adhesion (for example the ICAM molecules) and intracellular signalling (for example, receptor molecules, such as the PDGF receptor).
  • the present invention is applicable to all immunoglobulin superfamily molecules which possess binding domains.
  • the present invention relates to antibodies.
  • Variable domains according to the invention are combined to form a group of domains; for example, complementary domains may be combined, such as V L domains being combined with V H domains. Non-complementary domains may also be combined. Domains may be combined in a number of ways, involving linkage of the domains by covalent or non-covalent means.
  • Domain A domain is a folded protein structure which retains its tertiary structure independently of the rest of the protein. Generally, domains are responsible for discrete functional properties of proteins, and in many cases may be added, removed or transferred to other proteins without loss of function of the remainder of the protein and/or of the domain.
  • single antibody variable domain is meant a folded polypeptide domain comprising sequences characteristic of antibody variable domains. It therefore includes complete antibody variable domains and modified variable domains, for example in which one or more loops have been replaced by sequences which are not characteristic of antibody variable domains, or antibody variable domains which have been truncated or comprise N- or C-terminal extensions, as well as folded fragments of variable domains which retain at least in part the binding activity and specificity of the full-length domain.
  • Repertoire A collection of diverse variants, for example polypeptide variants which differ in their primary sequence.
  • a library used in the present invention will encompass a repertoire of polypeptides comprising at least 1000 members.
  • library refers to a mixture of heterogeneous polypeptides or nucleic acids.
  • the library is composed of members, each of which have a single polypeptide or nucleic acid sequence. To this extent, library is synonymous with repertoire. Sequence differences between library members are responsible for the diversity present in the library.
  • the library may take the form of a simple mixture of polypeptides or nucleic acids, or may be in the form of orgamsms or cells, for example bacteria, viruses, animal or plant cells and the like, transformed with a library of nucleic acids.
  • each individual organism or cell contains only one or a limited number of library members.
  • a library may take the form of a population of host organisms, each organism containing one or more copies of an expression vector containing a single member of the library in nucleic acid form which can be expressed to produce its corresponding polypeptide member.
  • the population of host organisms has the potential to encode a large repertoire of genetically diverse polypeptide variants.
  • a 'closed conformation multi-specific ligand' describes a multi-specific ligand as herein defined comprising at least two epitope binding domains as herein defined.
  • the term 'closed conformation' means that the epitope binding domains of the ligand are arranged such that epitope binding by one epitope binding domain competes with epitope binding by another epitope binding domain. That is, cognate epitopes may be bound by each epitope binding domain individually but not simultaneosuly.
  • the closed conformation of the ligand can be achieved using methods herein described.
  • Antibody An antibody (for example IgG, IgM, IgA, IgD or IgE) or fragment (such as a Fab , F(ab') , Fv, disulphide linked Fv, scFv, closed conformation multispecific antibody, disulphide-linked scFv, diabody) whether derived from any species naturally producing an antibody, or created by recombinant DNA technology; whether isolated from serum, B-cells, hybridomas, transfectomas, yeast or bacteria).
  • an antibody for example IgG, IgM, IgA, IgD or IgE
  • fragment such as a Fab , F(ab') , Fv, disulphide linked Fv, scFv, closed conformation multispecific antibody, disulphide-linked scFv, diabody
  • Dual-specific ligand A ligand comprising a first immunoglobulin single variable domain and a second immunoglobulin single variable domain as herein defined, wherein the variable regions are capable of binding to two different antigens or two epitopes on the same antigen which are not normally bound by a monospecific immunoglobulin.
  • the two epitopes may be on the same hapten, but are not the same epitope or sufficiently adjacent to be bound by a monospecific ligand.
  • the dual specific ligands according to the invention are composed of variable domains which have different specificities, and do not contain mutually complementary variable domain pairs which have the same specificity.
  • Antigen A molecule that is bound by a ligand according to the present invention.
  • antigens are bound by antibody ligands and are capable of raising an antibody response in vivo. It may be a polypeptide, protein, nucleic acid or other molecule.
  • the dual specific ligands according to the invention are selected for target specificity against a particular antigen.
  • the antibody binding site defined by the variable loops (LI, L2, L3 and HI, H2, H3) is capable of binding to the antigen.
  • Epitope A unit of structure conventionally bound by an immunoglobulin V H /V L pair. Epitopes define the minimum binding site for an antibody, and thus represent the target of specificity of an antibody. In the case of a single domain antibody, an epitope represents the unit of structure bound by a variable domain in isolation.
  • Universal framework A single antibody framework sequence corresponding to the regions of an antibody conserved in sequence as defined by Kabat ("Sequences of Proteins of Immunological Interest", US Department of Health and Human Services) or corresponding to the human germline immunoglobulin repertoire or structure as defined by Chothia and Lesk, (1987) J. Mol. Biol. 196:910-917.
  • the invention provides for the use of a single framework, or a set of such frameworks, which has been found to permit the derivation of virtually any binding specificity though variation in the hypervariable regions alone.
  • Half-life The time taken for the serum concentration of the ligand to reduce by 50%, in vivo, for example due to degradation of the ligand and/or clearance or sequestration of the ligand by natural mechanisms.
  • the ligands of the invention are stabilised in vivo and their half-life increased by binding to molecules which resist degradation and/or clearance or sequestration. Typically, such molecules are naturally occurring proteins which themselves have a long half-life in vivo.
  • the half-life of a ligand is increased if its functional activity persists, in vivo, for a longer period than a similar ligand which is not specific for the half-life increasing molecule.
  • a ligand specific for HSA and a target molecule is compared with the same ligand wherein the specificity for HSA is not present, that it does not bind HSA but binds another molecule. For example, it may bind a second epitope on the target molecule.
  • the half life is increased by 10%, 20%, 30%, 40%, 50% or more. Increases in the range of 2x, 3x, 4x, 5x, lOx, 20x, 30x, 40x, 5 Ox or more of the half life are possible. Alternatively, or in addition, increases in the range of up to 30x, 40x, 50x, 60x, 70x, 80x, 90x, lOOx, 150x of the half life are possible.
  • homologous immunoassay An immunoassay in which analyte is detected without need for a step of separating bound and un-bound reagents.
  • Substantially identical A first amino acid or nucleotide sequence that contains a sufficient number of identical or equivalent (e.g., with a similar side chain, e.g., conserved amino acid substitutions) amino acid residues or nucleotides to a second amino acid or nucleotide sequence such that the first and second amino acid or nucleotide sequences have similar activities.
  • the second antibody has the same binding specificity and has at least 50% of the affinity of the same.
  • the terms "low stringency,” “medium stringency,” “high stringency,” or “very high stringency conditions” describe conditions for nucleic acid hybridization and washing.
  • Guidance for performing hybridization reactions can be found in Current Protocols in Molecular Biology, John Wiley & Sons, N.Y. (1989), 6.3.1-6.3.6, which is inco ⁇ orated herein by reference in its entirety. Aqueous and nonaqueous methods are described in that reference and either can be used.
  • hybridization conditions referred to herein are as follows: (1) low stringency hybridization conditions in 6X sodium chloride/sodium citrate (SSC) at about 45°C, followed by two washes in 0.2X SSC, 0.1% SDS at least at 50°C (the temperature of the washes can be increased to 55°C for low stringency conditions); (2) medium stringency hybridization conditions in 6X SSC at about 45°C, followed by one or more washes in 0.2X SSC, 0.1% SDS at 60°C; (3) high stringency hybridization conditions in 6X SSC at about 45°C, followed by one or more washes in 0.2X SSC, 0.1% SDS at 65°C; and preferably (4) very high stringency hybridization conditions are 0.5M sodium phosphate, 7% SDS at 65°C, followed by one or more washes at 0.2X SSC, 1% SDS at 65°C. Very high stringency conditions (4) are the preferred conditions and the ones that should be used unless otherwise specified.
  • Dual specific ligands according to the invention may be prepared according to previously established techniques, used in the field of antibody engineering, for the preparation of scFv, "phage" antibodies and other engineered antibody molecules. Techniques for the preparation of antibodies, and in particular bispecific antibodies, are for example described in the following reviews and the references cited therein: Winter & Milstein, (1991) Nature 349:293-299; Plueckthun (1992) Immunological Reviews 130:151-188; Wright et al, (1992) Crti. Rev. Immunol.l2:125-168; Holliger, P. & Winter, G. (1993) Curr. Op. Biotechn.
  • the invention provides for the selection of variable domains against two different antigens or epitopes, and subsequent combination of the variable domains.
  • VH and/or V L libraries may be selected against target antigens or epitopes separately, in which case single domain binding is directly selected for, or together.
  • a preferred method for making a dual specific ligand according to the present invention comprises using a selection system in which a repertoire of variable domains is selected for binding to a first antigen or epitope and a repertoire of variable domains is selected for binding to a second antigen or epitope.
  • the selected variable first and second variable domains are then combined and the dual-specific ligand selected for binding to both first and second antigen or epitope. Closed conformation ligands are selected for binding both first and second antigen or epitope in isolation but not simultaneously.
  • Bacteriophage lambda expression systems may be screened directly as bacteriophage plaques or as colonies of lysogens, both as previously described (Huse et al. (1989) Science, 246: 1275; Caton and Koprowski (1990) Proc. Natl. Acad. Sci. U.S.A., 87; Mullinax et al. (1990) Proc. Natl Acad. Sci. U.S.A., 87: 8095; Persson et al. (1991) Proc. Natl. Acad. Sci. U.S.A., 88: 2432) and are of use in the invention. Whilst such expression systems can be used to screen up to 10 6 different members of a library, they are not really suited to screening of larger numbers (greater than 10 6 members).
  • selection display systems which enable a nucleic acid to be linked to the polypeptide it expresses.
  • a selection display system is a system that permits the selection, by suitable display means, of the individual members of the library by binding the generic and/or target ligands.
  • Selection protocols for isolating desired members of large libraries are known in the art, as typified by phage display techniques.
  • Such systems in which diverse peptide sequences are displayed on the surface of filamentous bacteriophage (Scott and Smith (1990) Science, 249: 386), have proven useful for creating libraries of antibody fragments (and the nucleotide sequences that encoding them) for the in vitro selection and amplification of specific antibody fragments that bind a target antigen (McCafferty et al, WO 92/01047).
  • the nucleotide sequences encoding the VH and V L regions are linked to gene fragments which encode leader signals that direct them to the periplasmic space of E.
  • phagebodies lambda phage capsids
  • An advantage of phage-based display systems is that, because they are biological systems, selected library members can be amplified simply by growing the phage containing the selected library member in bacterial cells. Furthermore, since the nucleotide sequence that encode the polypeptide library member is contained on a phage or phagemid vector, sequencing, expression and subsequent genetic manipulation is relatively straightforward.
  • RNA molecules are selected by alternate rounds of selection against a target ligand and
  • WO95/11922 (Affymax) use the polysomes to display polypeptides for selection.
  • a still further category of techniques involves the selection of repertoires in artificial compartments, which allow the linkage of a gene with its gene product.
  • a selection system in which nucleic acids encoding desirable gene products may be selected in microcapsules formed by water-in-oil emulsions is described in WO99/02671, WO00/40712 and Tawfik & Griffiths (1998) N ⁇ twre Biotechnol 16(7), 652-6.
  • Genetic elements encoding a gene product having a desired activity are compartmentalised into microcapsules and then transcribed and/or translated to produce their respective gene products (R ⁇ A or protein) within the microcapsules.
  • Genetic elements which produce gene product having desired activity are subsequently sorted. This approach selects gene products of interest by detecting the desired activity by a variety of means.
  • Libraries intended for selection may be constructed using techniques known in the art, for example as set forth above, or may be purchased from commercial sources. Libraries which are useful in the present invention are described, for example, in WO99/20749.
  • PCR polymerase chain reaction
  • PCR is performed using template DNA (at least lfg; more usefully, 1-1000 ng) and at least 25 pmol of oligonucleotide primers; it may be advantageous to use a larger amount of primer when the primer pool is heavily heterogeneous, as each sequence is represented by only a small fraction of the molecules of the pool, and amounts become limiting in the later amplification cycles.
  • a typical reaction mixture includes: 2 ⁇ l of DNA, 25 pmol of oligonucleotide primer, 2.5 ⁇ l of 10X PCR buffer 1 (Perkin-Elmer, Foster City, CA), 0.4 ⁇ l of 1.25 ⁇ M dNTP, 0.15 ⁇ l (or 2.5 units) of Taq DNA polymerase (Perkin Elmer, Foster City, CA) and deionized water to a total volume of 25 ⁇ l.
  • Mineral oil is overlaid and the PCR is performed using a programmable thermal cycler. The length and temperature of each step of a PCR cycle, as well as the number of cycles, is adjusted in accordance to the stringency requirements in effect.
  • Annealing temperature and timing are determined both by the efficiency with which a primer is expected to anneal to a template and the degree of mismatch that is to be tolerated; obviously, when nucleic acid molecules are simultaneously amplified and mutagenised, mismatch is required, at least in the first round of synthesis.
  • the ability to optimise the stringency of primer annealing conditions is well within the knowledge of one of moderate skill in the art.
  • An annealing temperature of between 30 °C and 72 °C is used.
  • Initial denaturation of the template molecules normally occurs at between 92°C and 99°C for 4 minutes, followed by 20-40 cycles consisting of denaturation (94-99°C for 15 seconds to 1 minute), annealing (temperature determined as discussed above; 1-2 minutes), and extension (72°C for 1-5 minutes, depending on the length of the amplified product).
  • Final extension is generally for 4 minutes at 72°C, and may be followed by an indefinite (0-24 hour) step at 4°C.
  • Domains useful in the invention may be combined by a variety of methods known in the art, including covalent and non-covalent methods.
  • Preferred methods include the use of polypeptide linkers, as described, for example, in connection with scFv molecules (Bird et al, (1988) Science 242:423-426). Discussion of suitable linkers is provided in Bird et al. Science 242, 423-426; Hudson et al , Journal Immunol Methods 231 (1999) 177-189; Hudson et al, Proc Nat Acad Sci USA 85, 5879- 5883. Linkers are preferably flexible, allowing the two single domains to interact.
  • the linkers used in diabodies, which are less flexible, may also be employed (Holliger et al, (1993) PNAS (USA) 90:6444-6448).
  • the linker employed is not an immunoglobulin hinge region.
  • Variable domains may be combined using methods other than linkers. For example, the use of disulphide bridges, provided through naturally-occurring or engineered cysteine residues, may be exploited to stabilise V H "V H> V L "V L or VH-VL dimers (Reiter et al, (1994) Protein Eng. 7:697-704) or by remodelling the interface between the variable domains to improve the "fit” and thus the stability of interaction (Ridgeway et al, (1996) Protein Eng. 7:617-621; Zhu et al, (1997) Protein Science 6:781-788).
  • variable domains of immunoglobulins and in particular antibody VH domains, may be employed as appropriate.
  • dual specific ligands can be in "closed” conformations in solution.
  • a “closed” configuration is that in which the two domains (for example V H and V L ) are present in associated form, such as that of an associated VH-VL pair which forms an antibody binding site.
  • scFv may be in a closed conformation, depending on the arrangement of the linker used to link the VH and VL domains. If this is sufficiently flexible to allow the domains to associate, or rigidly holds them in the associated position, it is likely that the domains will adopt a closed conformation.
  • V H domain pairs and V L domain pairs may exist in a closed conformation. Generally, this will be a function of close association of the domains, such as by a rigid linker, in the ligand molecule. Ligands in a closed conformation will be unable to bind both the molecule which increases the half-life of the ligand and a second target molecule. Thus, the ligand will typically only bind the second target molecule on dissociation from the molecule which increases the half-life of the ligand.
  • V H /V H , V L /V L or V H /V L dimers without linkers provides for competition between the domains.
  • Ligands according to the invention may moreover be in an open conformation. In such a conformation, the ligands will be able to simultaneously bind both the molecule which increases the half-life of the ligand and the second target molecule.
  • variable domains in an open configuration are (in the case of V H -V L pairs) held far enough apart for the domains not to interact and form an antibody binding site and not to compete for binding to their respective epitopes.
  • V H /VH or Vi/vY dimers the domains are not forced together by rigid linkers. Naturally, such domain pairings will not compete for antigen binding or form an antibody binding site.
  • Fab fragments and whole antibodies will exist primarily in the closed conformation, although it will be appreciated that open and closed dual specific ligands are likely to exist in a variety of equilibria under different circumstances. Binding of the ligand to a target is likely to shift the balance of the equilibrium towards the open configuration.
  • certain ligands according to the invention can exist in two conformations in solution, one of which (the open form) can bind two antigens or epitopes independently, whilst the alternative conformation (the closed form) can only bind one antigen or epitope; antigens or epitopes thus compete for binding to the ligand in this conformation.
  • the open form of the dual specific ligand may thus exist in equilibrium with the closed form in solution, it is envisaged that the equilibrium will favour the closed form; moreover, the open form can be sequestered by target binding into a closed conformation.
  • certain dual specific ligands of the invention are present in an equilibrium between two (open and closed) conformations.
  • Dual specific ligands according to the invention may be modified in order to favour an open or closed conformation.
  • stabilisation of V H -V L interactions with disulphide bonds stabilises the closed conformation.
  • linkers used to join the domains may be constructed such that the open from is favoured; for example, the linkers may sterically hinder the association of the domains, such as by incorporation of large amino acid residues in opportune locations, or the designing of a suitable rigid structure which will keep the domains physically spaced apart.
  • binding of the dual-specific ligand to its specific antigens or epitopes can be tested by methods which will be familiar to those skilled in the art and include ELISA. hi a preferred embodiment of the invention binding is tested using monoclonal phage ELISA.
  • Phage ELISA may be performed according to any suitable procedure: an exemplary protocol is set forth below.
  • phage produced at each round of selection can be screened for binding by ELISA to the selected antigen or epitope, to identify "polyclonal" phage antibodies. Phage from single infected bacterial colonies from these populations can then be screened by ELISA to identify "monoclonal” phage antibodies. It is also desirable to screen soluble antibody fragments for binding to antigen or epitope, and this can also be undertaken by ELISA using reagents, for example, against a C- or N-terminal tag (see for example Winter et al. (1994) Ann. Rev. Immunology 12, 433-55 and references cited therein.
  • the diversity of the selected phage monoclonal antibodies may also be assessed by gel electrophoresis of PCR products (Marks et al. 1991, supra; Nissim et al. 1994 supra), probing (Tomlinson et al, 1992) J. Mol. Biol. 227, 776) or by sequencing of the vector DNA.
  • an antibody is herein defined as an antibody (for example IgG, IgM, IgA, IgA, IgE) or fragment (Fab, Fv, disulphide linked Fv, scFv, diabody) which comprises at least one heavy and a light chain variable domain, at least two heavy chain variable domains or at least two light chain variable domains. It may be at least partly derived from any species naturally producing an antibody, or created by recombinant DNA technology; whether isolated from serum, B-cells, hybridomas, transfectomas, yeast or bacteria).
  • the dual-specific ligand comprises at least one single heavy chain variable domain of an antibody and one single light chain variable domain of an antibody, or two single heavy or light chain variable domains.
  • the ligand may comprise a V H /V L pair, a pair of VH domains or a pair of V L domains.
  • the first and the second variable domains of such a ligand may be on the same polypeptide chain. Alternatively they may be on separate polypeptide chains. In the case that they are on the same polypeptide chain they may be linked by a linker, which is preferentially a peptide sequence, as described above.
  • the first and second variable domains may be covalently or non-covalently associated.
  • the covalent bonds may be disulphide bonds.
  • variable domains are selected from V-gene repertoires selected for instance using phage display technology as herein described, then these variable domains comprise a universal framework region, such that is they may be recognised by a specific generic ligand as herein defined.
  • the use of universal frameworks, generic ligands and the like is described in WO99/20749.
  • variable domains are preferably located within the structural loops of the variable domains.
  • the polypeptide sequences of either variable domain may be altered by DNA shuffling or by mutation in order to enhance the interaction of each variable domain with its complementary pair.
  • DNA shuffling is known in the art and taught, for example, by Stemmer, 1994, Nature 370: 389-391 and U.S. Patent No. 6,297,053, both of which are incorporated herein by reference.
  • Other methods of mutagenesis are well known to those of skill in the art.
  • the 'dual-specific ligand' is a single chain Fv fragment. In an alternative embodiment of the invention, the 'dual-specific ligand' consists of a Fab format.
  • the present invention provides nucleic acid encoding at least a 'dual- specific ligand' as herein defined.
  • both antigens or epitopes may bind simultaneously to the same antibody molecule.
  • they may compete for binding to the same antibody molecule.
  • both variable domains of a dual specific ligand are able to independently bind their target epitopes.
  • the domains compete the one variable domain is capable of binding its target, but not at the same time as the other variable domain binds its cognate target; or the first variable domain is capable of binding its target, but not at the same time as the second variable domain binds its cognate target.
  • variable regions may be derived from antibodies directed against target antigens or epitopes. Alternatively they may be derived from a repertoire of single antibody domains such as those expressed on the surface of filamentous bacteriophage. Selection may be performed as described below.
  • nucleic acid molecules and vector constructs required for the performance of the present invention may be constructed and manipulated as set forth in standard laboratory manuals, such as Sambrook et al. (1989) Molecular Cloning: A Laboratory Manual, Cold Spring Harbor, USA.
  • nucleic acids useful in the present invention is typically carried out in recombinant vectors.
  • the present invention provides a vector comprising nucleic acid encoding at least a 'dual-specific ligand' as herein defined.
  • vector refers to a discrete element that is used to introduce heterologous DNA into cells for the expression and/or replication thereof.
  • Methods by which to select or construct and, subsequently, use such vectors are well known to one of ordinary skill in the art.
  • Numerous vectors are publicly available, including bacterial plasmids, bacteriophage, artificial chromosomes and episomal vectors. Such vectors may be used for simple cloning and mutagenesis; alternatively gene expression vector is employed.
  • a vector of use according to the invention may be selected to accommodate a polypeptide coding sequence of a desired size, typically from 0.25 kilobase (kb) to 40 kb or more in length
  • a suitable host cell is transformed with the vector after in vitro cloning manipulations.
  • Each vector contains various functional components, which generally include a cloning (or "polylinker") site, an origin of replication and at least one selectable marker gene. If given vector is an expression vector, it additionally possesses one or more of the following: enhancer element, promoter, transcription termination and signal sequences, each positioned in the vicinity of the cloning site, such that they are operatively linked to the gene encoding a ligand according to the invention.
  • Both cloning and expression vectors generally contain nucleic acid sequences that enable the vector to replicate in one or more selected host cells. Typically in cloning vectors, this sequence is one that enables the vector to replicate independently of the host chromosomal DNA and includes origins of replication or autonomously replicating sequences. Such sequences are well known for a variety of bacteria, yeast and viruses.
  • the origin of replication from the plasmid pBR322 is suitable for most Gram-negative bacteria, the 2 micron plasmid origin is suitable for yeast, and various viral origins (e.g.
  • SV 40 adenovirus
  • SV 40 adenovirus
  • the origin of replication is not needed for mammalian expression vectors unless these are used in mammalian cells able to replicate high levels of DNA, such as COS cells.
  • a cloning or expression vector may contain a selection gene also referred to as selectable marker.
  • This gene encodes a protein necessary for the survival or growth of transformed host cells grown in a selective culture medium. Host cells not transformed with the vector containing the selection gene will therefore not survive in the culture medium.
  • Typical selection genes encode proteins that confer resistance to antibiotics and other toxins, e.g. ampicillin, neomycin, methotrexate or tetracycline, complement auxotrophic deficiencies, or supply critical nutrients not available in the growth media.
  • an E. co/z-selectable marker for example, the ⁇ - lactamase gene that confers resistance to the antibiotic ampicillin.
  • E. coli plasmids such as pBR322 or a pUC plasmid such as pUC18 or pUC19.
  • Expression vectors usually contain a promoter that is recognised by the host organism and is operably linked to the coding sequence of interest. Such a promoter may be inducible or constitutive.
  • operably linked refers to a juxtaposition wherein the components described are in a relationship permitting them to function in their intended manner.
  • a control sequence "operably linked" to a coding sequence is ligated in such a way that expression of the coding sequence is achieved under conditions compatible with the control sequences.
  • Promoters suitable for use with prokaryotic hosts include, for example, the ⁇ -lactamase and lactose promoter systems, alkaline phosphatase, the tryptophan (trp) promoter system and hybrid promoters such as the tac promoter. Promoters for use in bacterial systems will also generally contain a Shine-Delgarno sequence operably linked to the coding sequence.
  • the preferred vectors are expression vectors that enables the expression of a nucleotide sequence corresponding to a polypeptide library member.
  • selection with the first and/or second antigen or epitope can be performed by separate propagation and expression of a single clone expressing the polypeptide library member or by use of any selection display system.
  • the preferred selection display system is bacteriophage display.
  • phage or phagemid vectors may be used, eg pITl or pIT2.
  • Leader sequences useful in the invention include pelB, stll, ompA, phoA, bla and pelA.
  • phagemid vectors which have an E. coli.
  • the vector contains a ⁇ -lactamase gene to confer selectivity on the phagemid and a lac promoter upstream of a expression cassette that consists (N to C terminal) of a pelB leader sequence (which directs the expressed polypeptide to the periplasmic space), a multiple cloning site (for cloning the nucleotide version of the library member), optionally, one or more peptide tag (for detection), optionally, one or more TAG stop codon and the phage protein pill.
  • a pelB leader sequence which directs the expressed polypeptide to the periplasmic space
  • a multiple cloning site for cloning the nucleotide version of the library member
  • one or more peptide tag for detection
  • TAG stop codon optionally, one or more TAG stop codon and the phage protein pill.
  • the vector is able to replicate as a plasmid with no expression, produce large quantities of the polypeptide library member only or produce phage, some of which contain at least one copy of the polypeptide-pIII fusion on their surface.
  • Construction of vectors encoding ligands according to the invention employs conventional ligation techniques. Isolated vectors or DNA fragments are cleaved, tailored, and religated in the form desired to generate the required vector. If desired, analysis to confirm that the correct sequences are present in the constructed vector can be performed in a known fashion. Suitable methods for constructing expression vectors, preparing in vitro transcripts, introducing DNA into host cells, and performing analyses for assessing expression and function are known to those skilled in the art.
  • telomere sequence The presence of a gene sequence in a sample is detected, or its amplification and/or expression quantified by conventional methods, such as Southern or Northern analysis, Western blotting, dot blotting of DNA, RNA or protein, in situ hybridisation, immunocytochemistry or sequence analysis of nucleic acid or protein molecules. Those skilled in the art will readily envisage how these methods may be modified, if desired.
  • the two or more non-complementary epitope binding domains are linked so that they are in a closed conformation as herein defined.
  • they may be further attached to a skeleton which may, as a alternative, or on addition to a linker described herein, facilitate the formation and/or maintenance of the closed conformation of the epitope binding sites with respect to one another.
  • Skeletons may be based on immunoglobulin molecules or may be non-immunoglobulin in origin as set forth above.
  • Preferred immunoglobulin skeletons as herein defined includes any one or more of those selected from the following: an immunoglobulin molecule comprising at least (i) the CL (kappa or lambda subclass) domain of an antibody; or (ii) the CHI domain of an antibody heavy chain; an immunoglobulin molecule comprising the CHI and CH2 domains of an antibody heavy chain; an immunoglobulin molecule comprising the CHI, CH2 and CH3 domains of an antibody heavy chain; or any of the subset (ii) in conjunction with the CL (kappa or lambda subclass) domain of an antibody.
  • a hinge region domain may also be included.
  • Such combinations of domains may, for example, mimic natural antibodies, such as IgG or IgM, or fragments thereof, such as Fv, scFv, Fab or F(ab') 2 molecules. Those skilled in the art will be aware that this list is not intended to be exhaustive.
  • Each epitope binding domain comprises a protein scaffold and one or more CDRs which are involved in the specific interaction of the domain with one or more epitopes.
  • an epitope binding domain according to the present invention comprises three CDRs.
  • Suitable protein scaffolds include any of those selected from the group consisting of the following: those based on immunoglobulin domains, those based on fibronectin, those based on affibodies, those based on CTLA4, those based on chaperones such as GroEL, those based on lipocallin and those based on the bacterial Fc receptors SpA and SpD. Those skilled in the art will appreciate that this list is not intended to be exhaustive.
  • the members of the immunoglobulin superfamily all share a similar fold for their polypeptide chain.
  • antibodies are highly diverse in terms of their primary sequence
  • comparison of sequences and crystallographic structures has revealed that, contrary to expectation, five of the six antigen binding loops of antibodies (HI, H2, LI, L2, L3) adopt a limited number of main-chain conformations, or canonical structures (Chothia and Lesk (1987) J. Mol. Biol, 196: 901; Chothia et al. (1989) Nature, 342: 877).
  • Analysis of loop lengths and key residues has therefore enabled prediction of the main- chain conformations of HI, H2, LI, L2 and L3 found in the majority of human antibodies (Chothia et al. (1992) J.
  • H3 region is much more diverse in terms of sequence, length and structure (due to the use of D segments), it also forms a limited number of main-chain conformations for short loop lengths which depend on the length and the presence of particular residues, or types of residue, at key positions in the loop and the antibody framework (Martin et al. (1996) J Mol. Biol, 263: 800; Shirai et al. (1996) FEBS Letters, 399: 1).
  • the dual specific ligands of the present invention are advantageously assembled from libraries of domains, such as libraries of VH domains and/or libraries of V L domains. Moreover, the dual specific ligands of the invention may themselves be provided in the form of libraries.
  • libraries of dual specific ligands and/or domains are designed in which certain loop lengths and key residues have been chosen to ensure that the main-chain conformation of the members is known.
  • these are real conformations of immunoglobulin superfamily molecules found in nature, to minimise the chances that they are non-functional, as discussed above.
  • Germline V gene segments serve as one suitable basic framework for constructing antibody or T-cell receptor libraries; other sequences are also of use. Variations may occur at a low frequency, such that a small number of functional members may possess an altered main-chain conformation, which does not affect its function.
  • Canonical structure theory is also of use to assess the number of different main-chain conformations encoded by ligands, to predict the main-chain conformation based on ligand sequences and to chose residues for diversification which do not affect the canonical structure. It is known that, in the human V ⁇ domain, the LI loop can adopt one of four canonical structures, the L2 loop has a single canonical structure and that 90% of human V ⁇ domains adopt one of four or five canonical structures for the L3 loop (Tomlinson et al. (1995) supra); thus, in the V ⁇ domain alone, different canonical structures can combine to create a range of different main-chain conformations.
  • V ⁇ domain encodes a different range of canonical structures for the LI, L2 and L3 loops and that V ⁇ and V ⁇ domains can pair with any VH domain which can encode several canonical structures for the HI and H2 loops
  • the number of canonical structure combinations observed for these five loops is very large. This implies that the generation of diversity in the main-chain conformation may be essential for the production of a wide range of binding specificities.
  • by constructing an antibody library based on a single known main-chain conformation it has been found, contrary to expectation, that diversity in the main-chain conformation is not required to generate sufficient diversity to target substantially all antigens.
  • the single main-chain conformation need not be a consensus structure - a single naturally occurring conformation can be used as the basis for an entire library.
  • the dual-specific ligands of the invention possess a single known main-chain conformation.
  • the single main-chain conformation that is chosen is preferably commonplace among molecules of the immunoglobulin superfamily type in question.
  • a conformation is commonplace when a significant number of naturally occurring molecules are observed to adopt it.
  • the natural occurrence of the different main-chain conformations for each binding loop of an immunoglobulin domain are considered separately and then a naturally occurring variable domain is chosen which possesses the desired combination of main-chain conformations for the different loops. If none is available, the nearest equivalent may be chosen.
  • the desired combination of main-chain conformations for the different loops is created by selecting germline gene segments which encode the desired main-chain conformations. It is more preferable, that the selected germline gene segments are frequently expressed in nature, and most preferable that they are the most frequently expressed of all natural germline gene segments.
  • the incidence of the different main- chain conformations for each of the six antigen binding loops may be considered separately.
  • HI, H2, LI, L2 and L3 a given conformation that is adopted by between 20% and 100% of the antigen binding loops of naturally occurring molecules is chosen.
  • its observed incidence is above 35% (i.e. between 35% and 100%) and, ideally, above 50% or even above 65%.
  • these segments can therefore be used in combination as a basis to construct a library with the desired single main-chain conformation.
  • the natural occurrence of combinations of main-chain conformations is used as the basis for choosing the single main-chain conformation.
  • the natural occurrence of canonical structure combinations for any two, three, four, five or for all six of the antigen binding loops can be determined.
  • the chosen conformation is commonplace in naturally occurring antibodies and most preferable that it observed most frequently in the natural repertoire.
  • dual specific ligands according to the invention or libraries for use in the invention can be constructed by varying the binding site of the molecule in order to generate a repertoire with structural and/or functional diversity. This means that variants are generated such that they possess sufficient diversity in their structure and/or in their function so that they are capable of providing a range of activities.
  • the desired diversity is typically generated by varying the selected molecule at one or more positions.
  • the positions to be changed can be chosen at random or are preferably selected.
  • the variation can then be achieved either by randomisation, during which the resident amino acid is replaced by any amino acid or analogue thereof, natural or synthetic, producing a very large number of variants or by replacing the resident amino acid with one or more of a defined subset of amino acids, producing a more limited number of variants.
  • H3 region of a human tetanus toxoid-binding Fab has been randomised to create a range of new binding specificities (Barbas et al. (1992) Proc. Natl. Acad. Sci. USA, 89: 4457). Random or semi-random H3 and L3 regions have been appended to germline V gene segments to produce large libraries with unmutated framework regions (Hoogenboom & Winter (1992) J Mol. Biol, 227: 381; Barbas et al. (1992) Proc. Natl. Acad. Sci. USA, 89: 4457; Nissim et al.
  • loop randomisation has the potential to create approximately more than 10 15 structures for H3 alone and a similarly large number of variants for the other five loops, it is not feasible using current transformation technology or even by using cell free systems to produce a library representing all possible combinations.
  • 6 x 10 10 different antibodies which is only a fraction of the potential diversity for a library of this design, were generated (Griffiths et al. (1994) supra).
  • the binding site for the target is most often the antigen binding site.
  • the invention provides libraries of or for the assembly of antibody dual-specific ligands in which only those residues in the antigen binding site are varied. These residues are extremely diverse in the human antibody repertoire and are known to make contacts in high-resolution antibody/antigen complexes. For example, in L2 it is known that positions 50 and 53 are diverse in naturally occurring antibodies and are observed to make contact with the antigen. In contrast, the conventional approach would have been to diversify all the residues in the corresponding Complementarity Determining Region (CDR1) as defined by Kabat et al. (1991, supra), some seven residues compared to the two diversified in the library for use according to the invention. This represents a significant improvement in terms of the functional diversity required to create a range of antigen binding specificities.
  • CDR1 Complementarity Determining Region
  • antibody diversity is the result of two processes: somatic recombination of germline V, D and J gene segments to create a naive primary repertoire (so called germline and junctional diversity) and somatic hypermutation of the resulting rearranged V genes.
  • somatic hypermutation spreads diversity to regions at the periphery of the antigen binding site that are highly conserved in the primary repertoire (see Tomlinson et al. (1996) J. Mol. Biol, 256: 813).
  • This complementarity has probably evolved as an efficient strategy for searching sequence space and, although apparently unique to antibodies, it can easily be applied to other polypeptide repertoires.
  • the residues which are varied are a subset of those that form the binding site for the target. Different (including overlapping) subsets of residues in the target binding site are diversified at different stages during selection, if desired.
  • an initial 'naive' repertoire is created where some, but not all, of the residues in the antigen binding site are diversified.
  • the term "naive” refers to antibody molecules that have no pre-determined target. These molecules resemble those which are encoded by the immunoglobulin genes of an individual who has not undergone immune diversification, as is the case with fetal and newborn individuals, whose immune systems have not yet been challenged by a wide variety of antigenic stimuli.
  • This repertoire is then selected against a range of antigens or epitopes. If required, further diversity can then be introduced outside the region diversified in the initial repertoire. This matured repertoire can be selected for modified function, specificity or affinity.
  • the invention provides two different naive repertoires of binding domains for the construction of dual specific ligands, or a na ⁇ ve library of dual specific ligands, in which some or all of the residues in the antigen binding site are varied.
  • the "primary" library mimics the natural primary repertoire, with diversity restricted to residues at the centre of the antigen binding site that are diverse in the germline V gene segments (germline diversity) or diversified during the recombination process (junctional diversity).
  • residues which are diversified include, but are not limited to, H50, H52, H52a, H53, H55, H56, H58, H95, H96, H97, H98, L50, L53, L91, L92, L93, L94 and L96.
  • diversity is restricted to residues that are diversified during the recombination process (junctional diversity) or are highly somatically mutated).
  • residues which are diversified include, but are not limited to: H31, H33, H35, H95, H96, H97, H98, L30, L31, L32, L34 and L96.
  • diversification of chosen positions is typically achieved at the nucleic acid level, by altering the coding sequence which specifies the sequence of the polypeptide such that a number of possible amino acids (all 20 or a subset thereof) can be incorporated at that position.
  • the most versatile codon is NNK, which encodes all amino acids as well as the TAG stop codon.
  • the NNK codon is preferably used in order to introduce the required diversity.
  • Other codons which achieve the same ends are also of use, including the NNN codon, which leads to the production of the additional stop codons TGA and TAA.
  • a feature of side-chain diversity in the antigen binding site of human antibodies is a pronounced bias which favours certain amino acid residues. If the amino acid composition of the ten most diverse positions in each of the V H , V K and V ⁇ regions are summed, more than 76% of the side-chain diversity comes from only seven different residues, these being, serine (24%), tyrosine (14%), asparagine (11%), glycine (9%), alanine (7%), aspartate (6%) and threonine (6%).
  • This bias towards hydrophilic residues and small residues which can provide main-chain flexibility probably reflects the evolution of surfaces which are predisposed to binding a wide range of antigens or epitopes and may help to explain the required promiscuity of antibodies in the primary repertoire.
  • the distribution of amino acids at the positions to be varied preferably mimics that seen in the antigen binding site of antibodies.
  • Such bias in the substitution of amino acids that permits selection of certain polypeptides (not just antibody polypeptides) against a range of target antigens is easily applied to any polypeptide repertoire.
  • There are various methods for biasing the amino acid distribution at the position to be varied including the use of tri-nucleotide mutagenesis, see WO97/08320), of which the preferred method, due to ease of synthesis, is the use of conventional degenerate codons.
  • the codons (AGT)(AGC)T, (AGT)(AGC)C and (AGT)(AGC)(CT) - that is, DVT, DVC and DVY, respectively using TUPAC nomenclature - are those closest to the desired amino acid profile: they encode 22% serine and 11% tyrosine, asparagine, glycine, alanine, aspartate, threonine and cysteine.
  • libraries are constructed using either the DVT, DVC or DVY codon at each of the diversified positions.
  • the dual specific ligands according to the invention are capable of binding to one or more molecules which can increase the half-life of the ligand in vivo.
  • molecules are polypeptides which occur naturally in vivo and which resist degradation or removal by endogenous mechanisms which remove unwanted material from the organism.
  • the molecule which increases the half-life of the organism may be selected from the following:
  • Proteins from the extracellular matrix for example collagen, laminins, integrins and fibronectin.
  • Collagens are the major proteins of the extracellular matrix.
  • about 15 types of collagen molecules are currently known, found in different parts of the body, eg type I collagen (accounting for 90% of body collagen) found in bone, skin, tendon, ligaments, cornea, internal organs or type II collagen found in cartilage, invertebral disc, notochord, vitreous humour of the eye.
  • Proteins found in blood including:
  • Plasma proteins such as fibrin, c-2 macroglobulin, serum albumin, fibrinogen A, fibrinogen B, serum amyloid protein A, heptaglobin, profilin, ubiquitin, uteroglobulin and /3-2-microglobulin;
  • Enzymes and inhibitors such as plasminogen, lysozyme, cystatin C, alpha- 1-antitrypsin and pancreatic trypsin inhibitor.
  • Plasminogen is the inactive precursor of the trypsin-like serine protease plasmin. It is normally found circulating through the blood stream. When plasminogen becomes activated and is converted to plasmin, it unfolds a potent enzymatic domain that dissolves the fibrinogen fibers that entgangle the blood cells in a blood clot. This is called fibrinolysis.
  • Immune system proteins such as IgE, IgG, IgM.
  • Transport proteins such as retinol binding protein, ⁇ :-l micro globulin.
  • Defensins such as beta-defensin 1, Neutrophil defensins 1,2 and 3.
  • Proteins found at the blood brain barrier or in neural tissues such as melanocortin receptor, myelin, ascorbate transporter.
  • brain capillary endothelial cell receptor transferrin, transferrin receptor, insulin, insulin-like growth factor 1 (IGF 1) receptor, insulin-like growth factor 2 (IGF 2) receptor, insulin receptor.
  • IGF 1 insulin-like growth factor 1
  • IGF 2 insulin-like growth factor 2
  • Proteins localised to the kidney such as polycystin, type IV collagen, organic anion transporter Kl, Heymann's antigen.
  • Proteins localised to the liver for example alcohol dehydrogenase, G250.
  • Proteins localised to the lung such as secretory component (binds IgA).
  • HSP 27 Proteins localised to the Heart, for example HSP 27. This is associated with dilated cardiomyopathy.
  • Proteins localised to the skin for example keratin.
  • Bone specific proteins such as bone morphogenic proteins (BMPs), which are a subset of the transforming growth factor ⁇ superfamily that demonstrate osteogenic activity.
  • BMPs bone morphogenic proteins
  • Examples include BMP -2, -4, -5, -6, -7 (also referred to as osteogenic protein (OP-1) and -8 (OP-2).
  • Tumour specific proteins including human trophoblast antigen, herceptin receptor, oestrogen receptor, cathepsins eg cathepsin B (found in liver and spleen).
  • Disease-specific proteins such as antigens expressed only on activated T-cells: including LAG-3 (lymphocyte activation gene), osteoprotegerin ligand (OPGL) see Nature 402, 304-309; 1999, OX40 (a member of the TNF receptor family, expressed on activated T cells and the only costimulatory T cell molecule known to be specifically up-regulated in human T cell leukaemia virus type-I (HTLV-I)-producing cells.) See J Immunol.
  • LAG-3 lymphocyte activation gene
  • osteoprotegerin ligand OPGL
  • OX40 a member of the TNF receptor family, expressed on activated T cells and the only costimulatory T cell molecule known to be specifically up-regulated in human T cell leukaemia virus type-I (HTLV-I)-producing cells.
  • Metalloproteases associated with arthritis/cancers, including CG6512 Drosophila, human paraplegin, human FtsH, human AFG3L2, murine ftsH; angiogenic growth factors, including acidic fibroblast growth factor (FGF-1), basic fibroblast growth factor (FGF-2), Vascular endothelial growth factor / vascular permeability factor (VEGF/VPF), transforming growth factor-a (TGF a), tumor necrosis factor-alpha (TNF- ⁇ ;), angiogenin, interleukin-3 (IL-3), interleukin-8 (IL-8), platelet- derived endothelial growth factor (PD-ECGF), placental growth factor (PIGF), midkine platelet-derived growth factor-BB (PDGF), fractalkine.
  • FGF-1 acidic fibroblast growth factor
  • FGF-2 basic fibroblast growth factor
  • VEGF/VPF Vascular endothelial growth factor / vascular permeability factor
  • HSPs are normally found intracellularly. When they are found extracellularly, it is an indicator that a cell has died and spilled out its contents. This unprogrammed cell death
  • extracellular HSPs trigger a response from the immune system that will fight infection and disease.
  • a dual specific which binds to extracellular HSP can be localised to a disease site.
  • Brambell receptor also known as FcRB
  • This Fc receptor has two functions, both of which are potentially useful for delivery
  • the functions are (1) The transport of IgG from mother to child across the placenta
  • Ligands according to the invention may designed to be specific for the above targets without requiring any increase in or increasing half life in vivo.
  • ligands according to the invention can be specific for targets selected from the foregoing which are tissue-specific, thereby enabling tissue-specific targeting of the dual specific ligand, or a dAb monomer that binds a tissue-specific therapeutically relevant target, irrespective of any increase in half-life, although this may result.
  • the ligand or dAb monomer targets kidney or liver, this may redirect the ligand or dAb monomer to an alternative clearance pathway in vivo (for example, the ligand may be directed away from liver clearance to kidney clearance).
  • Multispecific ligands according to the method of the second configuration of the present invention may be employed in in vivo therapeutic and prophylactic applications, in vitro and in vivo diagnostic applications, in vitro assay and reagent applications, and the like.
  • antibody molecules may be used in antibody based assay techniques, such as ELISA techniques, according to methods known to those skilled in the art.
  • the multispecific ligands according to the invention are of use in diagnostic, prophylactic and therapeutic procedures.
  • Multispecific antibodies according to the invention are of use diagnostically in Western analysis and in situ protein detection by standard immunohistochemical procedures; for use in these applications, the ligands may be labelled in accordance with techniques known to the art.
  • antibody polypeptides may be used preparatively in affinity chromatography procedures, when complexed to a chromatographic support, such as a resin. All such techniques are well known to one of skill in the art.
  • Diagnostic uses of the closed conformation multispecific ligands according to the invention include homogenous assays for analytes which exploit the ability of closed conformation multispecific ligands to bind two targets in competition, such that two targets cannot bind simultaneously (a closed conformation), or alternatively their ability to bind two targets simultaneously (an open conformation).
  • a true homogenous immunoassay format has been avidly sought by manufacturers of diagnostics and research assay systems used in drug discovery and development.
  • the main diagnostics markets include human testing in hospitals, doctor's offices and clinics, commercial reference laboratories, blood banks, and the home, non-human diagnostics (for example food testing, water testing, environmental testing, bio-defence, and veterinary testing), and finally research (including drug development; basic research and academic research).
  • an assay possesses fully quantitative read-outs with high sensitivity and a large dynamic range. Sensitivity is an important requirement, as is reducing the amount of sample required. Both of these features are features that a homogenous system offers. This is very important in point of care testing, and in drug development where samples are precious. Heterogenous systems, as currently available in the art, require large quantities of sample and expensive reagents
  • Tests for homogenous assays include cancer testing, where the biggest assay is that for Prostate Specific Antigen, used in screening men for prostate cancer.
  • Other applications include fertility testing, which provides a series of tests for women attempting to conceive including beta-hcg for pregnancy.
  • Tests for infectious diseases including hepatitis, HIV, rubella, and other viruses and microorganisms and sexually transmitted diseases. Tests are used by blood banks, especially tests for HIV, hepatitis A, B, C, non A non B.
  • Therapeutic drug monitoring tests include monitoring levels of prescribed drugs in patients for efficacy and to avoid toxicity, for example digoxin for arrhythmia, and phenobarbital levels in psychotic cases; theophylline for asthma. Diagnostic tests are moreover useful in abused drug testing, such as testing for cocaine, marijuana and the like. Metabolic tests are used for measuring thyroid function, anaemia and other physiological disorders and functions.
  • the homogenous immunoassay format is moreover useful in the manufacture of standard clinical chemistry assays.
  • the inclusion of immunoassays and chemistry assays on the same instrument is highly advantageous in diagnostic testing.
  • Suitable chemical assays include tests for glucose, cholesterol, potassium, and the like.
  • a further major application for homogenous immunoassays is drug discovery and development: high throughput screening includes testing combinatorial chemistry libraries versus targets in ultra high volume. Signal is detected, and positive groups then split into smaller groups, and eventually tested in cells and then animals. Homogenous assays may be used in all these types of test. In drug development, especially animal studies and clinical trials heavy use of immunoassays is made. Homogenous assays greatly accelerate and simplify these procedures.
  • Other Applications include food and beverage testing: testing meat and other foods for E. coli, salmonella, etc; water testing, including testing at water plants for all types of contaminants including E. coli; and veterinary testing.
  • the invention provides a binding assay comprising a detectable agent which is bound to a closed conformation multispecific ligand according to the invention, and whose detectable properties are altered by the binding of an analyte to said closed conformation multispecific ligand.
  • a binding assay comprising a detectable agent which is bound to a closed conformation multispecific ligand according to the invention, and whose detectable properties are altered by the binding of an analyte to said closed conformation multispecific ligand.
  • Such an assay may be configured in several different ways, each exploiting the above properties of closed conformation multispecific ligands.
  • the assay relies on the direct or indirect displacement of an agent by the analyte, resulting in a change in the detectable properties of the agent.
  • the agent is an enzyme which is capable of catalysing a reaction which has a detectable end-point
  • said enzyme can be bound by the ligand such as to obstruct its active site, thereby inactivating the enzyme.
  • the analyte which is also bound by the closed conformation multispecific ligand, displaces the enzyme, rendering it active through freeing of the active site.
  • the enzyme is then able to react with a substrate, to give rise to a detectable event.
  • the ligand may bind the enzyme outside of the active site, influencing the conformation of the enzyme and thus altering its activity.
  • the structure of the active site may be constrained by the binding of the ligand, or the binding of cofactors necessary for activity may be prevented.
  • the closed conformation multispecific ligand/enzyme complex may be provided on a test strip; the substrate may be provided in a different region of the test strip, and a solvent containing the analyte allowed to migrate through the ligand/enzyme complex, displacing the enzyme, and carrying it to the substrate region to produce a signal.
  • the ligand/enzyme complex may be provided on a test stick or other solid phase, and dipped into an analyte/substrate solution, releasing enzyme into the solution in response to the presence of analyte.
  • the assay is quantitative, with the strength of the signal generated in a given time being dependent on the concentration of analyte in the solution.
  • the closed conformation multispecific ligand may be configured to bind an enzyme in an allosteric site, thereby activating the enzyme.
  • the enzyme is active in the absence of analyte. Addition of the analyte displaces the enzyme and removes allosteric activation, thus inactivating the enzyme.
  • activation or inactivation of the enzyme refers to an increase or decrease in the activity of the enzyme, measured as the ability of the enzyme to catalyse a signal-generating reaction.
  • the enzyme may catalyse the conversion of an undetectable substrate to a detectable form thereof.
  • horseradish peroxidase is widely used in the art together with chromogenic or chemiluminescent substrates, which are available commercially.
  • the level of increase or decrease of the activity of the enzyme may between 10% and 100%, such as 20%, 30%, 40%, 50%, 60%, 70%, 80% or 90%; in the case of an increase in activity, the increase may be more than 100%, i.e. 200%, 300%, 500%) or more, or may not be measurable as a percentage if the baseline activity of the inhibited enzyme is undetectable.
  • the closed conformation multispecific ligand may bind the substrate of an enzyme/substrate pair, rather than the enzyme.
  • the substrate is therefore unavailable to the enzyme until released from the closed conformation multispecific ligand through binding of the analyte.
  • the implementations for this configuration are as for the configurations which bind enzyme.
  • the assay may be configured to bind a fluorescent molecule, such as a fluorescein or another fluorophore, in a conformation such that the fluorescence is quenched on binding to the ligand.
  • a fluorescent molecule such as a fluorescein or another fluorophore
  • binding of the analyte to the ligand will displace the fluorescent molecule, thus producing a signal.
  • fluorescent molecules which are useful in the present invention include luminescent agents, such as luciferin luciferase, and chromogenic agents, including agents commonly used in immunoassays such as HRP.
  • Multi-specificity can allow antibodies to bind to multimeric antigen with great avidity.
  • Multispecific ligands can allow thecross- linking of two antigens, for example in recruiting cytotoxic T-cells to mediate the killing of tumour cell lines.
  • Substantially pure ligands or binding proteins thereof, for example dAb monomers, of at least 90 to 95% homogeneity are preferred for administration to a mammal, and 98 to 99% or more homogeneity is most preferred for pharmaceutical uses, especially when the mammal is a human.
  • the ligands may be used diagnostically or therapeutically (including extracorporeally) or in developing and performing assay procedures, immunofluorescent stainings and the like (Lefkovite and Pernis, (1979 and 1981) Immuno logical Methods, Volumes I and II, Academic Press, NY).
  • the ligands or binding proteins thereof, for example dAb monomers, of the present invention will typically find use in preventing, suppressing or treating inflammatory states, allergic hypersensitivity, cancer, bacterial or viral infection, and autoimmune disorders (which include, but are not limited to, Type I diabetes, asthma, multiple sclerosis, rheumatoid arthritis, systemic lupus erythematosus, Crohn's disease and myasthenia gravis).
  • prevention involves administration of the protective composition prior to the induction of the disease.
  • suppression refers to administration of the composition after an inductive event, but prior to the clinical appearance of the disease.
  • Treatment involves administration of the protective composition after disease symptoms become manifest.
  • EAE in mouse and rat serves as a model for MS in human.
  • the demyelinating disease is induced by administration of myelin basic protein (see Paterson (1986) Textbook of Immunopathology, Mischer et al, eds., Grune and Stratum, New York, pp. 179-213; McFarlin et al. (1973) Science, 179: 478: and Satoh et al. (1987) J. Immunol, 138: 179).
  • the present ligands will be utilised in purified form together with pharmacologically appropriate carriers.
  • these carriers include aqueous or alcoholic/aqueous solutions, emulsions or suspensions, any including saline and/or buffered media.
  • Parenteral vehicles include sodium chloride solution, Ringer's dextrose, dextrose and sodium chloride and lactated Ringer's.
  • Suitable physiologically-acceptable adjuvants, if necessary to keep a polypeptide complex in suspension may be chosen from thickeners such as carboxymethylcellulose, polyvinylpyrrolidone, gelatin and alginates.
  • Intravenous vehicles include fluid and nutrient replenishers and electrolyte replenishers, such as those based on Ringer's dextrose. Preservatives and other additives, such as antimicrobials, antioxidants, chelating agents and inert gases, may also be present (Mack (1982) Remington 's Pharmaceutical Sciences, 16th Edition).
  • the ligands of the present invention may be used as separately administered compositions or in conjunction with other agents. These can include various immunotherapeutic drugs, such as cylcosporine, methotrexate, adriamycin or cisplatinum, and immunotoxins. Pharmaceutical compositions can include "cocktails" of various cytotoxic or other agents in conjunction with the ligands of the present invention, or even combinations of lignds according to the present invention having different specificities, such as ligands selected using different target antigens or epitopes, whether or not they are pooled prior to administration.
  • immunotherapeutic drugs such as cylcosporine, methotrexate, adriamycin or cisplatinum
  • Pharmaceutical compositions can include "cocktails" of various cytotoxic or other agents in conjunction with the ligands of the present invention, or even combinations of lignds according to the present invention having different specificities, such as ligands selected using different target antigens or epitopes,
  • the route of administration of pharmaceutical compositions according to the invention may be any of those commonly known to those of ordinary skill in the art.
  • the selected ligands thereof of the invention can be administered to any patient in accordance with standard techniques.
  • the administration can be by any appropriate mode, including parenterally, intravenously, intramuscularly, intraperitoneally, transdermally, via the pulmonary route, or also, appropriately, by direct infusion with a catheter.
  • the dosage and frequency of administration will depend on the age, sex and condition of the patient, concurrent administration of other drugs, counterindications and other parameters to be taken into account by the clinician.
  • the ligands ofthis invention can be lyophilised for storage and reconstituted in a suitable carrier prior to use. This technique has been shown to be effective with conventional immunoglobulins and art-known lyophilisation and reconstitution techniques can be employed. It will be appreciated by those skilled in the art that lyophilisation and reconstitution can lead to varying degrees of antibody activity loss (e.g. with conventional immunoglobulins, IgM antibodies tend to have greater activity loss than IgG antibodies) and that use levels may have to be adjusted upward to compensate.
  • compositions containing the present ligands or a cocktail thereof can be administered for prophylactic and/or therapeutic treatments, hi certain therapeutic applications, an adequate amount to accomplish at least partial inhibition, suppression, modulation, killing, or some other measurable parameter, of a population of selected cells is defined as a "therapeutically-effective dose”. Amounts needed to achieve this dosage will depend upon the severity of the disease and the general state of the patient's own immune system, but generally range from 0.005 to 5.0 mg of ligand, e.g. antibody, receptor (e.g. a T-cell receptor) or binding protein thereof per kilogram of body weight, with doses of 0.05 to 2.0 mg/kg/dose being more commonly used. For prophylactic applications, compositions containing the present ligands or cocktails thereof may also be administered in similar or slightly lower dosages.
  • Treatment performed using the compositions described herein is considered “effective” if one or more symptoms is reduced (e.g., by at least 10%> or at least one point on a clinical assessment scale), relative to such symptoms present before treatment, or relative to such symptoms in an individual (human or model animal) not treated with such composition. Symptoms will obviously vary depending upon the disease or disorder targeted, but can be measured by an ordinarily skilled clinician or technician.
  • Such symptoms can be measured, for example, by monitoring the level of one or more biochemical indicators of the disease or disorder (e.g., levels of an enzyme or metabolite correlated with the disease, affected cell numbers, etc.), by monitoring physical manifestations (e.g., inflammation, tumor size, etc.), or by an accepted clinical assessment scale, for example, the Expanded Disability Status Scale (for multiple sclerosis), the Irvine Inflammatory Bowel Disease Questionnaire (32 point assessment evaluates quality of life with respect to bowel function, systemic symptoms, social function and emotional status - score ranges from 32 to 224, with higher scores indicating a better quality of life), the Quality of Life Rheumatoid Arthritis Scale, or other accepted clinical assessment scale as known in the field.
  • biochemical indicators of the disease or disorder e.g., levels of an enzyme or metabolite correlated with the disease, affected cell numbers, etc.
  • physical manifestations e.g., inflammation, tumor size, etc.
  • an accepted clinical assessment scale for example, the Expande
  • a sustained (e.g., one day or more, preferably longer) reduction in disease or disorder symptoms by at least 10% or by one or more points on a given clinical scale is indicative of "effective” treatment.
  • prophylaxis performed using a composition as described herein is "effective” if the onset or severity of one or more symptoms is delayed, reduced or abolished relative to such symptoms in a similar individual (human or animal model) not treated with the composition.
  • a composition containing a ligand or cocktail thereof according to the present invention may be utilised in prophylactic and therapeutic settings to aid in the alteration, inactivation, killing or removal of a select target cell population in a mammal.
  • the selected repertoires of polypeptides described herein may be used extracorporeally or in vitro selectively to kill, deplete or otherwise effectively remove a target cell population from a heterogeneous collection of cells.
  • Blood from a mammal may be combined extracorporeally with the ligands, e.g. antibodies, cell-surface receptors or binding proteins thereof whereby the undesired cells are killed or otherwise removed from the blood for return to the mammal in accordance with standard techniques.
  • Dual-specific ligands according to the method of the present invention may be employed in in vivo therapeutic and prophylactic applications, in vivo diagnostic applications and the like.
  • Therapeutic and prophylactic uses of dual-specific ligands prepared according to the invention involve the administration of ligands according to the invention to a recipient mammal, such as a human.
  • Dual specific antibodies according to the invention comprise at least one specificity for a half-life enhancing molecule; one or more further specificities may be directed against target molecules.
  • a dual-specific IgG may be specific for four epitopes, one of which is on a half-life enhancing molecule.
  • Dual- specificity can allow antibodies to bind to multimeric antigen with great avidity.
  • Dual- specific antibodies can allow the cross-linking of two antigens, for example in recruiting cytotoxic T-cells to mediate the killing of tumour cell lines.
  • Substantially pure ligands or binding proteins thereof, such as dAb monomers, of at least 90 to 95%) homogeneity are preferred for administration to a mammal, and 98 to 99%> or more homogeneity is most preferred for pharmaceutical uses, especially when the mammal is a human.
  • the ligands may be used diagnostically or therapeutically (including extracorporeally) or in developing and performing assay procedures, immunofluorescent stainings and the like (Lefkovite and Pernis, (1979 and 1981) Immunological Methods, Volumes I and II, Academic Press, NY).
  • the ligands of the present invention will typically find use in preventing, suppressing or treating inflammatory states, allergic hypersensitivity, cancer, bacterial or viral infection, and autoimmune disorders (which include, but are not limited to, Type I diabetes, multiple sclerosis, rheumatoid arthritis, systemic lupus erythematosus, Crohn's disease and myasthenia gravis).
  • inflammatory states allergic hypersensitivity, cancer, bacterial or viral infection
  • autoimmune disorders which include, but are not limited to, Type I diabetes, multiple sclerosis, rheumatoid arthritis, systemic lupus erythematosus, Crohn's disease and myasthenia gravis.
  • prevention involves administration of the protective composition prior to the induction of the disease.
  • suppression refers to administration of the composition after an inductive event, but prior to the clinical appearance of the disease.
  • Treatment involves administration of the protective composition after disease symptoms become manifest.
  • Animal model systems which can be used to screen the effectiveness of the dual specific ligands in protecting against or treating the disease are available. Methods for the testing of systemic lupus erythematosus (SLE) in susceptible mice are known in the art (Knight et al. (1978) J. Exp. Med., 147: 1653; Reinersten et al. (1978) New Eng. J. Med., 299: 515).
  • MG Myasthenia Gravis
  • SJL/J female mice by inducing the disease with soluble AchR protein from another species
  • Arthritis is induced in a susceptible strain of mice by injection of Type II collagen (Stuart et al. (1984) Ann. Rev. Immunol, 42: 233).
  • a model by which adjuvant arthritis is induced in susceptible rats by injection of mycobacterial heat shock protein has been described (Van Eden et al. (1988) Nature, 331: 171).
  • Thyroiditis is induced in mice by administration of thyroglobulin as described (Maron et al. (1980) J. Exp.
  • IDM Insulin dependent diabetes mellitus
  • EAE in mouse and rat serves as a model for MS in human, hi this model, the demyelinating disease is induced by administration of myelin basic protein (see Paterson (1986) Textbook of Immunopathology, Mischer et al, eds., Grune and Srratton, New York, pp. 179-213; McFarlin et al. (1973) Science, 179: 478: and Satoh et al. (1987) J Immunol, 138: 179).
  • Dual specific ligands according to the invention and dAb monomers able to bind to extracellular targets involved in endocytosis enable dual specific ligands to be endocytosed, enabling another specificity able to bind to an intracellular target to be delivered to an intracellular environment.
  • This strategy requires a dual specific ligand with physical properties that enable it to remain functional inside the cell. Alternatively, if the final destination intracellular compartment is oxidising, a well folding ligand may not need to be disulphide free.
  • the present dual specific ligands will be utilised in purified form together with pharmacologically appropriate carriers.
  • these carriers include aqueous or alcoholic/aqueous solutions, emulsions or suspensions, any including saline and/or buffered media.
  • Parenteral vehicles include sodium chloride solution, Ringer's dextrose, dextrose and sodium chloride and lactated Ringer's.
  • Suitable physiologically-acceptable adjuvants, if necessary to keep a polypeptide complex in suspension may be chosen from thickeners such as carboxymethylcellulose, polyvinylpyrrolidone, gelatin and alginates.
  • Intravenous vehicles include fluid and nutrient replenishers and electrolyte replenishers, such as those based on Ringer's dextrose. Preservatives and other additives, such as antimicrobials, antioxidants, chelating agents and inert gases, may also be present (Mack (1982) Remington's Pharmaceutical Sciences, 16th Edition).
  • the ligands of the present invention may be used as separately administered compositions or in conjunction with other agents. These can include various immunotherapeutic drugs, such as cylcosporine, methotrexate, adriamycin or cisplatinum, and immunotoxins. Pharmaceutical compositions can include "cocktails" of various cytotoxic or other agents in conjunction with the ligands of the present invention.
  • the route of administration of pharmaceutical compositions according to the invention may be any of those commonly known to those of ordinary skill in the art.
  • the ligands of the invention can be administered to any patient in accordance with standard techniques.
  • the administration can be by any appropriate mode, including parenterally, intravenously, intramuscularly, intraperitoneally, transdermally, via the pulmonary route, or also, appropriately, by direct infusion with a catheter.
  • the dosage and frequency of administration will depend on the age, sex and condition of the patient, concurrent administration of other drugs, counterindications and other parameters to be taken into account by the clinician.
  • the ligands of the invention can be lyophilised for storage and reconstituted in a suitable carrier prior to use. This technique has been shown to be effective with conventional immunoglobulins and art-known lyophilisation and reconstitution techniques can be employed. It will be appreciated by those skilled in the art that lyophilisation and reconstitution can lead to varying degrees of antibody activity loss (e.g. with conventional immunoglobulins, IgM antibodies tend to have greater activity loss than IgG antibodies) and that use levels may have to be adjusted upward to compensate.
  • the compositions containing the present ligands or a cocktail thereof can be administered for prophylactic and/or therapeutic treatments.
  • an adequate amount to accomplish at least partial inhibition, suppression, modulation, killing, or some other measurable parameter, of a population of selected cells is defined as a "therapeutically-effective dose”. Amounts needed to achieve this dosage will depend upon the severity of the disease and the general state of the patient's own immune system, but generally range from 0.005 to 5.0 mg of ligand per kilogram of body weight, with doses of 0.05 to 2.0 mg/kg/dose being more commonly used. For prophylactic applications, compositions containing the present ligands or cocktails thereof may also be administered in similar or slightly lower dosages.
  • a composition containing a ligand according to the present invention may be utilised in prophylactic and therapeutic settings to aid in the alteration, inactivation, killing or removal of a select target cell population in a mammal.
  • the selected repertoires of polypeptides described herein may be used extracorporeally or in vitro selectively to kill, deplete or otherwise effectively remove a target cell population from a heterogeneous collection of cells.
  • Blood from a mammal may be combined extracorporeally with the ligands, e.g. antibodies, cell- surface receptors or binding proteins thereof whereby the undesired cells are killed or otherwise removed from the blood for return to the mammal in accordance with standard techniques.
  • TAR1 human TNF ⁇ receptor 1 (p55 receptor) is referred to as TAR2.
  • Example 1 Selection of a dual specific scFv antibody (K8) directed against human serum albumin (HSA) and ⁇ -galactosidase ( ⁇ -gal)
  • This example explains a method for making a dual specific antibody directed against ⁇ - gal and HSA in which a repertoire of V ⁇ variable domains linked to a germline (dummy) VH domain is selected for binding to ⁇ -gal and a repertoire of VH variable domains linked to a germline (dummy) V ⁇ domain is selected for binding to HSA.
  • the selected variable V H HSA and V ⁇ ⁇ -gal domains are then combined and the antibodies selected for binding to ⁇ -gal and HSA.
  • HSA is a half-life increasing protein found in human blood.
  • Library 1 and Library 2 contain a dummy V ⁇ sequence, whereas the sequence of V H is diversified at positions H50, H52, H52a, H53, H55, H56, H58, H95, H96, H97 and H98
  • the libraries are in phagemid pIT2/ScFv format ( Figure 2) and have been preselected for binding to generic ligands, Protein A and Protein L, so that the majority of clones in the unselected libraries are functional.
  • the sizes of the libraries shown above correspond to the sizes after preselection.
  • Library 1 and Library 2 were mixed prior to selections on antigen to yield a single V H /dummy V ⁇ library and Library 3 and Library 4 were mixed to form a single V ⁇ /dummy V H library.
  • trypsin cleaves the pill proteins derived from the helper phage (but not those from the phagemid) and elutes bound scFv-phage fusions by cleavage in the c-myc tag ( Figure 2), thereby providing a further enrichment for phages expressing functional scFvs and a corresponding reduction in background (Kristensen & Winter, Folding & Design 3: 321-328, Jul 9, 1998). Selections were performed using immunorubes coated with either HSA or ⁇ -gal at lOO ⁇ g/ml concentration.
  • DNA preps were made from V H /dummy V ⁇ library selected on HSA and from
  • V ⁇ /dummy V H library selected on ⁇ -gal using the QIAprep Spin Miniprep kit (Qiagen).
  • DNA preps were made from each of the three rounds of selections and then pulled together for each of the antigens. DNA preps were then digested with Sali/Not/ overnight at 37°C. Following gel purification of the fragments, V ⁇ chains from the V ⁇ /dummy V H library selected on ⁇ -gal were ligated in place of a dummy V ⁇ chain of the V H /dummy V ⁇ library selected on HSA creating a library of 3.3 x l ⁇ 9 clones.
  • This library was then either selected on HSA (first round) and ⁇ -gal (second round), HSA/ ⁇ -gal selection, or on ⁇ -gal (first round) and HSA (second round), ⁇ -gal/HSA selection. Selections were performed as described above. In each case after the second round 48 clones were tested for binding to HSA and ⁇ -gal by the monoclonal phage ELISA (as described above) and by ELISA of the soluble scFv fragments. Soluble antibody fragments were produced as described by Harrison et al, (1996), and standard ELISA protocol was followed Hoogenboom et al.
  • the binding properties of the K8 antibody were characterised by the monoclonal phage ELISA.
  • a 96-well plate was coated with lOO ⁇ l of HSA and ⁇ -gal alongside with alkaline phosphatase (APS), bovine serum albumin (BSA), peanut agglutinin, lysozyme and cytochrome c (to check for cross-reactivity) at lO ⁇ g/ml concentration in PBS overnight at 4°C.
  • the phagemid from K8 clone was rescued with KM13 as described by Harrison et al, (1996) and the supernatant (50 ⁇ l) containing phage assayed directly.
  • a soluble scFv ELISA was then performed using known concentrations of the K8 antibody fragment.
  • a 96-well plate was coated with lOO ⁇ l of HSA, BSA and ⁇ -gal at lO ⁇ g/ml and lOO ⁇ l of Protein A at 1 ⁇ g/ml concentration.
  • 50 ⁇ l of the serial dilutions of the K8 scFv was applied and the bound antibody fragments were detected with Protein L- HRP.
  • ELISA results confirmed the dual specific nature of the K8 antibody ( Figure 5).
  • V HSA/BSA by the V H domain of the K8 scFv antibody, the V ⁇ domain was cut out from K8 scFv DNA by SalZ/Not/ digestion and ligated into a Sal//Not/ digested pIT2 vector containing dummy VH chain ( Figures 1 and 2).
  • Binding characteristics of the resulting clone K8V K /dummy V H were analysed by soluble scFv ELISA. Production of the soluble scFv fragments was induced by LPTG as described by Harrison et al, (1996) and the supernatant (50 ⁇ ) containing scFvs assayed directly.
  • Soluble scFv ELISA was performed as described in Example 1 and the bound scFvs were detected with Protein L-HRP. The ELISA results revealed that this clone was still able to bind ⁇ -gal, whereas binding to BSA was abolished ( Figure 6).
  • Example 3 Selection of single VJJ domain antibodies antigens A and B and single V ⁇ domain antibodies directed against antigens C and D.
  • This example describes a method for making single VJJ domain antibodies directed against antigens A and B and single V ⁇ domain antibodies directed against antigens C and D by selecting repertoires of virgin single antibody variable domains for binding to these antigens in the absence of the complementary variable domains. Selections and characterisation of the binding clones is performed as described previously (see Example 5, PCT/GB 02/003014). Four clones are chosen for further work:
  • Examples 1-3 may be used, in a similar manner as that described, to produce dimer molecules comprising combinations of VH domains (i.e., V H - V H ligands) and cominations of V L domains (V L -V L ligands).
  • VHl single domain is excised from variable domain vector 1 ( Figure 7) by Nc ⁇ llXh ⁇ l digestion and ligated into NcollXhol digested variable domain vector 2 ( Figure 7) to create VHl/ variable domain vector 2.
  • VH2 single domain is PCR amplified from variable domain vector 1 using primers to introduce Sail restriction site to the 5' end and Notl restriction site to the 3' end. The PCR product is then digested with SaWNotl and ligated into SaWNotl digested VHl/ variable domain vector 2 to create VH1/VH2/ variable domain vector 2.
  • VK1/VK2/ variable domain vector 2 is created in a similar way.
  • the dual specific nature of the produced VH1/VH2 ScFv and VK1/VK2 ScFv is tested in a soluble ScFv ELISA as described previously (see Example 6, PCT/GB 02/003014). Competition ELISA is performed as described previously (see Example 8, PCT/GB 02/003014).
  • VK1/VK2 ScFv cannot bind to antigen D
  • Example 5 Construction of dual specific VH1/VH2 Fab and VK1/VK2 Fab and analysis of their binding properties.
  • VH1/VH2 Fab VHl single domain is ligated into Nc ⁇ UXliol digested CH vector ( Figure 8) to create VH1/CH and VH2 single domain is ligated into SaWNotl digested CK vector ( Figure 9) to create VH2/CK. Plasmid DNA from VH1/CH and VH2/CK is used to co-transform competent E. coli cells as described previously (see Example 8, PCT/GB02/003014).
  • VH1/CH and VH2/CK plasmids are then induced by L?TG to produce soluble VH1/VH2 Fab as described previously (see Example 8, PCT/GB 02/003014).
  • VK1/VK2 Fab is produced in a similar way.
  • Binding properties of the produced Fabs are tested by competition ELISA as described previously (see Example 8, PCT/GB 02/003014).
  • -VH1/VH2 Fab is able to bind antigens A and B simultaneously -VK1/VK2 Fab is able to bind antigens C and D simultaneously -VH1/VH2 Fab binding is competitive (when bound to antigen A, VH1/VH2 Fab cannot bind to antigen B)
  • VK1/VK2 Fab binding is competitive (when bound to antigen C, VK1/VK2 Fab cannot bind to antigen D)
  • VH and VK homo-dimers are created in a dAb-linker-dAb format using flexible polypeptide linkers.
  • Vectors were created in the dAb linker-dAb format containing glycine-serine linkers of different lengths 3U:(Gly 4 Ser) 3 , 5U:(Gly Ser) 5 , 7U:(Gly 4 Ser) 7.
  • Dimer libraries were created using guiding dAbs upstream of the linker: TAR1-5 (VK), TAR1-27(VK), TAR2-5(VH) or TAR2-6(VK) and a library of corresponding second dAbs after the linker. Using this method, novel dimeric dAbs were selected. The effect of dimerisation on antigen binding was determined by ELISA and BIAcore studies and in cell neutralisation and receptor binding assays. Dimerisation of both TAR1-5 and TAR1- 27 resulted in significant improvement in binding affinity and neutralisation levels.
  • pEDA3U, pEDA5U and pEDA7U vectors were designed to introduce different linker lengths compatible with the dAb-linker-dAb format.
  • sense and anti-sense 73-base pair oligo linkers were annealed using a slow annealing program (95°C-5mins, 80°C-10mins, 70°C-15mins, 56°C-15mins, 42°C until use) in buffer containing O.lMNaCl, lOmM Tris-HCl pH7.4 and cloned using the Xltol and Notl restriction sites.
  • the linkers encompassed 3 (Gly 4 Ser) units and a stuffer region housed between Sail and Notl cloning sites (scheme 1).
  • the stuffer region was designed to include 3 stop codons, a Sacl restriction site and a frame shift mutation to put the region out of frame when no second dAb was present.
  • overlapping oligo-linkers were designed for each vector, annealed and elongated using Klenow. The fragment was then purified and digested using the appropriate enzymes before cloning using X eXhol and Notl restriction sites.
  • VH genes have existing compatible sites, however cloning VK genes required the introduction of suitable restriction sites. This was achieved by using modifying PCR primers (VK-DLIBF: 5' cggccatggcgtcaacggacat; VKXholR: 5' atgtgcgctcgagcgtttgattt 3') in 30 cycles of PCR amplification using a 2:1 mixture of SuperTaq (HTBiotechnology Ltd)and pfu turbo (Stratagene).
  • TARl-5 (VK), TAR1-27(VK), TAR2-5(VH) or TAR2-6(VK) a library of corresponding second dAbs were cloned after the linker.
  • the complimentary dAb libraries were PCR amplified from phage recovered from round 1 selections of either a VK library against Human TNF ⁇ (at approximately 1 x 10 6 diversity after round 1) when TARl-5 or TARl-27 are the guiding dAbs, or a VH or VK library against human p55 TNF receptor (both at approximately 1 x 10 5 diversity after round 1) when TAR2-5 or TAR2-6 respectively are the guiding dAbs.
  • VK libraries PCR amplification was conducted using primers in 30 cycles of PCR amplification using a 2:1 mixture of SuperTaq and pfu turbo.
  • VH libraries were PCR amplified using primers in order to introduce a Sail restriction site at the 5' end of the gene.
  • the dAb library PCRs were digested with the appropriate restriction enzymes, ligated into the corresponding vectors down stream of the linker, using Sall/Notl restriction sites and electroporated into freshly prepared competent TGI cells.
  • the first round selections were carried out in immunotubes using human TNF ⁇ coated at 1 ⁇ g/ml or 20 ⁇ g/ml with 20 washes in PBS 0.1%Tween.
  • TGI cells are infected with the eluted phage and the titres are determined (eg, Marks et al J Mol Biol. 1991 Dec 5;222(3):581-97, Richmann et al Biochemistry. 1993 Aug 31;32(34):8848-55).
  • the second round selections were carried out using 3 different methods.
  • TARl-27 selections were carried out as described previously with the following modifications.
  • the first round selections were carried out in immunotubes using human TNF ⁇ coated at 1 ⁇ g/ml or 20 ⁇ g/ml with 20 washes in PBS 0.1%Tween.
  • the second round selections were carried out in immunotubes using 20 washes with overnight incubation followed by a further 20 washes. Single clones from round 2 selections were picked into 96 well plates and crude supernatant preps were made in 2ml 96 well plate format.
  • TARl-27 titres are as follows:
  • TNFRECEPTOR 1 (p55 RECEPTOR; TAR2)
  • TAR2h-5 titres are as follows:
  • Clones were grown in 2xTY with lOO ⁇ g/ml ampicillin and 1% glucose overnight at 37°C. A 1/100 dilution of this culture was inoculated into 2mls of 2xTY with lOO ⁇ g/ml ampicillin and 0.1 %> glucose in 2ml, 96 well plate format and grown at 37°C shaking until OD600 was approximately 0.9. The culture was then induced with ImM IPTG overnight at 30°C. The supernatants were clarified by centrifugation at 4000rpm for 15 mins in a sorval plate centrifuge. The supernatant preps the used for initial screening.
  • Binding activity of dimeric recombinant proteins was compared to monomer by Protein A/L ELISA or by antigen ELISA. Briefly, a 96 well plate is coated with antigen or Protein A L overnight at 4°C. The plate washed with 0.05% Tween-PBS, blocked for 2hrs with 2% Tween-PBS. The sample is added to the plate incubated for 1 hr at room temperature. The plate is washed and incubated with the secondary reagent for lhr at room temperature. The plate is washed and developed with TMB substrate. Protein A/L- HRP or India-HRP was used as a secondary reagent.
  • the antigen concentrations used were 1 ⁇ g/ml in PBS for Human TNF ⁇ and human THF receptor 1. Due to the presence of the guiding dAb in most cases dimers gave a positive ELISA signal therefore off rate determination was examined by BIAcore.
  • Human TNF ⁇ was coupled to a CM5 chip at high density (approximately 10000 RUs). 50 ⁇ l of Human TNF ⁇ (50 ⁇ g/ml) was coupled to the chip at 5 ⁇ l/min in acetate buffer - pH5.5. Regeneration of the chip following analysis using the standard methods is not possible due to the instability of Human TNF ⁇ , therefore after each sample was analysed, the chip was washed for lOmins with buffer.
  • clones supernatants from the round 2 selection were screened by BIAcore. 48 clones were screened from each of the 3U, 5U and 7U libraries obtained using the following selection methods:
  • RI 1 ⁇ g/ml human TNF ⁇ immunotube, R2 1 ⁇ g/ml human TNF ⁇ immunotube, overnight wash.
  • RI 20 ⁇ g/ml human TNFc immunotube, R2 20 ⁇ g/ml human TNF ⁇ immunotube, overnight wash.
  • RI 1 ⁇ g/ml human TNF ⁇ immunotube, R2 33 pmoles biotinylated human TNF ⁇ on beads.
  • human TNF ⁇ immunotube 20 ⁇ g/ml human TNF ⁇ immunotube, R2 33 pmoles biotinylated human TNF ⁇ beads.
  • human p55 TNF receptor was coupled to a CM5 chip at high density (approximately 4000 RUs). 100 ⁇ l of human p55 TNF receptor (10 ⁇ g/ml) was coupled to the chip at 5 ⁇ l/min in acetate buffer - pH5.5. Standard regeneration conditions were examined ( glycine pH2 or pH3) but in each case antigen was removed from the surface of the chip therefore as with TNF ⁇ , therefore after each sample was analysed, the chip was washed for lOmins with buffer.
  • clones supernatants from the round 2 selection were screened. 48 clones were screened from each of the 3U, 5U and 7U libraries, using the following selection methods :
  • RI l ⁇ g/ml human p55 TNF receptor immunotube, R2 l ⁇ g/ml human p55 TNF receptor immunotube, overnight wash.
  • RI lO ⁇ g/ml human p55 TNF receptor immunotube, R2 lO ⁇ g/ml human p55 TNF receptor immunotube, overnight wash.
  • Receptor binding Anti-TNF dAbs were tested for the ability to inhibit the binding of TNF to recombinant TNF receptor 1 (p55). Briefly, Maxisorp plates were incubated overnight with 30mg/ml anti-human Fc mouse monoclonal antibody (Zymed, San Francisco, USA). The wells were washed with phosphate buffered saline (PBS) containing 0.05% Tween-20 and then blocked with 1% BSA in PBS before being incubated with lOOng/ml TNF receptor 1 Fc fusion protein (R&D Systems, Minneapolis, USA). Anti-TNF dAb was mixed with TNF which was added to the washed wells at a final concentration of lOng/ml.
  • PBS phosphate buffered saline
  • TNF binding was detected with 0.2mg/ml biotinylated anti-TNF antibody (HyCult biotechnology, Uben, Netherlands) followed by 1 in 500 dilution of horse radish peroxidase labelled streptavidin (Amersham Biosciences, UK) and then incubation with TMB substrate (KPL, Gaithersburg, USA). The reaction was stopped by the addition of HCl and the absorbance was read at 450nm. Anti-TNF dAb activity lead to a decrease in TNF binding and therefore a decrease in absorbance compared with the TNF only control. L929 Cytotoxicity Assay
  • Anti-TNF dAbs were also tested for the ability to neutralise the cytotoxic activity of TNF on mouse L929 fibroblasts (Evans, T. (2000) Molecular Biotechnology 15, 243-248). Briefly, L929 cells plated in microtitre plates were incubated overnight with anti-TNF dAb, lOOpg/ml TNF and lmg/ml actinomycin D (Sigma, Poole, UK).
  • Anti-TNFRl or anti-TNF alpha dAbs were tested for the ability to neutralise the induction of IL-8 secretion by TNF in HeLa cells (method adapted from that of Akeson, L. et al (1996) Journal of Biological Chemistry 271, 30517-30523, describing the induction of IL-8 by IL-1 in HUVEC; here we look at induction by human TNF alpha and we use HeLa cells instead of the HUVEC cell line). Briefly, HeLa cells plated in microtitre plates were incubated overnight with dAb and 300pg/ml TNF. Post incubation the supernatant was aspirated off the cells and IL-8 concentration measured via a sandwich ELISA (R&D Systems). Anti-TNFRl dAb activity lead to a decrease in IL-8 secretion into the supernatant compared with the TNF only control.
  • the L929 assay is used throughout the following experiments; however, the use of the HeLa IL-8 assay is preferred to measure anti-TNF receptor 1 (p55) ligands; the presence of mouse p55 in the L929 assay poses certain limitations in its use.
  • the TARl-5 dimers that were shown to have good neutralisation properties were reformatted and analysed in the cell and receptor assays.
  • the TARl-5 guiding dab was substituted with the affinity matured clone TARl-5-19.
  • To achieve this TARl-5 was cloned out of the individual dimer pair and substituted with TARl-5-19 that had been amplified by PCR.
  • TARl-5-19 homodimers were also constructed in the 3U, 5U and 7U vectors.
  • the N terminal copy of the gene was amplified by PCR and cloned as described above and the C-terminal gene fragment was cloned using existing Sail and Notl restriction sites.
  • amber stop codon present in dAb2 one of the C-terminal dAbs in the TARl-5 dimer pairs was mutated to a glutamine by site-directed mutagenesis.
  • the dimers containing TARl-5 or TARl-5-19 were re-formatted into Fab expression vectors.
  • dAbs were cloned into expression vectors containing either the CK or CH genes using Sfil and Notl restriction sites and verified by sequence analysis.
  • the CK vector is derived from a pUC based ampicillin resistant vector and the CH vector is derived from a pACYC chloramphenicol resistant vector.
  • the dAb-CH and dAb-CK constructs were co-transformed into HB2151 cells and grown in 2xTY containing 0.1% glucose, lOO ⁇ g/ml ampicillin and lO ⁇ g/ml chloramphenicol.
  • Dimerisation of dAbs via cystine bond formation was examined.
  • a short sequence of amino acids EPKSGDKTHTCPPCP a modified form of the human IgGCl hinge was engineered at the C terminal region on the dAb.
  • An oligo linker encoding for this sequence was synfhesised and annealed, as described previously. The linker was cloned into the pEDA vector containing TARl-5-19 using Xhol and Notl restriction sites. Dimerisation occurs in situ in the periplasm.
  • Fab and cysteine hinge dimers were expressed as periplasmic proteins in HB2152 cells.
  • a 1/100 dilution of an overnight culture was inoculated into 2xTY with 0.1% glucose and the appropriate antibiotics and grown at 30°C shaking until OD600 was approximately 0.9.
  • the culture was then induced with ImM IPTG for 3-4 hours at 25°C.
  • the cells were harvested by centrifugation and the pellet resuspended in periplasmic preparation buffer (30mM Tris-HCl pH8.0, ImM EDTA, 20% sucrose). Following centrifugation the supernatant was retained and the pellet resuspended in 5mM MgSO 4 . The supernatant was harvested again by centrifugation, pooled and purified.
  • FPLC purification was carried out by FPLC analysis on the AKTA Explorer 100 system (Amersham Biosciences Ltd). TARl-5 and TARl-5-19 dimers were fractionated by cation exchange chromatography (1ml Resource S - Amersham Biosciences Ltd) eluted with a 0-1M NaCl gradient in 50mM acetate buffer pH4. Hinge dimers were purified by ion exchange (1ml Resource Q Amersham Biosciences Ltd) eluted with a 0-1M NaCl gradient in 25mMTris HCl pH 8.0.
  • Fabs were purified by size exclusion chromatography using a superose 12 (Amersham Biosciences Ltd ) column run at a flow rate of 0.5ml/min in PBS with 0.05% tween. Following purification samples were concentrated using vivaspin 5K cut off concentrators (Vivascience Ltd).
  • BIAcore analysis was conducted using the 2ml supernatants. BIAcore analysis revealed that the dimer Koff rates were vastly improved compared to monomeric TARl-5. Monomer Koff rate was in the range of 10 _1 M compared with dimer Koff rates which were in the range of 10 "3 - 10 "4 M. 16 clones that appeared to have very slow off rates were selected, these came from the 3U, 5U and 7U libraries and were sequenced. hi addition the supernatants were analysed for the ability to neutralise human TNF ⁇ in the receptor assay.
  • Protein was produced from the periplasm and supernatant, purified with protein L agarose and examined in the cell and receptor assays. The levels of neutralisation were variable (Table 1). The optimal conditions for protein preparation were determined. Protein produced from HB2151 cells as supernatants gave the highest yield (approximately lOmgs/L of culture). The supernatants were incubated with protein L agarose for 2hrs at room temperature or overnight at 4°C. The beads were washed with PBS/NaCl and packed onto an FPLC column using a peristaltic pump. The beads were washed with 10 column volumes of PBS/NaCl and eluted with 0.1M glycine pH2.5. In general, dimeric protein is eluted after the monomer.
  • TARl-5dl-6 dimers were purified by FPLC.
  • Three species were obtained, by FPLC purification and were identified by SDS PAGE. One species corresponds to monomer and the other two species corresponds to dimers of different sizes. The larger of the two species is possibly due to the presence of C terminal tags. These proteins were examined in the receptor assay.
  • Table 1 represents the optimum results obtained from the two dimeric species ( Figure 11)
  • the three second dAbs from the dimer pairs (ie, dAbl, dAb2 and dAb3) were cloned as monomers and examined by ELISA and in the cell and receptor assay. All three dAbs bind specifically to TNF by antigen ELISA and do not cross react with plastic or BSA. As monomers, none of the dAbs neutralise in the cell or receptor assays.
  • TARl-5-19 was substituted for TARl-5 in the 6 lead clones. Analysis of all TARl-5-19 dimers in the cell and receptor assays was conducted using total protein (protein L purified only) unless otherwise stated (Table 2). TARl-5-19d4 and TARl-5-19d3 have the best ND 50 ( ⁇ 5nM) in the cell assay, this is consistent with the receptor assay results and is an improvement over TARl-5-19 monomer (ND 50 ⁇ 30nM). Although purified TARl-5 dimers give variable results in the receptor and cell assays TARl-5-19 dimers were more consistent. Variability was shown when using different elution buffers during the protein purification. Elution using 0.1M Phosphate-citrate buffer pH2.6 or 0.2M Glycine pH2.5 although removing all protein from the protein L agarose in most cases rendered it less functional.
  • TARl-5-19d4 was expressed in the fermenter and purified on cation exchange FPLC to yield a completely pure dimer. As with TARl-5d4 three species were obtained, by FPLC purification corresponding to monomer and two dimer species. This dimer was amino acid sequenced. TARl-5-19 monomer and TARl-5-19d4 were then examined in the receptor assay and the resulting IC50 for monomer was 30nM and for dimer was 8nM. The results of the receptor assay comparing TARl-5-19 monomer, TARl-5-19d4 and TARl-5d4 is shown in figure 10.
  • TARl-5-19 homodimers were made in the 3U, 5U and 7U vectors, expressed and purified on Protein L.
  • the proteins were examined in the cell and receptor assays and the resulting IC 50 s (for receptor assay) and ND 50 s (for cell assay) were determined (table 3, figure 12).
  • TARl-5 and TARl-5-19 dimers were also cloned into Fab format, expressed and purified on protein L agarose. Fabs were assessed in the receptor assays (Table 4). The results showed that for both TARl-5-19 and TARl-5 dimers the neutralisation levels were similar to the original Gly 4 Ser linker dimers from which they were derived.
  • a TARl-5-19 Fab where TARl-5-19 was displayed on both CH and CK was expressed, protein L purified and assessed in the receptor assay. The resulting IC50 was approximately InM.
  • the 12 neutralising clones were expressed as 200ml supernatant preps and purified on protein L. These were assessed by protein L and antigen ELISA, BIAcore and in the receptor assay. Strong positive ELISA signals were obtained in all cases.
  • BIAcore analysis revealed all clones to have fast on and off rates. The off rates were improved compared to monomeric TARl-27, however the off rate of TARl-27 dimers was faster (Koff is approximately in the range of 10 "1 and 10 "2 M) than the TARl-5 dimers examined previously (Koff is approximately in the range of 10 "3 - 10 "4 M). The stability of the purified dimers was questioned and therefore in order to improve stability, the addition on 5%glycerol, 0.5% Triton X100 or 0.5% NP40 (Sigma) was included in the purification of
  • TARl-27 dimers (d2 and dl6). Addition of NP40 or Triton X100TM improved the yield of purified product approximately 2 fold. Both dimers were assessed in the receptor assay.
  • TARl-27d2 gave IC50 of ⁇ 30nM under all purification conditions.
  • TARl-27dl6 showed no neutralisation effect when purified without the use of stabilising agents but gave an IC50 of ⁇ 50nM when purified under stabilising conditions. No further analysis was conducted.
  • 3 x 96 clones were picked from the second round selections encompassing all the libraries and selection conditions. 2ml supernatant preps were made for analysis. Protein A and antigen ELISAs were conducted for each plate. 30 interesting clones were identified as having good off-rates by BIAcore (Koff ranges between 10 "2 - 10 "3 M). The clones were sequenced and 13 unique dimers were identified by sequence analysis.
  • dAb2 (Gly Ser) 3
  • d3 (Gly 4 Ser) 3
  • dAbl is the partner dAb to dimers 1, 5 and 6.
  • dAb3 is the partner dAb to dimer4. None of the partner dAbs neutralise alone.
  • FPLC purification is by cation exchange unless otherwise stated. The optimal dimeric species for each dimer obtained by FPLC was determined in these assays.
  • trimer protocol gives rise to a mixture of monomer, dimer and trimer.
  • the dimer was purified from the supernatant of the culture by capture on Protein L agarose as outlined in the example 8.
  • the mixed monomer/dimer sample was buffer exchanged into 50 mM sodium acetate buffer pH 4.0 using a PD-10 column (Amersham Pharmacia), following the manufacturer's guidelines. The sample was then applied to a ImL Resource S cation exchange column (Amersham Pharmacia), which had been pre- equilibrated with 50 mM sodium acetate pH 4.0. The monomer and dimer were separated using the following salt gradient in 50 mM sodium acetate pH 4.0:
  • TNF receptor assay and cell assay The affinity of the dimer for human TNFo; was determined using the TNF receptor and cell assay. IC50 in the receptor assay was approximately 0.3-0.8 nM; ND50 in the cell assay was approximately 3-8 nM.
  • Nektar Shearwater offer a range of bi-maleimide PEGs [mPEG2-(MAL)2 or mPEG-
  • the dimer has an affinity in the TNF receptor assay of - 1-3 nM.
  • the dimer can also be produced using TMEA (Tris[2- maleimidoethyl] amine) (Pierce Biotechnology) or other bi-functional linkers.
  • a free cysteine is required at the C-terminus of the protein.
  • the cysteine residue once reduced to give the free thiol, can then be used to specifically couple the protein to a trimeric maleimide molecule, for example TMEA (Tris[2- maleimido ethyl] amine) .
  • oligonucleotides were used to specifically PCR TARl-5-19 with a Sail and BamiTL sites for cloning and also to introduce a C-terminal cysteine residue:
  • the PCR reaction (50 ⁇ L volume) was set up as follows: 200 ⁇ M dNTPs, 0.4 ⁇ M of each primer, 5 ⁇ L of lOx wTurbo buffer (Sfratagene), 100 ng of template plasmid (encoding TARl-5-19), l ⁇ L of twTurbo enzyme (Sfratagene) and the volume adjusted to 50 ⁇ L using sterile water.
  • the following PCR conditions were used: initial denaturing step 94 °C for 2 mins, then 25 cycles of 94 °C for 30 sees, 64 °C for 30 sec and 72 °C for 30 sec. A final extension step was also included of 72 °C for 5 mins.
  • the PCR product was purified and digested with Sail and BamHI and ligated into the vector which had also been cut with the same restriction enzymes. Correct clones were verified by DNA sequencing. Expression and purification of TAR1-5-19CYS
  • TAR1-5-19CYS vector was transformed into BL21 (DE3) pLysS chemically competent cells (Novagen) following the manufacturer's protocol.
  • Cells carrying the dAb plasmid were selected for using lOO ⁇ g/mL carbenicillin and 37 ⁇ g/mL chloramphenicol. Cultures were set up in 2L baffled flasks containing 500 mL of terrific broth (Sigma- Aldrich), lOO ⁇ g/mL carbenicillin and 37 ⁇ g/mL chloramphenicol.
  • the cultures were grown at 30 °C at 200rpm to an O.D.600 of 1-1.5 and then induced with ImM IPTG (isopropyl-beta- D-thiogalactopyranoside, from Melford Laboratories).
  • the expression of the dAb was allowed to continue for 12-16 hrs at 30 °C. It was found that most of the dAb was present in the culture media. Therefore, the cells were separated from the media by centrifugation (8,000xg for 30 mins), and the supernatant used to purify the dAb. Per litre of supernatant, 30 mL of Protein L agarose (Affitech) was added and the dAb allowed to batch bind with stirring for 2 hours.
  • the resin was then allowed to settle under gravity for a further hour before the supernatant was siphoned off.
  • the agarose was then packed into a XK 50 column (Amersham Phamacia) and was washed with 10 column volumes of PBS.
  • the bound dAb was eluted with 100 mM glycine pH 2.0 and protein containing fractions were then neutralized by the addition of 1/5 volume of 1 M Tris pH 8.0. Per litre of culture supernatant 20 mg of pure protein was isolated, which contained a 50:50 ratio of monomer to dimer.
  • TMEA concentration of TMEA greater than 3:1 (molar ratio of dAb:TMEA) caused the rapid precipitation and cross-linking of the protein. Also the rate of precipitation and cross- linking was greater as the pH increased. Therefore using 100 ⁇ M reduced TAR1-5- 19CYS, 25 ⁇ M TMEA was added to trimerise the protein and the reaction allowed to proceed at room temperature for two hours. It was found that the addition of additives such as glycerol or ethylene glycol to 20% (v/v), significantly reduced the precipitation of the trimer as the coupling reaction proceeded. After coupling, SDS-PAGE analysis showed the presence of monomer, dimer and trimer in solution.
  • TNF receptor assay TNF receptor assay and cell assay
  • the affinity of the trimer for human TNFc was determined using the TNF receptor and cell assay.
  • IC50 in the receptor assay was 0.3nM;
  • ND50 in the cell assay was in the range of 3 to lOnM (eg, 3nM).
  • TAR1-5-19CYS may also be formatted into a trimer using the following reagents:
  • Nektar (Shearwater) offer a range of multi arm PEGs, which can be chemically modified at the terminal end of the PEG. Therefore using a PEG trimer with a maleimide functional group at the end of each arm would allow the trimerisation of the dAb in a manner similar to that outlined above using TMEA.
  • the PEG may also have the advantage in increasing the solubility of the trimer thus preventing the problem of aggregation.
  • each dAb has a C-terminal cysteine that is linked to a maleimide functional group, the maleimide functional groups being linked to a PEG trimer.
  • an immunoglobulin eg, IgG hinge region or random peptide sequence (eg, selected from a library of random peptide sequences) could be engineered between the dAb and the terminal cysteine residue.
  • an immunoglobulin eg, IgG hinge region or random peptide sequence (eg, selected from a library of random peptide sequences)
  • this would introduce a greater degree of flexibility and distance between the individual monomers, which may improve the binding characteristics to the target, eg a multisubunit target such as human TNF ⁇ .
  • dAbs single domain antibodies directed against human serum albumin (HSA) and mouse serum albumin (MSA).
  • This example explains a method for making a single domain antibody (dAb) directed against serum albumin. Selection of dAbs against both mouse serum albumin (MSA) and human serum albumin (HSA) is described.
  • MSA mouse serum albumin
  • HSA human serum albumin
  • Three human phage display antibody libraries were used in this experiment, each based on a single human framework for V H (see Figure 13: sequence of dummy V H based on V3-23/DP47 and JH4b) or VK (see Figure 15: sequence of dummy VK based on ol2/o2/DPK9 and Jkl) with side chain diversity encoded by NNK codons incorporated in complementarity determining regions (CDR1, CDR2 and CDR3).
  • V H Library 1 (V H ): Diversity at positions: H30, H31, H33, H35, H50, H52, H52a, H53, H55, H56, H58, H95, H97, H98. Library size: 6.2 x 10 9
  • V H Library 2
  • VK Library 3
  • V H and VK libraries have been preselected for binding to generic ligands protein A and protein L respectively so that the majority of clones in the unselected libraries are functional.
  • the sizes of the libraries shown above correspond to the sizes after preselection.
  • phage from each of the six first round selections was panned against (i) the same antigen again (eg 1 st round MSA, 2 nd round MSA) and (ii) against the reciprocal antigen (eg 1 st round MSA, 2 nd round HSA) resulting in a total of twelve 2 nd round selections, hi each case, after the second round of selection 48 clones were tested for binding to HSA and MSA.
  • Soluble dAb fragments were produced as described for scFv fragments by Harrison et al, Methods Enzymol. 1996;267:83-109 and standard ELISA protocol was followed (Hoogenboom et al.
  • ELISA protocol (as above) was followed except that ELISA plate was coated with l ⁇ g/ml protein L (for the VK clones) and l ⁇ g/ml protein A (for the V H clones). Soluble dAb was captured from solution as in the protocol and detection was with biotinylated MSA or HSA and streptavidin HRP. The biotinylated MSA and HSA had been prepared according to the manufacturer's instructions, with the aim of achieving an average of 2 biotins per serum albumin molecule. Twenty four clones were identified that captured biotinylated MSA from solution in the ELISA. Two of these (clones 2 and 38 below) also captured biotinylated HSA. Next, the dAbs were tested for their ability to bind MSA coated on a CM5 biacore chip. Eight clones were found that bound MSA on the biacore.
  • MSA16 and MSA26 Two clones that are highly expressed in E. coli (clones MSA16 and MSA26) were chosen for further study (see example 10). Full nucleotide and amino acid sequences for MSA16 and 26 are given in figure 16.
  • MSA16 and MSA26 were expressed in the periplasm of E. coli and purified using batch absorbtion to protein L-agarose affinity resin (Affitech, Norway) followed by elution with glycine at pH 2.2. The purified dAbs were then analysed by inhibition biacore to determine K d . Briefly, purified MSA16 and MSA26 were tested to determine the concentration of dAb required to achieve 200RUs of response on a biacore CM5 chip coated with a high density of MSA. Once the required concentrations of dAb had been determined, MSA antigen at a range of concentrations around the expected K d was premixed with the dAb and incubated overnight.
  • Binding to the MSA coated biacore chip of dAb in each of the premixes was then measured at a high flow-rate of 30 ⁇ l minute.
  • the resulting curves were used to create Klotz plots, which gave an estimated K d of 200nM for MSA16 and 70nM for MSA 26 ( Figure 17 A & B).
  • MSA16 and MSA26 were cloned into an expression vector with the HA tag (nucleic acid sequence: TATCCTTATGATGTTCCTGATTATGCA and amino acid sequence: YPYDVPDYA) and 2-10 mg quantities were expressed in E. coli and purified from the supernatant with protein L-agarose affinity resin (Affitech, Norway) and eluted with glycine at pH2.2. Serum half life of the dAbs was determined in mouse. MSA26 and MSA16 were dosed as single i.v. injections at approx 1.5mg/kg into CD1 mice.
  • This example describes a method for making V H - VH and V ⁇ -V ⁇ dual specifics as Fab like fragments.
  • dAbs that bind to targets of choice were first selected from dAb libraries similar to those described in example 9.
  • a V H dAb, HEL4, that binds to hen egg lysozyme (Sigma) was isolated and a second V H dAb (TAR2h-5) that binds to TNFo; receptor (R and D systems) was also isolated.
  • the sequences of these are given in the sequence listing.
  • a VK dAb that binds TNF ⁇ (TARl-5-19) was isolated by selection and affinity maturation and the sequence is also set forth in the sequence listing.
  • a second VK dAb (MSA 26) described in example 9 whose sequence is in figure 17B was also used in these experiments.
  • DNA from expression vectors containing the four dAbs described above was digested with enzymes Sail and Notl to excise the DNA coding for the dAb.
  • a band of the expected size (300-400bp) was purified by running the digest on an agarose gel and excising the band, followed by gel purification using the Qiagen gel purification kit (Qiagen, UK).
  • the DNA coding for the dAbs was then inserted into either the C H or CK vectors (Figs 8 and 9) as indicated in the table below.
  • V H CH and VH CK constructs were cotransformed into HB2151 cells. Separately, the VK C H and VK CK constructs were cotransformed into HB2151 cells. Cultures of each of the cotransformed cell lines were grown overnight (in 2xTy containing 5% glucose, lO ⁇ g/ml chloramphenicol and 100 ⁇ g/ml ampicillin to maintain antibiotic selection for both C H and CK plasmids). The overnight cultures were used to inoculate fresh media (2xTy, lO ⁇ g/ml chloramphenicol and lOO ⁇ g/ml ampicillin) and grown to OD 0.7-0.9 before induction by the addition of IPTG to express their C H and CK constructs. Expressed Fab like fragment was then purified from the periplasm by protein A purification (for the contransformed V H C H and V H CK) and MSA affinity resin purification (for the contransformed VK CH and VK CK).
  • V H C H and V H CK dual specific were tested by running the protein on a gel. The gel was blotted and a band the expected size for the Fab fragment could be detected on the Western blot via both the myc tag and the flag tag, indicating that both the V H C H and V H CK parts of the Fab like fragment were present.
  • an ELISA plate was coated overnight at 4°C with 100 ⁇ l per well of hen egg lysozyme (HEL) at 3 mg/ml in sodium bicarbonate buffer.
  • HEL hen egg lysozyme
  • the plate was then blocked (as described in example 1) with 2%> tween PBS followed by incubation with the V H C H /V H CK dual specific Fab like fragment. Detection of binding of the dual specific to the HEL was via the non cognate chain using 9el0 (a monoclonal antibody that binds the myc tag, Roche) and anti mouse IgG-HRP (Amersham Pharmacia Biotech).
  • 9el0 a monoclonal antibody that binds the myc tag, Roche
  • IgG-HRP Amersham Pharmacia Biotech.
  • the signal for the V H C H /V H CK dual specific Fab like fragment was 0.154 compared to a background signal of 0.069 for the V H CK chain expressed alone. This demonstrates that the Fab like fragment has binding specificity for target antigen.
  • the resulting protein was used to probe an ELISA plate coated with 1/xg/ml TNFo: and an ELISA plate coated with lO ⁇ g/ml MSA. As predicted, there was signal above background when detected with protein L-HRP on bot ELISA plates (data not shown). This indicated that the fraction of protein able to bind to MSA (and therefore purified on the MSA affinity column) was also able to bind TNFo: in a subsequent
  • TNFc 1 ⁇ g/ml TNFc was probed with dual specific VK CH and VK CK Fab like fragment and also with a control TNF ⁇ binding dAb at a concentration calculated to give a similar signal on the ELISA. Both the dual specific and control dAb were used to probe the
  • This example describes a method for making a dual specific antibody fragment specific for both mouse serum albumin and TNFo; by chemical coupling via a disulphide bond.
  • MSA16 from example 1
  • TARl-5-19 dAbs were recloned into a pET based vector with a C terminal cysteine and no tags.
  • the two dAbs were expressed at 4-10 mg levels and purified from the supernatant using protein L-agarose affinity resin (Affitiech, Norway).
  • the cysteine tagged dAbs were then reduced with dithiothreitol.
  • the TARl-5- 19 dAb was then coupled with dithiodipyridine to block reformation of disulphide bonds resulting in the formation of PEP 1-5-19 homodimers.
  • the two different dAbs were then mixed at pH 6.5 to promote disulphide bond formation and the generation of TARl-5-19, MSA16 cys bonded heterodimers.
  • This method for producing conjugates of two unlike proteins was originally described by King et al. (King TP, Li Y Kochoumian L Biochemistry. 1978 voll7: 1499-506 Preparation of protein conjugates via intermolecular disulfide bond formation.) Heterodimers were separated from monomeric species by cation exchange. Separation was confirmed by the presence of a band of the expected size on a SDS gel. The resulting heterodimeric species was tested in the TNF receptor assay and found to have an IC50 for neutralising TNF of approximately 18 nM.
  • the receptor assay was repeated with a constant concentration of heterodimer (18nM) and a dilution series of MSA and HSA.
  • HSA at a range of concentrations (up to 2 mg/ml) did not cause a reduction in the ability of the dimer to inhibit TNF ⁇ : .
  • MSA caused a dose dependant reduction in the ability of the dimer to inhibit TNFa (figure 20). This demonstrates that MSA and TNF ⁇ compete for binding to the cys bonded TARl-5-19, MSA16 dimer.
  • Annex 1 polypeptides which enhance half-life in vivo.
  • Alpha- 1 Glycoprotein (Orosomucoid) (AAG)
  • ACT Alpha- 1 Antichyromotrypsin
  • AAT Alpha- 1 Antitrypsin
  • Alpha-2 Macroglobulin A2M
  • Antithrombin III (AT III)
  • Apolipoprotein A-l (Apo A-l) Apoliprotein B (Apo B)
  • Beta-2-microglobulin (B2M)
  • CRP C-Reactive Protein
  • Fibrinogen Fibrinogen (FIB) Fibronectin (FN)
  • Immunoglobulin A (IgA)
  • Immunoglobulin D (IgD) Immunoglobulin E (IgE)
  • Immunoglobulin G (IgG)
  • Immunoglobulin M (IgM)
  • Myoglobin (Myo) Myoglobin
  • PAL Prealbumin (Transthyretin)
  • RBP Retinol-binding protein
  • RF Rheomatoid Factor
  • SAA Serum Amyloid A
  • SAA Soluble Tranferrin Receptor
  • TfR Transferrin

Abstract

The invention provides a dual-specific ligand comprising a first immunoglobulin variable domain having a first binding specificity and a complementary or non-complementary immunoglobulin variable domain having a second binding specificity.

Description

Ligand
The present invention relates to dual specific ligands. In particular, the invention provides a method for the preparation of dual-specific ligands comprising a first immunoglobulin single variable domain binding to a first antigen or epitope, and a second immunoglobulin single variable domain binding to a second antigen or epitope. More particularly, the invention relates to dual-specific ligands wherein binding to at least one of the first and second antigens or epitopes acts to increase the half-life of the ligand in vivo. Open and closed conformation ligands comprising more than one binding specificity are described.
Introduction
The antigen binding domain of an antibody comprises two separate regions: a heavy chain variable domain (vH) ∞d a light chain variable domain (V : which can be either K or Vχ). The antigen binding site itself is formed by six polypeptide loops: three from VH domain (HI, H2 and H3) and three from vL domain (LI, L2 and L3). A diverse primary repertoire of V genes that encode the VH and VL domains is produced by the combinatorial rearrangement of gene segments. The VH Sene *s produced by the recombination of three gene segments, VH> D and JH- TΏ. humans, there are approximately 51 functional VH segments (Cook and Tomlinson (1995) Immunol Today, 16: 237), 25 functional D segments (Corbett et al. (1997) J. Mol. Biol, 268: 69) and 6 functional JJJ segments (Ravetch et al. (1981) Cell, 27: 583), depending on the haplotype. The VH segment encodes the region of the polypeptide chain which forms the first and second antigen binding loops of the vH domain (HI and H2), whilst the VH> D an JH segments combine to form the third antigen binding loop of the VH domain (H3). The A Sene is produced by the recombination of only two gene segments, V an ^L- In humans, there are approximately 40 functional Vκ segments (Schable and Zachau (1993) Biol. Chem. Hoppe-Seyler, 374: 1001), 31 functional Vχ segments (Williams et al (1996) J. Mol. Biol, 264: 220; Kawasaki et al. (1997) Genome Res., 7: 250), 5 functional Jκ segments (Hieter et al. (1982) J Biol. Chem., 257: 1516) and 4 functional j segments (Vasicek and Leder (1990) J Exp. Med., 172: 609), depending on the haplotype. The γL segment encodes the region of the polypeptide chain which forms the first and second antigen binding loops of the yL domain (LI and L2), whilst the VL and JL segments combine to form the third antigen binding loop of the V domain (L3). Antibodies selected from this primary repertoire are believed to be sufficiently diverse to bind almost all antigens with at least moderate affinity. High affinity antibodies are produced by "affinity maturation" of the rearranged genes, in which point mutations are generated and selected by the immune system on the basis of improved binding.
Analysis of the structures and sequences of antibodies has shown that five of the six antigen binding loops (HI, H2, LI, L2, L3) possess a limited number of main-chain conformations or canonical structures (Chothia and Lesk (1987) J Mol. Biol, 196: 901; Chothia et al. (1989) Nature, 342: 877). The main-chain conformations are determined by (i) the length of the antigen binding loop, and (ii) particular residues, or types of residue, at certain key position in the antigen binding loop and the antibody framework. Analysis of the loop lengths and key residues has enabled us to the predict the main-chain conformations of HI, H2, LI, L2 and L3 encoded by the majority of human antibody sequences (Chothia et al. (1992) J. Mol. Biol, 227: 799; Tomlinson et al. (1995) EMBO J, 14: 4628; Williams et al. (1996) J. Mol. Biol, 264: 220). Although the H3 region is much more diverse in terms of sequence, length and structure (due to the use of D segments), it also forms a limited number of main-chain conformations for short loop lengths which depend on the length and the presence of particular residues, or types of residue, at key positions in the loop and the antibody framework (Martin et al. (1996) J. Mol. Biol, 263: 800; Shirai et al. (1996) FEBS Letters, 399: 1.
Bispecific antibodies comprising complementary pairs of VH an NL regions are known in the art. These bispecific antibodies must comprise two pairs of VH and VLs, eac VH^VL pair binding to a single antigen or epitope. Methods described involve hybrid hybridomas (Milstein & Cuello AC, Nature 305:537-40), minibodies (Hu et al, (1996) Cancer Res 56:3055-3061;), diabodies (Holliger et al, (1993) Proc. Natl. Acad. Sci. USA 90, 6444- 6448; WO 94/13804), chelating recombinant antibodies (CRAbs; (Neri et al, (1995) j. Mol. Biol. 246, 367-373), biscFv (e.g. Atwell et al, (1996) Mol. Immunol. 33, 1301- 1312), "knobs in holes" stabilised antibodies (Carter et al, (1997) Protein Sci. 6, 781- 788). In each case each antibody species comprises two antigen-binding sites, each fashioned by a complementary pair of VH and V domains. Each antibody is thereby able to bind to two different antigens or epitopes at the same time, with the binding to EACH antigen or epitope mediated by a VH an it complementary VL domain. Each of these techniques presents its particular disadvantages; for instance in the case of hybrid hybridomas, inactive VH^VL Parrs can greatly reduce the fraction of bispecific IgG.
Furthermore, most bispecific approaches rely on the association of the different VHL pairs or the association of VH and V chains to recreate the two different Yu/ L binding sites. It is therefore impossible to control the ratio of binding sites to each antigen or epitope in the assembled molecule and thus many of the assembled molecules will bind to one antigen or epitope but not the other. In some cases it has been possible to engineer the heavy or light chains at the sub-unit interfaces (Carter et al, 1997) in order to improve the number of molecules which have binding sites to both antigens or epitopes but this never results in all molecules having binding to both antigens or epitopes.
There is some evidence that two different antibody binding specificities might be incorporated into the same binding site, but these generally represent two or more specificities that correspond to structurally related antigens or epitopes or to antibodies that are broadly cross-reactive.. For example, cross-reactive antibodies have been described, usually where the two antigens are related in sequence and structure, such as hen egg white lysozyme and turkey lysozyme (McCafferty et al., WO 92/01047) or to free hapten and to hapten conjugated to carrier (Griffiths AD et al. EMBO J 1994 13:14 3245-60). In a further example, WO 02/02773 (Abbott Laboratories) describes antibody molecules with "dual specificity". The antibody molecules referred to are antibodies raised or selected against multiple antigens, such that their specificity spans more than a single antigen. Each complementary VH/VL pair in the antibodies of WO 02/02773 specifies a single binding specificity for two or more structurally related antigens; the VH and VL domains in such complementary pairs do not each possess a separate specificity. The antibodies thus have a broad single specificity which encompasses two antigens, which are structurally related. Furthermore natural auto antibodies have been described that are polyreactive (Casali & Notkins, Ann. Rev. Immunol. 7, 515-531), reacting with at least two (usually more) different antigens or epitopes that are not structurally related. It has also been shown that selections of random peptide repertoires using phage display technology on a monoclonal antibody will identify a range of peptide sequences that fit the antigen binding site. Some of the sequences are highly related, fitting a consensus sequence, whereas others are very different and have been termed mimotopes (Lane & Stephen, Current Opinion in Immunology, 1993, 5, 268-271). It is therefore clear that a natural four-chain antibody, comprising associated and complementary VH and VL domains, has the potential to bind to many different antigens from a large universe of known antigens. It is less clear how to create a binding site to two given antigens in the same antibody, particularly those which are not necessarily structurally related.
Protein engineering methods have been suggested that may have a bearing on this. For example it has also been proposed that a catalytic antibody could be created with a binding activity to a metal ion through one variable domain, and to a hapten (substrate) through contacts with the metal ion and a complementary variable domain (Barbas et al., 1993 Proc. Natl. Acad. Sci USA 90, 6385-6389). However in this case, the binding and catalysis of the substrate (first antigen) is proposed to require the binding of the metal ion (second antigen). Thus the binding to the VH^VL pairing relates to a single but multi- component antigen.
Methods have been described for the creation of bispecific antibodies from camel antibody heavy chain single domains in which binding contacts for one antigen are created in one variable domain, and for a second antigen in a second variable domain. However the variable domains were not complementary. Thus a first heavy chain variable domain is selected against a first antigen, and a second heavy chain variable domain against a second antigen, and then both domains are linked together on the same chain to give a bispecific antibody fragment (Conrath et al., J. Biol. Chem. 270, 27589-27594). However the camel heavy chain single domains are unusual in that they are derived from natural camel antibodies which have no light chains, and indeed the heavy chain single domains are unable to associate with camel light chains to form complementary VH and V pairs.
Single heavy chain variable domains have also been described, derived from natural antibodies which are normally associated with light chains (from monoclonal antibodies or from repertoires of domains; see EP-A-0368684). These heavy chain variable domains have been shown to interact specifically with one or more related antigens but have not been combined with other heavy or light chain variable domains to create a ligand with a specificity for two or more different antigens . Furthermore, these single domains have been shown to have a very short in vivo half-life. Therefore such domains are of limited therapeutic value.
It has been suggested to make bispecific antibody fragments by linking heavy chain variable domains of different specificity together (as described above). The disadvantage with this approach is that isolated antibody variable domains may have a hydrophobic interface that normally makes interactions with the light chain and is exposed to solvent and may be "sticky" allowing the single domain to bind to hydrophobic surfaces. Furthermore, in the absence of a partner light chain the combination of two or more different heavy chain variable domains and their association, possibly via their hydrophobic interfaces, may prevent them from binding to one in not both of the ligands they are able to bind in isolation. Moreover, in this case the heavy chain variable domains would not be associated with complementary light chain variable domains and thus may be less stable and readily unfold (Worn & Pluckthun, 1998 Biochemistry 37, 13120-7).
Summary of the invention
The inventors have described, in their copending international patent application WO 03/002609 as well as copending unpublished UK patent application 0230203.2, dual specific immunoglobulin ligands which comprise immunoglobulin single variable domains which each have different specificities. The domains may act in competition with each other or independently to bind antigens or epitopes on target molecules.
In a first configuration, the present invention provides a further improvement in dual specific ligands as developed by the present inventors, in which one specificity of the ligand is directed towards a protein or polypeptide present in vivo in an organism which can act to increase the half-life of the ligand by binding to it. Accordingly, in a first aspect, there is provided a dual-specific ligand comprising a first immunoglobulin single variable domain having a binding specificity to a first antigen or epitope and a second complementary immunoglobulin single variable domain having a binding activity to a second antigen or epitope, wherein one or both of said antigens or epitopes acts to increase the half-life of the ligand in vivo and wherein said first and second domains lack mutually complementary domains which share the same specificity, provided that said dual specific ligand does not consist of an anti-HSA VH domain and an anti-β galactosidase Vκ domain. Preferably, that neither of the first or second variable domains binds to human serum albumin (HSA).
Antigens or epitopes which increase the half-life of a ligand as described herein are advantageously present on proteins or polypeptides found in an organism in vivo. Examples include extracellular matrix proteins, blood proteins, and proteins present in various tissues in the organism. The proteins act to reduce the rate of ligand clearance from the blood, for example by acting as bulking agents, or by anchoring the ligand to a desired site of action. Examples of antigens/epitopes which increase half-life in vivo are given in Annex 1 below.
Increased half-life is useful in in vivo applications of immunoglobulins, especially antibodies and most especially antibody fragments of small size. Such fragments (Fvs, disulphide bonded Fvs, Fabs, scFvs, dAbs) suffer from rapid clearance from the body; thus, whilst they are able to reach most parts of the body rapidly, and are quick to produce and easier to handle, their in vivo applications have been limited by their only brief persistence in vivo. The invention solves this problem by providing increased half-life of the ligands in vivo and consequently longer persistence times in the body of the functional activity of the ligand.
Methods for pharmacokinetic analysis and determination of ligand half-life will be familiar to those skilled in the art. Details may be found in Kenneth, A et al: Chemical Stability of Pharmaceuticals: A Handbook for Pharmacists and in Peters et al, Pharmacokinetc analysis: A Practical Approach (1996). Reference is also made to "Pharmacokinetics", M Gibaldi & D Perron, published by Marcel Dekker, 2nd Rev. ex edition (1982), which describes pharmacokinetic parameters such as t alpha and t beta half lives and area under the curve (AUC).
Half lives (ilA alpha and XlA beta) and AUC can be determined from a curve of serum concentration of ligand against time. The WinNonlin analysis package (available from Pharsight Corp., Mountain View, CA94040, USA) can be used, for example, to model the curve. In a first phase (the alpha phase) the ligand is undergoing mainly distribution in the patient, with some elimination. A second phase (beta phase) is the terminal phase when the ligand has been distributed and the serum concentration is decreasing as the ligand is cleared from the patient. The t alpha half life is the half life of the first phase and the t beta half life is the half life of the second phase. Thus, advantageously, the present invention provides a ligand or a composition comprising a ligand according to the invention having a tα half-life in the range of 15 minutes or more. In one embodiment, the lower end of the range is 30 minutes, 45 minutes, 1 hour, 2 hours, 3 hours, 4 hours, 5 hours, 6 hours, 7 hours, 10 hours, 11 hours or 12 hours. In addition, or alternatively,a ligand or composition according to the invention will have a tα half life in the range of up to and including 12 hours. In one embodiment, the upper end of the range is 11, 10, 9, 8, 7, 6 or 5 hours. An example of a suitable range is 1 to 6 hours, 2 to 5 hours or 3 to 4 hours.
Advantageously, the present invention provides a ligand or a composition comprising a ligand according to the invention having a tβ half-life in the range of 2.5 hours or more. In one embodiment, the lower end of the range is 3 hours, 4 hours, 5 hours, 6 hours, 7 hours, 10 hours , 11 hours, or 12 hours. In addition, or alternatively, a ligand or composition according to the invention has a tβ half-life in the range of up to and including 21 days. In one embodiment, the upper end of the range is 12 hours, 24 hours, 2 days, 3 days, 5 days, 10 days, 15 days or 20 days. Advantageously a ligand or composition according to the invention will have a tβ half life in the range 12 to 60 hours. In a further embodiment, it will be in the range 12 to 48 hours. In a further embodiment still, it will be in the range 12 to 26 hours.
In addition, or alternatively to the above criteria, the present invention provides a ligand or a composition comprising a ligand according to the invention having an AUC value (area under the curve) in the range of 1 mg.min/ml or more. In one embodiment, the lower end of the range is 5, 10, 15, 20, 30, 100, 200 or 300mg.min ml. In addition, or alternatively, a ligand or composition according to the invention has an AUC in the range of up to 600 mg.min/ml. In one embodiment, the upper end of the range is 500, 400, 300, 200, 150, 100, 75 or 50 mg.min ml. Advantageously a ligand according to the invention will have a AUC in the range selected from the group consisting of the following: 15 to 150mg.min/ml, 15 to 100 mg.min/ml, 15 to 75 mg.min/ml, and 15 to 50mg.min/ml.
In a first embodiment, the dual specific ligand comprises two complementary variable domains, i.e. two variable domains that, in their natural environment, are capable of operating together as a cognate pair or group even if in the context of the present invention they bind separately to their cognate epitopes. For example, the complementary variable domains may be immunoglobulin heavy chain and light chain variable domains
(VH and VL). VH and VL domains are advantageously provided by scFv or Fab antibody fragments. Variable domains may be linked together to form multivalent ligands by, for example: provision of a hinge region at the C-terminus of each V domain and disulphide bonding between cysteines in the hinge regions; or provision of dAbs each with a cysteine at the C-terminus of the domain, the cysteines being disulphide bonded together; or production of V-CH & V-CL to produce a Fab format; or use of peptide linkers (for example Gly4Ser linkers discussed hereinbelow) to produce dimers, trimers and further multimers.
The inventors have found that the use of complementary variable domains allows the two domain surfaces to pack together and be sequestered from the solvent. Furthermore the complementary domains are able to stabilise each other. In addition, it allows the creation of dual-specific IgG antibodies without the disadvantages of hybrid hybridomas as used in the prior art, or the need to engineer heavy or light chains at the sub-unit interfaces. The dual-specific ligands of the first aspect of the present invention have at least one VH/VL pair. A bispecific IgG according to this invention will therefore comprise two such pairs, one pair on each arm of the Y-shaped molecule. Unlike conventional bispecific antibodies or diabodies, therefore, where the ratio of chains used is determinative in the success of the preparation thereof and leads to practical difficulties, the dual specific ligands of the invention are free from issues of chain balance. Chain imbalance in conventional bi-specific antibodies results from the association of two different VL chains with two different VH chains, where VL chain 1 together with VH chain 1 is able to bind to antigen or epitope 1 and V chain 2 together with VH chain 2 is able to bind to antigen or epitope 2 and the two correct pairings are in some way linked to one another. Thus, only when VL chain 1 is paired with VH chain 1 and VL chain 2 is paired with VH chain 2 in a single molecule is bi-specificity created. Such bi-specific molecules can be created in two different ways. Firstly, they can be created by association of two existing VH/V pairings that each bind to a different antigen or epitope (for example, in a bi-specific IgG). In this case the VH/VL pairings must come all together in a 1:1 ratio in order to create a population of molecules all of which are bi-specific. This never occurs (even when complementary CH domain is enhanced by "knobs into holes" engineering) leading to a mixture of bi-specific molecules and molecules that are only able to bind to one antigen or epitope but not the other. The second way of creating a bispecific antibody is by the simultaneous association of two different VH chain with two different VL chains (for example in a bi-specific diabody). In this case, although there tends to be a preference for VL chain 1 to pair with VH chain 1 and VL chain 2 to pair with VH chain 2 (which can be enhanced by "knobs into holes" engineering of the VL and VH domains), this paring is never achieved in all molecules, leading to a mixed formulation whereby incorrect pairings occur that are unable to bind to either antigen or epitope.
Bi-specific antibodies constructed according to the dual-specific ligand approach according to the first aspect of the present invention overcome all of these problems because the binding to antigen or epitope 1 resides within the VH or VL domain and the binding to antigen or epitope 2 resides with the complementary VL or VH domain, respectively. Since VH and VL domains pair on a 1:1 basis all VH/VL pairings will be bispecific and thus all formats constructed using these VH/VL pairings (Fv, scFvs, Fabs, minibodies, IgGs etc) will have 100% bi-specific activity.
In the context of the present invention, first and second "epitopes" are understood to be epitopes which are not the same and are not bound by a single monospecific ligand. In the first configuration of the invention, they are advantageously on different antigens, one of which acts to increase the half-life of the ligand in vivo. Likewise, the first and second antigens are advantageously not the same. The dual specific ligands of the invention do not include ligands as described in WO 02/02773. Thus, the ligands of the present invention do not comprise complementary VH/VL pairs which bind any one or more antigens or epitopes co-operatively. Instead, the ligands according to the first aspect of the invention comprise a VH VL complementary pair, wherein the V domains have different specificities.
Moreover, the ligands according to the first aspect of the invention comprise VH/VL complementary pairs having different specificities for non-structurally related epitopes or antigens. Structurally related epitopes or antigens are epitopes or antigens which possess sufficient structural similarity to be bound by a conventional VH VL complementary pair which acts in a co-operative manner to bind an antigen or epitope; in the case of structurally related epitopes, the epitopes are sufficiently similar in structure that they "fit" into the same binding pocket formed at the antigen binding site of the VH/VL dimer.
In a second aspect, the present invention provides a ligand comprising a first immunoglobulin variable domain having a first antigen or epitope binding specificity and a second immunoglobulin variable domain having a second antigen or epitope binding specificity wherein one or both of said first and second variable domains bind to an antigen which increases the half-life of the ligand in vivo, and the variable domains are not complementary to one another.
hi one embodiment, binding to one variable domain modulates the binding of the ligand to the second variable domain.
In this embodiment, the variable domains may be, for example, pairs of VH domains or pairs of VL domains. Binding of antigen at the first site may modulate, such as enhance or inhibit, binding of an antigen at the second site. For example, binding at the first site at least partially inhibits binding of an antigen at a second site. In such an embodiment, the ligand may for example be maintained in the body of a subject organism in vivo through binding to a protein which increases the half-life of the ligand until such a time as it becomes bound to the second target antigen and dissociates from the half-life increasing protein. Modulation of binding in the above context is achieved as a consequence of the structural proximity of the antigen binding sites relative to one another. Such structural proximity can be achieved by the nature of the structural components linking the two or more antigen binding sites, eg by the provision of a ligand with a relatively rigid structure that holds the antigen binding sites in close proximity. Advantageously, the two or more antigen binding sites are in physically close proximity to one another such that one site modulates the binding of antigen at another site by a process which involves steric hindrance and/or conformational changes within the immunoglobulin molecule.
The first and the second antigen binding domains may be associated either covalently or non-covalently. In the case that the domains are covalently associated, then the association may be mediated for example by disulphide bonds or by a polypeptide linker such as (Gly Ser)n, where n = from 1 to 8, eg, 2, 3, 4, 5 or 7.
Ligands according to the invention may be combined into non-immunoglobulin multi- ligand structures to form multivalent complexes, which bind target molecules with the same antigen, thereby providing superior avidity, while at least one variable domain binds an antigen to increase the half life of the multimer. For example natural bacterial receptors such as SpA have been used as scaffolds for the grafting of CDRs to generate ligands which bind specifically to one or more epitopes. Details of this procedure are described in US 5,831,012. Other suitable scaffolds include those based on fibronectin and affibodies. Details of suitable procedures are described in WO 98/58965. Other suitable scaffolds include lipocallin and CTLA4, as described in van den Beuken et al, J. Mol. Biol. (2001) 310, 591-601, and scaffolds such as those described in WO0069907 (Medical Research Council), which are based for example on the ring structure of bacterial GroEL or other chaperone polypeptides.
Protein scaffolds may be combined; for example, CDRs may be grafted on to a CTLA4 scaffold and used together with immunoglobulin VH or VL domains to form a ligand. Likewise, fibronectin, lipocallin and other scaffolds may be combined. In the case that the variable domains are selected from V-gene repertoires selected for instance using phage display technology as herein described, then these variable domains can comprise a universal framework region, such that is they may be recognised by a specific generic ligand as herein defined. The use of universal frameworks, generic ligands and the like is described in WO99/20749. In the present invention, reference to phage display includes the use of both phage and/or phagemids.
Where V-gene repertoires are used variation in polypeptide sequence is preferably located within the structural loops of the variable domains. The polypeptide sequences of either variable domain may be altered by DNA shuffling or by mutation in order to enhance the interaction of each variable domain with its complementary pair.
hi a preferred embodiment of the invention the 'dual-specific ligand' is a single chain Fv fragment. In an alternative embodiment of the invention, the 'dual-specific ligand' consists of a Fab region of an antibody. The term "Fab region" includes a Fab-like region where two VH or two VL domains are used.
The variable regions may be derived from antibodies directed against target antigens or epitopes. Alternatively they may be derived from a repertoire of single antibody domains such as those expressed on the surface of filamentous bacteriophage. Selection may be performed as described below.
In a third aspect, the invention provides a method for producing a ligand comprising a first immunoglobulin single variable domain having a first binding specificity and a second single immunoglobulin single variable domain having a second (different) binding specificity, one or both of the binding specificities being specific for an antigen which increases the half-life of the ligand in vivo, the method comprising the steps of:
(a) selecting a first variable domain by its ability to bind to a first epitope,
(b) selecting a second variable region by its ability to bind to a second epitope, (c) combining the variable domains; and
(d) selecting the ligand by its ability to bind to said first epitope and to said second epitope. The ligand can bind to the first and second epitopes either simultaneously or, where there is competition between the binding domains for epitope binding, the binding of one domain may preclude the binding of another domain to its cognate epitope. h one embodiment, therefore, step (d) above requires simultaneous binding to both first and second (and possibly further) epitopes; in another embodiment, the binding to the first and second epitoes is not simultaneous.
The epitopes are preferably on separate antigens.
Ligands advantageously comprise VH/VL combinations, or VH/VH or VL/VL combinations of immunoglobulin variable domains, as described above. The ligands may moreover comprise camelid VHH domains, provided that the VHH domain which is specific for an antigen which increases the half-life of the ligand in vivo does not bind Hen egg white lysozyme (HEL), porcine pancreatic alpha-amylase or NmC-A; hcg, BSA-linked RR6 azo dye or S. mutans HG982 cells, as described in Conrath et al, (2001) JBC 276:7346-7350 and WO99/23221, neither of which describe the use of a specificity for an antigen which increases half-life to increase the half life of the ligand in vivo.
In one embodiment, said first variable domain is selected for binding to said first epitope in absence of a complementary variable domain. In a further embodiment, said first variable domain is selected for binding to said first epitope/antigen in the presence of a third variable domain in which said third variable domain is different from said second variable domain and is complementary to the first domain. Similarly, the second domain may be selected in the absence or presence of a complementary variable domain.
The antigens or epitopes targeted by the ligands of the invention, in addition to the half- life enhancing protein, may be any antigen or epitope but advantageously is an antigen or epitope that is targeted with therapeutic benefit. The invention provides ligands, including open conformation, closed conformation and isolated dAb monomer ligands, specific for any such target, particularly those targets further identified herein. Such targets may be, or be part of, polypeptides, proteins or nucleic acids, which may be naturally occurring or synthetic. In this respect, the ligand of the invention may bind the epiotpe or antigen and act as an antagonist or agonist (eg, EPO receptor agonist). One skilled in the art will appreciate that the choice is large and varied. They may be for instance human or animal proteins, cytokines, cytokine receptors, enzymes co-factors for enzymes or DNA binding proteins. Suitable cytokines and growth factors include but are not limited to: ApoE, Apo-SAA, BDNF, Cardiotrophin-1, EGF, EGF receptor, ENA-78, Eotaxin, Eotaxin-2, Exodus-2, EpoR, FGF-acidic, FGF-basic, fibroblast growth factor- 10, FLT3 ligand, Fractalkine (CX3C), GDNF, G-CSF, GM-CSF, GF-βl, insulin, IFN-γ, IGF-I, IGF-II, IL-lα, IL-lβ, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8 (72 a.a.), IL-8 (77 a.a.), IL-9, IL-10, IL-11, IL-12, IL-13, IL-15, IL-16, IL-17, IL-18 (IGIF), Inhibin α, Inhibin β, IP- 10, keratinocyte growth factor-2 (KGF-2), KGF, Leptin, LIF, Lymphotactin, Mullerian inhibitory substance, monocyte colony inhibitory factor, monocyte attractant protein, M-CSF, MDC (67 a.a.), MDC (69 a.a.), MCP-1 (MCAF), MCP-2, MCP-3, MCP- 4, MDC (67 a.a.), MDC (69 a.a.), MIG, MIP-lα, MIP-lβ, MTP-3α, MLP-3β, MIP-4, myeloid progenitor inhibitor factor-1 (MPJJF-l), NAP -2, Neurturin, Nerve growth factor, β-NGF, NT-3, NT-4, Oncostatin M, PDGF-AA, PDGF-AB, PDGF-BB, PF-4, RANTES, SDFlα, SDFlβ, SCF, SCGF, stem cell factor (SCF), TARC, TGF-α, TGF-β, TGF-β2, TGF-β3, tumour necrosis factor (TNF), TNF-α, TNF-β, TNF receptor I, TNF receptor II, TNIL-1, TPO, VEGF, VEGF receptor 1, VEGF receptor 2, VEGF receptor 3, GCP-2, GRO/MGSA, GRO-β, GRO-γ, HCC1, 1-309, HER 1, HER 2, HER 3 and HER 4. Cytokine receptors include receptors for the foregoing cytokines. It will be appreciated that this list is by no means exhaustive.
In one embodiment of the invention, the variable domains are derived from a respective antibody directed against the antigen or epitope. In a preferred embodiment the variable domains are derived from a repertoire of single variable antibody domains.
In a further aspect, the present invention provides one or more nucleic acid molecules encoding at least a dual-specific ligand as herein defined. The dual specific ligand may be encoded on a single nucleic acid molecule; alternatively, each domain may be encoded by a separate nucleic acid molecule. Where the ligand is encoded by a single nucleic acid molecule, the domains may be expressed as a fusion polypeptide, in the manner of a scFv molecule, or may be separately expressed and subsequently linked together, for example using chemical linking agents. Ligands expressed from separate nucleic acids will be linked together by appropriate means. The nucleic acid may further encode a signal sequence for export of the polypeptides from a host cell upon expression and may be fused with a surface component of a filamentous bacteriophage particle (or other component of a selection display system) upon expression.
In a further aspect the present invention provides a vector comprising nucleic acid encoding a dual specific ligand according to the present invention.
In a yet further aspect, the present invention provides a host cell transfected with a vector encoding a dual specific ligand according to the present invention.
Expression from such a vector may be configured to produce, for example on the surface of a bacteriophage particle, variable domains for selection. This allows selection of displayed variable regions and thus selection of 'dual-specific ligands' using the method of the present invention.
The present invention further provides a kit comprising at least a dual-specific ligand according to the present invention.
Dual-Specific ligands according to the present invention preferably comprise combinations of heavy and light chain domains. For example, the dual specific ligand may comprise a VH domain and a VL domain, which may be linked together in the form of an scFv. In addition, the ligands may comprise one or more CH or CL domains. For example, the ligands may comprise a CHI domain, CH2 or CH3 domain, and/or a CL domain, Cμl, Cμ2, Cμ3 or Cμ4 domains, or any combination thereof. A hinge region domain may also be included. Such combinations of domains may, for example, mimic natural antibodies, such as IgG or IgM, or fragments thereof, such as Fv, scFv, Fab or F(ab')2 molecules. Other structures, such as a single arm of an IgG molecule comprising VH, VL, CHI and CL domains, are envisaged.
In a preferred embodiment of the invention, the variable regions are selected from single domain V gene repertoires. Generally the repertoire of single antibody domains is displayed on the surface of filamentous bacteriophage. In a preferred embodiment each single antibody domain is selected by binding of a phage repertoire to antigen.
In a preferred embodiment of the invention each single variable domain may be selected for binding to its target antigen or epitope in the absence of a complementary variable region. In an alternative embodiment, the single variable domains may be selected for binding to its target antigen or epitope in the presence of a complementary variable region. Thus the first single variable domain may be selected in the presence of a third complementary variable domain, and the second variable domain may be selected in the presence of a fourth complementary variable domain. The complementary third or fourth variable domain may be the natural cognate variable domain having the same specificity as the single domain being tested, or a non-cognate complementary domain - such as a "dummy" variable domain.
Preferably, the dual specific ligand of the invention comprises only two variable domains although several such ligands may be incorporated together into the same protein, for example two such ligands can be incorporated into an IgG or a multimeric immunoglobulin, such as IgM. Alternatively, in another embodiment a plurality of dual specific ligands are combined to form a multimer. For example, two different dual specific ligands are combined to create a tetra-specific molecule.
It will be appreciated by one skilled in the art that the light and heavy variable regions of a dual-specific ligand produced according to the method of the present invention may be on the same polypeptide chain, or alternatively, on different polypeptide chains. In the case that the variable regions are on different polypeptide chains, then they may be linked via a linker, generally a flexible linker (such as a polypeptide chain), a chemical linking group, or any other method known in the art.
In a further aspect, the present invention provides a composition comprising a dual- specific ligand, obtainable by a method of the present invention, and a pharmaceutically acceptable carrier, diluent or excipient. Moreover, the present invention provides a method for the treatment and/or prevention of disease using a 'dual-specific ligand' or a composition according to the present invention.
In a second configuration, the present invention provides multispecific ligands which comprise at least two non-complementary variable domains. For example, the ligands may comprise a pair of VH domains or a pair of VL domains. Advantageously, the domains are of non-camelid origin; preferably they are human domains or comprise human framework regions (FWs) and one or more heterologous CDRs. CDRs and framework regions are those regions of an immunoglobulin variable domain as defined in the Kabat database of Sequences of Proteins of rmmunological Interest.
Preferred human framework regions are those encoded by germline gene segments DP47 and DPK9. Advantageously, FWl, FW2 and FW3 of a VH or VL domain have the sequence of FWl, FW2 or FW3 from DP47 or DPK9. The human frameworks may optionally contain mutations, for example up to about 5 amino acid changes or up to about 10 amino acid changes collectively in the human frameworks used in the ligands of the invention.
The variable domains in the multispecific ligands according to the second configuration of the invention may be arranged in an open or a closed conformation; that is, they may be arranged such that the variable domains can bind their cognate ligands independently and simultaneously, or such that only one of the variable domains may bind its cognate ligand at any one time.
The inventors have realised that under certain structural conditions, non-complementary variable domains (for example two light chain variable domains or two heavy chain variable domains) may be present in a ligand such that binding of a first epitope to a first variable domain inhibits the binding of a second epitope to a second variable domain, even though such non-complementary domains do not operate together as a cognate pair. Advantageously, the ligand comprises two or more pairs of variable domains; that is, it comprises at least four variable domains. Advantageously, the four variable domains comprise frameworks of human origin.
In a preferred embodiment, the human frameworks are identical to those of human germline sequences.
The present inventors consider that such antibodies will be of particular use in ligand binding assays for therapeutic and other uses.
Thus, in a first aspect of the second configuration, the present invention provides a method for producing a multispecific ligand comprising the steps of: a) selecting a first epitope binding domain by its ability to bind to a first epitope, b) selecting a second epitope binding domain by its ability to bind to a second epitope, c) combining the epitope binding domains; and d) selecting the closed conformation multispecific ligand by its ability to bind to said first second epitope and said second epitope.
In a further aspect of the second configuration, the invention provides method for preparing a closed conformation multi-specific ligand comprising a first epitope binding domain having a first epitope binding specificity and a non-complementary second epitope binding domain having a second epitope binding specificity, wherein the first and second binding specificities compete for epitope binding such that the closed conformation multi-specific ligand may not bind both epitopes simultaneously, said method comprising the steps of:
a) selecting a first epitope binding domain by its ability to bind to a first epitope, b) selecting a second epitope binding domain by its ability to bind to a second epitope, c) combining the epitope binding domains such that the domains are in a closed conformation; and d) selecting the closed conformation multispecific ligand by its ability to bind to said first second epitope and said second epitope, but not to both said first and second epitopes simultaneously.
Moreover, the invention provides a closed conformation multi-specific ligand comprising a first epitope binding domain having a first epitope binding specificity and a non- complementary second epitope binding domain having a second epitope binding specificity, wherein the first and second binding specificities compete for epitope binding such that the closed conformation multi-specific ligand may not bind both epitopes simultaneously.
An alternative embodiment of the above aspect of the of the second configuration of the invention optionally comprises a further step (bl) comprising selecting a third or further epitope binding domain. In this way the multi-specific ligand produced, whether of open or closed conformation, comprises more than two epitope binding specificities. In a preferred aspect of the second configuration of the invention, where the multi-specific ligand comprises more than two epitope binding domains, at least two of said domains are in a closed conformation and compete for binding; other domains may compete for binding or may be free to associate independently with their cognate epitope(s).
According to the present invention the term 'multi-specific ligand' refers to a ligand which possesses more than one epitope binding specificity as herein defined.
As herein defined the term 'closed conformation' (multi-specific ligand) means that the epitope binding domains of the ligand are attached to or associated with each other, optionally by means of a protein skeleton, such that epitope binding by one epitope binding domain competes with epitope binding by another epitope binding domain. That is, cognate epitopes may be bound by each epitope binding domain individually but not simultaneosuly. The closed conformation of the ligand can be achieved using methods herein described.
"Open conformation" means that the epitope binding domains of the ligand are attached to or associated with each other, optionally by means of a protein skeleton, such that epitope binding by one epitope binding domain does not compete with epitope binding by another epitope binding domain.
As referred to herein, the term 'competes' means that the binding of a first epitope to its cognate epitope binding domain is inhibited when a second epitope is bound to its cognate epitope binding domain. For example, binding may be inhibited sterically, for example by physical blocking of a binding domain or by alteration of the structure or environment of a binding domain such that its affinity or avidity for an epitope is reduced.
In a further embodiment of the second configuration of the invention, the epitopes may displace each other on binding. For example, a first epitope may be present on an antigen which, on binding to its cognate first binding domain, causes steric hindrance of a second binding domain, or a coformational change therein, which displaces the epitope bound to the second binding domain.
Advantageously, binding is reduced by 25% or more, advantageously 40%, 50%, 60%, 70%, 80%, 90% or more, and preferably up to 100% or nearly so, such that binding is completely inhibited. Binding of epitopes can be measured by conventional antigen binding assays, such as ELISA, by fluorescence based techniques, including FRET, or by techniques such as suface plasmon resonance which measure the mass of molecules.
According to the method of the present invention, advantageously, each epitope binding domain is of a different epitope binding specificity.
In the context of the present invention, first and second "epitopes" are understood to be epitopes which are not the same and are not bound by a single monospecific ligand. They may be on different antigens or on the same antigen, but separated by a sufficient distance that they do not form a single entity that could be bound by a single mono-specific VH/VL binding pair of a conventional antibody. Experimentally, if both of the individual variable domains in single chain antibody form (domain antibodies or dAbs) are separately competed by a monospecific VH/VL ligand against two epitopes then those two epitopes are not sufficiently far apart to be considered separate epitopes according to the present invention. The closed conformation multispecific ligands of the invention do not include ligands as described in WO 02/02773. Thus, the ligands of the present invention do not comprise complementary VH^VL Pairs which bind any one or more antigens or epitopes cooperatively. Instead, the ligands according to the invention preferably comprise non- complementary VH'VH or VL"VL pairs. Advantageously, each VH or VL domain in each VH"VH or VL-VL Parr h s a different epitope binding specificity, and the epitope binding sites are so arranged that the binding of an epitope at one site competes with the binding of an epitope at another site.
According to the present invention, advantageously, each epitope binding domain comprises an immunoglobulin variable domain. More advantageously, each immunoglobulin variable domain will be either a variable light chain domain (V ) or a variable heavy chain domain VH- *1 the second configuration of the present invention, the immunoglobulin domains when present on a ligand according to the present invention are non-complementary, that is they do not associate to form a VH VL antigen binding site. Thus, multi-specific ligands as defined in the second configuration of the invention comprise immunoglobulin domains of the same sub-type, that is either variable light chain domains (y-) or variable heavy chain domains (vH)- Moreover, where the ligand according to the invention is in the closed conformation, the immunoglobulin domains may be of the camelid VHH type.
In an alternative embodiment, the ligand(s) according to the invention do not comprise a camelid VHH domain. More particularly, the ligand(s) of the invention do not comprise one or more amino acid residues that are specific to camelid VHH domains as compared to human VH domains.
Advantageously, the single variable domains are derived from antibodies selected for binding activity against different antigens or epitopes. For example, the variable domains may be isolated at least in part by human immunisation. Alternative methods are known in the art, including isolation from human antibody libraries and synthesis of artificial antibody genes. The variable domains advantageously bind superantigens, such as protein A or protein L. Binding to superantigens is a property of correctly folded antibody variable domains, and allows such domains to be isolated from, for example, libraries of recombinant or mutant domains.
Epitope binding domains according to the present invention comprise a protein scaffold and epitope interaction sites (which are advantageously on the surface of the protein scaffold).
Epitope binding domains may also be based on protein scaffolds or skeletons other than immunoglobulin domains. For example natural bacterial receptors such as SpA have been used as scaffolds for the grafting of CDRs to generate ligands which bind specifically to one or more epitopes. Details of this procedure are described in US 5,831,012. Other suitable scaffolds include those based on fibronectin and affibodies. Details of suitable procedures are described in WO 98/58965. Other suitable scaffolds include lipocallin and CTLA4, as described in van den Beuken et al, J. Mol. Biol. (2001) 310, 591-601, and scaffolds such as those described in WO0069907 (Medical Research Council), which are based for example on the ring structure of bacterial GroEL or other chaperone polypeptides.
Protein scaffolds may be combined; for example, CDRs may be grafted on to a CTLA4 scaffold and used together with immunoglobulin VH or VL domains to form a multivalent ligand. Likewise, fibronectin, lipocallin and other scaffolds maybe combined.
It will be appreciated by one skilled in the art that the epitope binding domains of a closed conformation multispecific ligand produced according to the method of the present invention may be on the same polypeptide chain, or alternatively, on different polypeptide chains. In the case that the variable regions are on different polypeptide chains, then they may be linked via a linker, advantageously a flexible linker (such as a polypeptide chain), a chemical linking group, or any other method known in the art. The first and the second epitope binding domains may be associated either covalently or non-covalently. In the case that the domains are covalently associated, then the association may be mediated for example by disulphide bonds.
In the second configuation of the invention, the first and the second epitopes are preferably different. They may be, or be part of, polypeptides, proteins or nucleic acids, which may be naturally occurring or synthetic. In this respect, the ligand of the invention may bind an epiotpe or antigen and act as an antagonist or agonist (eg, EPO receptor agonist). The epitope binding domains of the ligand in one embodiment have the same epitope specificity, and may for example simultaneously bind their epitope when multiple copies of the epitope are present on the same antigen. In another embodiment, these epitopes are provided on different antigens such that the ligand can bind the epitopes and bridge the antigens. One skilled in the art will appreciate that the choice of epitopes and antigens is large and varied. They may be for instance human or animal proteins, cytokines, cytokine receptors, enzymes co-factors for enzymes or DNA binding proteins. Suitable cytokines and growth factors include but are not limited to: ApoE, Apo-SAA, BDNF, Cardiotrophin-1, EGF, EGF receptor, ENA-78, Eotaxin, Eotaxin-2, Exodus-2, EpoR, FGF-acidic, FGF-basic, fibroblast growth factor- 10, FLT3 ligand, Fractalkine (CX3C), GDNF, G-CSF, GM-CSF, GF-βl, insulin, IFN-γ, IGF-L IGF-IL IL-lα, IL-lβ, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8 (72 a.a.), IL-8 (77 a.a.), IL-9, IL-10, IL-11, IL-12, IL-13, IL-15, IL-16, IL-17, IL-18 (IGTF), Inhibin α, Inhibin β, TP-10, keratinocyte growth factor-2 (KGF-2), KGF, Leptin, LIF, Lymphotactin, Mullerian inhibitory substance, monocyte colony inhibitory factor, monocyte attractant protein, M-CSF, MDC (67 a.a.), MDC (69 a.a.), MCP-1 (MCAF), MCP-2, MCP-3, MCP-4, MDC (67 a.a.), MDC (69 a.a.), MIG, MIP-lα, MTP-lβ, MIP-3α, MTP-3β, MIP-4, myeloid progenitor inhibitor factor-1 (MPLF-1), NAP-2, Neurturin, Nerve growth factor, β-NGF, NT-3, NT-4, Oncostatin M, PDGF-AA, PDGF-AB, PDGF-BB, PF-4, RANTES, SDFlα, SDFlβ, SCF, SCGF, stem cell factor (SCF), TARC, TGF-α, TGF-β, TGF-β2, TGF-β3, tumour necrosis factor (TNF), TNF-α, TNF-β, TNF receptor I, TNF receptor II, TNIL-1, TPO, VEGF, VEGF receptor 1, VEGF receptor 2, VEGF receptor 3, GCP-2, GRO/MGSA, GRO-β, GRO-γ, HCC1, 1-309, HER 1, HER 2, HER 3, HER 4, TACE recognition site, TNF BP-I and TNF BP-II, as well as any target disclosed in Annex 2 or Annex 3 hereto, whether in combination as set forth in the Annexes, in a different combination or individually. Cytokine receptors include receptors for the foregoing cytokines, e.g. IL-1 RI; IL-6R;
IL-10R; IL-18R, as well as receptors for cytokines set forth in Annex 2 or Annex 3 and also receptors disclosed in Annex 2 and 3. It will be appreciated that this list is by no means exhaustive. Where the multispecific ligand binds to two epitopes (on the same or different antigens), the antigen(s) may be selected from this list.
Advantageously, dual specific ligands may be used to target cytokines and other molecules which cooperate synergistically in therapeutic situations in the body of an organism. The invention therefore provides a method for synergising the activity of two or more cytokines, comprising administering a dual specific ligand capable of binding to said two or more cytokines. In this aspect of the invention, the dual specific ligand may be any dual specific ligand, including a ligand composed of complementary and/or non- complementary domains, a ligand in an open conformation, and a ligand in a closed conformation. For example, this aspect of the invention relates to combinations of VH domains and VL domains, VH domains only and VL domains only.
Synergy in a therapeutic context may be achieved in a number of ways. For example, target combinations may be therapeutically active only if both targets are targeted by the ligand, whereas targeting one target alone is not therapeutically effective, hi another embodiment, one target alone may provide some low or minimal therapeutic effect, but together with a second target the combination provides a synergistic increase in therapeutic effect.
Preferably, the cytokines bound by the dual specific ligands ofthis aspect of the invention are sleeted from the list shown in Annex 2.
Moreover, dual specific ligands may be used in oncology applications, where one specificity targets CD89, which is expressed by cytotoxic cells, and the other is tumour specific. Examples of tumour antigens which may be targetted are given in Annex 3.
In one embodiment of the second configuration of the invention, the variable domains are derived from an antibody directed against the first and/or second antigen or epitope. In a preferred embodiment the variable domains are derived from a repertoire of single variable antibody domains. In one example, the repertoire is a repertoire that is not created in an animal or a synthetic repertoire. In another example, the single variable domains are not isolated (at least in part) by animal immunisation. Thus, the single domains can be isolated from a naive library.
The second configuration of the invention, in another aspect, provides a multi-specific ligand comprising a first epitope binding domain having a first epitope binding specificity and a non-complementary second epitope binding domain having a second epitope binding specificity. The first and second binding specificities may be the same or different.
In a further aspect, the present invention provides a closed conformation multi-specific ligand comprising a first epitope binding domain having a first epitope binding specificity and a non-complementary second epitope binding domain having a second epitope binding specificity wherein the first and second binding specificities are capable of competing for epitope binding such that the closed conformation multi-specific ligand cannot bind both epitopes simultaneously.
In a still further aspect, the invention provides open conformation ligands comprising non-complementary binding domains, wherein the deomains are specific for a different epitope on the same target. Such ligands bind to targets with increased avidity. Similarly, the invention provides multivalent ligands comprising non-complementary binding domains specific for the same epitope and directed to targets which comprise multiple copies of said epitope, such as IL-5, PDGF-AA, PDGF-BB, TGF beta, TGF beta2, TGF beta3 and TNFα, for eample human TNF Receptor 1 and human TNFα.
In a similar aspect, ligands according to the invention can be configured to bind individual epitopes with low affinity, such that binding to individual epitopes is not therapeutically significant; but the increased avidity resulting from binding to two epitopes provides a theapeutic benefit. In a perticular example, epitopes may be targetted which are present individually on normal cell types, but present together only on abnormal or diseased cells, such as tumour cells. In such a situaton, only the abnormal or diseased cells are effectively targetted by the bispecific ligands according to the invention.
Ligand specific for multiple copies of the same epitope, or adjacent epitopes, on the same target (known as chelating dAbs) may also be trimeric or polymeric (tertrameric or more) ligands comprising three, four or more non-complementary binding domains. For example, ligands may be constructed comprising three or four VH domains or VL domains.
Moreover, ligands are provided which bind to multisubunit targets, wherein each binding domain is specific for a subunit of said target. The ligand may be dimeric, trimeric or polymeric.
Preferably, the multi-specific ligands according to the above aspects of the invention are obtainable by the method of the first aspect of the invention.
According to the above aspect of the second configuration of the invention, advantageously the first epitope binding domain and the second epitope binding domains are non-complementary immunoglobulin variable domains, as herein defined. That is either VH"VH or VL"VL variable domains.
Chelating dAbs in particular may be prepared according to a preferred aspect of the invention, namely the use of anchor dAbs, in which a library of dimeric, trimeric or multimeric dAbs is constructed using a vector which comprises a constant dAb upstream or downstream of a linker sequence, with a repertoire of second, third and further dAbs being inserted on the other side of the linker. For example, the anchor or guiding dAb maybe TAR1-5 (VK), TARl-27(Vκ), TAR2h-5(VH) or TAR2h-6(Vκ).
In alternative methodologies, the use of linkers may be avoided, for example by the use of non-covalent bonding or naturall affinity between binding domains such as VH and Vκ. The invention accordingly provides a method for preparing a chelating multimeric ligand comprising the steps of: (a) providing a vector comprising a nucleic acid sequence encoding a single binding domain specific for a first epitope on a target;
(b) providing a vector encoding a repertoire comprising second binding domains specific for a second epitope on said target, which epitope can be the same or different to the first epitope, said second epitope being adjacent to said first epitope; and
(c) expressing said first and second binding domains; and
(d) isolating those combinations of first and second binding domains which combine together to produce a target-binding dimer.
The first and second epitopes are adjacent such that a multimeric ligand is capable of binding to both epitopes simultaneously. This provides the ligand with the advantages of increased avidity if binding. Where the epitopes are the same, the increased avidity is obtained by the presence of multiple copies of the epitope on the target, allowing at least two copies to be simultaneously bound in order to obtain the increased avidity effect.
The binding domains may be associated by several methods, as well as the use of linkers. For example, the binding domains may comprise cys residues, avidin and streptavidin groups or other means for non-covalent attachment post-synthesis; those combinations which bind to the target efficiently will be isolated. Alternatively, a linker may be present between the first and second binding domains, which are expressed as a single polypeptide from a single vector, which comprises the first binding domain, the linker and a repertoire of second binding domains, for instance as described above.
In a preferred aspect, the first and second binding domains associate naturally when bound to antigen; for example, VH and Vκ domains, when bound to adjacent epitopes, will naturally associate in a three-way interaction to form a stable dimer. Such associated proteins can be isolated in a target binding assay. An advantage of this procedure is that only binding domains which bind to closely adjacent epitopes, in the correct conformation, will associate and thus be isolated as a result of their increased avidity for the target.
In an alternative embodiment of the above aspect of the second configuration of the invention, at least one epitope binding domain comprises a non-immunoglobulin 'protein scaffold' or 'protein skeleton' as herein defined. Suitable non-immunoglobulin protein scaffolds include but are not limited to any of those selected from the group consisting of: SpA, fibronectin, GroEL and other chaperones, lipocallin, CCTLA4 and affibodies, as set forth above.
According to the above aspect of the second configuration of the invention, advantageously, the epitope binding domains are attached to a 'protein skeleton'. Advantageously, a protein skeleton according to the invention is an immunoglobulin skeleton.
According to the present invention, the term 'immunoglobulin skeleton' refers to a protein which comprises at least one immunoglobulin fold and which acts as a nucleus for one or more epitope binding domains, as defined herein.
Preferred immunoglobulin skeletons as herein defined includes any one or more of those selected from the following: an immunoglobulin molecule comprising at least (i) the CL (kappa or lambda subclass) domain of an antibody; or (ii) the CHI domain of an antibody heavy chain; an immunoglobulin molecule comprising the CHI and CH2 domains of an antibody heavy chain; an immunoglobulin molecule comprising the CHI, CH2 and CH3 domains of an antibody heavy chain; or any of the subset (ii) in conjunction with the CL (kappa or lambda subclass) domain of an antibody. A hinge region domain may also be included. Such combinations of domains may, for example, mimic natural antibodies, such as IgG or IgM, or fragments thereof, such as Fv, scFv, Fab or F(ab')2 molecules. Those skilled in the art will be aware that this list is not intended to be exhaustive.
Linking of the skeleton to the epitope binding domains, as herein defined may be achieved at the polypeptide level, that is after expression of the nucleic acid encoding the skeleton and/or the epitope binding domains. Alternatively, the linking step may be performed at the nucleic acid level. Methods of linking a protein skeleton according to the present invention, to the one or more epitope binding domains include the use of protein chemistry and/or molecular biology techniques which will be familiar to those skilled in the art and are described herein. Advantageously, the closed conformation multispecific ligand may comprise a first domain capable of binding a target molecule, and a second domain capable of binding a molecule or group which extends the half-life of the ligand. For example, the molecule or group may be a bulky agent, such as HSA or a cell matrix protein. As used herein, the phrase "molecule or group which extends the half-life of a ligand" refers to a molecule or chemical group which, when bound by a dual-specific ligand as described herein increases the in vivo half-life of such dual specific ligand when admimstered to an animal, relative to a ligand that does not bind that molecule or group. Examples of molecules or groups that extend the half-life of a ligand are described hereinbelow. In a preferred embodiment, the closed conformation multispecific ligand may be capable of binding the target molecule only on displacement of the half-life enhancing molecule or group. Thus, for example, a closed conformation multispecific ligand is maintained in circulation in the bloodstream of a subject by a bulky molecule such as HSA. When a target molecule is encountered, competition between the binding domains of the closed conformation multispecific ligand results in displacement of the HSA and binding of the target.
Ligands according to any aspect of the present invention, as well as dAb monomers useful in constructing such ligands, may advantageously dissociate from their cognate target(s) with a Kd of 300nM to 5pM (ie, 3 x 10"7 to 5 x 10"12M), preferably 50nM to20ρM, or 5nM to 200pM or InM to lOOpM, 1 x 10"7 M or less, 1 x 10"8 M or less, 1 x 10"9 M or less, 1 x 10"10 M or less, 1 x 10"11 M or less; and/or a Koff rate constant of 5 x 10"1 to 1 x 10"7 S"1, preferably 1 x 10"2 to 1 x 10"6 S"1, or 5 x 10"3 to 1 x 10"5 S'1, or 5 x 10"1 S"1 or less, or 1 x 10"2 S"1 or less, or 1 x 10"3 S"1 or less, or 1 x 10"4 S"1 or less, or 1 x 10"5 S"1 or less, or 1 x 10"6 S"1 or less as determined by surface plasmon resonance. The Kd rate constand is defined as Koff/Kon-
In particular the invention provides an anti-TNFα dAb monomer (or dual specific ligand comprising such a dAb), homodimer, heterodimer or homotrimer ligand, wherein each dAb binds TNFα. The ligand binds to TNFα with a Kd of 300nM to 5ρM (ie, 3 x 10-7 to 5 x 10"12M), preferably 50nM to 20pM, more preferably 5nM to 200pM and most preferably InM to lOOpM; expressed in an alternative manner, the Kd is 1 x 10"7 M or less, preferably 1 x 10"8 M or less, more preferably 1 x 10"9 M or less, advantageously 1 x 10"10 M or less and most preferably 1 x 10"11 M or less; and/or a Koff rate constant of 5 x
10"1 to 1 x 10"7 S"1, preferably 1 x 10"2 to 1 x 10"6 S"1, more preferably 5 x 10"3 to 1 x 10"5
S"1, for example 5 x 10"1 S"1 or less, preferably 1 x 10"2 S"1 or less, more preferably 1 x 10"
3 S"1 or less, advantageously 1 x 10"4 S"1 or less, further advantageously 1 x 10"5 S~l or less, and most preferably 1 x 10"6 S"1 or less, as determined by surface plasmon resonance.
Preferably, the ligand neutralises TNFα in a standard L929 assay with an ND50 of 500nM to 50pM, preferably or lOOnM to 50pM, advantageously lOnM to lOOpM, more preferably InM to lOOpM; for example 50nM or less, preferably 5nM or less, advantageously 500pM or less, more preferably 200pM or less and most preferably lOOpM or less.
Preferably, the ligand inhibits binding of TNF alpha to TNF alpha Receptor I (p55 receptor) with an IC50 of 500nM to 50pM, preferably lOOnM to 50pM, more preferably lOnM to lOOpM, advantageously InM to lOOpM; for example 50nM or less, preferably 5nM or less, more preferably 500pM or less, advantageously 200pM or less, and most preferably lOOpM or less. Preferably, the TNFα is Human TNFα.
Furthermore, the invention provides a an anti-TNF Receptor I dAb monomer, or dual specific ligand comprising such a dAb, that binds to TNF Receptor I with a Kd of 300nM to 5pM (ie, 3 x 10"7 to 5 x 10"12M), preferably 50nM to20pM, more preferably 5nM to 200pM and most preferably InM to lOOpM, for example 1 x 10"7 M or less, preferably 1 x 10"8 M or less, more preferably 1 x 10"9 M or less, advantageously 1 x 10"10 M or less and most preferably 1 x 10"11 M or less; and/or a Kof rate constant of 5 x 10"1 to 1 x 10"7 S"\ preferably 1 x 10"2 to 1 x 10"6 S"1, more preferably 5 x 10"3 to 1 x 10~5 S"1, for example 5 x 10"1 S"1 or less, preferably 1 x 10"2 S"1 or less, advantageously 1 x 10"3 S"1 or less, more preferably 1 x 10"4 S"1 or less, still more preferably 1 x 10"5 S" or less, and most preferably 1 x 10"6 S"1 or less as determined by surface plasmon resonance.
Preferably, the dAb monomeror ligand neutralises TNFα in a standard assay (eg, the L929 or HeLa assays described herein) with an ND50 of 500nM to 50pM, preferably lOOnM to 50pM, more preferably lOnM to lOOpM, advantageously InM to lOOpM; for example 50nM or less, preferably 5nM or less, more preferably 500pM or less, advantageously 200pM or less, and most preferably lOOpM or less.
Preferably, the dAb monomer or ligand inhibits binding of TNF alpha to TNF alpha Receptor I (p55 receptor) with an IC50 of 500nM to 50pM, preferably lOOnM to 50pM, more preferably lOnM to lOOpM, advantageously InM to lOOpM; for example 50nM or less, preferably 5nM or less, more preferably 500pM or less, advantageously 200pM or less, and most preferably lOOpM or less. Preferably, the TNF Receptor I target is Human TNFα.
Furthermore, the invention provides a dAb monomer(or dual specific ligand comprising such a dAb) that binds to serum albumin (SA) with a Kd of InM to 50 μM (ie, x 10"9 to 5 x 10"4), preferably lOOnM to lOμM. Preferably, for a dual specific ligand comprising a first anti-SA dAb and a second dAb to another target, the affinity (eg Kd and/or Koff as measured by surface plasmon resonance, eg using BiaCore) of the second dAb for its target is from 1 to 100000 times (preferably 100 to 100000, more preferably 1000 to 100000, or 10000 to 100000 times) the affinity of the first dAb for SA. For example, the first dAb binds SA with an affinity of approximately lOμM, while the second dAb binds its target with an affinity of lOOpM. Preferably, the serum albumin is human serum albumin (HSA).
In one embodiment, the first dAb (or a dAb monomer) binds SA (eg, HSA) with a K of approximately 50, preferably 70, and more preferably 100, 150 or 200 nM.
The invention moreover provides dimers, trimers and polymers of the aforementioned dAb monomers, in accordance with the foregoing aspect of the present invention.
Ligands according to the invention, including dAb monomers, dimers and trimers, can be linked to an antibody Fc region, comprising one or both of CH2 and CH3 domains, and optionally a hinge region. For example, vectors encoding ligands linked as a single nucleotide sequence to an Fc region may be used to prepare such polypeptides. In a further aspect of the second configuration of the invention, the present invention provides one or more nucleic acid molecules encoding at least a multispecific ligand as herein defined. In one embodiment, the ligand is a closed conformation ligand. In another embodiment, it is an open conformation ligand. The multispecific ligand may be encoded on a single nucleic acid molecule; alternatively, each epitope binding domain may be encoded by a separate nucleic acid molecule. Where the ligand is encoded by a single nucleic acid molecule, the domains may be expressed as a fusion polypeptide, or may be separately expressed and subsequently linked together, for example using chemical linking agents. Ligands expressed from separate nucleic acids will be linked together by appropriate means.
The nucleic acid may further encode a signal sequence for export of the polypeptides from a host cell upon expression and may be fused with a surface component of a filamentous bacteriophage particle (or other component of a selection display system) upon expression. Leader sequences, which may be used in bacterial expresion and/or phage or phagemid display, include pelB, stll, ompA, phoA, bla and pelA.
In a further aspect of the second configuration of the invention the present invention provides a vector comprising nucleic acid according to the present invention.
In a yet further aspect, the present invention provides a host cell transfected with a vector according to the present invention.
Expression from such a vector may be configured to produce, for example on the surface of a bacteriophage particle, epitope binding domains for selection. This allows selection of displayed domains and thus selection of 'multispecific ligands' using the method of the present invention.
hi a preferred embodiment of the second configuration of the invention, the epitope binding domains are immunoglobulin variable regions and are selected from single domain V gene repertoires. Generally the repertoire of single antibody domains is displayed on the surface of filamentous bacteriophage. In a preferred embodiment each single antibody domain is selected by binding of a phage repertoire to antigen. The present invention further provides a kit comprising at least a multispecific ligand according to the present invention, which may be an open conformation or closed conformation ligand. Kits according to the invention may be, for example, diagnostic kits, therapeutic kits, kits for the detection of chemical or biological species, and the like.
In a further aspect still of the second configuration of the invention, the present invention provides a homogenous immunoassay using a ligand according to the present invention.
In a further aspect still of the second configuration of the invention, the present invention provides a composition comprising a closed conformation multispecific ligand, obtainable by a method of the present invention, and a pharmaceutically acceptable carrier, diluent or excipient.
Moreover, the present invention provides a method for the treatment of disease using a 'closed conformation multispecific ligand' or a composition according to the present invention.
In a preferred embodiment of the invention the disease is cancer or an inflammatory disease, eg rheumatoid arthritis, asthma or Crohn's disease.
In a further aspect of the second configuration of the invention, the present invention provides a method for the diagnosis, including diagnosis of disease using a closed conformation multispecific ligand, or a composition according to the present invention. Thus in general the binding of an analyte to a closed conformation multispecific ligand may be exploited to displace an agent, which leads to the generation of a signal on displacement. For example, binding of analyte (second antigen) could displace an enzyme (first antigen) bound to the antibody providing the basis for an immunoassay, especially if the enzyme were held to the antibody through its active site.
Thus in a final aspect of the second configuration, the present invention provides a method for detecting the presence of a target molecule, comprising: (a) providing a closed conformation multispecific ligand bound to an agent, said ligand being specific for the target molecule and the agent, wherein the agent which is bound by the ligand leads to the generation of a detectable signal on displacement from the ligand; (b) exposing the closed conformation multispecific ligand to the target molecule; and (c) detecting the signal generated as a result of the displacement of the agent.
According to the above aspect of the second configuration of the invention, advantageously, the agent is an enzyme, which is inactive when bound by the closed conformation multi-specific ligand. Alternatively, the agent may be any one or more selected from the group consisting of the following: the substrate for an enzyme, and a fluorescent, luminescent or chromogenic molecule which is inactive or quenched when bound by the ligand.
Sequences similar or homologous (e.g., at least about 70% sequence identity) to the sequences disclosed herein are also part of the invention. In some embodiments, the sequence identity at the amino acid level can be about 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or higher. At the nucleic acid level, the sequence identity can be about 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%) or higher. Alternatively, substantial identity exists when the nucleic acid segments will hybridize under selective hybridization conditions (e.g., very high stringency hybridization conditions), to the complement of the strand. The nucleic acids may be present in whole cells, in a cell lysate, or in a partially purified or substantially pure form.
Calculations of "homology" or "sequence identity" or "similarity" between two sequences (the terms are used interchangeably herein) are performed as follows. The sequences are aligned for optimal comparison purposes (e.g., gaps can be introduced in one or both of a first and a second amino acid or nucleic acid sequence for optimal alignment and non-homologous sequences can be disregarded for comparison purposes). In a preferred embodiment, the length of a reference sequence aligned for comparison purposes is at least 30%, preferably at least 40%, more preferably at least 50%, even more preferably at least 60%, and even more preferably at least 70%, 80%, 90%, 100% of the length of the reference sequence. The amino acid residues or nucleotides at corresponding amino acid positions or nucleotide positions are then compared. When a position in the first sequence is occupied by the same amino acid residue or nucleotide as the corresponding position in the second sequence, then the molecules are identical at that position (as used herein amino acid or nucleic acid "homology" is equivalent to amino acid or nucleic acid "identity"). The percent identity between the two sequences is a function of the number of identical positions shared by the sequences, taking into account the number of gaps, and the length of each gap, which need to be introduced for optimal alignment of the two sequences.
Advantageously, the BLAST algorithm (version 2.0) is employed for sequence alignment, with parameters set to default values. The BLAST algorithm is described in detail at the world wide web site ("www") of the National Center for Biotechnology Information (".ncbi") of the National Institutes of Health ("nih") of the U.S. government (".gov"), in the "/Blast/" directory, in the "blast_help.html" file. The search parameters are defined as follows, and are advantageously set to the defined default parameters.
BLAST (Basic Local Alignment Search Tool) is the heuristic search algorithm employed by the programs blastp, blastn, blastx, tblastn, and tblastx; these programs ascribe significance to their findings using the statistical methods of Karlin and Altschul, 1990, Proc. Natl. Acad. Sci. USA 87(6):2264-8 (see the "blast_help.html" file, as described above) with a few enhancements. The BLAST programs were tailored for sequence similarity searching, for example to identify homologues to a query sequence. The programs are not generally useful for motif-style searching. For a discussion of basic issues in similarity searching of sequence databases, see Altschul et al. (1994).
The five BLAST programs available at the National Center for Biotechnology Information web site perform the following tasks:
"blastp" compares an amino acid query sequence against a protein sequence database; "blastn" compares a nucleotide query sequence against a nucleotide sequence database; "blastx" compares the six-frame conceptual translation products of a nucleotide query sequence (both strands) against a protein sequence database;
"tblastn" compares a protein query sequence against a nucleotide sequence database dynamically translated in all six reading frames (both strands). "tblastx" compares the six-frame translations of a nucleotide query sequence against the six-frame translations of a nucleotide sequence database. BLAST uses the following search parameters:
HISTOGRAM Display a histogram of scores for each search; default is yes. (See parameter H in the BLAST Manual).
DESCRIPTIONS Restricts the number of short descriptions of matching sequences reported to the number specified; default limit is 100 descriptions. (See parameter V in the manual page). See also EXPECT and CUTOFF. ALIGNMENTS Restricts database sequences to the number specified for which high- scoring segment pairs (HSPs) are reported; the default limit is 50. If more database sequences than this happen to satisfy the statistical significance threshold for reporting (see EXPECT and CUTOFF below), only the matches ascribed the greatest statistical significance are reported. (See parameter B in the BLAST Manual). EXPECT The statistical significance threshold for reporting matches against database sequences; the default value is 10, such that 10 matches are expected to be found merely by chance, according to the stochastic model of Karlin and Altschul (1990). If the statistical significance ascribed to a match is greater than the EXPECT threshold, the match will not be reported. Lower EXPECT thresholds are more stringent, leading to fewer chance matches being reported. Fractional values are acceptable. (See parameter E in the BLAST Manual).
CUTOFF Cutoff score for reporting high-scoring segment pairs. The default value is calculated from the EXPECT value (see above). HSPs are reported for a database sequence only if the statistical significance ascribed to them is at least as high as would be ascribed to a lone HSP having a score equal to the CUTOFF value. Higher CUTOFF values are more stringent, leading to fewer chance matches being reported. (See parameter S in the BLAST Manual). Typically, significance thresholds can be more intuitively managed using EXPECT.
MATRIX Specify an alternate scoring matrix for BLASTP, BLASTX, TBLASTN and TBLASTX. The default matrix is BLOSUM62 (Henikoff & Henikoff, 1992, Proc. Natl. Aacad. Sci. USA 89(22):10915-9). The valid alternative choices include: PAM40, PAM120, PAM250 and IDENTITY. No alternate scoring matrices are available for BLASTN; specifying the MATRIX directive in BLASTN requests returns an error response. STRAND Restrict a TBLASTN search to just the top or bottom strand of the database sequences; or restrict a BLASTN, BLASTX or TBLASTX search to just reading frames on the top or bottom strand of the query sequence.
FILTER Mask off segments of the query sequence that have low compositional complexity, as determined by the SEG program of Wootton & Federhen (1993) Computers and Chemistry 17:149-163, or segments consisting of short-periodicity internal repeats, as determined by the XNU program of Claverie & States, 1993, Computers and Chemistry 17:191-201, or, for BLASTN, by the DUST program of Tarusov and Lipman (see the world wide web site of the NCBI). Filtering can eliminate statistically significant but biologically uninteresting reports from the blast output (e.g., hits against common acidic-, basic- or proline-rich regions), leaving the more biologically interesting regions of the query sequence available for specific matching against database sequences.
Low complexity sequence found by a filter program is substituted using the letter "N" in nucleotide sequence (e.g., "N" repeated 13 times) and the letter "X" in protein sequences (e.g., "X" repeated 9 times).
Filtering is only applied to the query sequence (or its translation products), not to database sequences. Default filtering is DUST for BLASTN, SEG for other programs.
It is not unusual for nothing at all to be masked by SEG, XNU, or both, when applied to sequences in SWISS-PROT, so filtering should not be expected to always yield an effect. Furthermore, in some cases, sequences are masked in their entirety, indicating that the statistical significance of any matches reported against the unfiltered query sequence should be suspect.
NCBI-gi Causes NCBI gi identifiers to be shown in the output, in addition to the accession and/or locus name.
Most preferably, sequence comparisons are conducted using the simple BLAST search algorithm provided at the NCBI world wide web site described above, in the "/BLAST" directory. Brief Description of the Figures
Figure 1 shows the diversification of VH/HSA at positions H50, H52, H52a, H53,
H55, H56, H58, H95, H96, H97, H98 (DVT or NNK encoded respectively) which are in the antigen binding site of VH HSA. The sequence of VK. is diversified at positions L50, L53.
Figure 2 shows Library 1 : Germline Vκ/DVT VH,
Library 2: Germline Vκ /NNK VH, Library 3 : Germline VH/DVT Vκ
Library 4: Germline VH/NNK VK In phage display/ScFv format. These libraries were pre-selected for binding to generic ligands protein A and protein L so that the majority of the clones and selected libraries are functional. Libraries were selected on HSA (first round) and /3-gal (second round) or HSA 3-gal selection or on
/3-gal (first round) and HSA (second round) /3-gal HSA selection. Soluble scFv from these clones of PCR are amplified in the sequence. One clone encoding a dual specific antibody K8 was chosen for further work.
Figure 3 shows an alignment of VH chains and Vκ chains.
Figure 4 shows the characterisation of the binding properties of the K8 antibody, the binding properties of the K8 antibody characterised by monoclonal phage ELISA, the dual specific K8 antibody was found to bind HSA and /3-gal and displayed on the surface of the phage with absorbant signals greater than 1.0. No cross reactivity with other proteins was detected.
Figure 5 shows soluble scFv ELISA performed using known concentrations of the
K8 antibody fragment. A 96-well plate was coated with lOOμg of HSA, BSA and /3-gal at lOμg/ml and lOOμg/ml of Protein A at 1 μg/ml concentration. 50μg of the serial dilutions of the K8 scFv was applied and the bound antibody fragments were detected with Protein L-HRP. ELISA results confirm the dual specific nature of the K8 antibody. Figure 6 shows the binding characteristics of the clone K8Vκ/dummy VH analysed using soluble scFv ELISA. Production of the soluble scFv fragments was induced by IPTG as described by Harrison et al, Methods Enzymol. 1996;267:83-109 and the supernatant containing scFv assayed directly.
Soluble scFv ELISA is performed as described in example 1 and the bound scFvs were detected with Protein L-HRP. The ELISA results revealed that this clone was still able to bind /3-gal, whereas binding BSA was abolished.
Figure 7 shows the sequence of variable domain vectors 1 and 2.
Figure 8 is a map of the CH vector used to construct a VH1/VH2 multipsecific ligand.
Figure 9 is a map of the Vκ vector used to construct a Vκl/Vκ2 multispecific ligand.
Figure 10 TNF receptor assay comparing TAR1-5 dimer 4, TAR1-5-19 dimer 4 and TAR1-5-19 monomer.
Figure 11 TNF receptor assay comparing TAR1-5 dimers 1-6. All dimers have been FPLC purified and the results for the optimal dimeric species are shown.
Figure 12 TNF receptor assay of TAR1-5 19 homodimers in different formats: dAb- linker-dAb format with 3U, 5U or 7U linker, Fab format and cysteine hinge linker format.
Figure 13 Dummy VH sequence for library 1. The sequence of the VH framework based on germline sequence DP47 - JH4b. Positions where NNK randomisation (N=A or T or C or G nucleotides; K = G or T nucleotides) has been incorporated into library 1 are indicated in bold underlined text.
Figure 14 Dummy VH sequence for library 2. The sequence of the VH framework based on germline sequence DP47 - JH4b. Positions where NNK randomisation (N=A or T or C or G nucleotides; K = G or T nucleotides) has been incorporated into library 2 are indicated in bold underlined text.
Figure 15 Dummy VK sequence for library 3. The sequence of the V/c framework based on germline sequence DPκ9 - J κl- Positions where NNK randomisation (N=A or T or C or G nucleotides; K = G or T nucleotides) has been incorporated into library 3 are indicated in bold underlined text.
Figure 16 Nucleotide and amino acid sequence of anti MSA dAbs MSA 16 and MSA 26.
Figure 17 Inhibition biacore of MSA 16 and 26. Purified dAbs MSA16 and MSA26 were analysed by inhibition biacore to determine Kd. Briefly, the dAbs were tested to determine the concentration of dAb required to achieve 200RUs of response on a biacore CM5 chip coated with a high density of
MSA. Once the required concentrations of dAb had been determined, MSA antigen at a range of concentrations around the expected Kd was premixed with the dAb and incubated overnight. Binding to the MSA coated biacore chip of dAb in each of the premixes was then measured at a high flow-rate of 30 μl/minute.
Figure 18 Serum levels of MSA16 following injection. Serum half life of the dAb MSA16 was determined in mouse. MSA16 was dosed as single i.v. injections at approx 1.5mg/kg into CD1 mice. Modelling with a 2 compartment model showed MSA16 had a tl/2α of 0.98hr, a tl/2/3 of
36.5hr and an AUC of 913hr.mg/ml. MSA16 had a considerably lengthened half life compared with HEL4 (an anti-hen egg white lysozyme dAb) which had a tl/2α of 0.06hr and a tl/2/3 of 0.34hr.
Figure 19 ELISA (a) and TNF receptor assay (c) showing inhibition of TNF binding with a Fab-like fragment comprising MSA26Ck and TAR1-5-19CH. Addition of MSA with the Fab-like fragment reduces the level of inhibition. An ELISA plate coated with 1 μg/ml TNFα; was probed with dual specific VK CH and VK CK Fab like fragment and also with a control TNFα: binding dAb at a concentration calculated to give a similar signal on the ELISA. Both the dual specific and control dAb were used to probe the ELISA plate in the presence and in the absence of 2mg/ml MSA. The signal in the dual specific well was reduced by more than 50% but the signal in the dAb well was not reduced at all (see figure 19a). The same dual specific protein was also put into the receptor assay with and without MSA and competition by MSA was also shown (see figure 19c). This demonstrates that binding of MSA to the dual specific is competitive with binding to TNFα!.
Figure 20 TNF receptor assay showing inhibiton of TNF binding with a disulphide bonded heterodimer of TAR1-5-19 dAb and MSA16 dAb. Addition of MSA with the dimer reduces the level of inhibiton in a dose dependant manner. The TNF receptor assay (figure 19 (b)) was conducted in the presence of a constant concentration of heterodimer (18nM) and a dilution series of MSA and HSA. The presence of HSA at a range of concentrations (up to 2 mg/ml) did not cause a reduction in the ability of the dimer to inhibit TNFα; . However, the addition of MSA caused a dose dependant reduction in the ability of the dimer to inhibit TNFo; (figure
19a) .This demonstrates that MSA and TNFo; compete for binding to the cys bonded TAR1-5-19, MSA16 dimer. MSA and HSA alone did not have an effect on the TNF binding level in the assay.
Detailed Description of the Invention
Definitions
Complementary Two immunoglobulin domains are "complementary" where they belong to families of structures which form cognate pairs or groups or are derived from such families and retain this feature. For example, a H domain and a VL domain of an antibody are complementary; two VH domains are not complementary, and two domains are not complementary. Complementary domains may be found in other members of the immunoglobulin superfamily, such as the Vα and Vβ (or γ and δ) domains of the T-cell receptor. In the context of the second configuration of the present invention, non-complementary domains do not bind a target molecule cooperatively, but act independently on different target epitopes which may be on the same or different molecules. Domains which are artificial, such as domains based on protein scaffolds which do not bind epitopes unless engineered to do so, are non-complementary. Likewise, two domains based on (for example) an immunoglobulin domain and a fibronectin domain are not complementary.
Immunoglobulin This refers to a family of polypeptides which retain the immunoglobulin fold characteristic of antibody molecules, which contains two β sheets and, usually, a conserved disulphide bond. Members of the immunoglobulin superfamily are involved in many aspects of cellular and non-cellular interactions in vivo, including widespread roles in the immune system (for example, antibodies, T-cell receptor molecules and the like), involvement in cell adhesion (for example the ICAM molecules) and intracellular signalling (for example, receptor molecules, such as the PDGF receptor). The present invention is applicable to all immunoglobulin superfamily molecules which possess binding domains. Preferably, the present invention relates to antibodies.
Combining Variable domains according to the invention are combined to form a group of domains; for example, complementary domains may be combined, such as VL domains being combined with VH domains. Non-complementary domains may also be combined. Domains may be combined in a number of ways, involving linkage of the domains by covalent or non-covalent means.
Domain A domain is a folded protein structure which retains its tertiary structure independently of the rest of the protein. Generally, domains are responsible for discrete functional properties of proteins, and in many cases may be added, removed or transferred to other proteins without loss of function of the remainder of the protein and/or of the domain. By single antibody variable domain is meant a folded polypeptide domain comprising sequences characteristic of antibody variable domains. It therefore includes complete antibody variable domains and modified variable domains, for example in which one or more loops have been replaced by sequences which are not characteristic of antibody variable domains, or antibody variable domains which have been truncated or comprise N- or C-terminal extensions, as well as folded fragments of variable domains which retain at least in part the binding activity and specificity of the full-length domain.
Repertoire A collection of diverse variants, for example polypeptide variants which differ in their primary sequence. A library used in the present invention will encompass a repertoire of polypeptides comprising at least 1000 members.
Library The term library refers to a mixture of heterogeneous polypeptides or nucleic acids. The library is composed of members, each of which have a single polypeptide or nucleic acid sequence. To this extent, library is synonymous with repertoire. Sequence differences between library members are responsible for the diversity present in the library. The library may take the form of a simple mixture of polypeptides or nucleic acids, or may be in the form of orgamsms or cells, for example bacteria, viruses, animal or plant cells and the like, transformed with a library of nucleic acids. Preferably, each individual organism or cell contains only one or a limited number of library members. Advantageously, the nucleic acids are incorporated into expression vectors, in order to allow expression of the polypeptides encoded by the nucleic acids. In a preferred aspect, therefore, a library may take the form of a population of host organisms, each organism containing one or more copies of an expression vector containing a single member of the library in nucleic acid form which can be expressed to produce its corresponding polypeptide member. Thus, the population of host organisms has the potential to encode a large repertoire of genetically diverse polypeptide variants.
A 'closed conformation multi-specific ligand' describes a multi-specific ligand as herein defined comprising at least two epitope binding domains as herein defined. The term 'closed conformation' (multi-specific ligand) means that the epitope binding domains of the ligand are arranged such that epitope binding by one epitope binding domain competes with epitope binding by another epitope binding domain. That is, cognate epitopes may be bound by each epitope binding domain individually but not simultaneosuly. The closed conformation of the ligand can be achieved using methods herein described. Antibody An antibody (for example IgG, IgM, IgA, IgD or IgE) or fragment (such as a Fab , F(ab') , Fv, disulphide linked Fv, scFv, closed conformation multispecific antibody, disulphide-linked scFv, diabody) whether derived from any species naturally producing an antibody, or created by recombinant DNA technology; whether isolated from serum, B-cells, hybridomas, transfectomas, yeast or bacteria).
Dual-specific ligand A ligand comprising a first immunoglobulin single variable domain and a second immunoglobulin single variable domain as herein defined, wherein the variable regions are capable of binding to two different antigens or two epitopes on the same antigen which are not normally bound by a monospecific immunoglobulin. For example, the two epitopes may be on the same hapten, but are not the same epitope or sufficiently adjacent to be bound by a monospecific ligand. The dual specific ligands according to the invention are composed of variable domains which have different specificities, and do not contain mutually complementary variable domain pairs which have the same specificity.
Antigen A molecule that is bound by a ligand according to the present invention.
Typically, antigens are bound by antibody ligands and are capable of raising an antibody response in vivo. It may be a polypeptide, protein, nucleic acid or other molecule. Generally, the dual specific ligands according to the invention are selected for target specificity against a particular antigen. In the case of conventional antibodies and fragments thereof, the antibody binding site defined by the variable loops (LI, L2, L3 and HI, H2, H3) is capable of binding to the antigen.
Epitope A unit of structure conventionally bound by an immunoglobulin VH/VL pair. Epitopes define the minimum binding site for an antibody, and thus represent the target of specificity of an antibody. In the case of a single domain antibody, an epitope represents the unit of structure bound by a variable domain in isolation.
Generic ligand A ligand that binds to all members of a repertoire. Generally, not bound through the antigen binding site as defined above. Non-limiting examples include protein A, protein L and protein G. Selecting Derived by screening, or derived by a Darwinian selection process, in which binding interactions are made between a domain and the antigen or epitope or between an antibody and an antigen or epitope. Thus a first variable domain may be selected for binding to an antigen or epitope in the presence or in the absence of a complementary variable domain.
Universal framework A single antibody framework sequence corresponding to the regions of an antibody conserved in sequence as defined by Kabat ("Sequences of Proteins of Immunological Interest", US Department of Health and Human Services) or corresponding to the human germline immunoglobulin repertoire or structure as defined by Chothia and Lesk, (1987) J. Mol. Biol. 196:910-917. The invention provides for the use of a single framework, or a set of such frameworks, which has been found to permit the derivation of virtually any binding specificity though variation in the hypervariable regions alone.
Half-life The time taken for the serum concentration of the ligand to reduce by 50%, in vivo, for example due to degradation of the ligand and/or clearance or sequestration of the ligand by natural mechanisms. The ligands of the invention are stabilised in vivo and their half-life increased by binding to molecules which resist degradation and/or clearance or sequestration. Typically, such molecules are naturally occurring proteins which themselves have a long half-life in vivo. The half-life of a ligand is increased if its functional activity persists, in vivo, for a longer period than a similar ligand which is not specific for the half-life increasing molecule. Thus, a ligand specific for HSA and a target molecule is compared with the same ligand wherein the specificity for HSA is not present, that it does not bind HSA but binds another molecule. For example, it may bind a second epitope on the target molecule. Typically, the half life is increased by 10%, 20%, 30%, 40%, 50% or more. Increases in the range of 2x, 3x, 4x, 5x, lOx, 20x, 30x, 40x, 5 Ox or more of the half life are possible. Alternatively, or in addition, increases in the range of up to 30x, 40x, 50x, 60x, 70x, 80x, 90x, lOOx, 150x of the half life are possible.
Homogeneous immunoassay An immunoassay in which analyte is detected without need for a step of separating bound and un-bound reagents. Substantially identical (or "substantially homologous") A first amino acid or nucleotide sequence that contains a sufficient number of identical or equivalent (e.g., with a similar side chain, e.g., conserved amino acid substitutions) amino acid residues or nucleotides to a second amino acid or nucleotide sequence such that the first and second amino acid or nucleotide sequences have similar activities. In the case of antibodies, the second antibody has the same binding specificity and has at least 50% of the affinity of the same.
As used herein, the terms "low stringency," "medium stringency," "high stringency," or "very high stringency conditions" describe conditions for nucleic acid hybridization and washing. Guidance for performing hybridization reactions can be found in Current Protocols in Molecular Biology, John Wiley & Sons, N.Y. (1989), 6.3.1-6.3.6, which is incoφorated herein by reference in its entirety. Aqueous and nonaqueous methods are described in that reference and either can be used. Specific hybridization conditions referred to herein are as follows: (1) low stringency hybridization conditions in 6X sodium chloride/sodium citrate (SSC) at about 45°C, followed by two washes in 0.2X SSC, 0.1% SDS at least at 50°C (the temperature of the washes can be increased to 55°C for low stringency conditions); (2) medium stringency hybridization conditions in 6X SSC at about 45°C, followed by one or more washes in 0.2X SSC, 0.1% SDS at 60°C; (3) high stringency hybridization conditions in 6X SSC at about 45°C, followed by one or more washes in 0.2X SSC, 0.1% SDS at 65°C; and preferably (4) very high stringency hybridization conditions are 0.5M sodium phosphate, 7% SDS at 65°C, followed by one or more washes at 0.2X SSC, 1% SDS at 65°C. Very high stringency conditions (4) are the preferred conditions and the ones that should be used unless otherwise specified.
Detailed Description of the Invention
Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art (e.g., in cell culture, molecular genetics, nucleic acid chemistry, hybridisation techniques and biochemistry). Standard techniques are used for molecular, genetic and biochemical methods (see generally, Sambrook et al, Molecular Cloning: A Laboratory Manual, 2d ed. (1989) Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. and Ausubel et al, Short Protocols in Molecular Biology (1999) 4th Ed, John Wiley & Sons, Inc. which are incorporated herein by reference) and chemical methods.
Preparation of immunoglobulin based multi-specific ligands
Dual specific ligands according to the invention, whether open or closed in conformation according to the desired configuration of the invention, may be prepared according to previously established techniques, used in the field of antibody engineering, for the preparation of scFv, "phage" antibodies and other engineered antibody molecules. Techniques for the preparation of antibodies, and in particular bispecific antibodies, are for example described in the following reviews and the references cited therein: Winter & Milstein, (1991) Nature 349:293-299; Plueckthun (1992) Immunological Reviews 130:151-188; Wright et al, (1992) Crti. Rev. Immunol.l2:125-168; Holliger, P. & Winter, G. (1993) Curr. Op. Biotechn. 4, 446-449; Carter, et al. (1995) J. Hematother. 4, 463-470; Chester, K.A. & Hawkins, R.E. (1995) Trends Biotechn. 13, 294-300; Hoogenboom, H.R. (1997) Nature Biotechnol. 15, 125-126; Fearon, D. (1997) Nature Biotechnol. 15, 618-619; Plϋckthun, A. & Pack, P. (1997) hnmunotechnology 3, 83-105; Carter, P. & Merchant, A.M. (1997) Curr. Opin. Biotechnol. 8, 449-454; Holliger, P. & Winter, G. (1997) Cancer Immunol, hnmunother. 45,128-130.
The invention provides for the selection of variable domains against two different antigens or epitopes, and subsequent combination of the variable domains.
The techniques employed for selection of the variable domains employ libraries and selection procedures which are known in the art. Natural libraries (Marks et al. (1991) J. Mol. Biol, 222: 581; Vaughan et al. (1996) Nature Biotech., 14: 309) which use rearranged V genes harvested from human B cells are well known to those skilled in the art. Synthetic libraries (Hoogenboom & Winter (1992) J Mol. Biol, 111: 381; Barbas et al (1992) Proc. Natl Acad. Sci. USA, 89: 4457; Nissim et al. (1994) EMBO J, 13: 692; Griffiths et al (1994) EMBO J, 13: 3245; De Kruif et al (1995) J Mol Biol, 248: 97) are prepared by cloning immunoglobulin V genes, usually using PCR. Errors in the PCR process can lead to a high degree of randomisation. VH and/or VL libraries may be selected against target antigens or epitopes separately, in which case single domain binding is directly selected for, or together.
A preferred method for making a dual specific ligand according to the present invention comprises using a selection system in which a repertoire of variable domains is selected for binding to a first antigen or epitope and a repertoire of variable domains is selected for binding to a second antigen or epitope. The selected variable first and second variable domains are then combined and the dual-specific ligand selected for binding to both first and second antigen or epitope. Closed conformation ligands are selected for binding both first and second antigen or epitope in isolation but not simultaneously.
A. Library vector systems
A variety of selection systems are known in the art which are suitable for use in the present invention. Examples of such systems are described below.
Bacteriophage lambda expression systems may be screened directly as bacteriophage plaques or as colonies of lysogens, both as previously described (Huse et al. (1989) Science, 246: 1275; Caton and Koprowski (1990) Proc. Natl. Acad. Sci. U.S.A., 87; Mullinax et al. (1990) Proc. Natl Acad. Sci. U.S.A., 87: 8095; Persson et al. (1991) Proc. Natl. Acad. Sci. U.S.A., 88: 2432) and are of use in the invention. Whilst such expression systems can be used to screen up to 106 different members of a library, they are not really suited to screening of larger numbers (greater than 106 members).
Of particular use in the construction of libraries are selection display systems, which enable a nucleic acid to be linked to the polypeptide it expresses. As used herein, a selection display system is a system that permits the selection, by suitable display means, of the individual members of the library by binding the generic and/or target ligands.
Selection protocols for isolating desired members of large libraries are known in the art, as typified by phage display techniques. Such systems, in which diverse peptide sequences are displayed on the surface of filamentous bacteriophage (Scott and Smith (1990) Science, 249: 386), have proven useful for creating libraries of antibody fragments (and the nucleotide sequences that encoding them) for the in vitro selection and amplification of specific antibody fragments that bind a target antigen (McCafferty et al, WO 92/01047). The nucleotide sequences encoding the VH and VL regions are linked to gene fragments which encode leader signals that direct them to the periplasmic space of E. coli and as a result the resultant antibody fragments are displayed on the surface of the bacteriophage, typically as fusions to bacteriophage coat proteins (e.g., pill or pVIII). Alternatively, antibody fragments are displayed externally on lambda phage capsids (phagebodies). An advantage of phage-based display systems is that, because they are biological systems, selected library members can be amplified simply by growing the phage containing the selected library member in bacterial cells. Furthermore, since the nucleotide sequence that encode the polypeptide library member is contained on a phage or phagemid vector, sequencing, expression and subsequent genetic manipulation is relatively straightforward.
Methods for the construction of bacteriophage antibody display libraries and lambda phage expression libraries are well known in the art (McCafferty et al. (1990) Nature, 348: 552; Kang et al (1991) Proc. Natl. Acad. Sci. U.S.A., 88: 4363; Clackson et al.
(1991) Nature, 352: 624; Lowman et al. (1991) Biochemistry, 30: 10832; Burton et al. (1991) Proc. Natl Acad. Sci U.S.A., 88: 10134; Hoogenboom et al. (1991) Nucleic Acids
Res., 19: 4133; Chang et al. (1991) J. Immunol, 147: 3610; Breifling et al. (1991) Gene, 104: 147; Marks et al. (1991) supra; Barbas et al. (1992) supra; Hawkins and Winter
(1992) J. Immunol, 22: 867; Marks et al, 1992, J. Biol. Chem., 267: 16007; Lerner et al. (1992) Science, 258: 1313, incorporated herein by reference).
One particularly advantageous approach has been the use of scFv phage-libraries (Huston et al, 1988, Proc. Natl. Acad. Sci U.S.A., 85: 5879-5883; Chaudhary et al. (1990) Proc. Natl. Acad. Sci U.S.A., 87: 1066-1070; McCafferty et al (1990) supra; Clackson et al. (1991) Nature, 352: 624; Marks et al. (1991) J. Mol Biol, 222: 581; Chiswell et al. (1992) Trends Biotech., 10: 80; Marks et al. (1992) J. Biol Chem., 267). Various embodiments of scFv libraries displayed on bacteriophage coat proteins have been described. Refinements of phage display approaches are also known, for example as described in WO96/06213 and WO92/01047 (Medical Research Council et al) and WO97/08320 (Morphosys), which are incorporated herein by reference.
Other systems for generating libraries of polypeptides involve the use of cell-free enzymatic machinery for the in vitro synthesis of the library members. In one method,
RNA molecules are selected by alternate rounds of selection against a target ligand and
PCR amplification (Tuerk and Gold (1990) Science, 249: 505; Ellington and Szostak
(1990) Nature, 346: 818). A similar technique may be used to identify DNA sequences which bind a predetermined human transcription factor (Thiesen and Bach (1990) Nucleic Acids Res., 18: 3203; Beaudry and Joyce (1992) Science, 257: 635; WO92/05258 and
WO92/14843). In a similar way, in vitro translation can be used to synthesise polypeptides as a method for generating large libraries. These methods which generally comprise stabilised polysome complexes, are described further in WO88/08453,
WO90/05785, WO90/07003, WO91/02076, WO91/05058, and WO92/02536. Alternative display systems which are not phage-based, such as those disclosed in WO95/22625 and
WO95/11922 (Affymax) use the polysomes to display polypeptides for selection.
A still further category of techniques involves the selection of repertoires in artificial compartments, which allow the linkage of a gene with its gene product. For example, a selection system in which nucleic acids encoding desirable gene products may be selected in microcapsules formed by water-in-oil emulsions is described in WO99/02671, WO00/40712 and Tawfik & Griffiths (1998) Nαtwre Biotechnol 16(7), 652-6. Genetic elements encoding a gene product having a desired activity are compartmentalised into microcapsules and then transcribed and/or translated to produce their respective gene products (RΝA or protein) within the microcapsules. Genetic elements which produce gene product having desired activity are subsequently sorted. This approach selects gene products of interest by detecting the desired activity by a variety of means.
B. Library Construction.
Libraries intended for selection, may be constructed using techniques known in the art, for example as set forth above, or may be purchased from commercial sources. Libraries which are useful in the present invention are described, for example, in WO99/20749. Once a vector system is chosen and one or more nucleic acid sequences encoding polypeptides of interest are cloned into the library vector, one may generate diversity within the cloned molecules by undertaking mutagenesis prior to expression; alternatively, the encoded proteins may be expressed and selected, as described above, before mutagenesis and additional rounds of selection are performed. Mutagenesis of nucleic acid sequences encoding structurally optimised polypeptides is carried out by standard molecular methods. Of particular use is the polymerase chain reaction, or PCR, (Mullis and Faloona (1987) Methods Enzymol, 155: 335, herein incorporated by reference). PCR, which uses multiple cycles of DNA replication catalysed by a thermostable, DNA-dependent DNA polymerase to amplify the target sequence of interest, is well known in the art. The construction of various antibody libraries has been discussed in Winter et al. (1994) Ann. Rev. Immunology 12, 433-55, and references cited therein.
PCR is performed using template DNA (at least lfg; more usefully, 1-1000 ng) and at least 25 pmol of oligonucleotide primers; it may be advantageous to use a larger amount of primer when the primer pool is heavily heterogeneous, as each sequence is represented by only a small fraction of the molecules of the pool, and amounts become limiting in the later amplification cycles. A typical reaction mixture includes: 2μl of DNA, 25 pmol of oligonucleotide primer, 2.5 μl of 10X PCR buffer 1 (Perkin-Elmer, Foster City, CA), 0.4 μl of 1.25 μM dNTP, 0.15 μl (or 2.5 units) of Taq DNA polymerase (Perkin Elmer, Foster City, CA) and deionized water to a total volume of 25 μl. Mineral oil is overlaid and the PCR is performed using a programmable thermal cycler. The length and temperature of each step of a PCR cycle, as well as the number of cycles, is adjusted in accordance to the stringency requirements in effect. Annealing temperature and timing are determined both by the efficiency with which a primer is expected to anneal to a template and the degree of mismatch that is to be tolerated; obviously, when nucleic acid molecules are simultaneously amplified and mutagenised, mismatch is required, at least in the first round of synthesis. The ability to optimise the stringency of primer annealing conditions is well within the knowledge of one of moderate skill in the art. An annealing temperature of between 30 °C and 72 °C is used. Initial denaturation of the template molecules normally occurs at between 92°C and 99°C for 4 minutes, followed by 20-40 cycles consisting of denaturation (94-99°C for 15 seconds to 1 minute), annealing (temperature determined as discussed above; 1-2 minutes), and extension (72°C for 1-5 minutes, depending on the length of the amplified product). Final extension is generally for 4 minutes at 72°C, and may be followed by an indefinite (0-24 hour) step at 4°C.
C. Combining single variable domains
Domains useful in the invention, once selected, may be combined by a variety of methods known in the art, including covalent and non-covalent methods.
Preferred methods include the use of polypeptide linkers, as described, for example, in connection with scFv molecules (Bird et al, (1988) Science 242:423-426). Discussion of suitable linkers is provided in Bird et al. Science 242, 423-426; Hudson et al , Journal Immunol Methods 231 (1999) 177-189; Hudson et al, Proc Nat Acad Sci USA 85, 5879- 5883. Linkers are preferably flexible, allowing the two single domains to interact. One linker example is a (Gly4 Ser)n linker, where n=l to 8, eg, 2, 3, 4, 5 or 7. The linkers used in diabodies, which are less flexible, may also be employed (Holliger et al, (1993) PNAS (USA) 90:6444-6448).
In one embodiment, the linker employed is not an immunoglobulin hinge region.
Variable domains may be combined using methods other than linkers. For example, the use of disulphide bridges, provided through naturally-occurring or engineered cysteine residues, may be exploited to stabilise VH"VH>VL"VL or VH-VL dimers (Reiter et al, (1994) Protein Eng. 7:697-704) or by remodelling the interface between the variable domains to improve the "fit" and thus the stability of interaction (Ridgeway et al, (1996) Protein Eng. 7:617-621; Zhu et al, (1997) Protein Science 6:781-788).
Other techniques for joining or stabilising variable domains of immunoglobulins, and in particular antibody VH domains, may be employed as appropriate.
In accordance with the present invention, dual specific ligands can be in "closed" conformations in solution. A "closed" configuration is that in which the two domains (for example VH and VL) are present in associated form, such as that of an associated VH-VL pair which forms an antibody binding site. For example, scFv may be in a closed conformation, depending on the arrangement of the linker used to link the VH and VL domains. If this is sufficiently flexible to allow the domains to associate, or rigidly holds them in the associated position, it is likely that the domains will adopt a closed conformation.
Similarly, VH domain pairs and VL domain pairs may exist in a closed conformation. Generally, this will be a function of close association of the domains, such as by a rigid linker, in the ligand molecule. Ligands in a closed conformation will be unable to bind both the molecule which increases the half-life of the ligand and a second target molecule. Thus, the ligand will typically only bind the second target molecule on dissociation from the molecule which increases the half-life of the ligand.
Moreover, the construction of VH/VH, VL/VL or VH/VL dimers without linkers provides for competition between the domains.
Ligands according to the invention may moreover be in an open conformation. In such a conformation, the ligands will be able to simultaneously bind both the molecule which increases the half-life of the ligand and the second target molecule. Typically, variable domains in an open configuration are (in the case of VH-VL pairs) held far enough apart for the domains not to interact and form an antibody binding site and not to compete for binding to their respective epitopes. In the case of VH/VH or Vi/vY dimers, the domains are not forced together by rigid linkers. Naturally, such domain pairings will not compete for antigen binding or form an antibody binding site.
Fab fragments and whole antibodies will exist primarily in the closed conformation, although it will be appreciated that open and closed dual specific ligands are likely to exist in a variety of equilibria under different circumstances. Binding of the ligand to a target is likely to shift the balance of the equilibrium towards the open configuration. Thus, certain ligands according to the invention can exist in two conformations in solution, one of which (the open form) can bind two antigens or epitopes independently, whilst the alternative conformation (the closed form) can only bind one antigen or epitope; antigens or epitopes thus compete for binding to the ligand in this conformation.
Although the open form of the dual specific ligand may thus exist in equilibrium with the closed form in solution, it is envisaged that the equilibrium will favour the closed form; moreover, the open form can be sequestered by target binding into a closed conformation. Preferably, therefore, certain dual specific ligands of the invention are present in an equilibrium between two (open and closed) conformations.
Dual specific ligands according to the invention may be modified in order to favour an open or closed conformation. For example, stabilisation of VH-VL interactions with disulphide bonds stabilises the closed conformation. Moreover, linkers used to join the domains, including VH domain and VL domain pairs, may be constructed such that the open from is favoured; for example, the linkers may sterically hinder the association of the domains, such as by incorporation of large amino acid residues in opportune locations, or the designing of a suitable rigid structure which will keep the domains physically spaced apart.
D. Characterisation of the dual-specific ligand.
The binding of the dual-specific ligand to its specific antigens or epitopes can be tested by methods which will be familiar to those skilled in the art and include ELISA. hi a preferred embodiment of the invention binding is tested using monoclonal phage ELISA.
Phage ELISA may be performed according to any suitable procedure: an exemplary protocol is set forth below.
Populations of phage produced at each round of selection can be screened for binding by ELISA to the selected antigen or epitope, to identify "polyclonal" phage antibodies. Phage from single infected bacterial colonies from these populations can then be screened by ELISA to identify "monoclonal" phage antibodies. It is also desirable to screen soluble antibody fragments for binding to antigen or epitope, and this can also be undertaken by ELISA using reagents, for example, against a C- or N-terminal tag (see for example Winter et al. (1994) Ann. Rev. Immunology 12, 433-55 and references cited therein.
The diversity of the selected phage monoclonal antibodies may also be assessed by gel electrophoresis of PCR products (Marks et al. 1991, supra; Nissim et al. 1994 supra), probing (Tomlinson et al, 1992) J. Mol. Biol. 227, 776) or by sequencing of the vector DNA.
Structure of 'Dual-specific ligands'.
As described above, an antibody is herein defined as an antibody (for example IgG, IgM, IgA, IgA, IgE) or fragment (Fab, Fv, disulphide linked Fv, scFv, diabody) which comprises at least one heavy and a light chain variable domain, at least two heavy chain variable domains or at least two light chain variable domains. It may be at least partly derived from any species naturally producing an antibody, or created by recombinant DNA technology; whether isolated from serum, B-cells, hybridomas, transfectomas, yeast or bacteria).
In a preferred embodiment of the invention the dual-specific ligand comprises at least one single heavy chain variable domain of an antibody and one single light chain variable domain of an antibody, or two single heavy or light chain variable domains. For example, the ligand may comprise a VH/VL pair, a pair of VH domains or a pair of VL domains.
The first and the second variable domains of such a ligand may be on the same polypeptide chain. Alternatively they may be on separate polypeptide chains. In the case that they are on the same polypeptide chain they may be linked by a linker, which is preferentially a peptide sequence, as described above.
The first and second variable domains may be covalently or non-covalently associated. In the case that they are covalently associated, the covalent bonds may be disulphide bonds.
In the case that the variable domains are selected from V-gene repertoires selected for instance using phage display technology as herein described, then these variable domains comprise a universal framework region, such that is they may be recognised by a specific generic ligand as herein defined. The use of universal frameworks, generic ligands and the like is described in WO99/20749.
Where V-gene repertoires are used variation in polypeptide sequence is preferably located within the structural loops of the variable domains. The polypeptide sequences of either variable domain may be altered by DNA shuffling or by mutation in order to enhance the interaction of each variable domain with its complementary pair. DNA shuffling is known in the art and taught, for example, by Stemmer, 1994, Nature 370: 389-391 and U.S. Patent No. 6,297,053, both of which are incorporated herein by reference. Other methods of mutagenesis are well known to those of skill in the art.
In a preferred embodiment of the invention the 'dual-specific ligand' is a single chain Fv fragment. In an alternative embodiment of the invention, the 'dual-specific ligand' consists of a Fab format.
In a further aspect, the present invention provides nucleic acid encoding at least a 'dual- specific ligand' as herein defined.
One skilled in the art will appreciate that, depending on the aspect of the invention, both antigens or epitopes may bind simultaneously to the same antibody molecule. Alternatively, they may compete for binding to the same antibody molecule. For example, where both epitopes are bound simultaneously, both variable domains of a dual specific ligand are able to independently bind their target epitopes. Where the domains compete, the one variable domain is capable of binding its target, but not at the same time as the other variable domain binds its cognate target; or the first variable domain is capable of binding its target, but not at the same time as the second variable domain binds its cognate target.
The variable regions may be derived from antibodies directed against target antigens or epitopes. Alternatively they may be derived from a repertoire of single antibody domains such as those expressed on the surface of filamentous bacteriophage. Selection may be performed as described below. In general, the nucleic acid molecules and vector constructs required for the performance of the present invention may be constructed and manipulated as set forth in standard laboratory manuals, such as Sambrook et al. (1989) Molecular Cloning: A Laboratory Manual, Cold Spring Harbor, USA.
The manipulation of nucleic acids useful in the present invention is typically carried out in recombinant vectors.
Thus in a further aspect, the present invention provides a vector comprising nucleic acid encoding at least a 'dual-specific ligand' as herein defined.
As used herein, vector refers to a discrete element that is used to introduce heterologous DNA into cells for the expression and/or replication thereof. Methods by which to select or construct and, subsequently, use such vectors are well known to one of ordinary skill in the art. Numerous vectors are publicly available, including bacterial plasmids, bacteriophage, artificial chromosomes and episomal vectors. Such vectors may be used for simple cloning and mutagenesis; alternatively gene expression vector is employed. A vector of use according to the invention may be selected to accommodate a polypeptide coding sequence of a desired size, typically from 0.25 kilobase (kb) to 40 kb or more in length A suitable host cell is transformed with the vector after in vitro cloning manipulations. Each vector contains various functional components, which generally include a cloning (or "polylinker") site, an origin of replication and at least one selectable marker gene. If given vector is an expression vector, it additionally possesses one or more of the following: enhancer element, promoter, transcription termination and signal sequences, each positioned in the vicinity of the cloning site, such that they are operatively linked to the gene encoding a ligand according to the invention.
Both cloning and expression vectors generally contain nucleic acid sequences that enable the vector to replicate in one or more selected host cells. Typically in cloning vectors, this sequence is one that enables the vector to replicate independently of the host chromosomal DNA and includes origins of replication or autonomously replicating sequences. Such sequences are well known for a variety of bacteria, yeast and viruses. The origin of replication from the plasmid pBR322 is suitable for most Gram-negative bacteria, the 2 micron plasmid origin is suitable for yeast, and various viral origins (e.g.
SV 40, adenovirus) are useful for cloning vectors in mammalian cells. Generally, the origin of replication is not needed for mammalian expression vectors unless these are used in mammalian cells able to replicate high levels of DNA, such as COS cells.
Advantageously, a cloning or expression vector may contain a selection gene also referred to as selectable marker. This gene encodes a protein necessary for the survival or growth of transformed host cells grown in a selective culture medium. Host cells not transformed with the vector containing the selection gene will therefore not survive in the culture medium. Typical selection genes encode proteins that confer resistance to antibiotics and other toxins, e.g. ampicillin, neomycin, methotrexate or tetracycline, complement auxotrophic deficiencies, or supply critical nutrients not available in the growth media.
Since the replication of vectors encoding a ligand according to the present invention is most conveniently performed in E. coli, an E. co/z-selectable marker, for example, the β- lactamase gene that confers resistance to the antibiotic ampicillin, is of use. These can be obtained from E. coli plasmids, such as pBR322 or a pUC plasmid such as pUC18 or pUC19.
Expression vectors usually contain a promoter that is recognised by the host organism and is operably linked to the coding sequence of interest. Such a promoter may be inducible or constitutive. The term "operably linked" refers to a juxtaposition wherein the components described are in a relationship permitting them to function in their intended manner. A control sequence "operably linked" to a coding sequence is ligated in such a way that expression of the coding sequence is achieved under conditions compatible with the control sequences.
Promoters suitable for use with prokaryotic hosts include, for example, the β-lactamase and lactose promoter systems, alkaline phosphatase, the tryptophan (trp) promoter system and hybrid promoters such as the tac promoter. Promoters for use in bacterial systems will also generally contain a Shine-Delgarno sequence operably linked to the coding sequence.
The preferred vectors are expression vectors that enables the expression of a nucleotide sequence corresponding to a polypeptide library member. Thus, selection with the first and/or second antigen or epitope can be performed by separate propagation and expression of a single clone expressing the polypeptide library member or by use of any selection display system. As described above, the preferred selection display system is bacteriophage display. Thus, phage or phagemid vectors may be used, eg pITl or pIT2. Leader sequences useful in the invention include pelB, stll, ompA, phoA, bla and pelA. One example are phagemid vectors which have an E. coli. origin of replication (for double stranded replication) and also a phage origin of replication (for production of single-stranded DNA). The manipulation and expression of such vectors is well known in the art (Hoogenboom and Winter (1992) supra; Nissim et al. (1994) supra). Briefly, the vector contains a β-lactamase gene to confer selectivity on the phagemid and a lac promoter upstream of a expression cassette that consists (N to C terminal) of a pelB leader sequence (which directs the expressed polypeptide to the periplasmic space), a multiple cloning site (for cloning the nucleotide version of the library member), optionally, one or more peptide tag (for detection), optionally, one or more TAG stop codon and the phage protein pill. Thus, using various suppressor and non-suppressor strains of E. coli and with the addition of glucose, iso-propyl thio-β-D-galactoside (IPTG) or a helper phage, such as VCS Ml 3, the vector is able to replicate as a plasmid with no expression, produce large quantities of the polypeptide library member only or produce phage, some of which contain at least one copy of the polypeptide-pIII fusion on their surface.
Construction of vectors encoding ligands according to the invention employs conventional ligation techniques. Isolated vectors or DNA fragments are cleaved, tailored, and religated in the form desired to generate the required vector. If desired, analysis to confirm that the correct sequences are present in the constructed vector can be performed in a known fashion. Suitable methods for constructing expression vectors, preparing in vitro transcripts, introducing DNA into host cells, and performing analyses for assessing expression and function are known to those skilled in the art. The presence of a gene sequence in a sample is detected, or its amplification and/or expression quantified by conventional methods, such as Southern or Northern analysis, Western blotting, dot blotting of DNA, RNA or protein, in situ hybridisation, immunocytochemistry or sequence analysis of nucleic acid or protein molecules. Those skilled in the art will readily envisage how these methods may be modified, if desired.
Structure of closed conformation multispecific lisands
According to one aspect of the second configuration of the invention present invention, the two or more non-complementary epitope binding domains are linked so that they are in a closed conformation as herein defined. Advantageously, they may be further attached to a skeleton which may, as a alternative, or on addition to a linker described herein, facilitate the formation and/or maintenance of the closed conformation of the epitope binding sites with respect to one another.
CD Skeletons
Skeletons may be based on immunoglobulin molecules or may be non-immunoglobulin in origin as set forth above. Preferred immunoglobulin skeletons as herein defined includes any one or more of those selected from the following: an immunoglobulin molecule comprising at least (i) the CL (kappa or lambda subclass) domain of an antibody; or (ii) the CHI domain of an antibody heavy chain; an immunoglobulin molecule comprising the CHI and CH2 domains of an antibody heavy chain; an immunoglobulin molecule comprising the CHI, CH2 and CH3 domains of an antibody heavy chain; or any of the subset (ii) in conjunction with the CL (kappa or lambda subclass) domain of an antibody. A hinge region domain may also be included.. Such combinations of domains may, for example, mimic natural antibodies, such as IgG or IgM, or fragments thereof, such as Fv, scFv, Fab or F(ab')2 molecules. Those skilled in the art will be aware that this list is not intended to be exhaustive.
(II) Protein scaffolds
Each epitope binding domain comprises a protein scaffold and one or more CDRs which are involved in the specific interaction of the domain with one or more epitopes. Advantageously, an epitope binding domain according to the present invention comprises three CDRs. Suitable protein scaffolds include any of those selected from the group consisting of the following: those based on immunoglobulin domains, those based on fibronectin, those based on affibodies, those based on CTLA4, those based on chaperones such as GroEL, those based on lipocallin and those based on the bacterial Fc receptors SpA and SpD. Those skilled in the art will appreciate that this list is not intended to be exhaustive.
F: Scaffolds for use in Constructing Dual Specific Ligands
i. Selection of the main-chain conformation
The members of the immunoglobulin superfamily all share a similar fold for their polypeptide chain. For example, although antibodies are highly diverse in terms of their primary sequence, comparison of sequences and crystallographic structures has revealed that, contrary to expectation, five of the six antigen binding loops of antibodies (HI, H2, LI, L2, L3) adopt a limited number of main-chain conformations, or canonical structures (Chothia and Lesk (1987) J. Mol. Biol, 196: 901; Chothia et al. (1989) Nature, 342: 877). Analysis of loop lengths and key residues has therefore enabled prediction of the main- chain conformations of HI, H2, LI, L2 and L3 found in the majority of human antibodies (Chothia et al. (1992) J. Mol. Biol, 227: 799; Tomlinson et al. (1995) EMBO J, 14: 4628; Williams et al. (1996) J. Mol. Biol, 264: 220). Although the H3 region is much more diverse in terms of sequence, length and structure (due to the use of D segments), it also forms a limited number of main-chain conformations for short loop lengths which depend on the length and the presence of particular residues, or types of residue, at key positions in the loop and the antibody framework (Martin et al. (1996) J Mol. Biol, 263: 800; Shirai et al. (1996) FEBS Letters, 399: 1).
The dual specific ligands of the present invention are advantageously assembled from libraries of domains, such as libraries of VH domains and/or libraries of VL domains. Moreover, the dual specific ligands of the invention may themselves be provided in the form of libraries. In one aspect of the present invention, libraries of dual specific ligands and/or domains are designed in which certain loop lengths and key residues have been chosen to ensure that the main-chain conformation of the members is known. Advantageously, these are real conformations of immunoglobulin superfamily molecules found in nature, to minimise the chances that they are non-functional, as discussed above. Germline V gene segments serve as one suitable basic framework for constructing antibody or T-cell receptor libraries; other sequences are also of use. Variations may occur at a low frequency, such that a small number of functional members may possess an altered main-chain conformation, which does not affect its function.
Canonical structure theory is also of use to assess the number of different main-chain conformations encoded by ligands, to predict the main-chain conformation based on ligand sequences and to chose residues for diversification which do not affect the canonical structure. It is known that, in the human Vκ domain, the LI loop can adopt one of four canonical structures, the L2 loop has a single canonical structure and that 90% of human Vκ domains adopt one of four or five canonical structures for the L3 loop (Tomlinson et al. (1995) supra); thus, in the Vκ domain alone, different canonical structures can combine to create a range of different main-chain conformations. Given that the Vλ domain encodes a different range of canonical structures for the LI, L2 and L3 loops and that Vκ and Vλ domains can pair with any VH domain which can encode several canonical structures for the HI and H2 loops, the number of canonical structure combinations observed for these five loops is very large. This implies that the generation of diversity in the main-chain conformation may be essential for the production of a wide range of binding specificities. However, by constructing an antibody library based on a single known main-chain conformation it has been found, contrary to expectation, that diversity in the main-chain conformation is not required to generate sufficient diversity to target substantially all antigens. Even more surprisingly, the single main-chain conformation need not be a consensus structure - a single naturally occurring conformation can be used as the basis for an entire library. Thus, in a preferred aspect, the dual-specific ligands of the invention possess a single known main-chain conformation.
The single main-chain conformation that is chosen is preferably commonplace among molecules of the immunoglobulin superfamily type in question. A conformation is commonplace when a significant number of naturally occurring molecules are observed to adopt it. Accordingly, in a preferred aspect of the invention, the natural occurrence of the different main-chain conformations for each binding loop of an immunoglobulin domain are considered separately and then a naturally occurring variable domain is chosen which possesses the desired combination of main-chain conformations for the different loops. If none is available, the nearest equivalent may be chosen. It is preferable that the desired combination of main-chain conformations for the different loops is created by selecting germline gene segments which encode the desired main-chain conformations. It is more preferable, that the selected germline gene segments are frequently expressed in nature, and most preferable that they are the most frequently expressed of all natural germline gene segments.
In designing dual specific ligands or libraries thereof the incidence of the different main- chain conformations for each of the six antigen binding loops may be considered separately. For HI, H2, LI, L2 and L3, a given conformation that is adopted by between 20% and 100% of the antigen binding loops of naturally occurring molecules is chosen. Typically, its observed incidence is above 35% (i.e. between 35% and 100%) and, ideally, above 50% or even above 65%. Since the vast majority of H3 loops do not have canonical structures, it is preferable to select a main-chain conformation which is commonplace among those loops which do display canonical structures. For each of the loops, the conformation which is observed most often in the natural repertoire is therefore selected. In human antibodies, the most popular canonical structures (CS) for each loop are as follows: HI - CS 1 (79% of the expressed repertoire), H2 - CS 3 (46%), LI - CS 2 of Vκ (39%), L2 - CS 1 (100%), L3 - CS 1 of Vκ (36%) (calculation assumes a κ:λ ratio of 70:30, Hood et al. (1967) Cold Spring Harbor Symp. Quant. Biol, 48: 133). For H3 loops that have canonical structures, a CDR3 length (Kabat et al. (1991) Sequences of proteins of immunological interest, U.S. Department of Health and Human Services) of seven residues with a salt-bridge from residue 94 to residue 101 appears to be the most common. There are at least 16 human antibody sequences in the EMBL data library with the required H3 length and key residues to form this conformation and at least two crystallographic structures in the protein data bank which can be used as a basis for antibody modelling (2cgr and ltet). The most frequently expressed germline gene segments that this combination of canonical structures are the VH segment 3-23 (DP-47), the JH segment JH4b, the Vκ segment O2/O12 (DPK9) and the Jκ segment Jκl. VH segments DP45 and DP38 are also suitable. These segments can therefore be used in combination as a basis to construct a library with the desired single main-chain conformation. Alternatively, instead of choosing the single main-chain conformation based on the natural occurrence of the different main-chain conformations for each of the binding loops in isolation, the natural occurrence of combinations of main-chain conformations is used as the basis for choosing the single main-chain conformation. In the case of antibodies, for example, the natural occurrence of canonical structure combinations for any two, three, four, five or for all six of the antigen binding loops can be determined. Here, it is preferable that the chosen conformation is commonplace in naturally occurring antibodies and most preferable that it observed most frequently in the natural repertoire. Thus, in human antibodies, for example, when natural combinations of the five antigen binding loops, HI, H2, LI, L2 and L3, are considered, the most frequent combination of canonical structures is determined and then combined with the most popular conformation for the H3 loop, as a basis for choosing the single main-chain conformation.
ii. Diversification of the canonical sequence
Having selected several known main-chain conformations or, preferably a single known main-chain conformation, dual specific ligands according to the invention or libraries for use in the invention can be constructed by varying the binding site of the molecule in order to generate a repertoire with structural and/or functional diversity. This means that variants are generated such that they possess sufficient diversity in their structure and/or in their function so that they are capable of providing a range of activities.
The desired diversity is typically generated by varying the selected molecule at one or more positions. The positions to be changed can be chosen at random or are preferably selected. The variation can then be achieved either by randomisation, during which the resident amino acid is replaced by any amino acid or analogue thereof, natural or synthetic, producing a very large number of variants or by replacing the resident amino acid with one or more of a defined subset of amino acids, producing a more limited number of variants.
Various methods have been reported for introducing such diversity. Error-prone PCR (Hawkins et al. (1992) J. Mol. Biol, 226: 889), chemical mutagenesis (Deng et al (1994) J Biol. Chem., 269: 9533) or bacterial mutator strains (Low et al. (1996) J. Mol. Biol, 260: 359) can be used to introduce random mutations into the genes that encode the molecule. Methods for mutating selected positions are also well known in the art and include the use of mismatched oligonucleotides or degenerate oligonucleotides, with or without the use of PCR. For example, several synthetic antibody libraries have been created by targeting mutations to the antigen binding loops. The H3 region of a human tetanus toxoid-binding Fab has been randomised to create a range of new binding specificities (Barbas et al. (1992) Proc. Natl. Acad. Sci. USA, 89: 4457). Random or semi-random H3 and L3 regions have been appended to germline V gene segments to produce large libraries with unmutated framework regions (Hoogenboom & Winter (1992) J Mol. Biol, 227: 381; Barbas et al. (1992) Proc. Natl. Acad. Sci. USA, 89: 4457; Nissim et al. (1994) EMBO J, 13: 692; Griffiths et al. (1994) EMBO J, 13: 3245; De Kraif et al. (1995) J. Mol. Biol, 248: 97). Such diversification has been extended to include some or all of the other antigen binding loops (Crameri et al. (1996) Nature Med., 2: 100; Riechmann et al. (1995) Bio/Technology, 13: 475; Morphosys, WO97/08320, supra).
Since loop randomisation has the potential to create approximately more than 1015 structures for H3 alone and a similarly large number of variants for the other five loops, it is not feasible using current transformation technology or even by using cell free systems to produce a library representing all possible combinations. For example, in one of the largest libraries constructed to date, 6 x 1010 different antibodies, which is only a fraction of the potential diversity for a library of this design, were generated (Griffiths et al. (1994) supra).
In a preferred embodiment, only those residues which are directly involved in creating or modifying the desired function of the molecule are diversified. For many molecules, the function will be to bind a target and therefore diversity should be concentrated in the target binding site, while avoiding changing residues which are crucial to the overall packing of the molecule or to maintaining the chosen main-chain conformation. Diversification of the canonical sequence as it applies to antibody domains
In the case of antibody dual-specific ligands, the binding site for the target is most often the antigen binding site. Thus, in a highly preferred aspect, the invention provides libraries of or for the assembly of antibody dual-specific ligands in which only those residues in the antigen binding site are varied. These residues are extremely diverse in the human antibody repertoire and are known to make contacts in high-resolution antibody/antigen complexes. For example, in L2 it is known that positions 50 and 53 are diverse in naturally occurring antibodies and are observed to make contact with the antigen. In contrast, the conventional approach would have been to diversify all the residues in the corresponding Complementarity Determining Region (CDR1) as defined by Kabat et al. (1991, supra), some seven residues compared to the two diversified in the library for use according to the invention. This represents a significant improvement in terms of the functional diversity required to create a range of antigen binding specificities.
In nature, antibody diversity is the result of two processes: somatic recombination of germline V, D and J gene segments to create a naive primary repertoire (so called germline and junctional diversity) and somatic hypermutation of the resulting rearranged V genes. Analysis of human antibody sequences has shown that diversity in the primary repertoire is focused at the centre of the antigen binding site whereas somatic hypermutation spreads diversity to regions at the periphery of the antigen binding site that are highly conserved in the primary repertoire (see Tomlinson et al. (1996) J. Mol. Biol, 256: 813). This complementarity has probably evolved as an efficient strategy for searching sequence space and, although apparently unique to antibodies, it can easily be applied to other polypeptide repertoires. The residues which are varied are a subset of those that form the binding site for the target. Different (including overlapping) subsets of residues in the target binding site are diversified at different stages during selection, if desired.
In the case of an antibody repertoire, an initial 'naive' repertoire is created where some, but not all, of the residues in the antigen binding site are diversified. As used herein in this context, the term "naive" refers to antibody molecules that have no pre-determined target. These molecules resemble those which are encoded by the immunoglobulin genes of an individual who has not undergone immune diversification, as is the case with fetal and newborn individuals, whose immune systems have not yet been challenged by a wide variety of antigenic stimuli. This repertoire is then selected against a range of antigens or epitopes. If required, further diversity can then be introduced outside the region diversified in the initial repertoire. This matured repertoire can be selected for modified function, specificity or affinity.
The invention provides two different naive repertoires of binding domains for the construction of dual specific ligands, or a naϊve library of dual specific ligands, in which some or all of the residues in the antigen binding site are varied. The "primary" library mimics the natural primary repertoire, with diversity restricted to residues at the centre of the antigen binding site that are diverse in the germline V gene segments (germline diversity) or diversified during the recombination process (junctional diversity). Those residues which are diversified include, but are not limited to, H50, H52, H52a, H53, H55, H56, H58, H95, H96, H97, H98, L50, L53, L91, L92, L93, L94 and L96. In the "somatic" library, diversity is restricted to residues that are diversified during the recombination process (junctional diversity) or are highly somatically mutated). Those residues which are diversified include, but are not limited to: H31, H33, H35, H95, H96, H97, H98, L30, L31, L32, L34 and L96. All the residues listed above as suitable for diversification in these libraries are known to make contacts in one or more antibody-antigen complexes. Since in both libraries, not all of the residues in the antigen binding site are varied, additional diversity is incorporated during selection by varying the remaining residues, if it is desired to do so. It shall be apparent to one skilled in the art that any subset of any of these residues (or additional residues which comprise the antigen binding site) can be used for the initial and/or subsequent diversification of the antigen binding site.
In the construction of libraries for use in the invention, diversification of chosen positions is typically achieved at the nucleic acid level, by altering the coding sequence which specifies the sequence of the polypeptide such that a number of possible amino acids (all 20 or a subset thereof) can be incorporated at that position. Using the TUPAC nomenclature, the most versatile codon is NNK, which encodes all amino acids as well as the TAG stop codon. The NNK codon is preferably used in order to introduce the required diversity. Other codons which achieve the same ends are also of use, including the NNN codon, which leads to the production of the additional stop codons TGA and TAA.
A feature of side-chain diversity in the antigen binding site of human antibodies is a pronounced bias which favours certain amino acid residues. If the amino acid composition of the ten most diverse positions in each of the VH, VK and Vχ regions are summed, more than 76% of the side-chain diversity comes from only seven different residues, these being, serine (24%), tyrosine (14%), asparagine (11%), glycine (9%), alanine (7%), aspartate (6%) and threonine (6%). This bias towards hydrophilic residues and small residues which can provide main-chain flexibility probably reflects the evolution of surfaces which are predisposed to binding a wide range of antigens or epitopes and may help to explain the required promiscuity of antibodies in the primary repertoire.
Since it is preferable to mimic this distribution of amino acids, the distribution of amino acids at the positions to be varied preferably mimics that seen in the antigen binding site of antibodies. Such bias in the substitution of amino acids that permits selection of certain polypeptides (not just antibody polypeptides) against a range of target antigens is easily applied to any polypeptide repertoire. There are various methods for biasing the amino acid distribution at the position to be varied (including the use of tri-nucleotide mutagenesis, see WO97/08320), of which the preferred method, due to ease of synthesis, is the use of conventional degenerate codons. By comparing the amino acid profile encoded by all combinations of degenerate codons (with single, double, triple and quadruple degeneracy in equal ratios at each position) with the natural amino acid use it is possible to calculate the most representative codon. The codons (AGT)(AGC)T, (AGT)(AGC)C and (AGT)(AGC)(CT) - that is, DVT, DVC and DVY, respectively using TUPAC nomenclature - are those closest to the desired amino acid profile: they encode 22% serine and 11% tyrosine, asparagine, glycine, alanine, aspartate, threonine and cysteine. Preferably, therefore, libraries are constructed using either the DVT, DVC or DVY codon at each of the diversified positions. G: Antigens capable of increasing ligand half-life
The dual specific ligands according to the invention, in one configuration thereof, are capable of binding to one or more molecules which can increase the half-life of the ligand in vivo. Typically, such molecules are polypeptides which occur naturally in vivo and which resist degradation or removal by endogenous mechanisms which remove unwanted material from the organism. For example, the molecule which increases the half-life of the organism may be selected from the following:
Proteins from the extracellular matrix; for example collagen, laminins, integrins and fibronectin. Collagens are the major proteins of the extracellular matrix. About 15 types of collagen molecules are currently known, found in different parts of the body, eg type I collagen (accounting for 90% of body collagen) found in bone, skin, tendon, ligaments, cornea, internal organs or type II collagen found in cartilage, invertebral disc, notochord, vitreous humour of the eye.
Proteins found in blood, including:
Plasma proteins such as fibrin, c-2 macroglobulin, serum albumin, fibrinogen A, fibrinogen B, serum amyloid protein A, heptaglobin, profilin, ubiquitin, uteroglobulin and /3-2-microglobulin;
Enzymes and inhibitors such as plasminogen, lysozyme, cystatin C, alpha- 1-antitrypsin and pancreatic trypsin inhibitor. Plasminogen is the inactive precursor of the trypsin-like serine protease plasmin. It is normally found circulating through the blood stream. When plasminogen becomes activated and is converted to plasmin, it unfolds a potent enzymatic domain that dissolves the fibrinogen fibers that entgangle the blood cells in a blood clot. This is called fibrinolysis.
Immune system proteins, such as IgE, IgG, IgM.
Transport proteins such as retinol binding protein, α:-l micro globulin. Defensins such as beta-defensin 1, Neutrophil defensins 1,2 and 3.
Proteins found at the blood brain barrier or in neural tissues, such as melanocortin receptor, myelin, ascorbate transporter.
Transferrin receptor specific ligand-neuropharmaceutical agent fusion proteins (see US5977307);
brain capillary endothelial cell receptor, transferrin, transferrin receptor, insulin, insulin- like growth factor 1 (IGF 1) receptor, insulin-like growth factor 2 (IGF 2) receptor, insulin receptor.
Proteins localised to the kidney, such as polycystin, type IV collagen, organic anion transporter Kl, Heymann's antigen.
Proteins localised to the liver, for example alcohol dehydrogenase, G250.
Blood coagulation factor X αl antitrypsin HNF lo!
Proteins localised to the lung, such as secretory component (binds IgA).
Proteins localised to the Heart, for example HSP 27. This is associated with dilated cardiomyopathy.
Proteins localised to the skin, for example keratin.
Bone specific proteins, such as bone morphogenic proteins (BMPs), which are a subset of the transforming growth factor β superfamily that demonstrate osteogenic activity.
Examples include BMP -2, -4, -5, -6, -7 (also referred to as osteogenic protein (OP-1) and -8 (OP-2). Tumour specific proteins, including human trophoblast antigen, herceptin receptor, oestrogen receptor, cathepsins eg cathepsin B (found in liver and spleen).
Disease-specific proteins, such as antigens expressed only on activated T-cells: including LAG-3 (lymphocyte activation gene), osteoprotegerin ligand (OPGL) see Nature 402, 304-309; 1999, OX40 (a member of the TNF receptor family, expressed on activated T cells and the only costimulatory T cell molecule known to be specifically up-regulated in human T cell leukaemia virus type-I (HTLV-I)-producing cells.) See J Immunol. 2000 Jul l;165(l):263-70; Metalloproteases (associated with arthritis/cancers), including CG6512 Drosophila, human paraplegin, human FtsH, human AFG3L2, murine ftsH; angiogenic growth factors, including acidic fibroblast growth factor (FGF-1), basic fibroblast growth factor (FGF-2), Vascular endothelial growth factor / vascular permeability factor (VEGF/VPF), transforming growth factor-a (TGF a), tumor necrosis factor-alpha (TNF-α;), angiogenin, interleukin-3 (IL-3), interleukin-8 (IL-8), platelet- derived endothelial growth factor (PD-ECGF), placental growth factor (PIGF), midkine platelet-derived growth factor-BB (PDGF), fractalkine.
Stress proteins (heat shock proteins)
HSPs are normally found intracellularly. When they are found extracellularly, it is an indicator that a cell has died and spilled out its contents. This unprogrammed cell death
(necrosis) only occurs when as a result of trauma, disease or injury and therefore in vivo, extracellular HSPs trigger a response from the immune system that will fight infection and disease. A dual specific which binds to extracellular HSP can be localised to a disease site.
Proteins involved in Fc transport
Brambell receptor (also known as FcRB)
This Fc receptor has two functions, both of which are potentially useful for delivery
The functions are (1) The transport of IgG from mother to child across the placenta
(2) the protection of IgG from degradation thereby prolonging its serum half life of IgG. It is thought that the receptor recycles IgG from endosome. See Holliger et al, Nat Biotechnol 1997 Jul;15(7):632-6.
Ligands according to the invention may designed to be specific for the above targets without requiring any increase in or increasing half life in vivo. For example, ligands according to the invention can be specific for targets selected from the foregoing which are tissue-specific, thereby enabling tissue-specific targeting of the dual specific ligand, or a dAb monomer that binds a tissue-specific therapeutically relevant target, irrespective of any increase in half-life, although this may result. Moreover, where the ligand or dAb monomer targets kidney or liver, this may redirect the ligand or dAb monomer to an alternative clearance pathway in vivo (for example, the ligand may be directed away from liver clearance to kidney clearance).
H: Use of multispecific ligands according to the second configuration of the invention
Multispecific ligands according to the method of the second configuration of the present invention may be employed in in vivo therapeutic and prophylactic applications, in vitro and in vivo diagnostic applications, in vitro assay and reagent applications, and the like. For example antibody molecules may be used in antibody based assay techniques, such as ELISA techniques, according to methods known to those skilled in the art.
As alluded to above, the multispecific ligands according to the invention are of use in diagnostic, prophylactic and therapeutic procedures. Multispecific antibodies according to the invention are of use diagnostically in Western analysis and in situ protein detection by standard immunohistochemical procedures; for use in these applications, the ligands may be labelled in accordance with techniques known to the art. In addition, such antibody polypeptides may be used preparatively in affinity chromatography procedures, when complexed to a chromatographic support, such as a resin. All such techniques are well known to one of skill in the art.
Diagnostic uses of the closed conformation multispecific ligands according to the invention include homogenous assays for analytes which exploit the ability of closed conformation multispecific ligands to bind two targets in competition, such that two targets cannot bind simultaneously (a closed conformation), or alternatively their ability to bind two targets simultaneously (an open conformation).
A true homogenous immunoassay format has been avidly sought by manufacturers of diagnostics and research assay systems used in drug discovery and development. The main diagnostics markets include human testing in hospitals, doctor's offices and clinics, commercial reference laboratories, blood banks, and the home, non-human diagnostics (for example food testing, water testing, environmental testing, bio-defence, and veterinary testing), and finally research (including drug development; basic research and academic research).
At present all these markets utilise immunoassay systems that are built around chemiluminescent, ELISA, fluorescence or in rare cases radio-immunoassay technologies. Each of these assay formats requires a separation step (separating bound from un-bound reagents). In some cases, several separation steps are required. Adding these additional steps adds reagents and automation, takes time, and affects the ultimate outcome of the assays. In human diagnostics, the separation step may be automated, which masks the problem, but does not remove it. The robotics, additional reagents, additional incubation times, and the like add considerable cost and complexity. In drug development, such as high throughput screening, where literally millions of samples are tested at once, with very low levels of test molecule, adding additional separation steps can eliminate the ability to perform a screen. However, avoiding the separation creates too much noise in the read out. Thus, there is a need for a true homogenous format that provides sensitivities at the range obtainable from present assay formats. Advantageously, an assay possesses fully quantitative read-outs with high sensitivity and a large dynamic range. Sensitivity is an important requirement, as is reducing the amount of sample required. Both of these features are features that a homogenous system offers. This is very important in point of care testing, and in drug development where samples are precious. Heterogenous systems, as currently available in the art, require large quantities of sample and expensive reagents
Applications for homogenous assays include cancer testing, where the biggest assay is that for Prostate Specific Antigen, used in screening men for prostate cancer. Other applications include fertility testing, which provides a series of tests for women attempting to conceive including beta-hcg for pregnancy. Tests for infectious diseases, including hepatitis, HIV, rubella, and other viruses and microorganisms and sexually transmitted diseases. Tests are used by blood banks, especially tests for HIV, hepatitis A, B, C, non A non B. Therapeutic drug monitoring tests include monitoring levels of prescribed drugs in patients for efficacy and to avoid toxicity, for example digoxin for arrhythmia, and phenobarbital levels in psychotic cases; theophylline for asthma. Diagnostic tests are moreover useful in abused drug testing, such as testing for cocaine, marijuana and the like. Metabolic tests are used for measuring thyroid function, anaemia and other physiological disorders and functions.
The homogenous immunoassay format is moreover useful in the manufacture of standard clinical chemistry assays. The inclusion of immunoassays and chemistry assays on the same instrument is highly advantageous in diagnostic testing. Suitable chemical assays include tests for glucose, cholesterol, potassium, and the like.
A further major application for homogenous immunoassays is drug discovery and development: high throughput screening includes testing combinatorial chemistry libraries versus targets in ultra high volume. Signal is detected, and positive groups then split into smaller groups, and eventually tested in cells and then animals. Homogenous assays may be used in all these types of test. In drug development, especially animal studies and clinical trials heavy use of immunoassays is made. Homogenous assays greatly accelerate and simplify these procedures. Other Applications include food and beverage testing: testing meat and other foods for E. coli, salmonella, etc; water testing, including testing at water plants for all types of contaminants including E. coli; and veterinary testing.
In a broad embodiment, the invention provides a binding assay comprising a detectable agent which is bound to a closed conformation multispecific ligand according to the invention, and whose detectable properties are altered by the binding of an analyte to said closed conformation multispecific ligand. Such an assay may be configured in several different ways, each exploiting the above properties of closed conformation multispecific ligands. The assay relies on the direct or indirect displacement of an agent by the analyte, resulting in a change in the detectable properties of the agent. For example, where the agent is an enzyme which is capable of catalysing a reaction which has a detectable end-point, said enzyme can be bound by the ligand such as to obstruct its active site, thereby inactivating the enzyme. The analyte, which is also bound by the closed conformation multispecific ligand, displaces the enzyme, rendering it active through freeing of the active site. The enzyme is then able to react with a substrate, to give rise to a detectable event. In an alternative embodiment, the ligand may bind the enzyme outside of the active site, influencing the conformation of the enzyme and thus altering its activity. For example, the structure of the active site may be constrained by the binding of the ligand, or the binding of cofactors necessary for activity may be prevented.
The physical implementation of the assay may take any form known in the art. For example, the closed conformation multispecific ligand/enzyme complex may be provided on a test strip; the substrate may be provided in a different region of the test strip, and a solvent containing the analyte allowed to migrate through the ligand/enzyme complex, displacing the enzyme, and carrying it to the substrate region to produce a signal. Alternatively, the ligand/enzyme complex may be provided on a test stick or other solid phase, and dipped into an analyte/substrate solution, releasing enzyme into the solution in response to the presence of analyte.
Since each molecule of analyte potentially releases one enzyme molecule, the assay is quantitative, with the strength of the signal generated in a given time being dependent on the concentration of analyte in the solution.
Further configurations using the analyte in a closed conformation are possible. For example, the closed conformation multispecific ligand may be configured to bind an enzyme in an allosteric site, thereby activating the enzyme. In such an embodiment, the enzyme is active in the absence of analyte. Addition of the analyte displaces the enzyme and removes allosteric activation, thus inactivating the enzyme. In the context of the above embodiments which employ enzyme activity as a measure of the analyte concentration, activation or inactivation of the enzyme refers to an increase or decrease in the activity of the enzyme, measured as the ability of the enzyme to catalyse a signal-generating reaction. For example, the enzyme may catalyse the conversion of an undetectable substrate to a detectable form thereof. For example, horseradish peroxidase is widely used in the art together with chromogenic or chemiluminescent substrates, which are available commercially. The level of increase or decrease of the activity of the enzyme may between 10% and 100%, such as 20%, 30%, 40%, 50%, 60%, 70%, 80% or 90%; in the case of an increase in activity, the increase may be more than 100%, i.e. 200%, 300%, 500%) or more, or may not be measurable as a percentage if the baseline activity of the inhibited enzyme is undetectable.
In a further configuration, the closed conformation multispecific ligand may bind the substrate of an enzyme/substrate pair, rather than the enzyme. The substrate is therefore unavailable to the enzyme until released from the closed conformation multispecific ligand through binding of the analyte. The implementations for this configuration are as for the configurations which bind enzyme.
Moreover, the assay may be configured to bind a fluorescent molecule, such as a fluorescein or another fluorophore, in a conformation such that the fluorescence is quenched on binding to the ligand. In this case, binding of the analyte to the ligand will displace the fluorescent molecule, thus producing a signal. Alternatives to fluorescent molecules which are useful in the present invention include luminescent agents, such as luciferin luciferase, and chromogenic agents, including agents commonly used in immunoassays such as HRP.
Therapeutic and prophylactic uses of multispecific ligands prepared according to the invention involve the administration of ligands according to the invention to a recipient mammal, such as a human. Multi-specificity can allow antibodies to bind to multimeric antigen with great avidity. Multispecific ligands can allow thecross- linking of two antigens, for example in recruiting cytotoxic T-cells to mediate the killing of tumour cell lines. Substantially pure ligands or binding proteins thereof, for example dAb monomers, of at least 90 to 95% homogeneity are preferred for administration to a mammal, and 98 to 99% or more homogeneity is most preferred for pharmaceutical uses, especially when the mammal is a human. Once purified, partially or to homogeneity as desired, the ligands may be used diagnostically or therapeutically (including extracorporeally) or in developing and performing assay procedures, immunofluorescent stainings and the like (Lefkovite and Pernis, (1979 and 1981) Immuno logical Methods, Volumes I and II, Academic Press, NY).
The ligands or binding proteins thereof, for example dAb monomers, of the present invention will typically find use in preventing, suppressing or treating inflammatory states, allergic hypersensitivity, cancer, bacterial or viral infection, and autoimmune disorders (which include, but are not limited to, Type I diabetes, asthma, multiple sclerosis, rheumatoid arthritis, systemic lupus erythematosus, Crohn's disease and myasthenia gravis).
hi the instant application, the term "prevention" involves administration of the protective composition prior to the induction of the disease. "Suppression" refers to administration of the composition after an inductive event, but prior to the clinical appearance of the disease. "Treatment" involves administration of the protective composition after disease symptoms become manifest.
Animal model systems which can be used to screen the effectiveness of the antibodies or binding proteins thereof in protecting against or treating the disease are available. Methods for the testing of systemic lupus erythematosus (SLE) in susceptible mice are known in the art (Knight et al. (1978) J. Exp. Med., 147: 1653; Reinersten et al. (1978) New Eng. J. Med., 299: 515). Myasthenia Gravis (MG) is tested in SJL/J female mice by inducing the disease with soluble AchR protein from another species (Lindstrom et al. (1988) Adv. Immunol, 42: 233). Arthritis is induced in a susceptible strain of mice by injection of Type II collagen (Stuart et al. (1984) Ann. Rev. Immunol, 42: 233). A model by which adjuvant arthritis is induced in susceptible rats by injection of mycobacterial heat shock protein has been described (Van Eden et al. (1988) Nature, 331: 171). Thyroiditis is induced in mice by administration of thyroglobulin as described (Maron et al (1980) J. Exp. Med., 152: 1115). Insulin dependent diabetes mellitus (JDDM) occurs naturally or can be induced in certain strains of mice such as those described by Kanasawa et al. (1984) Diabetologia, 27: 113. EAE in mouse and rat serves as a model for MS in human. In this model, the demyelinating disease is induced by administration of myelin basic protein (see Paterson (1986) Textbook of Immunopathology, Mischer et al, eds., Grune and Stratum, New York, pp. 179-213; McFarlin et al. (1973) Science, 179: 478: and Satoh et al. (1987) J. Immunol, 138: 179).
Generally, the present ligands will be utilised in purified form together with pharmacologically appropriate carriers. Typically, these carriers include aqueous or alcoholic/aqueous solutions, emulsions or suspensions, any including saline and/or buffered media. Parenteral vehicles include sodium chloride solution, Ringer's dextrose, dextrose and sodium chloride and lactated Ringer's. Suitable physiologically-acceptable adjuvants, if necessary to keep a polypeptide complex in suspension, may be chosen from thickeners such as carboxymethylcellulose, polyvinylpyrrolidone, gelatin and alginates.
Intravenous vehicles include fluid and nutrient replenishers and electrolyte replenishers, such as those based on Ringer's dextrose. Preservatives and other additives, such as antimicrobials, antioxidants, chelating agents and inert gases, may also be present (Mack (1982) Remington 's Pharmaceutical Sciences, 16th Edition).
The ligands of the present invention may be used as separately administered compositions or in conjunction with other agents. These can include various immunotherapeutic drugs, such as cylcosporine, methotrexate, adriamycin or cisplatinum, and immunotoxins. Pharmaceutical compositions can include "cocktails" of various cytotoxic or other agents in conjunction with the ligands of the present invention, or even combinations of lignds according to the present invention having different specificities, such as ligands selected using different target antigens or epitopes, whether or not they are pooled prior to administration.
The route of administration of pharmaceutical compositions according to the invention may be any of those commonly known to those of ordinary skill in the art. For therapy, including without limitation immunotherapy, the selected ligands thereof of the invention can be administered to any patient in accordance with standard techniques. The administration can be by any appropriate mode, including parenterally, intravenously, intramuscularly, intraperitoneally, transdermally, via the pulmonary route, or also, appropriately, by direct infusion with a catheter. The dosage and frequency of administration will depend on the age, sex and condition of the patient, concurrent administration of other drugs, counterindications and other parameters to be taken into account by the clinician.
The ligands ofthis invention can be lyophilised for storage and reconstituted in a suitable carrier prior to use. This technique has been shown to be effective with conventional immunoglobulins and art-known lyophilisation and reconstitution techniques can be employed. It will be appreciated by those skilled in the art that lyophilisation and reconstitution can lead to varying degrees of antibody activity loss (e.g. with conventional immunoglobulins, IgM antibodies tend to have greater activity loss than IgG antibodies) and that use levels may have to be adjusted upward to compensate.
The compositions containing the present ligands or a cocktail thereof can be administered for prophylactic and/or therapeutic treatments, hi certain therapeutic applications, an adequate amount to accomplish at least partial inhibition, suppression, modulation, killing, or some other measurable parameter, of a population of selected cells is defined as a "therapeutically-effective dose". Amounts needed to achieve this dosage will depend upon the severity of the disease and the general state of the patient's own immune system, but generally range from 0.005 to 5.0 mg of ligand, e.g. antibody, receptor (e.g. a T-cell receptor) or binding protein thereof per kilogram of body weight, with doses of 0.05 to 2.0 mg/kg/dose being more commonly used. For prophylactic applications, compositions containing the present ligands or cocktails thereof may also be administered in similar or slightly lower dosages.
Treatment performed using the compositions described herein is considered "effective" if one or more symptoms is reduced (e.g., by at least 10%> or at least one point on a clinical assessment scale), relative to such symptoms present before treatment, or relative to such symptoms in an individual (human or model animal) not treated with such composition. Symptoms will obviously vary depending upon the disease or disorder targeted, but can be measured by an ordinarily skilled clinician or technician. Such symptoms can be measured, for example, by monitoring the level of one or more biochemical indicators of the disease or disorder (e.g., levels of an enzyme or metabolite correlated with the disease, affected cell numbers, etc.), by monitoring physical manifestations (e.g., inflammation, tumor size, etc.), or by an accepted clinical assessment scale, for example, the Expanded Disability Status Scale (for multiple sclerosis), the Irvine Inflammatory Bowel Disease Questionnaire (32 point assessment evaluates quality of life with respect to bowel function, systemic symptoms, social function and emotional status - score ranges from 32 to 224, with higher scores indicating a better quality of life), the Quality of Life Rheumatoid Arthritis Scale, or other accepted clinical assessment scale as known in the field. A sustained (e.g., one day or more, preferably longer) reduction in disease or disorder symptoms by at least 10% or by one or more points on a given clinical scale is indicative of "effective" treatment. Similarly, prophylaxis performed using a composition as described herein is "effective" if the onset or severity of one or more symptoms is delayed, reduced or abolished relative to such symptoms in a similar individual (human or animal model) not treated with the composition.
A composition containing a ligand or cocktail thereof according to the present invention may be utilised in prophylactic and therapeutic settings to aid in the alteration, inactivation, killing or removal of a select target cell population in a mammal. In addition, the selected repertoires of polypeptides described herein may be used extracorporeally or in vitro selectively to kill, deplete or otherwise effectively remove a target cell population from a heterogeneous collection of cells. Blood from a mammal may be combined extracorporeally with the ligands, e.g. antibodies, cell-surface receptors or binding proteins thereof whereby the undesired cells are killed or otherwise removed from the blood for return to the mammal in accordance with standard techniques.
I: Use of half-life enhanced dual-specific ligands according to the invention
Dual-specific ligands according to the method of the present invention may be employed in in vivo therapeutic and prophylactic applications, in vivo diagnostic applications and the like. Therapeutic and prophylactic uses of dual-specific ligands prepared according to the invention involve the administration of ligands according to the invention to a recipient mammal, such as a human. Dual specific antibodies according to the invention comprise at least one specificity for a half-life enhancing molecule; one or more further specificities may be directed against target molecules. For example, a dual-specific IgG may be specific for four epitopes, one of which is on a half-life enhancing molecule. Dual- specificity can allow antibodies to bind to multimeric antigen with great avidity. Dual- specific antibodies can allow the cross-linking of two antigens, for example in recruiting cytotoxic T-cells to mediate the killing of tumour cell lines.
Substantially pure ligands or binding proteins thereof, such as dAb monomers, of at least 90 to 95%) homogeneity are preferred for administration to a mammal, and 98 to 99%> or more homogeneity is most preferred for pharmaceutical uses, especially when the mammal is a human. Once purified, partially or to homogeneity as desired, the ligands may be used diagnostically or therapeutically (including extracorporeally) or in developing and performing assay procedures, immunofluorescent stainings and the like (Lefkovite and Pernis, (1979 and 1981) Immunological Methods, Volumes I and II, Academic Press, NY).
The ligands of the present invention will typically find use in preventing, suppressing or treating inflammatory states, allergic hypersensitivity, cancer, bacterial or viral infection, and autoimmune disorders (which include, but are not limited to, Type I diabetes, multiple sclerosis, rheumatoid arthritis, systemic lupus erythematosus, Crohn's disease and myasthenia gravis).
In the instant application, the term "prevention" involves administration of the protective composition prior to the induction of the disease. "Suppression" refers to administration of the composition after an inductive event, but prior to the clinical appearance of the disease. "Treatment" involves administration of the protective composition after disease symptoms become manifest. Animal model systems which can be used to screen the effectiveness of the dual specific ligands in protecting against or treating the disease are available. Methods for the testing of systemic lupus erythematosus (SLE) in susceptible mice are known in the art (Knight et al. (1978) J. Exp. Med., 147: 1653; Reinersten et al. (1978) New Eng. J. Med., 299: 515). Myasthenia Gravis (MG) is tested in SJL/J female mice by inducing the disease with soluble AchR protein from another species (Lindstrom et al. (1988) Adv. Immunol, 42: 233). Arthritis is induced in a susceptible strain of mice by injection of Type II collagen (Stuart et al. (1984) Ann. Rev. Immunol, 42: 233). A model by which adjuvant arthritis is induced in susceptible rats by injection of mycobacterial heat shock protein has been described (Van Eden et al. (1988) Nature, 331: 171). Thyroiditis is induced in mice by administration of thyroglobulin as described (Maron et al. (1980) J. Exp. Med., 152: 1115). Insulin dependent diabetes mellitus (IDDM) occurs naturally or can be induced in certain strains of mice such as those described by Kanasawa et al. (1984) Diabetologia, 27: 113. EAE in mouse and rat serves as a model for MS in human, hi this model, the demyelinating disease is induced by administration of myelin basic protein (see Paterson (1986) Textbook of Immunopathology, Mischer et al, eds., Grune and Srratton, New York, pp. 179-213; McFarlin et al. (1973) Science, 179: 478: and Satoh et al. (1987) J Immunol, 138: 179).
Dual specific ligands according to the invention and dAb monomers able to bind to extracellular targets involved in endocytosis (e.g. Clathrin) enable dual specific ligands to be endocytosed, enabling another specificity able to bind to an intracellular target to be delivered to an intracellular environment. This strategy requires a dual specific ligand with physical properties that enable it to remain functional inside the cell. Alternatively, if the final destination intracellular compartment is oxidising, a well folding ligand may not need to be disulphide free.
Generally, the present dual specific ligands will be utilised in purified form together with pharmacologically appropriate carriers. Typically, these carriers include aqueous or alcoholic/aqueous solutions, emulsions or suspensions, any including saline and/or buffered media. Parenteral vehicles include sodium chloride solution, Ringer's dextrose, dextrose and sodium chloride and lactated Ringer's. Suitable physiologically-acceptable adjuvants, if necessary to keep a polypeptide complex in suspension, may be chosen from thickeners such as carboxymethylcellulose, polyvinylpyrrolidone, gelatin and alginates.
Intravenous vehicles include fluid and nutrient replenishers and electrolyte replenishers, such as those based on Ringer's dextrose. Preservatives and other additives, such as antimicrobials, antioxidants, chelating agents and inert gases, may also be present (Mack (1982) Remington's Pharmaceutical Sciences, 16th Edition).
The ligands of the present invention may be used as separately administered compositions or in conjunction with other agents. These can include various immunotherapeutic drugs, such as cylcosporine, methotrexate, adriamycin or cisplatinum, and immunotoxins. Pharmaceutical compositions can include "cocktails" of various cytotoxic or other agents in conjunction with the ligands of the present invention.
The route of administration of pharmaceutical compositions according to the invention may be any of those commonly known to those of ordinary skill in the art. For therapy, including without limitation immunotherapy, the ligands of the invention can be administered to any patient in accordance with standard techniques. The administration can be by any appropriate mode, including parenterally, intravenously, intramuscularly, intraperitoneally, transdermally, via the pulmonary route, or also, appropriately, by direct infusion with a catheter. The dosage and frequency of administration will depend on the age, sex and condition of the patient, concurrent administration of other drugs, counterindications and other parameters to be taken into account by the clinician.
The ligands of the invention can be lyophilised for storage and reconstituted in a suitable carrier prior to use. This technique has been shown to be effective with conventional immunoglobulins and art-known lyophilisation and reconstitution techniques can be employed. It will be appreciated by those skilled in the art that lyophilisation and reconstitution can lead to varying degrees of antibody activity loss (e.g. with conventional immunoglobulins, IgM antibodies tend to have greater activity loss than IgG antibodies) and that use levels may have to be adjusted upward to compensate. The compositions containing the present ligands or a cocktail thereof can be administered for prophylactic and/or therapeutic treatments. In certain therapeutic applications, an adequate amount to accomplish at least partial inhibition, suppression, modulation, killing, or some other measurable parameter, of a population of selected cells is defined as a "therapeutically-effective dose". Amounts needed to achieve this dosage will depend upon the severity of the disease and the general state of the patient's own immune system, but generally range from 0.005 to 5.0 mg of ligand per kilogram of body weight, with doses of 0.05 to 2.0 mg/kg/dose being more commonly used. For prophylactic applications, compositions containing the present ligands or cocktails thereof may also be administered in similar or slightly lower dosages.
A composition containing a ligand according to the present invention may be utilised in prophylactic and therapeutic settings to aid in the alteration, inactivation, killing or removal of a select target cell population in a mammal.
In addition, the selected repertoires of polypeptides described herein may be used extracorporeally or in vitro selectively to kill, deplete or otherwise effectively remove a target cell population from a heterogeneous collection of cells. Blood from a mammal may be combined extracorporeally with the ligands, e.g. antibodies, cell- surface receptors or binding proteins thereof whereby the undesired cells are killed or otherwise removed from the blood for return to the mammal in accordance with standard techniques.
The invention is further described, for the purposes of illustration only, in the following examples. As used herein, for the purposes of dAb nomenclature, human TNFα is referred to as TAR1 and human TNFα receptor 1 (p55 receptor) is referred to as TAR2.
Example 1. Selection of a dual specific scFv antibody (K8) directed against human serum albumin (HSA) and β-galactosidase (β -gal)
This example explains a method for making a dual specific antibody directed against β- gal and HSA in which a repertoire of Vκ variable domains linked to a germline (dummy) VH domain is selected for binding to β-gal and a repertoire of VH variable domains linked to a germline (dummy) Vκ domain is selected for binding to HSA. The selected variable VH HSA and Vκ β-gal domains are then combined and the antibodies selected for binding to β-gal and HSA. HSA is a half-life increasing protein found in human blood.
Four human phage antibody libraries were used in this experiment.
Library 1 Germline VK/DVT VH 8.46 x lO7
Library 2 Germline VK/NNK VH 9.64 x lO7 Library 3 Germline VH/DVT Vκ 1.47 x 108
Library 4 Germline VH/NNK Vκ 1.45 x 108
All libraries are based on a single human framework for VH (V3-23/DP47 and JH4b) and Vκ (O12/O2/DPK9 and Jκl) with side chain diversity incorporated in complementarity determining regions (CDR2 and CDR3).
Library 1 and Library 2 contain a dummy Vκ sequence, whereas the sequence of VH is diversified at positions H50, H52, H52a, H53, H55, H56, H58, H95, H96, H97 and H98
(DVT or NNK encoded, respectively) (Figure 1). Library 3 and Library 4 contain a dummy VH sequence, whereas the sequence of Vκ is diversified at positions L50, L53,
L91, L92, L93, L94 and L96 (DVT or NNK encoded, respectively) (Figure 1). The libraries are in phagemid pIT2/ScFv format (Figure 2) and have been preselected for binding to generic ligands, Protein A and Protein L, so that the majority of clones in the unselected libraries are functional. The sizes of the libraries shown above correspond to the sizes after preselection. Library 1 and Library 2 were mixed prior to selections on antigen to yield a single VH/dummy Vκ library and Library 3 and Library 4 were mixed to form a single Vκ/dummy VH library.
Three rounds of selections were performed on β-gal using Vκ/dummy VH library and three rounds of selections were performed on HSA using VH/du my Vκ library. In the case of β-gal the phage titres went up from 1.1 x 10°" in the first round to 2.0 x 108 in the third round. In the case of HSA the phage titres went up from 2 x 10^ in the first round to
1.4 x 10^ in the third round. The selections were performed as described by Griffith et al, (1993), except that KM13 helper phage (which contains a pill protein with a protease cleavage site between the D2 and D3 domains) was used and phage were eluted with 1 mg/ml trypsin in PBS. The addition of trypsin cleaves the pill proteins derived from the helper phage (but not those from the phagemid) and elutes bound scFv-phage fusions by cleavage in the c-myc tag (Figure 2), thereby providing a further enrichment for phages expressing functional scFvs and a corresponding reduction in background (Kristensen & Winter, Folding & Design 3: 321-328, Jul 9, 1998). Selections were performed using immunorubes coated with either HSA or β-gal at lOOμg/ml concentration.
To check for binding, 24 colonies from the third round of each selection were screened by monoclonal phage ELISA. Phage particles were produced as described by Harrison et al, Methods Enzymol. 1996;267:83-109. 96-well ELISA plates were coated with lOOμl of HSA or β-gal at lOμg/ml concentration in PBS overnight at 4°C. A standard ELISA protocol was followed (Hoogenboom et al, 1991) using detection of bound phage with anti-M13-HRP conjugate. A selection of clones gave ELISA signals of greater than 1.0 with 50μl supernatant.
Next, DNA preps were made from VH/dummy Vκ library selected on HSA and from
Vκ/dummy VH library selected on β-gal using the QIAprep Spin Miniprep kit (Qiagen).
To access most of the diversity, DNA preps were made from each of the three rounds of selections and then pulled together for each of the antigens. DNA preps were then digested with Sali/Not/ overnight at 37°C. Following gel purification of the fragments, Vκ chains from the Vκ/dummy VH library selected on β-gal were ligated in place of a dummy Vκ chain of the VH/dummy Vκ library selected on HSA creating a library of 3.3 x lθ9 clones.
This library was then either selected on HSA (first round) and β-gal (second round), HSA/β-gal selection, or on β-gal (first round) and HSA (second round), β-gal/HSA selection. Selections were performed as described above. In each case after the second round 48 clones were tested for binding to HSA and β-gal by the monoclonal phage ELISA (as described above) and by ELISA of the soluble scFv fragments. Soluble antibody fragments were produced as described by Harrison et al, (1996), and standard ELISA protocol was followed Hoogenboom et al. (1991) Nucleic Acids Res., 19: 4133, except that 2% Tween/PBS was used as a blocking buffer and bound scFvs were detected with Protein L-HRP. Three clones (E4, E5 and E8) from the HSA/β-gal selection and two clones (K8 and K10) from the β-gal/HSA selection were able to bind both antigens. scFvs from these clones were PCR amplified and sequenced as described by Ignatovich et al, (1999) J Mol Biol 1999 Nov 26;294(2):457-65, using the primers LMB3 and pHENseq. Sequence analysis revealed that all clones were identical. Therefore, only one clone encoding a dual specific antibody (K8) was chosen for further work (Figure 3).
Example 2. Characterisation of the binding properties of the K8 antibody.
Firstly, the binding properties of the K8 antibody were characterised by the monoclonal phage ELISA. A 96-well plate was coated with lOOμl of HSA and β-gal alongside with alkaline phosphatase (APS), bovine serum albumin (BSA), peanut agglutinin, lysozyme and cytochrome c (to check for cross-reactivity) at lOμg/ml concentration in PBS overnight at 4°C. The phagemid from K8 clone was rescued with KM13 as described by Harrison et al, (1996) and the supernatant (50μl) containing phage assayed directly. A standard ELISA protocol was followed (Hoogenboom et al, 1991) using detection of bound phage with anti-M13-HRP conjugate. The dual specific K8 antibody was found to bind to HSA and β-gal when displayed on the surface of the phage with absorbance signals greater than 1.0 (Figure 4). Strong binding to BSA was also observed (Figure 4). Since HSA and BSA are 76% homologous on the amino acid level, it is not surprising that K8 antibody recognised both ofthese structurally related proteins. No cross-reactivity with other proteins was detected (Figure 4).
Secondly, the binding properties of the K8 antibody were tested in a soluble scFv ELISA. Production of the soluble scFv fragment was induced by JRTG as described by Harrison et al, (1996). To determine the expression levels of K8 scFv, the soluble antibody fragments were purified from the supernatant of 50ml inductions using Protein A-
Sepharose columns as described by Harlow and Lane, Antibodies: a Laboratory Manual,
(1988) Cold Spring Harbor. OD28O was then measured and the protein concentration calculated as described by Sambrook et al, (1989). K8 scFv was produced in supernatant at 19mg/l.
A soluble scFv ELISA was then performed using known concentrations of the K8 antibody fragment. A 96-well plate was coated with lOOμl of HSA, BSA and β-gal at lOμg/ml and lOOμl of Protein A at 1 μg/ml concentration. 50μl of the serial dilutions of the K8 scFv was applied and the bound antibody fragments were detected with Protein L- HRP. ELISA results confirmed the dual specific nature of the K8 antibody (Figure 5).
To confirm that binding to β-gal is determined by the Vκ domain and binding to
HSA/BSA by the VH domain of the K8 scFv antibody, the Vκ domain was cut out from K8 scFv DNA by SalZ/Not/ digestion and ligated into a Sal//Not/ digested pIT2 vector containing dummy VH chain (Figures 1 and 2). Binding characteristics of the resulting clone K8VK/dummy VH were analysed by soluble scFv ELISA. Production of the soluble scFv fragments was induced by LPTG as described by Harrison et al, (1996) and the supernatant (50μ) containing scFvs assayed directly. Soluble scFv ELISA was performed as described in Example 1 and the bound scFvs were detected with Protein L-HRP. The ELISA results revealed that this clone was still able to bind β-gal, whereas binding to BSA was abolished (Figure 6).
Example 3. Selection of single VJJ domain antibodies antigens A and B and single Vκ domain antibodies directed against antigens C and D.
This example describes a method for making single VJJ domain antibodies directed against antigens A and B and single Vκ domain antibodies directed against antigens C and D by selecting repertoires of virgin single antibody variable domains for binding to these antigens in the absence of the complementary variable domains. Selections and characterisation of the binding clones is performed as described previously (see Example 5, PCT/GB 02/003014). Four clones are chosen for further work:
VHl - Anti A VH VH2 - Anti B Vπ VKl -Anti C VK VK2 -Anti D VK
The procedures described above in Examples 1-3 may be used, in a similar manner as that described, to produce dimer molecules comprising combinations of VH domains (i.e., VH- VH ligands) and cominations of VL domains (VL-VL ligands).
Example 4. Creation and characterisation of the dual specific ScFv antibodies (VH1/VH2 directed against antigens A and B and VK1/VK2 directed against antigens C and D).
This example demonstrates that dual specific ScFv antibodies (VH1/VH2 directed against antigens A and B and VK1/VK2 directed against antigens C and D) could be created by combining Vκ and VH single domains selected against respective antigens in a ScFv vector.
To create dual specific antibody VH1/VH2, VHl single domain is excised from variable domain vector 1 (Figure 7) by NcόllXhόl digestion and ligated into NcollXhol digested variable domain vector 2 (Figure 7) to create VHl/ variable domain vector 2. VH2 single domain is PCR amplified from variable domain vector 1 using primers to introduce Sail restriction site to the 5' end and Notl restriction site to the 3' end. The PCR product is then digested with SaWNotl and ligated into SaWNotl digested VHl/ variable domain vector 2 to create VH1/VH2/ variable domain vector 2.
VK1/VK2/ variable domain vector 2 is created in a similar way. The dual specific nature of the produced VH1/VH2 ScFv and VK1/VK2 ScFv is tested in a soluble ScFv ELISA as described previously (see Example 6, PCT/GB 02/003014). Competition ELISA is performed as described previously (see Example 8, PCT/GB 02/003014).
Possible outcomes: -VH1/VH2 ScFv is able to bind antigens A and B simultaneously -VK1/VK2 ScFv is able to bind antigens C and D simultaneously -VH1/VH2 ScFv binding is competitive (when bound to antigen A, VH1/VH2 ScFv cannot bind to antigen B)
-VK1/VK2 ScFv binding is competitive (when bound to antigen C, VK1/VK2 ScFv cannot bind to antigen D)
Example 5. Construction of dual specific VH1/VH2 Fab and VK1/VK2 Fab and analysis of their binding properties.
To create VH1/VH2 Fab, VHl single domain is ligated into NcόUXliol digested CH vector (Figure 8) to create VH1/CH and VH2 single domain is ligated into SaWNotl digested CK vector (Figure 9) to create VH2/CK. Plasmid DNA from VH1/CH and VH2/CK is used to co-transform competent E. coli cells as described previously (see Example 8, PCT/GB02/003014).
The clone containing VH1/CH and VH2/CK plasmids is then induced by L?TG to produce soluble VH1/VH2 Fab as described previously (see Example 8, PCT/GB 02/003014).
VK1/VK2 Fab is produced in a similar way.
Binding properties of the produced Fabs are tested by competition ELISA as described previously (see Example 8, PCT/GB 02/003014).
Possible outcomes:
-VH1/VH2 Fab is able to bind antigens A and B simultaneously -VK1/VK2 Fab is able to bind antigens C and D simultaneously -VH1/VH2 Fab binding is competitive (when bound to antigen A, VH1/VH2 Fab cannot bind to antigen B)
-VK1/VK2 Fab binding is competitive (when bound to antigen C, VK1/VK2 Fab cannot bind to antigen D)
Example 6
Chelating dAb Dimers
Summary
VH and VK homo-dimers are created in a dAb-linker-dAb format using flexible polypeptide linkers. Vectors were created in the dAb linker-dAb format containing glycine-serine linkers of different lengths 3U:(Gly4Ser)3, 5U:(Gly Ser)5, 7U:(Gly4Ser)7. Dimer libraries were created using guiding dAbs upstream of the linker: TAR1-5 (VK), TAR1-27(VK), TAR2-5(VH) or TAR2-6(VK) and a library of corresponding second dAbs after the linker. Using this method, novel dimeric dAbs were selected. The effect of dimerisation on antigen binding was determined by ELISA and BIAcore studies and in cell neutralisation and receptor binding assays. Dimerisation of both TAR1-5 and TAR1- 27 resulted in significant improvement in binding affinity and neutralisation levels.
1.0 Methods
1.1 Library generation 1.1.1 Vectors pEDA3U, pEDA5U and pEDA7U vectors were designed to introduce different linker lengths compatible with the dAb-linker-dAb format. For pEDA3U, sense and anti-sense 73-base pair oligo linkers were annealed using a slow annealing program (95°C-5mins, 80°C-10mins, 70°C-15mins, 56°C-15mins, 42°C until use) in buffer containing O.lMNaCl, lOmM Tris-HCl pH7.4 and cloned using the Xltol and Notl restriction sites. The linkers encompassed 3 (Gly4Ser) units and a stuffer region housed between Sail and Notl cloning sites (scheme 1). In order to reduce the possibility of monomeric dAbs being selected for by phage display, the stuffer region was designed to include 3 stop codons, a Sacl restriction site and a frame shift mutation to put the region out of frame when no second dAb was present. For pEDA5U and 7U due to the length of the linkers required, overlapping oligo-linkers were designed for each vector, annealed and elongated using Klenow. The fragment was then purified and digested using the appropriate enzymes before cloning using X eXhol and Notl restriction sites.
Linker 3U
Ncol - Xhol - ban i Oti
5U
7U
Still er 1 Stuffer 2
Scheme 1
1.1.2 Library preparation
The N-terminal V gene corresponding to the guiding dAb was cloned upstream of the linker using Ncol and Xhol restriction sites. VH genes have existing compatible sites, however cloning VK genes required the introduction of suitable restriction sites. This was achieved by using modifying PCR primers (VK-DLIBF: 5' cggccatggcgtcaacggacat; VKXholR: 5' atgtgcgctcgagcgtttgattt 3') in 30 cycles of PCR amplification using a 2:1 mixture of SuperTaq (HTBiotechnology Ltd)and pfu turbo (Stratagene). This maintained the Ncol site at the 5' end while destroying the adjacent Sail site and introduced the Xhol site at the 3' end. 5 guiding dAbs were cloned into each of the 3 dimer vectors: TARl-5 (VK), TAR1-27(VK), TAR2-5(VH), TAR2-6(VK) and TAR2-7(VK). All constructs were verified by sequence analysis.
Having cloned the guiding dAbs upstream of the linker in each of the vectors (pEDA3U, 5U and 7U): TARl-5 (VK), TAR1-27(VK), TAR2-5(VH) or TAR2-6(VK) a library of corresponding second dAbs were cloned after the linker. To achieve this, the complimentary dAb libraries were PCR amplified from phage recovered from round 1 selections of either a VK library against Human TNFα (at approximately 1 x 106 diversity after round 1) when TARl-5 or TARl-27 are the guiding dAbs, or a VH or VK library against human p55 TNF receptor (both at approximately 1 x 105 diversity after round 1) when TAR2-5 or TAR2-6 respectively are the guiding dAbs. For VK libraries PCR amplification was conducted using primers in 30 cycles of PCR amplification using a 2:1 mixture of SuperTaq and pfu turbo. VH libraries were PCR amplified using primers in order to introduce a Sail restriction site at the 5' end of the gene. The dAb library PCRs were digested with the appropriate restriction enzymes, ligated into the corresponding vectors down stream of the linker, using Sall/Notl restriction sites and electroporated into freshly prepared competent TGI cells.
The titres achieved for each library are as follows: TARl-5: pEDA3U = 4xl08, pEDA5U = 8xl07, pEDA7U = 1x10s TARl-27: pEDA3U = 6.2xl08, pEDA5U = 1x10s, pEDA7U = lxlO9 TAR2h-5: pEDA3U = 4xl07, pEDA5U = 2 x 108, pEDA7U = 8xl07 TAR2h-6: pEDA3U = 7.4xl08, pEDA5U = 1.2 x 108, pEDA7U = 2.2xl08
1.2 Selections 1.2.1 TNFα
Selections were conducted using human TNFα passively coated on immunotubes. Briefly, Immunotubes are coated overnight with l-4mls of the required antigen. The immunotubes were then washed 3 times with PBS and blocked with 2%milk powder in PBS for l-2hrs and washed a further 3 times with PBS. The phage solution is diluted in 2%milk powder in PBS and incubated at room temperature for 2hrs. The tubes are then washed with PBS and the phage eluted with lmg/ml trypsin-PBS. Three selection strategies were investigated for the TARl-5 dimer libraries. The first round selections were carried out in immunotubes using human TNFα coated at 1 μg/ml or 20μg/ml with 20 washes in PBS 0.1%Tween. TGI cells are infected with the eluted phage and the titres are determined (eg, Marks et al J Mol Biol. 1991 Dec 5;222(3):581-97, Richmann et al Biochemistry. 1993 Aug 31;32(34):8848-55).
The titres recovered were: pEDA3U = 2.8xl07 (1 μg ml TNF) 1.5x108 (20μg/mlTNF), pEDA5U = 1.8xl07 (1 μg/ml TNF), 1.6xl08 (20μg/ml TNF) pEDA7U = 8xl06 (1 μg ml TNF), 7xl07 (20μg/ml TNF).
The second round selections were carried out using 3 different methods.
1. In immunotubes, 20 washes with overnight incubation followed by a further 10 washes. 2. In immunotubes, 20 washes followed by lhr incubation at RT in wash buffer with (1 μg/ml TNFα) and 10 further washes.
3. Selection on streptavidin beads using 33 pmoles biotinylated human TNFα (Henderikx et al, 2002, Selection of antibodies against biotinylated antigens. Antibody Phage Display : Methods and protocols, Ed. O'Brien and Atkin, Humana Press). Single clones from round 2 selections were picked into 96 well plates and crude supernatant preps were made in 2ml 96 well plate format.
Figure imgf000095_0001
For TARl-27, selections were carried out as described previously with the following modifications. The first round selections were carried out in immunotubes using human TNFα coated at 1 μg/ml or 20μg/ml with 20 washes in PBS 0.1%Tween. The second round selections were carried out in immunotubes using 20 washes with overnight incubation followed by a further 20 washes. Single clones from round 2 selections were picked into 96 well plates and crude supernatant preps were made in 2ml 96 well plate format.
TARl-27 titres are as follows:
Figure imgf000096_0001
1.2.2 TNFRECEPTOR 1 (p55 RECEPTOR; TAR2)
Selections were conducted as described previously for the TAR2h-5 libraries only. 3 rounds of selections were carried out in immunotubes using either 1 μg/ml human p55 TNF receptor or lOμg/ml human p55 TNF receptor with 20 washes in PBS 0.1%oTween with overnight incubation followed by a further 20 washes. Single clones from round 2 and 3 selections were picked into 96 well plates and crude supernatant preps were made in 2ml 96 well plate format.
TAR2h-5 titres are as follows:
Figure imgf000096_0002
1.3 Screening
Single clones from round 2 or 3 selections were picked from each of the 3U, 5U and 7U libraries from the different selections methods, where appropriate. Clones were grown in 2xTY with lOOμg/ml ampicillin and 1% glucose overnight at 37°C. A 1/100 dilution of this culture was inoculated into 2mls of 2xTY with lOOμg/ml ampicillin and 0.1 %> glucose in 2ml, 96 well plate format and grown at 37°C shaking until OD600 was approximately 0.9. The culture was then induced with ImM IPTG overnight at 30°C. The supernatants were clarified by centrifugation at 4000rpm for 15 mins in a sorval plate centrifuge. The supernatant preps the used for initial screening.
1.3.1 ELISA
Binding activity of dimeric recombinant proteins was compared to monomer by Protein A/L ELISA or by antigen ELISA. Briefly, a 96 well plate is coated with antigen or Protein A L overnight at 4°C. The plate washed with 0.05% Tween-PBS, blocked for 2hrs with 2% Tween-PBS. The sample is added to the plate incubated for 1 hr at room temperature. The plate is washed and incubated with the secondary reagent for lhr at room temperature. The plate is washed and developed with TMB substrate. Protein A/L- HRP or India-HRP was used as a secondary reagent. For antigen ELISAs, the antigen concentrations used were 1 μg/ml in PBS for Human TNFα and human THF receptor 1. Due to the presence of the guiding dAb in most cases dimers gave a positive ELISA signal therefore off rate determination was examined by BIAcore.
1.3.2 BIAcore
BIAcore analysys was conducted for TARl-5 and TAR2h-5 clones. For screening,
Human TNFαwas coupled to a CM5 chip at high density (approximately 10000 RUs). 50 μl of Human TNFα(50 μg/ml) was coupled to the chip at 5μl/min in acetate buffer - pH5.5. Regeneration of the chip following analysis using the standard methods is not possible due to the instability of Human TNFα, therefore after each sample was analysed, the chip was washed for lOmins with buffer.
For TARl-5, clones supernatants from the round 2 selection were screened by BIAcore. 48 clones were screened from each of the 3U, 5U and 7U libraries obtained using the following selection methods:
RI: 1 μg/ml human TNFα immunotube, R2 1 μg/ml human TNFα immunotube, overnight wash.
RI: 20 μg/ml human TNFc immunotube, R2 20μg/ml human TNFα immunotube, overnight wash.
RI: 1 μg/ml human TNFα immunotube, R2 33 pmoles biotinylated human TNFα on beads.
RI: 20μg/ml human TNFα immunotube, R2 33 pmoles biotinylated human TNFα beads. For screening, human p55 TNF receptor was coupled to a CM5 chip at high density (approximately 4000 RUs). 100 μl of human p55 TNF receptor (10 μg/ml) was coupled to the chip at 5μl/min in acetate buffer - pH5.5. Standard regeneration conditions were examined ( glycine pH2 or pH3) but in each case antigen was removed from the surface of the chip therefore as with TNFα, therefore after each sample was analysed, the chip was washed for lOmins with buffer.
For TAR2-5, clones supernatants from the round 2 selection were screened. 48 clones were screened from each of the 3U, 5U and 7U libraries, using the following selection methods :
RI: lμg/ml human p55 TNF receptor immunotube, R2 lμg/ml human p55 TNF receptor immunotube, overnight wash.
RI: lOμg/ml human p55 TNF receptor immunotube, R2 lOμg/ml human p55 TNF receptor immunotube, overnight wash.
1.3.3 Receptor and Cell Assays
The ability of the dimers to neutralise in the receptor assay was conducted as follows:
Receptor binding Anti-TNF dAbs were tested for the ability to inhibit the binding of TNF to recombinant TNF receptor 1 (p55). Briefly, Maxisorp plates were incubated overnight with 30mg/ml anti-human Fc mouse monoclonal antibody (Zymed, San Francisco, USA). The wells were washed with phosphate buffered saline (PBS) containing 0.05% Tween-20 and then blocked with 1% BSA in PBS before being incubated with lOOng/ml TNF receptor 1 Fc fusion protein (R&D Systems, Minneapolis, USA). Anti-TNF dAb was mixed with TNF which was added to the washed wells at a final concentration of lOng/ml. TNF binding was detected with 0.2mg/ml biotinylated anti-TNF antibody (HyCult biotechnology, Uben, Netherlands) followed by 1 in 500 dilution of horse radish peroxidase labelled streptavidin (Amersham Biosciences, UK) and then incubation with TMB substrate (KPL, Gaithersburg, USA). The reaction was stopped by the addition of HCl and the absorbance was read at 450nm. Anti-TNF dAb activity lead to a decrease in TNF binding and therefore a decrease in absorbance compared with the TNF only control. L929 Cytotoxicity Assay
Anti-TNF dAbs were also tested for the ability to neutralise the cytotoxic activity of TNF on mouse L929 fibroblasts (Evans, T. (2000) Molecular Biotechnology 15, 243-248). Briefly, L929 cells plated in microtitre plates were incubated overnight with anti-TNF dAb, lOOpg/ml TNF and lmg/ml actinomycin D (Sigma, Poole, UK). Cell viability was measured by reading absorbance at 490nm following an incubation with [3-(4,5- dimethylthiazol-2-yl)-5-(3-carbboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium (Promega, Madison, USA). Anti-TNF dAb activity lead to a decrease in TNF cytotoxicity and therefore an increase in absorbance compared with the TNF only control.
In the initial screen, supernatants prepared for BIAcore analysis, described above, were also used in the receptor assay. Further analysis of selected dimers was also conducted in the receptor and cell assays using purified proteins.
HeLa IL-8 assay
Anti-TNFRl or anti-TNF alpha dAbs were tested for the ability to neutralise the induction of IL-8 secretion by TNF in HeLa cells (method adapted from that of Akeson, L. et al (1996) Journal of Biological Chemistry 271, 30517-30523, describing the induction of IL-8 by IL-1 in HUVEC; here we look at induction by human TNF alpha and we use HeLa cells instead of the HUVEC cell line). Briefly, HeLa cells plated in microtitre plates were incubated overnight with dAb and 300pg/ml TNF. Post incubation the supernatant was aspirated off the cells and IL-8 concentration measured via a sandwich ELISA (R&D Systems). Anti-TNFRl dAb activity lead to a decrease in IL-8 secretion into the supernatant compared with the TNF only control.
The L929 assay is used throughout the following experiments; however, the use of the HeLa IL-8 assay is preferred to measure anti-TNF receptor 1 (p55) ligands; the presence of mouse p55 in the L929 assay poses certain limitations in its use.
1.4 Sequence analysis
Dimers that proved to have interesting properties in the BIAcore and the receptor assay screens were sequenced. Sequences are detailed in the sequence listing. 1.5 Formatting
1.5.1 TARl-5-19 dimers
The TARl-5 dimers that were shown to have good neutralisation properties were reformatted and analysed in the cell and receptor assays. The TARl-5 guiding dab was substituted with the affinity matured clone TARl-5-19. To achieve this TARl-5 was cloned out of the individual dimer pair and substituted with TARl-5-19 that had been amplified by PCR. In addition, TARl-5-19 homodimers were also constructed in the 3U, 5U and 7U vectors. The N terminal copy of the gene was amplified by PCR and cloned as described above and the C-terminal gene fragment was cloned using existing Sail and Notl restriction sites.
1.5.2 Mutagenesis
The amber stop codon present in dAb2, one of the C-terminal dAbs in the TARl-5 dimer pairs was mutated to a glutamine by site-directed mutagenesis.
1.5.3 Fabs
The dimers containing TARl-5 or TARl-5-19 were re-formatted into Fab expression vectors. dAbs were cloned into expression vectors containing either the CK or CH genes using Sfil and Notl restriction sites and verified by sequence analysis. The CK vector is derived from a pUC based ampicillin resistant vector and the CH vector is derived from a pACYC chloramphenicol resistant vector. For Fab expression the dAb-CH and dAb-CK constructs were co-transformed into HB2151 cells and grown in 2xTY containing 0.1% glucose, lOOμg/ml ampicillin and lOμg/ml chloramphenicol.
1.5.3 Hinge dimerisation
Dimerisation of dAbs via cystine bond formation was examined. A short sequence of amino acids EPKSGDKTHTCPPCP a modified form of the human IgGCl hinge was engineered at the C terminal region on the dAb. An oligo linker encoding for this sequence was synfhesised and annealed, as described previously. The linker was cloned into the pEDA vector containing TARl-5-19 using Xhol and Notl restriction sites. Dimerisation occurs in situ in the periplasm.
1.6 Expression and purification 1.6.1 Expression
Supernatants were prepared in the 2ml, 96-well plate format for the initial screening as described previously. Following the initial screening process selected dimers were analysed further. Dimer constructs were expressed in TOPI OF' or HB2151 cells as supernatants. Briefly, an individual colony from a freshly streaked plate was grown overnight at 37°C in 2xTY with lOOμg/ml ampicillin and 1% glucose. A 1/100 dilution ofthis culture was inoculated into 2xTY with lOOμg/ml ampicillin and 0.1%o glucose and grown at 37°C shaking until OD600 was approximately 0.9. The culture was then induced with ImM IPTG overnight at 30°C. The cells were removed by centrifugation and the supernatant purified with protein A or L agarose.
Fab and cysteine hinge dimers were expressed as periplasmic proteins in HB2152 cells. A 1/100 dilution of an overnight culture was inoculated into 2xTY with 0.1% glucose and the appropriate antibiotics and grown at 30°C shaking until OD600 was approximately 0.9. The culture was then induced with ImM IPTG for 3-4 hours at 25°C. The cells were harvested by centrifugation and the pellet resuspended in periplasmic preparation buffer (30mM Tris-HCl pH8.0, ImM EDTA, 20% sucrose). Following centrifugation the supernatant was retained and the pellet resuspended in 5mM MgSO4. The supernatant was harvested again by centrifugation, pooled and purified.
1.6.2 Protein A/L purification
Optimisation of the purification of dimer proteins from Protein L agarose (Affitech, Norway) or Protein A agarose (Sigma, UK) was examined. Protein was eluted by batch or by column elution using a peristaltic pump. Three buffers were examined 0.1M Phosphate-citrate buffer pH2.6, 0.2M Glycine pH2.5 and 0.1M Glycine pH2.5. The optimal condition was determined to be under peristaltic pump conditions using 0.1M Glycine pH2.5 over 10 column volumes. Purification from protein A was conducted peristaltic pump conditions using 0.1M Glycine pH2.5.
1.6.3 FPLC purification Further purification was carried out by FPLC analysis on the AKTA Explorer 100 system (Amersham Biosciences Ltd). TARl-5 and TARl-5-19 dimers were fractionated by cation exchange chromatography (1ml Resource S - Amersham Biosciences Ltd) eluted with a 0-1M NaCl gradient in 50mM acetate buffer pH4. Hinge dimers were purified by ion exchange (1ml Resource Q Amersham Biosciences Ltd) eluted with a 0-1M NaCl gradient in 25mMTris HCl pH 8.0. Fabs were purified by size exclusion chromatography using a superose 12 (Amersham Biosciences Ltd ) column run at a flow rate of 0.5ml/min in PBS with 0.05% tween. Following purification samples were concentrated using vivaspin 5K cut off concentrators (Vivascience Ltd).
2.0 Results
2.1 TARl-5 dimers
6 x 96 clones were picked from the round 2 selection encompassing all the libraries and selection conditions. Supernatant preps were made and assayed by antigen and Protein L ELISA, BIAcore and in the receptor assays. In ELISAs, positive binding clones were identified from each selection method and were distributed between 3U, 5U and 7U libraries. However, as the guiding dAb is always present it was not possible to discriminate between high and low affinity binders by this method therefore BIAcore analysis was conducted.
BIAcore analysis was conducted using the 2ml supernatants. BIAcore analysis revealed that the dimer Koff rates were vastly improved compared to monomeric TARl-5. Monomer Koff rate was in the range of 10_1M compared with dimer Koff rates which were in the range of 10"3 - 10"4M. 16 clones that appeared to have very slow off rates were selected, these came from the 3U, 5U and 7U libraries and were sequenced. hi addition the supernatants were analysed for the ability to neutralise human TNFα in the receptor assay.
6 lead clones (dl-d6 below) that neutralised in these assays and have been sequenced. The results shows that out of the 6 clones obtained there are only 3 different second dAbs (dAbl, dAb2 and dAb3) however where the second dAb is found more than once they are linked with different length linkers.
TARl-5dl : 3U linker 2nd dAb=dAbl - 1 μg/ml Ag immunotube overnight wash TARl-5d2: 3U linker 2nd dAb=dAb2 - 1 μg/ml Ag immunotube overnight wash TARl-5d3: 5U linker 2nd dAb=dAb2 - 1 μg/ml Ag immunotube overnight wash TARl-5d4: 5U linker 2nd dAb=dAb3 - 20μg/ml Ag immunotube overnight wash TARl-5d5: 5U linker 2nd dAb=dAbl - 20μg/ml Ag immunotube overnight wash TARl-5d6: 7U linker 2nd dAb=dAbl- Rl:l μg/ml Ag immunotube overnight wash, R2:beads
The 6 lead clones were examined further. Protein was produced from the periplasm and supernatant, purified with protein L agarose and examined in the cell and receptor assays. The levels of neutralisation were variable (Table 1). The optimal conditions for protein preparation were determined. Protein produced from HB2151 cells as supernatants gave the highest yield (approximately lOmgs/L of culture). The supernatants were incubated with protein L agarose for 2hrs at room temperature or overnight at 4°C. The beads were washed with PBS/NaCl and packed onto an FPLC column using a peristaltic pump. The beads were washed with 10 column volumes of PBS/NaCl and eluted with 0.1M glycine pH2.5. In general, dimeric protein is eluted after the monomer.
TARl-5dl-6 dimers were purified by FPLC. Three species were obtained, by FPLC purification and were identified by SDS PAGE. One species corresponds to monomer and the other two species corresponds to dimers of different sizes. The larger of the two species is possibly due to the presence of C terminal tags. These proteins were examined in the receptor assay. The data presented in table 1 represents the optimum results obtained from the two dimeric species (Figure 11)
The three second dAbs from the dimer pairs (ie, dAbl, dAb2 and dAb3) were cloned as monomers and examined by ELISA and in the cell and receptor assay. All three dAbs bind specifically to TNF by antigen ELISA and do not cross react with plastic or BSA. As monomers, none of the dAbs neutralise in the cell or receptor assays.
2.1.2 TARl-5-19 dimers
TARl-5-19 was substituted for TARl-5 in the 6 lead clones. Analysis of all TARl-5-19 dimers in the cell and receptor assays was conducted using total protein (protein L purified only) unless otherwise stated (Table 2). TARl-5-19d4 and TARl-5-19d3 have the best ND50 (~5nM) in the cell assay, this is consistent with the receptor assay results and is an improvement over TARl-5-19 monomer (ND50~30nM). Although purified TARl-5 dimers give variable results in the receptor and cell assays TARl-5-19 dimers were more consistent. Variability was shown when using different elution buffers during the protein purification. Elution using 0.1M Phosphate-citrate buffer pH2.6 or 0.2M Glycine pH2.5 although removing all protein from the protein L agarose in most cases rendered it less functional.
TARl-5-19d4 was expressed in the fermenter and purified on cation exchange FPLC to yield a completely pure dimer. As with TARl-5d4 three species were obtained, by FPLC purification corresponding to monomer and two dimer species. This dimer was amino acid sequenced. TARl-5-19 monomer and TARl-5-19d4 were then examined in the receptor assay and the resulting IC50 for monomer was 30nM and for dimer was 8nM. The results of the receptor assay comparing TARl-5-19 monomer, TARl-5-19d4 and TARl-5d4 is shown in figure 10.
TARl-5-19 homodimers were made in the 3U, 5U and 7U vectors, expressed and purified on Protein L. The proteins were examined in the cell and receptor assays and the resulting IC50s (for receptor assay) and ND50s (for cell assay) were determined (table 3, figure 12).
2.2 Fabs
TARl-5 and TARl-5-19 dimers were also cloned into Fab format, expressed and purified on protein L agarose. Fabs were assessed in the receptor assays (Table 4). The results showed that for both TARl-5-19 and TARl-5 dimers the neutralisation levels were similar to the original Gly4Ser linker dimers from which they were derived. A TARl-5-19 Fab where TARl-5-19 was displayed on both CH and CK was expressed, protein L purified and assessed in the receptor assay. The resulting IC50 was approximately InM.
2.3 TARl-27 dimers
3 x 96 clones were picked from the round 2 selection encompassing all the libraries and selection conditions. 2ml supernatant preps were made for analysis in ELISA and bioassays. Antigen ELISA gave 71 positive clones. The receptor assay of crude supernatants yielded 42 clones with inhibitory properties (TNF binding 0-60%). In the majority of cases inhibitory properties correlated with a strong ELISA signal. 42 clones were sequenced, 39 of these have unique second dAb sequences. The 12 dimers that gave the best inhibitory properties were analysed further.
The 12 neutralising clones were expressed as 200ml supernatant preps and purified on protein L. These were assessed by protein L and antigen ELISA, BIAcore and in the receptor assay. Strong positive ELISA signals were obtained in all cases. BIAcore analysis revealed all clones to have fast on and off rates. The off rates were improved compared to monomeric TARl-27, however the off rate of TARl-27 dimers was faster (Koff is approximately in the range of 10"1 and 10"2M) than the TARl-5 dimers examined previously (Koff is approximately in the range of 10"3 - 10"4M). The stability of the purified dimers was questioned and therefore in order to improve stability, the addition on 5%glycerol, 0.5% Triton X100 or 0.5% NP40 (Sigma) was included in the purification of
2 TARl-27 dimers (d2 and dl6). Addition of NP40 or Triton X100™ improved the yield of purified product approximately 2 fold. Both dimers were assessed in the receptor assay. TARl-27d2 gave IC50 of ~30nM under all purification conditions. TARl-27dl6 showed no neutralisation effect when purified without the use of stabilising agents but gave an IC50 of ~50nM when purified under stabilising conditions. No further analysis was conducted.
2.4 TAR2-5 dimers
3 x 96 clones were picked from the second round selections encompassing all the libraries and selection conditions. 2ml supernatant preps were made for analysis. Protein A and antigen ELISAs were conducted for each plate. 30 interesting clones were identified as having good off-rates by BIAcore (Koff ranges between 10"2 - 10"3M). The clones were sequenced and 13 unique dimers were identified by sequence analysis.
Table 1: TARl-5 dimers
Figure imgf000105_0001
Figure imgf000106_0001
*note dimer 2 and dimer 3 have the same second dAb (called dAb2), however have different linker lengths (d2 = (Gly Ser)3, d3 = (Gly4Ser)3). dAbl is the partner dAb to dimers 1, 5 and 6. dAb3 is the partner dAb to dimer4. None of the partner dAbs neutralise alone. FPLC purification is by cation exchange unless otherwise stated. The optimal dimeric species for each dimer obtained by FPLC was determined in these assays.
Table 2: TARl-5-19 dimers
Figure imgf000106_0002
Table 3: TARl-5-19 homodimers
Figure imgf000106_0003
Figure imgf000107_0001
Table 4: TAR1-5/TAR1-5-19 Fabs
Figure imgf000107_0002
PCR construction of TAR1-5-19CYS dimer
See example 8 describing the dAb trimer. The trimer protocol gives rise to a mixture of monomer, dimer and trimer.
Expression and purification of TAR1-5-19CYS dimer
The dimer was purified from the supernatant of the culture by capture on Protein L agarose as outlined in the example 8.
Separation of TAR1-5-19CYS monomer from the TAR1-5-19CYS dimer Prior to cation exchange separation, the mixed monomer/dimer sample was buffer exchanged into 50 mM sodium acetate buffer pH 4.0 using a PD-10 column (Amersham Pharmacia), following the manufacturer's guidelines. The sample was then applied to a ImL Resource S cation exchange column (Amersham Pharmacia), which had been pre- equilibrated with 50 mM sodium acetate pH 4.0. The monomer and dimer were separated using the following salt gradient in 50 mM sodium acetate pH 4.0:
150 to 200 mM sodium chloride over 15 column volumes 200 to 450 mM sodium chloride over 10 column volumes 450 to 1000 mM sodium chloride over 15 column volumes
Fractions containing dimer only were identified using SDS-PAGE and then pooled and the pH increased to 8 by the addition of 1/5 volume of IM Tris pH 8.0.
In vitro functional binding assay: TNF receptor assay and cell assay The affinity of the dimer for human TNFo; was determined using the TNF receptor and cell assay. IC50 in the receptor assay was approximately 0.3-0.8 nM; ND50 in the cell assay was approximately 3-8 nM.
Other possible TAR1-5-19CYS dimer formats
PEG dimers and custom synthetic maleimide dimers
Nektar (Shearwater) offer a range of bi-maleimide PEGs [mPEG2-(MAL)2 or mPEG-
(MAL)2] which would allow the monomer to be formatted as a dimer, with a small linker separating the dAbs and both being linked to a PEG ranging in size from 5 to 40 kDa. It has been shown that the 5kDa mPEG-(MAL)2 (ie, [TARl-5-19]-Cys-maleimide-PEG x
2, wherein the maleimides are linked together in the dimer) has an affinity in the TNF receptor assay of - 1-3 nM. Also the dimer can also be produced using TMEA (Tris[2- maleimidoethyl] amine) (Pierce Biotechnology) or other bi-functional linkers.
It is also possible to produce the disulphide dimer using a chemical coupling procedure using 2,2'-dithiodipyridine (Sigma Aldrich) and the reduced monomer.
Addition of a polypeptide linker or hinge to the C-terminus of the dAb.
A small linker, either (Gly4Ser)n where n=l to 10, eg, 1, 2, 3, 4, 5, 6 or 7, an immunoglobulin (eg, IgG hinge region or random peptide sequence (eg, selected from a library of random peptide sequences) can be engineered between the dAb and the terminal cysteine residue. This can then be used to make dimers as outlined above.
Example 8 dAb trimerisation
Summary
For dAb trimerisation, a free cysteine is required at the C-terminus of the protein. The cysteine residue, once reduced to give the free thiol, can then be used to specifically couple the protein to a trimeric maleimide molecule, for example TMEA (Tris[2- maleimido ethyl] amine) .
PCR construction of TAR1-5-19CYS
The following oligonucleotides were used to specifically PCR TARl-5-19 with a Sail and BamiTL sites for cloning and also to introduce a C-terminal cysteine residue:
Sail
Trp Ser Ala Ser Thr Asp* Ile Gin Met Thr Gin Ser Pro Ser Ser Leu Ser Ala Ser Val 1 TGG AGC GCG TCG ACG GAC ATC CAG ATG ACC CAG TCT CCA TCC TCT CTG TCT GCA TCT GTA ACC TCG CGC AGC TGC CTG TAG GTC TAC TGG GTC AGA GGT AGG AGA GAC AGA CGT AGA CAT
Gly Asp Arg Val Thr Ile Thr Cys Arg Ala Ser Gin Ser Ile Asp Ser Tyr Leu His Trp SI GGA GAC CGT GTC ACC ATC ACT TGC CGG GCA AGT CAG AGC ATT GAT AGT TAT TTA CAT TGG CCT CTG GCA CAG TGG TAG TGA ACG GCC CGT TCA GTC TCG TAA CTA TCA ATA AAT GTA ACC
Tyr Gin Gin Lys Pro Gly Lys Ala Pro Lys Leu Leu Ile Tyr Ser Ala Ser Glu Leu Gin 121 TAC CAG CAG AAA CCA GGG AAA GCC CCT AAG CTC CTG ATC TAT AGT GCA TCC GAG TTG CAA
ATG GTC GTC TTT GGT CCC TTT CGG GGA TTC GAG GAC TAG ATA TCA CGT AGG CTC AAC GTT
Ser Gly Val Pro Ser Arg Phe Ser Gly Ser Gly Ser Gly Thr Asp Phe Thr Leu Thr lie
181 AGT GGG GTC CCA TCA CGT TTC AGT GGC AGT GGA TCT GGG ACA GAT TTC ACT CTC ACC ATC TCA CCC CAG GGT AGT GCA AAG TCA CCG TCA CCT AGA CCC TGT CTA AAG TGA GAG TGG TAG
Ser Ser Leu Gin Pro Glu Asp Phe Ala Thr Tyr Tyr Cys Gin Gin Val Val Trp Arg Pro 241 AGC AGT CTG CAA CCT GAA GAT TTT GCT ACG TAC TAC TGT CAA CAG GTT GTG TGG CGT CCT TCG TCA GAC GTT GGA CTT CTA AAA CGA TGC ATG ATG ACA GTT GTC CAA CAC ACC GCA GGA BamHI
Phe Thr Phe Gly Gin Gly Thr Lys Val Glu Ile Lys Arg Cys *** *** Gly Ser Gly
301 TTT ACG TTC GGC CAA GGG ACC AAG GTG GAA ATC AAA CGG TGC TAA TAA GGA TCC GGC
AAA TGC AAG CCG GTT CCC TGG TTC CAC CTT TAG TTT GCC ACG ATT ATT CCT AGG CCG
(* start of TAR1-5-19CYS sequence)
Forward primer
5 '-TGGAGCGCGTCGACGGACATCCAGATGACCCAGTCTCCA-3 '
Reverse primer
5 '-TTAGCAGCCGGATCCTTATTAGCACCGTTTGATTTCCAC-3 '
The PCR reaction (50μL volume) was set up as follows: 200μM dNTPs, 0.4μM of each primer, 5 μL of lOx wTurbo buffer (Sfratagene), 100 ng of template plasmid (encoding TARl-5-19), lμL of twTurbo enzyme (Sfratagene) and the volume adjusted to 50μL using sterile water. The following PCR conditions were used: initial denaturing step 94 °C for 2 mins, then 25 cycles of 94 °C for 30 sees, 64 °C for 30 sec and 72 °C for 30 sec. A final extension step was also included of 72 °C for 5 mins. The PCR product was purified and digested with Sail and BamHI and ligated into the vector which had also been cut with the same restriction enzymes. Correct clones were verified by DNA sequencing. Expression and purification of TAR1-5-19CYS
TAR1-5-19CYS vector was transformed into BL21 (DE3) pLysS chemically competent cells (Novagen) following the manufacturer's protocol. Cells carrying the dAb plasmid were selected for using lOOμg/mL carbenicillin and 37 μg/mL chloramphenicol. Cultures were set up in 2L baffled flasks containing 500 mL of terrific broth (Sigma- Aldrich), lOOμg/mL carbenicillin and 37 μg/mL chloramphenicol. The cultures were grown at 30 °C at 200rpm to an O.D.600 of 1-1.5 and then induced with ImM IPTG (isopropyl-beta- D-thiogalactopyranoside, from Melford Laboratories). The expression of the dAb was allowed to continue for 12-16 hrs at 30 °C. It was found that most of the dAb was present in the culture media. Therefore, the cells were separated from the media by centrifugation (8,000xg for 30 mins), and the supernatant used to purify the dAb. Per litre of supernatant, 30 mL of Protein L agarose (Affitech) was added and the dAb allowed to batch bind with stirring for 2 hours. The resin was then allowed to settle under gravity for a further hour before the supernatant was siphoned off. The agarose was then packed into a XK 50 column (Amersham Phamacia) and was washed with 10 column volumes of PBS. The bound dAb was eluted with 100 mM glycine pH 2.0 and protein containing fractions were then neutralized by the addition of 1/5 volume of 1 M Tris pH 8.0. Per litre of culture supernatant 20 mg of pure protein was isolated, which contained a 50:50 ratio of monomer to dimer.
Trimerisation of TAR1-5-19CYS
2.5 ml of 100 μM TAR1-5-19CYS was reduce with 5 mM dithiothreitol and left at room temperature for 20 minutes. The sample was then buffer exchanged using a PD-10 column (Amersham Pharmacia). The column had been pre-equilibrated with 5 mM EDTA, 50 mM sodium phosphate pH 6.5, and the sample applied and eluted following the manufactures guidelines. The sample was placed on ice until required. TMEA (Tris[2- maleimidoethyl] amine) was purchased from Pierce Biotechnology. A 20 M stock solution of TMEA was made in 100% DMSO (dimethyl sulphoxide). It was found that a concentration of TMEA greater than 3:1 (molar ratio of dAb:TMEA) caused the rapid precipitation and cross-linking of the protein. Also the rate of precipitation and cross- linking was greater as the pH increased. Therefore using 100 μM reduced TAR1-5- 19CYS, 25 μM TMEA was added to trimerise the protein and the reaction allowed to proceed at room temperature for two hours. It was found that the addition of additives such as glycerol or ethylene glycol to 20% (v/v), significantly reduced the precipitation of the trimer as the coupling reaction proceeded. After coupling, SDS-PAGE analysis showed the presence of monomer, dimer and trimer in solution.
Purification of the trimeric TAR1-5-19CYS
40 μL of 40% glacial acetic acid was added per mL of the TMEA-TARl-5-19cys reaction to reduce the pH to ~4. The sample was then applied to a ImL Resource S cation exchange column (Amersham Pharmacia), which had been pre-equilibrated with 50 mM sodium acetate pH 4.0. The dimer and trimer were partially separated using a salt gradient of 340 to 450 mM Sodium chloride, 50 mM sodium acetate pH 4.0 over 30 column volumes. Fractions containing trimer only were identified using SDS-PAGE and then pooled and the pH increased to 8 by the addition of 1/5 volume of IM Tris pH 8.0. To prevent precipitation of the trimer during concentration steps (using 5K cut off Viva spin concentrators; Vivascience), 10% glycerol was added to the sample.
In vitro functional binding assay: TNF receptor assay and cell assay
The affinity of the trimer for human TNFc was determined using the TNF receptor and cell assay. IC50 in the receptor assay was 0.3nM; ND50 in the cell assay was in the range of 3 to lOnM (eg, 3nM).
Other possible TAR1-5-19CYS trimer formats
TAR1-5-19CYS may also be formatted into a trimer using the following reagents:
PEG trimers and custom synthetic maleimide trimers Nektar (Shearwater) offer a range of multi arm PEGs, which can be chemically modified at the terminal end of the PEG. Therefore using a PEG trimer with a maleimide functional group at the end of each arm would allow the trimerisation of the dAb in a manner similar to that outlined above using TMEA. The PEG may also have the advantage in increasing the solubility of the trimer thus preventing the problem of aggregation. Thus, one could produce a dAb trimer in which each dAb has a C-terminal cysteine that is linked to a maleimide functional group, the maleimide functional groups being linked to a PEG trimer. Addition of a polypeptide linker or hinge to the C-terminus of the dAb A small linker, either (Gly4Ser)n where n= 1 to 10, eg, 1, 2, 3, 4, 5, 6 or 7 , an immunoglobulin (eg, IgG hinge region or random peptide sequence (eg, selected from a library of random peptide sequences) could be engineered between the dAb and the terminal cysteine residue. When used to make multimers (eg, dimers or trimers), this again would introduce a greater degree of flexibility and distance between the individual monomers, which may improve the binding characteristics to the target, eg a multisubunit target such as human TNFα.
Example 9.
Selection of a collection of single domain antibodies (dAbs) directed against human serum albumin (HSA) and mouse serum albumin (MSA).
This example explains a method for making a single domain antibody (dAb) directed against serum albumin. Selection of dAbs against both mouse serum albumin (MSA) and human serum albumin (HSA) is described. Three human phage display antibody libraries were used in this experiment, each based on a single human framework for VH (see Figure 13: sequence of dummy VH based on V3-23/DP47 and JH4b) or VK (see Figure 15: sequence of dummy VK based on ol2/o2/DPK9 and Jkl) with side chain diversity encoded by NNK codons incorporated in complementarity determining regions (CDR1, CDR2 and CDR3).
Library 1 (VH): Diversity at positions: H30, H31, H33, H35, H50, H52, H52a, H53, H55, H56, H58, H95, H97, H98. Library size: 6.2 x 109
Library 2 (VH): Diversity at positions: H30, H31, H33, H35, H50, H52, H52a, H53, H55, H56, H58, H95, H97, H98, H99, H100, HlOOa, HlOOb. Library size: 4.3 x 109 Library 3 (VK):
Diversity at positions: L30, L31, L32, L34, L50, L53, L91, L92, L93, L94, L96 Library size: 2 x 109
The VH and VK libraries have been preselected for binding to generic ligands protein A and protein L respectively so that the majority of clones in the unselected libraries are functional. The sizes of the libraries shown above correspond to the sizes after preselection.
Two rounds of selection were performed on serum albumin using each of the libraries separately. For each selection, antigen was coated on immunotube (nunc) in 4ml of PBS at a concentration of lOOμg/ml. In the first round of selection, each of the three libraries was panned separately against HSA (Sigma) and MSA (Sigma). In the second round of selection, phage from each of the six first round selections was panned against (i) the same antigen again (eg 1st round MSA, 2nd round MSA) and (ii) against the reciprocal antigen (eg 1st round MSA, 2nd round HSA) resulting in a total of twelve 2nd round selections, hi each case, after the second round of selection 48 clones were tested for binding to HSA and MSA. Soluble dAb fragments were produced as described for scFv fragments by Harrison et al, Methods Enzymol. 1996;267:83-109 and standard ELISA protocol was followed (Hoogenboom et al. (1991) Nucleic Acids Res., 19: 4133) except that 2% tween PBS was used as a blocking buffer and bound dAbs were detected with either protein L-HRP (Sigma) (for the V/ s) and protein A -HRP (Amersham Pharmacia Biotech) (for the VHS).
dAbs that gave a signal above background indicating binding to MSA, HSA or both were tested in ELISA insoluble form for binding to plastic alone but all were specific for serum albumin. Clones were then sequenced (see table below) revealing that 21 unique dAb sequences had been identified. The minimum similarity (at the amino acid level) between the VK dAb clones selected was 86.25%) ((69/80)xl00; the result when all the diversified residues are different, eg clones 24 and 34). The minimum similarity between the VH dAb clones selected was 94 % ((127/136)xl00). Next, the serum albumin binding dAbs were tested for their ability to capture biotinylated antigen from solution. ELISA protocol (as above) was followed except that ELISA plate was coated with lμg/ml protein L (for the VK clones) and lμg/ml protein A (for the VH clones). Soluble dAb was captured from solution as in the protocol and detection was with biotinylated MSA or HSA and streptavidin HRP. The biotinylated MSA and HSA had been prepared according to the manufacturer's instructions, with the aim of achieving an average of 2 biotins per serum albumin molecule. Twenty four clones were identified that captured biotinylated MSA from solution in the ELISA. Two of these (clones 2 and 38 below) also captured biotinylated HSA. Next, the dAbs were tested for their ability to bind MSA coated on a CM5 biacore chip. Eight clones were found that bound MSA on the biacore.
dAb (all Binds capture MSA Captures biotinylated H in biotinylated
MSA) or K CDR1 CDR2 CDR3 biacore? HSA?
VK library 3 template
(dummy) K XXXLX XASXLQS QQXXXXPXT
2, 4, 7, 41, K SSY N RASPLQS QQTYSVPPT ^all 4 bind
38,54 K SSYLN RASPLQS QQTYRIPPT both bind
46, 47, 52, 56 K FKSLK NASYLQS QQWYWPVT
13,15 K. YYHLK KASTLQS QQVRKVPRT
30,35 K RRYLK QASVLQS QQGLYPPIT
19, K YNW K RASSLQS QQNWIPRT
22, K L H R HAS QS QQSAVYPKT
23, K FRYLA HASHLQS QQR LYPKT
24, K FYHLA PASKLQS QQRARWPRT
31, K I HLN RASRLQS QQVARVPRT
33, K YRYLR KASSLQS QQYVGYPRT
34, K LKYLK NASHLQS QQTTYYPIT
53, K RYLR KASWLQS QQVLYYPQT
11, K RSLK AASRLQS QQWYWPAT •
12, K FRHLK AASRLQS QQVALYPKT S
17, K RKYLR TASSLQS QQNLFWPRT
18, K RRYLN AASSLQS QQMLFYPKT
16, 21 K IKH K GASRLQS QQGARWPQT
25,26 K YYHLK KASTLQS QQVRKVPRT s
27, K YKHLK NASHLQS QQVGRYPKT
55, K FKSLK NASYLQS QQWYWPVT
VH library 1
(and 2) template
(dummy) H XXYXXX XIXXXGXXTXYADSVKG XXXX (XXXX) FDY
8,10 H WVYQMD SISAFGAKTLYADSV G SGKFDY
36, H WSYQMT SISSFGSST YADSVKG GRDHNYSLFDY In all cases the frameworks were identical to the frameworks in the corresponding dummy sequence, with diversity in the CDRs as indicated in the table above.
Of the eight clones that bound MSA on the biacore, two clones that are highly expressed in E. coli (clones MSA16 and MSA26) were chosen for further study (see example 10). Full nucleotide and amino acid sequences for MSA16 and 26 are given in figure 16.
Example 10.
Determination of affinity and serum half-life in mouse of MSA binding dAbs MSA16 and MSA26.
dAbs MSA16 and MSA26 were expressed in the periplasm of E. coli and purified using batch absorbtion to protein L-agarose affinity resin (Affitech, Norway) followed by elution with glycine at pH 2.2. The purified dAbs were then analysed by inhibition biacore to determine Kd. Briefly, purified MSA16 and MSA26 were tested to determine the concentration of dAb required to achieve 200RUs of response on a biacore CM5 chip coated with a high density of MSA. Once the required concentrations of dAb had been determined, MSA antigen at a range of concentrations around the expected Kd was premixed with the dAb and incubated overnight. Binding to the MSA coated biacore chip of dAb in each of the premixes was then measured at a high flow-rate of 30 μl minute. The resulting curves were used to create Klotz plots, which gave an estimated Kd of 200nM for MSA16 and 70nM for MSA 26 (Figure 17 A & B).
Next, clones MSA16 and MSA26 were cloned into an expression vector with the HA tag (nucleic acid sequence: TATCCTTATGATGTTCCTGATTATGCA and amino acid sequence: YPYDVPDYA) and 2-10 mg quantities were expressed in E. coli and purified from the supernatant with protein L-agarose affinity resin (Affitech, Norway) and eluted with glycine at pH2.2. Serum half life of the dAbs was determined in mouse. MSA26 and MSA16 were dosed as single i.v. injections at approx 1.5mg/kg into CD1 mice. Analysis of serum levels was by goat anti-HA (Abeam, UK) capture and protein L-HRP (invitrogen) detection ELISA which was blocked with 4%> Marvel. Washing was with 0.05%) tween PBS. Standard curves of known concentrations of dAb were set up in the presence of lxmouse serum to ensure comparability with the test samples. Modelling with a 2 compartment model showed MSA-26 had a tl/2α of 0.16hr, a tl/23 of 14.5hr and an area under the curve (AUC) of 465hr.mg/ml (data not shown) and MSA-16 had a tl/2α of 0.98hr, a tl/2/3 of 36.5hr and an AUC of 913hr.mg/ml (figure 18). Both anti-MSA clones had considerably lengthened half life compared with HEL4 (an anti-hen egg white lysozyme dAb) which had a tl/2α of 0.06hr, and a tl/2/3 of 0.34hr.
Example 11.
Creation of VH-VH and VK- VK dual specific Fab like fragments
This example describes a method for making VH- VH and Vκ-Vκ dual specifics as Fab like fragments. Before constructing each of the Fab like fragments described, dAbs that bind to targets of choice were first selected from dAb libraries similar to those described in example 9. A VH dAb, HEL4, that binds to hen egg lysozyme (Sigma) was isolated and a second VH dAb (TAR2h-5) that binds to TNFo; receptor (R and D systems) was also isolated. The sequences of these are given in the sequence listing. A VK dAb that binds TNFα (TARl-5-19) was isolated by selection and affinity maturation and the sequence is also set forth in the sequence listing. A second VK dAb (MSA 26) described in example 9 whose sequence is in figure 17B was also used in these experiments.
DNA from expression vectors containing the four dAbs described above was digested with enzymes Sail and Notl to excise the DNA coding for the dAb. A band of the expected size (300-400bp) was purified by running the digest on an agarose gel and excising the band, followed by gel purification using the Qiagen gel purification kit (Qiagen, UK). The DNA coding for the dAbs was then inserted into either the CH or CK vectors (Figs 8 and 9) as indicated in the table below.
Figure imgf000117_0001
The VH CH and VH CK constructs were cotransformed into HB2151 cells. Separately, the VK CH and VK CK constructs were cotransformed into HB2151 cells. Cultures of each of the cotransformed cell lines were grown overnight (in 2xTy containing 5% glucose, lOμg/ml chloramphenicol and 100μg/ml ampicillin to maintain antibiotic selection for both CH and CK plasmids). The overnight cultures were used to inoculate fresh media (2xTy, lOμg/ml chloramphenicol and lOOμg/ml ampicillin) and grown to OD 0.7-0.9 before induction by the addition of IPTG to express their CH and CK constructs. Expressed Fab like fragment was then purified from the periplasm by protein A purification (for the contransformed VH CH and VH CK) and MSA affinity resin purification (for the contransformed VK CH and VK CK).
VH-VH dual specific
Expression of the VH CH and VH CK dual specific was tested by running the protein on a gel. The gel was blotted and a band the expected size for the Fab fragment could be detected on the Western blot via both the myc tag and the flag tag, indicating that both the VH CH and VH CK parts of the Fab like fragment were present. Next, in order to determine whether the two halves of the dual specific were present in the same Fab-like fragment, an ELISA plate was coated overnight at 4°C with 100 μl per well of hen egg lysozyme (HEL) at 3 mg/ml in sodium bicarbonate buffer. The plate was then blocked (as described in example 1) with 2%> tween PBS followed by incubation with the VH CH /VH CK dual specific Fab like fragment. Detection of binding of the dual specific to the HEL was via the non cognate chain using 9el0 (a monoclonal antibody that binds the myc tag, Roche) and anti mouse IgG-HRP (Amersham Pharmacia Biotech). The signal for the VH CH /VH CK dual specific Fab like fragment was 0.154 compared to a background signal of 0.069 for the VH CK chain expressed alone. This demonstrates that the Fab like fragment has binding specificity for target antigen.
Vκ-Vκ dual specific
After purifying the contransformed VK CH and VK CK dual specific Fab like fragment on an MSA affinity resin, the resulting protein was used to probe an ELISA plate coated with 1/xg/ml TNFo: and an ELISA plate coated with lOμg/ml MSA. As predicted, there was signal above background when detected with protein L-HRP on bot ELISA plates (data not shown). This indicated that the fraction of protein able to bind to MSA (and therefore purified on the MSA affinity column) was also able to bind TNFo: in a subsequent
ELISA, confirming the dual specificity of the antibody fragment. This fraction of protein was then used for two subsequent experiments. Firstly, an ELISA plate coated with
, 1 μg/ml TNFc was probed with dual specific VK CH and VK CK Fab like fragment and also with a control TNFα binding dAb at a concentration calculated to give a similar signal on the ELISA. Both the dual specific and control dAb were used to probe the
ELISA plate in the presence and in the absence of 2mg/ml MSA. The signal in the dual specific well was reduced by more than 50% but the signal in the dAb well was not reduced at all (see figure 19a). The same protein was also put into the receptor assay with and without MSA and competition by MSA was also shown (see figure 19c). This demonstrates that binding of MSA to the dual specific is competitive with binding to
TNFα
Example 12.
Creation of a VK- VK dual specific cys bonded dual specific with specificity for mouse serum albumin and TNFα
This example describes a method for making a dual specific antibody fragment specific for both mouse serum albumin and TNFo; by chemical coupling via a disulphide bond. Both MSA16 (from example 1) and TARl-5-19 dAbs were recloned into a pET based vector with a C terminal cysteine and no tags. The two dAbs were expressed at 4-10 mg levels and purified from the supernatant using protein L-agarose affinity resin (Affitiech, Norway). The cysteine tagged dAbs were then reduced with dithiothreitol. The TARl-5- 19 dAb was then coupled with dithiodipyridine to block reformation of disulphide bonds resulting in the formation of PEP 1-5-19 homodimers. The two different dAbs were then mixed at pH 6.5 to promote disulphide bond formation and the generation of TARl-5-19, MSA16 cys bonded heterodimers. This method for producing conjugates of two unlike proteins was originally described by King et al. (King TP, Li Y Kochoumian L Biochemistry. 1978 voll7: 1499-506 Preparation of protein conjugates via intermolecular disulfide bond formation.) Heterodimers were separated from monomeric species by cation exchange. Separation was confirmed by the presence of a band of the expected size on a SDS gel. The resulting heterodimeric species was tested in the TNF receptor assay and found to have an IC50 for neutralising TNF of approximately 18 nM. Next, the receptor assay was repeated with a constant concentration of heterodimer (18nM) and a dilution series of MSA and HSA. The presence of HSA at a range of concentrations (up to 2 mg/ml) did not cause a reduction in the ability of the dimer to inhibit TNFα: . However, the addition of MSA caused a dose dependant reduction in the ability of the dimer to inhibit TNFa (figure 20).This demonstrates that MSA and TNFα compete for binding to the cys bonded TARl-5-19, MSA16 dimer.
Data Summary
A summary of data obtained in the experiments set forth in the foregoing examples is set forth in Annex 4.
All publications mentioned in the present specification, and references cited in said publications, are herein incorporated by reference. Various modifications and variations of the described methods and system of the invention will be apparent to those skilled in the art without departing from the scope and spirit of the invention. Although the invention has been described in connection with specific preferred embodiments, it should be understood that the invention as claimed should not be unduly limited to such specific embodiments. Indeed, various modifications of the described modes for carrying out the invention which are obvious to those skilled in molecular biology or related fields are intended to be within the scope of the following claims.
Annex 1; polypeptides which enhance half-life in vivo.
Alpha- 1 Glycoprotein (Orosomucoid) (AAG)
Alpha- 1 Antichyromotrypsin (ACT) Alpha- 1 Antitrypsin (AAT)
Alpha-1 Microglobulin (Protein HC) (AIM)
Alpha-2 Macroglobulin (A2M)
Antithrombin III (AT III)
Apolipoprotein A-l (Apo A-l) Apoliprotein B (Apo B)
Beta-2-microglobulin (B2M)
Ceruloplasmin (Cp)
Complement Component (C3)
Complement Component (C4) Cl Esterase Inhibitor (Cl INH)
C-Reactive Protein (CRP)
Cystatin C (Cys C)
Ferritin (FER)
Fibrinogen (FIB) Fibronectin (FN)
Haptoglobin (Hp)
Hemopexin (HPX)
Immunoglobulin A (IgA)
Immunoglobulin D (IgD) Immunoglobulin E (IgE)
Immunoglobulin G (IgG)
Immunoglobulin M (IgM)
Immunoglobulin Light Chains (kapa/lambda)
Liρoprotein(a) [Lp(a)] Mannose-bindign protein (MBP)
Myoglobin (Myo)
Plasminogen (PSM)
Prealbumin (Transthyretin) (PAL) Retinol-binding protein (RBP) Rheomatoid Factor (RF) Serum Amyloid A (SAA) Soluble Tranferrin Receptor (sTfR) Transferrin (Tf)
Annex 2
Figure imgf000123_0001
Figure imgf000124_0001
Fractalkine expression by human astrocytes.
Many papers on inflammatory responses - i.e. LPS, also macrophage activation.
Anti-TNF and anti-IFN-γ synergize to protect mice from lethal endotoxemia.
Figure imgf000125_0001
Figure imgf000126_0001
Figure imgf000127_0001
Figure imgf000128_0001
Figure imgf000129_0001
Figure imgf000130_0001
Annex 3: Oncology combinations
Figure imgf000131_0001
Figure imgf000132_0001
Annex 4
Data Summary
C co
m
CΛ m m
c m ro σ>
Figure imgf000133_0001
C ro
m
CΛ m m
TJ c m ro σ>
Figure imgf000134_0001

Claims

Claims
1. A dual-specific ligand comprising a first immunoglobulin single variable domain having a binding specificity to a first epitope or antigen and a second complementary immunoglobulin single variable domain having a binding activity to a second epitope or antigen, wherein one or both of said antigens or epitopes acts to increase the half-life of the ligand in vivo and wherein said first and second domains lack mutually complementary domains which share the same specificity, provided that said dual specific ligand does not consist of an anti-HSA NH domain and an anti-β galactosidase Nκ domain.
2. A dual-specific ligand according to claim 1, comprising at least one single heavy chain variable domain of an antibody and one complementary single light chain variable domain of an antibody such that the two regions are capable of associating to form a complementary VH/VL pair.
3. A dual specific ligand according to claim 2 wherein the VH and V are provided by an antibody scFv fragment.
4. A dual-specific ligand according to claim 2 wherein the VH and V are provided by an antibody Fab region.
5. A four chain IgG immunoglobulin ligand comprising a dual specific ligand of claim 2.
6. A four chain IgG immunoglobulin ligand according to claim 5, wherein said IgG comprises two dual specific ligands, said dual specific ligands being identical in their variable domains.
7. A four chain IgG immunoglobulin ligand according to claim 5, wherein said IgG comprises two dual specific ligands, said dual specific ligands being different in their variable domains.
8. A ligand comprising a first immunoglobulin variable domain having a first antigen or epitope binding specificity and a second immunoglobulin variable domain having a second antigen or epitope binding specificity wherein one or both of said first and second variable domains bind to an antigen which increases the half-life of the ligand in vivo, and the variable domains are not complementary to one another.
9. A ligand according to claim 8 wherein the first and the second immunoglobulin variable domains are heavy chain variable domains (VH).
10. A ligand according to claim 8 wherein the first and the second immunoglobulin variable domains are light chain variable domains (VL).
11. A ligand according to claim any preceding claim, wherein the first and second epitopes bind independently, such that the dual specific ligand may simultaneously bind both the first and second epitopes or antigens.
Figure imgf000135_0001
12. A ligand according to claim 11, wherein the dual specific ligand comprises a first form and a second form in equilibrium in solution, wherein both epitopes or antigens bind to the first form independently but compete for binding to the second form.
13. A ligand according to any preceding claim wherein the variable regions are derived from immunoglobulins directed against said epitopes or antigens.
14. A ligand according to any preceding claim, wherein said first and second epitopes are present on separate antigens.
15. A ligand according to any one of claims 1 to 11 , wherein said first and second epitopes are present on the same antigen.
16. A ligand according to any preceding claim comprising a variable domain that is derived from a repertoire of single antibody domains.
17. A ligand of claim 16 wherein said repertoire is displayed on the surface of filamentous bacteriophage and wherein the single antibody domains are selected by binding of the bacteriophage repertoire to antigen.
18. A ligand according to any preceding claim wherein the sequence of at least one variable domain is modified by mutation or DNA shuffling.
19. A dual-specific ligand according to any preceding claim wherein the variable regions are non-covalently associated.
20. A dual-specific ligand according to any one of claims 1 to 18 wherein the variable regions are covalently associated.
21. A dual-specific ligand according to claim 20 wherein the covalent association is mediated by disulphide bonds.
22. A dAb monomer ligand specific for TNFα, which dissociates from human TNFα with a dissociation constant (Kd) of 50nM to 20pM, and a K0ff rate constant of 5xl0_1 to lxlO"7 s"1, as determined by surface plasmon resonance.
23. A dAb monomer ligand specific for TNFα according to claim 22, wherein the dAb is a VK.
24. A dAb monomer ligand specific for TNF receptor 1 (p55), which dissociates from human TNF receptor 1 with a dissociation constant (Kd) of 50nM to 20pM, and a K0ff rate constant of 5x10"1 to lxl 0"7 s"1, as determined by surface plasmon resonance.
25. A dAb monomer ligand according to claim 22 to claim 24, wherein the monomer neutralises human TNFα or TNF receptor 1 in a standard cell assay with an ND50 of 500nM to 50pM.
Figure imgf000136_0001
26. A dAb monomer ligand specific for TNF receptor 1 (p55), wherein the dAb antagonises the activity of the TNF receptor 1 in a standard cell assay with an ND50 of ≤lOOnM, and at a concentration of <10μM the dAb agonises the activity of the TNF receptor 1 by <5% in the assay.
27. A dAb monomer ligand specific for serum albumin (SA) which dissociates from SA with a dissociation constant (Kd) of InM to 500μM, as determined by surface plasmon resonance.
28. A dAb monomer ligand according to claim 27, wherein the monomer binds SA in a standard ligand binding assay with an IC50 of InM to 500μM.
29. A dAb monomer ligand specific for TNFα, wherein the dAb comprises the amino acid sequence of TARl-5-19 or a sequence that is at least 80% homologous thereto.
30. A dAb monomer ligand specific for TNFα, wherein the dAb comprises the amino acid sequence of TARl-5 or a sequence that is at least 80% homologous thereto.
31. A dAb monomer ligand specific for TNFα, wherein the dAb comprises the amino acid sequence of TARl-27 or a sequence that is at least 80% homologous thereto.
32. A dAb monomer ligand specific for TNF receptor 1, wherein the dAb comprises the amino acid sequence of TAR2-10 or a sequence that is at least 80% homologous thereto.
33. A dAb monomer ligand specific for TNF receptor 1, wherein the dAb comprises the amino acid sequence of TAR2-10 or a sseeqquueennccee tthhaatt iiss aatt lleeaasstt 90% homologous thereto.
34. A dAb monomer ligand specific for TNF receptor 1 , wherein the dAb comprises the amino acid sequence of TAR2-5 or a sequence that is at least 80% homologous thereto.
35. A dAb monomer ligand specific for TNF receptor 1 , wherein the dAb comprises the amino acid sequence of TAR2-5 or a sequence that is at least 90% homologous thereto.
36. A dAb monomer ligand specific for SA, wherein the dAb comprises the amino acid sequence of MSA- 16 or a sequence that is at least 80% homologous thereto.
37. A dAb monomer ligand specific for SA, wherein the dAb comprises the amino acid sequence of MSA-26 or a sequence that is at least 80% homologous thereto.
Figure imgf000137_0001
38. A dAb monomer according to any one of claims 29 to 37, wherein the TNFα, TNF receptor 1 or S A is in human form.
39. A dAb monomer further comprising a terminal Cys residue.
40. A dAb monomer according to any one of claims 29 to 38, further comprising a terminal Cys residue.
41. A dual specific ligand comprising at least one dAb monomer according to any one of claims 22 to 40.
42. A dual specific ligand according to claim 41, which is a dimer.
43. A dual specific ligand according to claim 42, wherein the dimer comprises anti-human TNF alpha dAb according to any one of claims 22, 23 and 28-30, and an anti-SA dAb according to any one of claims 26, 27 and 34-36.
44. A dual specific ligand according to claim 42, wherein the dimer is a homo- or hetero-dimer comprising first and second anti-human TNF alpha dAbs, each dAb being according to any one of claims 22, 23 and 28-30.
45. A dual specific ligand according to claim 41, which is a trimer.
46. A dual specific ligand according to claim 45, which is a homotrimer comprising three copies of an anti-human TNF alpha dAb according to any one of claims 22, 23 and 29-31.
47. A ligand according to any preceding claim, which comprises a universal framework.
48. A ligand according to claim 47, wherein the universal framework comprises a VH framework selected from the group consisting of DP47, DP45 and DP38; and/or the VL framework is DPK9.
49. A ligand according to any preceding claim which comprises a binding site for a generic ligand.
50. The ligand of claim 49, wherein the generic ligand binding site is selected from the group consisting of protein A, protein L and protein G.
51. A ligand according to any preceding claim, wherein the ligand comprises a variable domain having one or more framework regions comprising an amino acid sequence that is the same as the amino acid sequence of a corresponding framework region encoded by a human germline antibody gene segment, or the amino acid sequences of one or more of said framework regions collectively comprises up to 5 amino acid differences relative to the amino acid sequence of said corresponding framework region encoded by a human germline antibody gene segment.
Figure imgf000138_0001
52. A ligand according to any one of claims 1 to 51, wherein the ligand comprises a variable domain, wherein the amino acid sequences of FWl, FW2, FW3 and FW4 are the same as the amino acid sequences of corresponding framework regions encoded by a human germline antibody gene segment, or the amino acid sequences of FWl, FW2, FW3 and FW4 collectively contain up to 10 amino acid differences relative to the amino acid sequences of corresponding framework regions encoded by said human germline antibody gene segment.
53. The ligand according to claim 51 or claim 52, which comprises an antibody variable domain comprising FWl, FW2 and FW3 regions, and the amino acid sequence of said FWl, FW2 and FW3 are the same as the amino acid sequences of corresponding framework regions encoded by human germline antibody gene segments.
54. The ligand according to any one of claims 51 to 53, wherein said human germline antibody gene segment is selected from the group consisting of DP47, DP45, DP48 and DPK9.
55. A ligand according to any preceding claim, comprising a VH domain that is not a Camelid immunoglobulin variable domain.
56. The ligand of Claim 55, comprising a VH domain that does not contain one or more amino acids that are specific to Camelid immunoglobulin variable domains as compared to human VH domains.
57. A method for producing a ligand comprising a first immunoglobulin single variable domain having a first binding specificity and a second single immunoglobulin single variable domain having a second binding specificity, one or both of the binding specificities being specific for a protein which increases the half-life of the ligand in vivo, the method comprising the steps of:
(a) selecting a first variable domain by its ability to bind to a first epitope,
(b) selecting a second variable region by its ability to bind to a second epitope,
(c) combining the variable regions; and
(d) selecting the ligand by its ability to bind to said first and second epitopes; wherein, when said variable domains are complementary, neither of said domains is a VH domain specific for HSA.
58. A method according to claim 57 wherein said first variable domain is selected for binding to said first epitope in absence of a complementary variable domain.
59. A method according to claim 57 wherein said first variable domain is selected for binding to said first epitope in the presence of a third complementary variable domain in which said third variable domain is different from said second variable domain.
60. Nucleic acid encoding a dual-specific ligand according to any one of claims 1 to 56.
61. A nucleic acid according to claim 60 which is specific for TNFα, comprising the nucleic acid sequence of TARl-5-19 or a sequence that is at least 70% homologous thereto.
62. A nucleic acid according to claim 60 which is specific for TNFα, comprising the nucleic acid sequence of TARl-5 or a sequence that is at least 70% homologous thereto.
63. A nucleic acid according to claim 60 which is specific for TNFα, comprising the nucleic acid sequence of TARl-27 or a sequence that is at least 70% homologous thereto.
64. A nucleic acid according to claim 60 which is specific for TNF receptor 1, comprising the nucleic acid sequence of TAR2-10 or a sequence that is at least 70% homologous thereto.
65. A nucleic acid according to claim 60 which is specific for TNF receptor 1, comprising the nucleic acid sequence of TAR2-10 or a sequence that is at least 80% homologous thereto.
66. A nucleic acid according to claim 60 which is specific for TNF receptor 1 , comprising the nucleic acid sequence of TAR2h-5 or a sequence that is at least 70% homologous thereto.
67. A nucleic acid according to claim 60 which is specific for TNF receptor 1, comprising the nucleic acid sequence of TAR2h-5 or a sequence that is at least 80% homologous thereto.
68. A nucleic acid according to claim 60 which is specific for SA, comprising the nucleic acid sequence of MSA- 16 or a sequence that is at least 70% homologous thereto.
69. A nucleic acid according to claim 60 which is specific for SA, comprising the nucleic acid sequence of MSA-26 or a sequence that is at least 70% homologous thereto.
70. A vector comprising nucleic acid according to any one of claims 60 to 69.
71. A vector according to claim 70, further comprising components necessary for the expression of a dual-specific ligand.
72. A host cell transfected with a vector according to claim 71.
73. A method for producing a closed conformation multi-specific ligand comprising a first single epitope binding domain having a first epitope binding specificity and a non-complementary second epitope binding domain having a second epitope binding specificity, wherein the first and second binding specificities are capable of competing for epitope binding such that the closed conformation multi-specific ligand may not bind both epitopes simultaneously, said method comprising the steps of: a) selecting a first epitope binding domain by its ability to bind to a first epitope, b) selecting a second epitope binding domain by its ability to bind to a second epitope, c) combining the epitope binding domains such that the domains are in a closed conformation; and d) selecting the closed conformation multispecific ligand by its ability to bind to said first second epitope and said second epitope, but not to both said first and second epitopes simultaneously.
74. A method according to claim 73 wherein the first and the second epitope binding domains are immunoglobulin variable heavy chain domains (VH)-
75. A method according to claim 73 wherein the first and the second immunoglobulin variable domains are immunoglobulin variable light chain domains (vL)-
76. A method according to any one of claims 73 to 75 wherein the immunoglobulin domains are derived from immunoglobulins directed against said epitopes.
77. A method according to any one of claims 73 to 76, wherein said first and second epitopes are present on separate antigens.
78. A method according to any one of claims 73 to 76, wherein said first and second epitopes are present on the same antigen.
79. A method according to any one of claims 73 to 78 wherein the variable domain is derived from a repertoire of single antibody domains.
80. A method of claim 79 wherein said repertoire is displayed on the surface of filamentous bacteriophage and wherein the single antibody domains are selected by binding of the bacteriophage repertoire to antigen.
81. A method of any one of claims 73 to 80 wherein the sequence of at least one immunoglobulin variable domain is modified by mutation or DNA shuffling.
82. A closed conformation multispecific ligand comprising a first epitope binding domain having a first epitope binding specificity and a non- complementary second epitope binding domain having a second epitope binding specificity wherein the first and second binding specificities are capable of competing for epitope binding such that the closed conformation multi-specific ligand cannot bind both epitopes simultaneously.
83. A closed conformation multispecific ligand according to claim 82, obtainable by a method according to any one of claims 73 to 80.
84. A closed conformation multispecific ligand according to claim 82 or claim 83, comprising more than one single heavy chain variable domain of an antibody or more than one light chain variable domain of an antibody.
85. A closed conformation multi-specific ligand according to claim 84 wherein the VH an<1 VL εaQ linked by a peptide linker.
86. A closed conformation multi-specific ligand according to claim 84 wherein the VH or VL are provided by an antibody Fab-like region.
87. A closed conformation multi-specific ligand according to any one of claims 82 to 84 wherein the variable regions are non-covalently associated.
88. A closed conformation multi-specific ligand according to any one of claims 82 to 84 wherein the variable regions are covalently associated.
89. A closed conformation multi-specific ligand according to claim 87 wherein the covalent association is mediated by disulphide bonds.
90. A closed conformation multi-specific ligand according to any of claims 82 to 89 which comprises a universal framework.
91. A closed conformation multi-specific ligand according to any of claims 82 to 90 which comprises a binding site for a generic ligand.
92. The closed conformation multi-specific ligand of claim 91 , wherein the generic ligand binding site is selected from the group consisting of protein A, protein L and protein G.
93. A closed conformation ligand according to any of claims 82 to 92, wherein the ligand comprises a variable domain having one or more framework regions comprising an amino acid sequence that is the same as the amino acid sequence of a corresponding framework region encoded by a human germline antibody gene segment, or the amino acid sequences of one or more of said framework regions collectively comprises up to 5 amino acid differences relative to the amino acid sequence of said corresponding framework region encoded by a human germline antibody gene segment.
94. The closed conformation ligand according to claim 93, wherein the ligand comprises a variable domain wherein the amino acid sequences of FWl, FW2, FW3 and FW4 are the same as the amino acid sequences of corresponding framework regions encoded by a human germline antibody gene segment, or the amino acid sequences of FWl, FW2, FW3 and FW4 collectively contain up to 10 amino acid differences relative to the amino acid sequences of corresponding framework regions encoded by said human germline antibody gene segment.
95. The closed conformation ligand according to claim 93 or claim 94, which comprises an antibody variable domain comprising FWl, FW2 and FW3 regions, and the amino acid sequence of said FWl, FW2 and FW3 are the same as the amino acid sequences of corresponding framework regions encoded by human germline antibody gene segments.
96. The closed conformation ligand according to any one of claims 92 to 95, wherein said human germline antibody gene segment is selected from the group consisting of DP47, DP45, DP48 and DPK9.
97. A closed conformation ligand according to any one of claims 92 to 96, comprising a VH domain that is not a Camelid immunoglobulin variable domain.
98. The closed conformation ligand of Claim 97, wherein the VH domain does not contain one or more amino acids that are specific to Camelid immunoglobulin variable domains as compared to human VH domains.
99. A closed conformation multi-specific ligand according to any one of claims 82 to 98, wherein one specificity thereof is for an agent effective to increase the half life of the ligand.
100. A kit comprising a closed conformation multi-specific ligand according to any one of claims 82 to 99.
101. Nucleic acid encoding at least a closed conformation multispecific ligand according to any one of claims 82 to 99.
102. A vector comprising nucleic acid according to claim 101.
103. A vector according to claim 102, further comprising components necessary for the expression of a closed conformation multispecific ligand.
104. A host cell transfected with a vector according to claim 103.
105. A method for detecting the presence of a target molecule, comprising:
(a) providing a closed conformation multispecific ligand bound to an agent, said ligand being specific for the target molecule and the agent, wherein the agent which is bound by the ligand leads to the generation of a detectable signal on displacement from the ligand;
(b) exposing the closed conformation multispecific ligand to the target molecule; and
(c) detecting the signal generated as a result of the displacement of the agent.
106. A method according to claim 105, wherein the agent is an enzyme, which is inactive when bound by the closed conformation multispecific ligand.
107. A method according to claim 105, wherein the agent is the substrate for an enzyme
108. A method according to claim 107, wherein the agent is a fluorescent, luminescent or chromogenic molecule which is inactive or quenched when bound by the ligand.
109. A kit for performing a method according to any one of claims 105-108, comprising a closed conformation multispecific ligand capable of binding to a target molecule, and optionally an agent and buffers suitable therefor.
110. A homogenous immunoassay incorporating a method according to any one of claims 105-108.
111. A ligand according to any one of claims 1 to 56 for use in therapy.
112. A pharmaceutical composition comprising a ligand according to any one of claims 1 to 56, and a pharmaceutically acceptable eccipient, carrier or diluent.
113. A method for preparing a chelating multimeric ligand comprising the steps of:
(a) providing a vector comprising a nucleic acid sequence encoding a single binding domain specific for a first epitope on a target;
(b) providing a vector encoding a repertoire comprising second binding domains specific for a second epitope on said target, which epitope can be the same or different to the first epitope, said second epitope being adjacent to said first epitope; and
(c) expressing said first and second binding domains; and
(d) isolating those combinations of first and second binding domains which combine together to produce a target-binding dimer.
114. A method according to claim 113, wherein the first and second binding domains are associated covalently through a linker.
115. A method according to claim 113, wherein the first and second binding domains are associated non-covalently.
116. A method according to claim 113, wherein the first and second binding domains are associated through natural association of the domains.
117. A method according to claim 116, wherein the binding domains comprise a VH domain and a Vκ domain.
PCT/GB2003/002804 2001-06-28 2003-06-30 Immunoglobin single variant antigen-binding domains and dual-specific constructs WO2004003019A2 (en)

Priority Applications (55)

Application Number Priority Date Filing Date Title
JP2004516979A JP2006512895A (en) 2002-06-28 2003-06-30 Ligand
AU2003244817A AU2003244817B2 (en) 2002-06-28 2003-06-30 Antigen-binding immunoglobulin single variable domains and dual-specific constructs
DE60305919T DE60305919T2 (en) 2002-06-28 2003-06-30 DUAL SPECIFIC LIGANDS WITH INCREASED HALF TIME
SI200330414T SI1517921T1 (en) 2002-06-28 2003-06-30 Dual specific ligands with increased serum half-life
EP03738294A EP1517921B1 (en) 2002-06-28 2003-06-30 Dual specific ligands with increased serum half-life
CA002492092A CA2492092A1 (en) 2002-06-28 2003-06-30 Immunoglobin single variant antigen-binding domains and dual-specific constructs
EP03782689A EP1578801A2 (en) 2002-12-27 2003-12-24 Dual specific single domain antibodies specific for a ligand and for the receptor of the ligand
JP2005509717A JP2006523090A (en) 2002-12-27 2003-12-24 Bispecific single domain antibody specific for ligand and for ligand receptor
CA002511910A CA2511910A1 (en) 2002-12-27 2003-12-24 Dual specific single domain antibodies specific for a ligand and for the receptor of the ligand
AU2003290330A AU2003290330A1 (en) 2002-12-27 2003-12-24 Dual specific single domain antibodies specific for a ligand and for the receptor of the ligand
PCT/GB2003/005646 WO2004058821A2 (en) 2002-12-27 2003-12-24 Dual specific single domain antibodies specific for a ligand and for the receptor of the ligand
ES04737302T ES2315664T3 (en) 2003-06-30 2004-06-30 SINGLE-DOMAIN ANTIBODIES (DAB) PEGILATED.
PT04737302T PT1639011E (en) 2003-06-30 2004-06-30 Pegylated single domain antibodies (dab)
PCT/GB2004/002829 WO2004081026A2 (en) 2003-06-30 2004-06-30 Polypeptides
JP2006518331A JP5087274B2 (en) 2003-06-30 2004-06-30 Polypeptide
AT04737302T ATE414106T1 (en) 2003-06-30 2004-06-30 PEGYLATED SINGLE DOMAIN ANTIBODIES (DAB)
PL04737302T PL1639011T3 (en) 2003-06-30 2004-06-30 Pegylated single domain antibodies (dAb)
CN2004800249396A CN1845938B (en) 2003-06-30 2004-06-30 Polypeptides
EP10177693A EP2267028A3 (en) 2003-06-30 2004-06-30 Dual specific single domain antibodies (dAb) conjugated to PEG
CA002529819A CA2529819A1 (en) 2003-06-30 2004-06-30 Pegylated single domain antibodies
AU2004220325A AU2004220325B2 (en) 2003-06-30 2004-06-30 Polypeptides
DK04737302T DK1639011T3 (en) 2003-06-30 2004-06-30 Pegylated Single-Domain Antibodies (dAb)
DE602004017726T DE602004017726D1 (en) 2003-06-30 2004-06-30 Pegylated single-domain antibodies (dAb)
SI200431021T SI1639011T1 (en) 2003-06-30 2004-06-30 Pegylated single domain antibodies (dAb)
EP08166692A EP2221317A3 (en) 2003-06-30 2004-06-30 PEGylated single domain antibodies (dAb)
EP04737302A EP1639011B1 (en) 2003-06-30 2004-06-30 Pegylated single domain antibodies (dAb)
US10/925,366 US20050271663A1 (en) 2001-06-28 2004-08-24 Compositions and methods for treating inflammatory disorders
US10/985,847 US20060002935A1 (en) 2002-06-28 2004-11-10 Tumor Necrosis Factor Receptor 1 antagonists and methods of use therefor
US11/023,959 US20060106203A1 (en) 2002-06-28 2004-12-28 Ligand
HK05103582A HK1070081A1 (en) 2002-06-28 2005-01-01 Dual specific ligands with increased serum half-life
GBGB0501752.0A GB0501752D0 (en) 2002-06-28 2005-01-27 Immunoglobin single variant antigen-binding domains and dual-specific constructs
US11/098,758 US20060073141A1 (en) 2001-06-28 2005-04-04 Compositions and methods for treating inflammatory disorders
US11/141,661 US20060257406A1 (en) 2002-12-27 2005-05-31 Ligand
US11/217,919 US20060063921A1 (en) 2002-06-28 2005-09-01 Ligand
US11/664,542 US20080008713A1 (en) 2002-06-28 2005-10-07 Single domain antibodies against tnfr1 and methods of use therefor
US11/667,393 US20070298041A1 (en) 2002-06-28 2005-11-10 Ligands That Enhance Endogenous Compounds
US11/315,848 US20060286066A1 (en) 2003-06-30 2005-12-22 Pegylated immunoglobulin variable region polypeptides
US11/338,863 US20070104710A1 (en) 2002-06-28 2006-01-24 Ligand that has binding specificity for IL-4 and/or IL-13
US11/501,522 US20070093651A1 (en) 2001-06-28 2006-08-08 Ligand
US11/501,546 US20100234570A1 (en) 2001-06-28 2006-08-08 Ligand
CY20061101247T CY1105191T1 (en) 2002-06-28 2006-09-04 DUAL SPECIFICATION CONNECTORS WITH INCREASED HALF-LIFE
ZA200610474A ZA200610474B (en) 2002-12-27 2006-12-13 Compositions and methods for treating inflammatory disorders
US11/704,832 US9321832B2 (en) 2002-06-28 2007-02-08 Ligand
IL183027A IL183027A0 (en) 2002-06-28 2007-05-07 Ligands that enhance endogenous compounds
US11/981,821 US20100081792A1 (en) 2001-06-28 2007-10-31 Ligand
US12/006,933 US20080241166A1 (en) 2002-06-28 2008-01-07 Ligands that bind a receptor
US12/150,567 US20080233130A1 (en) 2002-06-28 2008-04-29 Ligand that has binding specificity for IL-4 and/or IL-13
US12/150,487 US20080233129A1 (en) 2002-06-28 2008-04-29 Ligand that has binding specificity for IL-4 and/or lL-13
CY20091100035T CY1108691T1 (en) 2003-06-30 2009-01-14 Polypeptides
US12/409,617 US20090258012A1 (en) 2001-06-28 2009-03-24 Compositions and methods for treating inflammatory disorders
AU2011200930A AU2011200930B2 (en) 2003-06-30 2011-03-03 Polypeptides
US13/181,834 US9028822B2 (en) 2002-06-28 2011-07-13 Antagonists against TNFR1 and methods of use therefor
US13/733,675 US20130216538A1 (en) 2002-12-27 2013-01-03 Compositions and Methods for Treating Inflammatory Disorders
US14/173,204 US20150087813A1 (en) 2001-06-28 2014-02-05 Ligand
US14/924,971 US20170145080A1 (en) 2002-12-27 2015-10-28 Ligand

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
GBPCT/GB02/03014 2002-06-28
PCT/GB2002/003014 WO2003002609A2 (en) 2001-06-28 2002-06-28 Dual-specific ligand and its use
GB0230202.4 2002-12-27
GB0230202A GB0230202D0 (en) 2002-12-27 2002-12-27 Ligand

Related Parent Applications (5)

Application Number Title Priority Date Filing Date
PCT/GB2002/003014 Continuation-In-Part WO2003002609A2 (en) 2001-06-28 2002-06-28 Dual-specific ligand and its use
US10/744,774 Continuation-In-Part US20040219643A1 (en) 2001-06-28 2003-12-23 Dual-specific ligand
PCT/GB2004/002829 Continuation-In-Part WO2004081026A2 (en) 2001-06-28 2004-06-30 Polypeptides
US11/023,959 Continuation-In-Part US20060106203A1 (en) 2001-06-28 2004-12-28 Ligand
US11/141,661 Continuation-In-Part US20060257406A1 (en) 2001-06-28 2005-05-31 Ligand

Related Child Applications (5)

Application Number Title Priority Date Filing Date
US10/744,774 Continuation-In-Part US20040219643A1 (en) 2001-06-28 2003-12-23 Dual-specific ligand
PCT/GB2003/005646 Continuation-In-Part WO2004058821A2 (en) 2001-06-28 2003-12-24 Dual specific single domain antibodies specific for a ligand and for the receptor of the ligand
US10/925,366 Continuation-In-Part US20050271663A1 (en) 2001-06-28 2004-08-24 Compositions and methods for treating inflammatory disorders
US11/023,959 Continuation US20060106203A1 (en) 2001-06-28 2004-12-28 Ligand
US11/098,758 Continuation-In-Part US20060073141A1 (en) 2001-06-28 2005-04-04 Compositions and methods for treating inflammatory disorders

Publications (3)

Publication Number Publication Date
WO2004003019A2 true WO2004003019A2 (en) 2004-01-08
WO2004003019A3 WO2004003019A3 (en) 2004-09-10
WO2004003019A9 WO2004003019A9 (en) 2006-04-27

Family

ID=30001956

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB2003/002804 WO2004003019A2 (en) 2001-06-28 2003-06-30 Immunoglobin single variant antigen-binding domains and dual-specific constructs

Country Status (16)

Country Link
US (5) US20060106203A1 (en)
EP (4) EP1517921B1 (en)
JP (2) JP2006512895A (en)
CN (1) CN1678634A (en)
AT (1) ATE328906T1 (en)
AU (1) AU2003244817B2 (en)
CA (1) CA2492092A1 (en)
CY (1) CY1105191T1 (en)
DE (1) DE60305919T2 (en)
DK (1) DK1517921T3 (en)
ES (1) ES2263984T3 (en)
GB (1) GB0501752D0 (en)
HK (1) HK1070081A1 (en)
PT (1) PT1517921E (en)
SI (1) SI1517921T1 (en)
WO (1) WO2004003019A2 (en)

Cited By (443)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004081026A3 (en) * 2003-06-30 2005-01-27 Domantis Ltd Polypeptides
EP1517921A2 (en) 2002-06-28 2005-03-30 Domantis Limited Immunoglobulin single variable antigen-binding domains and dual-specific constructs thereof
EP1558650A2 (en) * 2002-11-08 2005-08-03 Ablynx N.V. Camelidae antibodies against immunoglobulin e and use thereof for the treatment of allergic disorders
WO2005118642A2 (en) * 2004-06-01 2005-12-15 Domantis Limited Bispecific fusion antibodies with enhanced serum half-life
WO2006030220A1 (en) * 2004-09-17 2006-03-23 Domantis Limited Compositions monovalent for cd40l binding and methods of use
WO2006038027A2 (en) 2004-10-08 2006-04-13 Domantis Limited SINGLE DOMAIN ANTIBODIES AGAINST TNFRl AND METHODS OF USE THEREFOR
WO2006003388A3 (en) * 2004-06-30 2006-04-27 Domantis Ltd Compositions and methods for treating inflammatory disorders
WO2006051288A2 (en) * 2004-11-10 2006-05-18 Domantis Limited Ligands that enhance endogenous compounds
WO2006059110A2 (en) * 2004-12-02 2006-06-08 Domantis Limited Plad domain peptides with increased serum half life due to conjugation to domain antibodies
WO2006059106A2 (en) * 2004-12-02 2006-06-08 Domantis Limited Bispecific domain antibodies targeting serum albumin and glp-1 or pyy
WO2006059108A2 (en) * 2004-12-02 2006-06-08 Domantis Limited ANTI-IL-IRl SINGLE DOMAIN ANTIBODIES AND THERAPEUTIC USES
WO2006089133A2 (en) 2005-02-15 2006-08-24 Duke University Anti-cd19 antibodies and uses in oncology
WO2006097689A2 (en) * 2005-03-18 2006-09-21 Domantis Limited Antibodies against candida antigens
WO2006122787A1 (en) * 2005-05-18 2006-11-23 Ablynx Nv Serum albumin binding proteins
WO2006129843A2 (en) * 2005-05-31 2006-12-07 Canon Kabushiki Kaisha Bispecific capturing molecule
JP2007008925A (en) * 2005-05-31 2007-01-18 Canon Inc Target substance-catching molecule
WO2007049017A2 (en) * 2005-10-24 2007-05-03 Domantis Limited Agents that bind a target in pulmonary tissue for treating respiratory diseases
EP1806365A1 (en) 2006-01-05 2007-07-11 Boehringer Ingelheim International GmbH Antibody molecules specific for fibroblast activation protein and immunoconjugates containing them
WO2007077001A2 (en) 2005-12-30 2007-07-12 Universidad Publica De Navarra Method and system of control of the converter of an electricity generation facility connected to an electricity network in the presence of voltage sags in said network
WO2007085815A2 (en) * 2006-01-24 2007-08-02 Domantis Limited Ligands that bind il-4 and/or il-13
WO2007063308A3 (en) * 2005-12-01 2007-09-20 Domantis Ltd Noncompetitive domain antibody formats that bind interleukin 1 receptor type 1
WO2007063311A3 (en) * 2005-12-01 2007-09-20 Domantis Ltd Competitive domain antibody formats that bind interleukin 1 receptor type 1
WO2007141280A2 (en) 2006-06-06 2007-12-13 Oxford Genome Sciences (Uk) Ltd Proteins
JP2008504356A (en) * 2004-06-30 2008-02-14 ドマンティス リミテッド Compositions and methods for treating inflammatory diseases
WO2008030611A2 (en) 2006-09-05 2008-03-13 Medarex, Inc. Antibodies to bone morphogenic proteins and receptors therefor and methods for their use
WO2008043821A1 (en) * 2006-10-11 2008-04-17 Ablynx N. V. Amino acid sequences that bind to serum proteins in a manner that is essentially independent of the ph, compounds comprising the same, and use thereof
WO2008054603A2 (en) 2006-10-02 2008-05-08 Amgen Inc. Il-17 receptor a antigen binding proteins
WO2008070569A2 (en) 2006-12-01 2008-06-12 Medarex, Inc. Human antibodies that bind cd22 and uses thereof
WO2008074004A2 (en) 2006-12-14 2008-06-19 Medarex, Inc. Human antibodies that bind cd70 and uses thereof
EP1945669A1 (en) * 2005-08-15 2008-07-23 Arana Therapeutics Limited Chimeric antibodies with new world primate regions
WO2008096158A3 (en) * 2007-02-08 2008-12-18 Domantis Ltd Antibody single variable domains against serum albumin
WO2008153926A2 (en) 2007-06-05 2008-12-18 Yale University Inhibitors of receptor tyrosine kinases and methods of use thereof
WO2009040562A1 (en) * 2007-09-26 2009-04-02 Ucb Pharma S.A. Dual specificity antibody fusions
US7521425B2 (en) 2005-03-03 2009-04-21 Covx Technologies Ireland Limited Anti-angiogenic compounds
WO2009054863A2 (en) 2006-12-13 2009-04-30 Medarex, Inc. Human antibodies that bind cd19 and uses thereof
JP2009518024A (en) * 2005-12-06 2009-05-07 ドマンティス リミテッド Ligands having binding specificity for EGFR and / or VEGF and methods of use thereof
JP2009518025A (en) * 2005-12-06 2009-05-07 ドマンティス リミテッド Bispecific ligands having binding specificity for cell surface targets and methods of use thereof
JP2009523162A (en) * 2006-01-11 2009-06-18 ドマンティス リミテッド Ligands having binding specificity for VEGF and / or EGFR and methods of use thereof
WO2009155420A1 (en) * 2008-06-18 2009-12-23 California Institute Of Technology Multi-ligand capture agents and related compositions, methods and systems
US20090324579A1 (en) * 2005-09-01 2009-12-31 Siyaram Pandey Anti-apoptotic protein antibodies
US20090324491A1 (en) * 2006-10-12 2009-12-31 Forerunner Pharma Research Co., Ltd. Diagnosis and Treatment of Cancer Using Anti-EREG Antibody
WO2010011944A2 (en) 2008-07-25 2010-01-28 Wagner Richard W Protein screeing methods
US20100061988A1 (en) * 2008-09-04 2010-03-11 Hansen Genevieve Monoclonal antibodies
US20100093977A1 (en) * 2006-09-29 2010-04-15 Ucb Pharma S.A. Altered Antibodies
US20100104573A1 (en) * 2007-03-22 2010-04-29 Ucb Parma S.A. Binding proteins, including antibodies, antibody derivatives and antibody fragments, that specifically bind cd154 and uses thereof
US20100104510A1 (en) * 2007-04-02 2010-04-29 The United States Of America, As Represented By The Secretary, Department Of Health And Human Selenocysteine mediated hybrid antibody molecules
WO2010056804A1 (en) 2008-11-12 2010-05-20 Medimmune, Llc Antibody formulation
US20100136627A1 (en) * 2007-05-04 2010-06-03 Technophage,Investigacao E Desenvolvimento Em Biotechnologia, Sa Engineered Rabbit Antibody Variable Domains and Uses Thereof
US7763244B2 (en) 2002-07-01 2010-07-27 Human Genome Sciences, Inc. Antibodies that specifically bind to Reg IV
WO2010084408A2 (en) 2009-01-21 2010-07-29 Oxford Biotherapeutics Ltd. Pta089 protein
WO2010085590A1 (en) 2009-01-23 2010-07-29 Biosynexus Incorporated Opsonic and protective antibodies specific for lipoteichoic acid gram positive bacteria
US20100197897A1 (en) * 2004-07-22 2010-08-05 Franklin Gerardus Grosveld Binding molecules
US7771724B2 (en) 2002-08-07 2010-08-10 Ablynx N.V. Modulation of platelet adhesion based on the surface-exposed beta-switch loop of platelet glycoprotein IB-alpha
EP2215125A1 (en) * 2007-11-27 2010-08-11 Ablynx N.V. Method for obtaining polypeptide constructs comprising two or more single domain antibodies
EP2221316A1 (en) 2005-05-05 2010-08-25 Duke University Anti-CD19 antibody therapy for autoimmune disease
WO2010094722A2 (en) 2009-02-19 2010-08-26 Glaxo Group Limited Improved anti-serum albumin binding variants
WO2010094720A2 (en) 2009-02-19 2010-08-26 Glaxo Group Limited Improved anti-tnfr1 polypeptides, antibody variable domains & antagonists
JP2010528646A (en) * 2007-06-06 2010-08-26 ドマンティス リミテッド Polypeptides, antibody variable domains, and antagonists
WO2010097385A1 (en) 2009-02-24 2010-09-02 Glaxo Group Limited Antigen-binding constructs
JP2010529841A (en) * 2007-06-06 2010-09-02 ドマンティス リミテッド Polypeptides, antibody variable domains and antagonists
WO2010102175A1 (en) 2009-03-05 2010-09-10 Medarex, Inc. Fully human antibodies specific to cadm1
WO2010100135A1 (en) 2009-03-05 2010-09-10 Ablynx N.V. Novel antigen binding dimer-complexes, methods of making/avoiding and uses thereof
US7807162B2 (en) 2005-05-20 2010-10-05 Ablynx N.V. Single domain VHH antibodies against von Willebrand factor
US20100255010A1 (en) * 2009-03-20 2010-10-07 Genentech, Inc. Anti-her antibodies
WO2010122090A1 (en) 2009-04-24 2010-10-28 Glaxo Group Limited Fgfr1c antibody combinations
US20100278842A1 (en) * 2005-01-31 2010-11-04 Genentech, Inc. Anti-ephb2 antibodies and methods using same
US7829084B2 (en) * 2001-01-17 2010-11-09 Trubion Pharmaceuticals, Inc. Binding constructs and methods for use thereof
US20100297153A1 (en) * 2004-10-12 2010-11-25 Crucell Holland B.V. Binding molecules for treatment and detection of cancer
US7846439B2 (en) 2006-02-01 2010-12-07 Cephalon Australia Pty Ltd Domain antibody construct
US20100330080A1 (en) * 2008-07-02 2010-12-30 Torsten Dreier Antigen binding polypeptides
US7867493B2 (en) * 2006-08-18 2011-01-11 Novartis Ag PRLR-specific antibody and uses thereof
WO2011006915A2 (en) 2009-07-16 2011-01-20 Glaxo Group Limited Improved anti-serum albumin binding single variable domains
WO2011006914A2 (en) 2009-07-16 2011-01-20 Glaxo Group Limited Antagonists, uses & methods for partially inhibiting tnfr1
US7875465B2 (en) 2005-05-31 2011-01-25 Canon Kabushiki Kaisha Target substance capturing molecule
WO2011012609A2 (en) 2009-07-29 2011-02-03 Glaxo Group Limited Ligands that bind tgf-beta receptor rii
DE112009000507T5 (en) 2008-03-05 2011-02-10 Ablynx Nv Novel antigen-binding dimer complexes, process for their preparation and their use
US7892543B2 (en) * 1996-10-04 2011-02-22 Chugai Seiyako Kabushiki Kaisha Reshaped human anti-HM 1.24 antibody
WO2010139808A3 (en) * 2009-06-05 2011-02-24 Ablynx Nv Monovalent, bivalent and trivalent anti human respiratory syncytial virus (hrsv) nanobody constructs for the prevention and/or treatment of respiratory tract infections
US20110059469A1 (en) * 2008-01-11 2011-03-10 Hiroyuki Aburatani Anti-cldn6 antibody
US7928214B2 (en) * 2005-04-26 2011-04-19 Agouron Pharmaceuticals, Inc. P-cadherin antibodies
WO2011047083A1 (en) 2009-10-13 2011-04-21 Oxford Biotherapeutics Ltd. Antibodies against epha10
WO2011046958A1 (en) 2009-10-12 2011-04-21 Amgen Inc. Use of il-17 receptor a antigen binding proteins
US20110104256A1 (en) * 2008-03-25 2011-05-05 Yaolin Wang Methods for treating or preventing colorectal cancer
WO2011051217A1 (en) 2009-10-27 2011-05-05 Glaxo Group Limited Stable anti-tnfr1 polypeptides, antibody variable domains & antagonists
US20110104163A1 (en) * 2008-01-07 2011-05-05 The United States Of America, As Represented By Th Anti-hiv domain antibodies and method of making and using same
WO2011054007A1 (en) 2009-11-02 2011-05-05 Oxford Biotherapeutics Ltd. Ror1 as therapeutic and diagnostic target
US7939277B2 (en) 2005-01-14 2011-05-10 Umc Utrecht Holding Bv Methods and assays for distinguishing between different forms of diseases and disorders characterized by thrombocytopenia and/or by spontaneous interaction between Von Willebrand Factor (vWF) and platelets
WO2011054893A2 (en) 2009-11-05 2011-05-12 Novartis Ag Biomarkers predictive of progression of fibrosis
EP2322554A1 (en) * 2004-06-30 2011-05-18 Domantis Limited Composition comprising an anti-TNF-alpha domain antibody for the treatment of rheumatoid arthritis
WO2011064382A1 (en) 2009-11-30 2011-06-03 Ablynx N.V. Improved amino acid sequences directed against human respiratory syncytial virus (hrsv) and polypeptides comprising the same for the prevention and/or treatment of respiratory tract infections
WO2011073180A1 (en) 2009-12-14 2011-06-23 Ablynx N.V. Single variable domain antibodies against ox40l, constructs and therapeutic use
US20110150905A1 (en) * 2006-12-14 2011-06-23 Regeneron Pharmaceuticals, Inc. Human antibodies to human delta like ligand 4
US20110158902A1 (en) * 2005-04-15 2011-06-30 Neogenix Oncology, Inc. Recombinant monoclonal antibodies and corresponding antigens for colon and pancreatic cancers
US20110158992A1 (en) * 2008-08-27 2011-06-30 Schering Corporation Engineered Anti-IL-23R Antibodies
WO2011083140A1 (en) 2010-01-08 2011-07-14 Ablynx Nv Immunoglobulin single variable domain directed against human cxcr4
WO2011089183A2 (en) 2010-01-20 2011-07-28 Boehringer Ingelheim International Gmbh Anticoagulant antidotes
WO2011095545A1 (en) 2010-02-05 2011-08-11 Ablynx Nv Peptides capable of binding to serum albumin and compounds, constructs and polypeptides comprising the same
WO2011098449A1 (en) 2010-02-10 2011-08-18 Novartis Ag Methods and compounds for muscle growth
WO2011098518A2 (en) 2010-02-11 2011-08-18 Ablynx Nv Delivery of immunoglobulin variable domains and constructs thereof
WO2011098520A1 (en) 2010-02-10 2011-08-18 Novartis Ag Agonist dr5 binding polypeptides
WO2011100403A1 (en) 2010-02-10 2011-08-18 Immunogen, Inc Cd20 antibodies and uses thereof
US20110207917A1 (en) * 2009-02-09 2011-08-25 Patrys Limited Sam-6 variants, target and methods of use
US8007794B2 (en) * 2005-11-14 2011-08-30 Rinat Neuroscience Corporation Antagonist antibodies directed against calcitonin gene-related peptide and methods using same
US8017118B2 (en) * 2008-03-17 2011-09-13 LivTech Inc. — Teikyo University Biotechnology Research Center Anti-hDlk-1 antibody having an antitumor activity in vivo
US8021661B2 (en) * 2005-01-05 2011-09-20 Biogen Idec Ma Inc. Cripto binding molecules
EP2366715A2 (en) 2005-11-14 2011-09-21 Amgen Inc. Rankl Antibody-PTH/PTHRP Chimeric Molecules
EP2368908A2 (en) 2006-11-10 2011-09-28 CovX Technologies Ireland Limited Anti- angiogenic compounds
US20110237779A1 (en) * 2004-02-19 2011-09-29 Genentech, Inc. Cdr-repaired antibodies
US8029790B2 (en) * 2004-06-29 2011-10-04 B.R.A.H.M.S. Gmbh Monoclonal thyroid stimulating or blocking antibodies, peptide sequences corresponding to their variable regions, and their uses in diagnostic, preventive and therapeutic medicine
WO2011131659A2 (en) 2010-04-21 2011-10-27 Glaxo Group Limited Binding domains
WO2011138391A1 (en) 2010-05-06 2011-11-10 Novartis Ag Compositions and methods of use for therapeutic low density lipoprotein - related protein 6 (lrp6) multivalent antibodies
US20110275787A1 (en) * 2008-10-01 2011-11-10 Microment AG Cross-species-specific single domain bispecific single chain antibody
WO2011144751A1 (en) 2010-05-20 2011-11-24 Glaxo Group Limited Improved anti-serum albumin binding variants
WO2011144749A1 (en) 2010-05-20 2011-11-24 Ablynx Nv Biological materials related to her3
WO2011161263A1 (en) 2010-06-25 2011-12-29 Ablynx Nv Pharmaceutical compositions for cutaneous administration
US8088384B2 (en) * 2002-12-24 2012-01-03 Rinat Neuroscience Corp. Anti-NGF antibodies and methods using same
US8088896B2 (en) * 2005-10-12 2012-01-03 Morphosys Ag Generation and profiling of fully human gold-derived therapeutic antibodies specific for human CD38
US8097251B2 (en) 2001-10-24 2012-01-17 Vlaams Interuniversitair Instituut Voor Biotechnologie Vzw Functional heavy chain antibodies, fragments thereof, library thereof and methods of production thereof
US20120020957A1 (en) * 2008-10-13 2012-01-26 Institute For Research In Biomedicine Dengue virus neutralizing antibodies and use thereof
US8105592B2 (en) * 2004-11-25 2012-01-31 Vhsquared Limited Heavy chain and single domain antibodies
US8106161B2 (en) 2001-01-17 2012-01-31 Emergent Product Development Seattle, Llc Binding domain-immunoglobulin fusion proteins
US8106162B2 (en) * 1997-06-12 2012-01-31 Novartis Ag Artificial antibody polypeptides
US20120034212A1 (en) * 2009-01-29 2012-02-09 Michael Bowen Human Anti-IL-6 Antibodies With Extended In Vivo Half-Life And Their Use In Treatment Of Oncology, Autoimmune Diseases And Inflammatory Diseases
WO2012020096A1 (en) 2010-08-13 2012-02-16 Medimmune Limited Monomeric polypeptides comprising variant fc regions and methods of use
WO2012020143A1 (en) 2010-08-13 2012-02-16 Glaxo Group Limited Improved anti-serum albumin binding variants
WO2012022814A1 (en) 2010-08-20 2012-02-23 Novartis Ag Antibodies for epidermal growth factor receptor 3 (her3)
WO2012022734A2 (en) 2010-08-16 2012-02-23 Medimmune Limited Anti-icam-1 antibodies and methods of use
WO2012022703A2 (en) 2010-08-20 2012-02-23 Glaxo Group Limited Improved anti-serum albumin binding variants
US8133488B2 (en) * 2008-01-18 2012-03-13 Genentech, Inc. Methods and compositions for targeting polyubiquitin
WO2012032433A1 (en) 2010-09-09 2012-03-15 Pfizer Inc. 4-1bb binding molecules
US8147835B2 (en) 2001-01-17 2012-04-03 Emergent Product Development Seattle, Llc Binding domain-immunoglobulin fusion proteins
US8158124B2 (en) * 2007-05-30 2012-04-17 Lpath, Inc. Compositions and methods for binding lysophosphatidic acid
EP2441775A1 (en) 2007-02-26 2012-04-18 Oxford Biotherapeutics Ltd. Protein
US8163552B2 (en) * 2003-11-14 2012-04-24 Patrys Limited Adenocarcinoma specific antibody SAM-6, and uses thereof
US8168183B2 (en) * 2008-04-28 2012-05-01 Kalobios Pharmaceuticals, Inc. Antibodies to granulocyte-macrophage colony-stimulating factor
EP2447719A1 (en) 2007-02-26 2012-05-02 Oxford Biotherapeutics Ltd. Proteins
WO2012062713A1 (en) 2010-11-08 2012-05-18 Novartis Ag Cxcr2 binding polypeptides
WO2012069654A1 (en) 2010-11-26 2012-05-31 Molecular Partners Ag Designed repeat proteins binding to serum albumin
US8193321B2 (en) 2008-09-03 2012-06-05 Genentech, Inc. Multispecific antibodies
WO2012072731A2 (en) 2010-12-01 2012-06-07 Glaxo Group Limited Improved anti-serum albumin binding single variable domains
US8206709B2 (en) * 2006-06-30 2012-06-26 Novo Nordisk A/S Anti-NKG2A antibodies and uses thereof
US20120164068A1 (en) * 2009-07-03 2012-06-28 Peter John Hudson Immuno-conjugates and methods for producing them
US20120171115A1 (en) * 2009-12-23 2012-07-05 Avipep Pty Limited Immuno-conjugates and methods for producing them
US8217140B2 (en) 2008-04-17 2012-07-10 Ablynx N.V. Peptides capable of binding to serum proteins and compounds, constructs and polypeptides comprising the same
WO2012093125A1 (en) 2011-01-06 2012-07-12 Glaxo Group Limited Ligands that bind tgf-beta receptor ii
US8221754B2 (en) * 2006-07-12 2012-07-17 Gene Techno Science Co., Ltd. Antihuman α9 integrin antibody and use of the same
US8227578B2 (en) * 2006-11-10 2012-07-24 Abe, Ikubo & Katayama Anti-human dlk-1 antibody showing anti-tumor activity in vivo
US8236931B2 (en) 2006-10-30 2012-08-07 Glaxo Group Limited Prevention of aggregation of immunoglobulin light or heavy chains
US20120202977A1 (en) * 2002-11-08 2012-08-09 Ablynx N.V. Single domain antibodies directed against tumor necrosis factor-alpha and uses therefor
WO2012104227A1 (en) 2011-02-02 2012-08-09 Glaxo Group Limited Novel antigen binding proteins
EP2486941A1 (en) 2006-10-02 2012-08-15 Medarex, Inc. Human antibodies that bind CXCR4 and uses thereof
WO2012109624A2 (en) 2011-02-11 2012-08-16 Zyngenia, Inc. Monovalent and multivalent multispecific complexes and uses thereof
US8263746B2 (en) 2004-02-06 2012-09-11 Morphosys Ag Anti-CD38 human antibodies and uses thereof
WO2012130834A1 (en) 2011-03-30 2012-10-04 Boehringer Ingelheim International Gmbh Anticoagulant antidotes
US8283294B2 (en) * 2007-03-01 2012-10-09 Symphogen A/S Method for cloning cognate antibodies
US8293233B2 (en) * 2005-03-25 2012-10-23 National Research Council Of Canada Method for isolation of soluble polypeptides
EP2514767A1 (en) 2006-12-19 2012-10-24 Ablynx N.V. Amino acid sequences directed against a metalloproteinase from the ADAM family and polypeptides comprising the same for the treatment of ADAM-related diseases and disorders
WO2012156219A1 (en) 2011-05-05 2012-11-22 Ablynx Nv Amino acid sequences directed against il-17a, il-17f and/or il17-a/f and polypeptides comprising the same
US8318159B2 (en) 2008-12-12 2012-11-27 Boehringer Ingelheim International Gmbh Anti-IGF antibodies
US8323645B2 (en) * 2005-03-24 2012-12-04 Millennium Pharmaceuticals, Inc. Antibodies that bind OV064 and methods of use therefor
WO2012166906A1 (en) 2011-05-31 2012-12-06 Massachusetts Institute Of Technology Cell-directed synthesis of multifunctional nanopatterns and nanomaterials
WO2012163887A1 (en) 2011-05-27 2012-12-06 Ablynx Nv Inhibition of bone resorption with rankl binding peptides
US8333966B2 (en) 2008-04-11 2012-12-18 Emergent Product Development Seattle, Llc CD37 immunotherapeutics and uses thereof
WO2012175740A1 (en) 2011-06-23 2012-12-27 Ablynx Nv Immunoglobulin single variable domains directed against ige
EP2540741A1 (en) 2006-03-06 2013-01-02 Aeres Biomedical Limited Humanized anti-CD22 antibodies and their use in treatment of oncology, transplantation and autoimmune disease
WO2013003625A2 (en) 2011-06-28 2013-01-03 Oxford Biotherapeutics Ltd. Antibodies
WO2013001369A2 (en) 2011-06-28 2013-01-03 Oxford Biotherapeutics Ltd. Therapeutic and diagnostic target
WO2013016220A1 (en) 2011-07-22 2013-01-31 Amgen Inc. Il-17 receptor a is required for il-17c biology
EP2557090A2 (en) 2006-12-19 2013-02-13 Ablynx N.V. Amino acid sequences directed against GPCRs and polypeptides comprising the same for the treatment of GPCR-related diseases and disorders
US8388972B2 (en) * 2006-09-15 2013-03-05 Advanced Accelerator Applications S.A. Human anti-folate receptor alpha antibodies and antibody fragments for the radioimmunotherapy of ovarian carcinoma
WO2013038191A2 (en) 2011-09-16 2013-03-21 Bioceros B.V. Anti-cd134 (ox40) antibodies and uses thereof
WO2013043933A2 (en) 2011-09-22 2013-03-28 Amgen Inc. Cd27l antigen binding proteins
US8409577B2 (en) 2006-06-12 2013-04-02 Emergent Product Development Seattle, Llc Single chain multivalent binding proteins with effector function
WO2013060872A1 (en) 2011-10-27 2013-05-02 Boehringer Ingelheim International Gmbh Anticancer combination therapy
WO2013067355A1 (en) 2011-11-04 2013-05-10 Novartis Ag Low density lipoprotein-related protein 6 (lrp6) - half life extender constructs
WO2013068571A1 (en) 2011-11-11 2013-05-16 Ucb Pharma S.A. Albumin binding antibodies and binding fragments thereof
EP2594590A1 (en) 2007-12-14 2013-05-22 Bristol-Myers Squibb Company Binding molecules to the human OX40 receptor
WO2013084147A2 (en) 2011-12-05 2013-06-13 Novartis Ag Antibodies for epidermal growth factor receptor 3 (her3)
WO2013084148A2 (en) 2011-12-05 2013-06-13 Novartis Ag Antibodies for epidermal growth factor receptor 3 (her3) directed to domain ii of her3
US8486391B2 (en) * 1998-07-13 2013-07-16 Board Of Regents, University Of Texas System Cancer treatment kits using antibodies to aminophospholipids
US8569460B2 (en) 2009-03-25 2013-10-29 Vet Therapeutics, Inc. Antibody constant domain regions and uses thereof
US8580254B2 (en) 2007-06-19 2013-11-12 Boehringer Ingelheim International Gmbh Anti-IGF antibodies
WO2013168108A2 (en) 2012-05-09 2013-11-14 Novartis Ag Chemokine receptor binding polypeptides
US8603475B2 (en) 2005-12-15 2013-12-10 Genentech, Inc. Methods and compositions for targeting polyubiquitin
US8603473B2 (en) 2003-06-27 2013-12-10 Biogen Idec Ma Inc. Modified binding molecules comprising connecting peptides
US8623592B2 (en) 2008-08-15 2014-01-07 Merrimack Pharmaceuticals, Inc. Methods and systems for predicting response of cells to a therapeutic agent
US8623366B2 (en) 2009-08-28 2014-01-07 Labrys Biologics, Inc. Methods for treating visceral pain by administering antagonist antibodies directed against calcitonin gene-related peptide
US8642035B2 (en) * 2008-01-17 2014-02-04 Irm Llc Anti-TrkB antibodies
WO2014020331A1 (en) 2012-08-01 2014-02-06 Oxford Biotherapeutics Ltd. Therapeutic and diagnostic target
US8647625B2 (en) 2004-07-26 2014-02-11 Biogen Idec Ma Inc. Anti-CD154 antibodies
US8652470B2 (en) 2010-03-04 2014-02-18 Vet Therapeutics, Inc. Monoclonal antibodies directed to CD52
EP2700651A1 (en) 2008-07-18 2014-02-26 Bristol-Myers Squibb Company Compositions monovalent for CD28 binding and methods of use
WO2014029752A1 (en) 2012-08-22 2014-02-27 Glaxo Group Limited Anti lrp6 antibodies
EP2703011A2 (en) 2007-05-07 2014-03-05 MedImmune, LLC Anti-icos antibodies and their use in treatment of oncology, transplantation and autoimmune disease
US20140066599A2 (en) * 2011-03-25 2014-03-06 Glenmark Pharmaceuticals S.A. Hetero-Dimeric Immunoglobulins
US8679490B2 (en) * 2005-11-07 2014-03-25 Genentech, Inc. Binding polypeptides with diversified and consensus VH/VL hypervariable sequences
US8691225B2 (en) * 2007-02-16 2014-04-08 Merrimack Pharmaceuticals, Inc. Antibodies against the ectodomain of ErbB3 and uses thereof
US8703131B2 (en) 2005-05-21 2014-04-22 Ablynx N.V. Nanobodies against tumor necrosis factor-alpha
US8703133B2 (en) 1998-11-18 2014-04-22 Genentech, Inc. Antibody variants
US20140112914A1 (en) * 2010-11-30 2014-04-24 Chugai Seiyaku Kabushiki Kaisha Cytotoxicity-inducing therapeutic agent
US8722859B2 (en) 2000-04-11 2014-05-13 Genentech, Inc. Multivalent antibodies and uses therefor
WO2014089335A2 (en) 2012-12-07 2014-06-12 Amgen Inc. Bcma antigen binding proteins
US8759490B2 (en) 2005-03-24 2014-06-24 Millennium Pharamaceuticals, Inc. Antibodies that bind OV064 and methods of use therefor
WO2014111550A1 (en) 2013-01-17 2014-07-24 Glaxosmithkline Intellectual Property Development Limited Modified anti-serum albumin binding proteins
EP2758434A2 (en) * 2011-09-23 2014-07-30 Technophage, Investigação E Desenvolvimento Em Biotecnologia, SA Anti-tumor necrosis factor-alpha agents and uses thereof
WO2014114801A1 (en) 2013-01-25 2014-07-31 Amgen Inc. Antibodies targeting cdh19 for melanoma
US8796427B2 (en) 2008-01-24 2014-08-05 Novo Nordisk A/S Humanized anti-human NKG2A monoclonal antibody
WO2014120916A1 (en) 2013-02-01 2014-08-07 Bristol-Myers Squibb Company Pegylated domain antibodies monovalent for cd28 binding and methods of use
EP2261672B1 (en) * 2004-07-14 2014-09-03 The Regents of The University of California Biomarkers for early detection of ovarian cancer
WO2014133855A1 (en) 2013-02-28 2014-09-04 Caprion Proteomics Inc. Tuberculosis biomarkers and uses thereof
WO2014148895A1 (en) 2013-03-18 2014-09-25 Biocerox Products B.V. Humanized anti-cd134 (ox40) antibodies and uses thereof
WO2014159239A2 (en) 2013-03-14 2014-10-02 Novartis Ag Antibodies against notch 3
US20140328836A1 (en) * 2008-04-02 2014-11-06 Macrogenics, Inc. HER2/neu-Specific Antibodies and Methods of Using Same
US8895001B2 (en) 2010-03-11 2014-11-25 Merrimack Pharmaceuticals, Inc. Use of ErbB3 inhibitors in the treatment of triple negative and basal-like breast cancers
US20140377781A1 (en) * 2006-12-14 2014-12-25 Chugai Seiyaku Kabushiki Kaisha Anti-Claudin 3 Monoclonal Antibody and Treatment and Diagnosis of Cancer Using the Same
US20150010567A1 (en) * 2012-02-03 2015-01-08 Hoffmann-La Roche Inc. Bispecific antibody molecules with antigen-transfected t-cells and their use in medicine
US20150017169A1 (en) * 2012-02-22 2015-01-15 Ucb Pharma S.A. Sequence Asymmetric Modified IgG4 Bispecific Antibodies
US20150018529A1 (en) * 2012-02-22 2015-01-15 Ucb Pharma S.A. Sequence Symmetric Modified IgG4 Bispecific Antibodies
WO2015015401A2 (en) 2013-08-02 2015-02-05 Pfizer Inc. Anti-cxcr4 antibodies and antibody-drug conjugates
US20150079088A1 (en) * 2013-07-25 2015-03-19 Cytomx Therapeutics, Inc. Multispecific antibodies, multispecific activatable antibodies and methods of using the same
US8992919B2 (en) 2010-04-15 2015-03-31 Genentech, Inc. Anti-polyubiquitin antibodies and methods of use
US8993319B2 (en) 2004-12-28 2015-03-31 Innate Pharma S.A. Monoclonal antibodies against NKG2A
WO2015051010A1 (en) 2013-10-02 2015-04-09 Medimmune, Llc Neutralizing anti-influenza a antibodies and uses thereof
WO2015050959A1 (en) 2013-10-01 2015-04-09 Yale University Anti-kit antibodies and methods of use thereof
US20150104443A1 (en) * 2012-05-08 2015-04-16 Chong Kun Dang Pharmaceutical Corp. Anti-erbb2 antibody variants
US9028816B2 (en) 2003-01-10 2015-05-12 Ablynx N.V. Polypeptides and polypeptide constructs comprising single domain antibodies directed against von Willebrand factor
EP1824884B1 (en) 2004-12-16 2015-05-13 Centre National De La Recherche Scientifique (Cnrs) Production of antibody formats and immunological applications of said formats
US20150133638A1 (en) * 2012-02-10 2015-05-14 Genentech, Inc. Single-chain antibodies and other heteromultimers
US20150152192A1 (en) * 2013-11-29 2015-06-04 Samsung Electronics Co., Ltd. Anti-c-met/anti-ang2 bispecific antibody
US20150166670A1 (en) * 2012-05-24 2015-06-18 Hoffmann-La Roche Inc. Multispecific antibodies
US9062101B2 (en) 2010-08-14 2015-06-23 AbbVie Deutschland GmbH & Co. KG Amyloid-beta binding proteins
US9072798B2 (en) 2009-02-18 2015-07-07 Ludwig Institute For Cancer Research Ltd. Specific binding proteins and uses thereof
US20150203591A1 (en) * 2012-08-02 2015-07-23 Regeneron Pharmaceuticals, Inc. Mutivalent antigen-binding proteins
US20150203586A1 (en) * 2006-08-18 2015-07-23 Armagen Technologies, Inc. Macromolecular compositions that cross the blood-brain barrier and methods of use thereof
US9090693B2 (en) 2007-01-25 2015-07-28 Dana-Farber Cancer Institute Use of anti-EGFR antibodies in treatment of EGFR mutant mediated disease
WO2015127134A2 (en) 2014-02-20 2015-08-27 Allergan, Inc. Complement component c5 antibodies
WO2015130826A1 (en) 2014-02-27 2015-09-03 Allergan, Inc. COMPLEMENT FACTOR Bb ANTIBODIES
US9127056B2 (en) 2011-10-17 2015-09-08 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Monospecific and bispecific human monoclonal antibodies targeting insulin-like growth factor II (IGF-II)
US20150259424A1 (en) * 2002-11-15 2015-09-17 Genmab A/S Human monoclonal antibodies against cd25
US20150283178A1 (en) * 2014-04-07 2015-10-08 Carl H. June Treatment of cancer using anti-cd19 chimeric antigen receptor
US9163091B2 (en) 2007-05-30 2015-10-20 Lpath, Inc. Compositions and methods for binding lysophosphatidic acid
US9169318B2 (en) 2008-03-28 2015-10-27 Sea Lane Biotechnologies, Inc. Neutralizing molecules to viral antigens
US9193780B2 (en) 2008-06-05 2015-11-24 Ablynx N.V. Amino acid sequences directed against envelope proteins of a virus and polypeptides comprising the same for the treatment of viral diseases
EP2947097A1 (en) 2008-04-07 2015-11-25 Ablynx N.V. Amino acid sequences directed against the Notch pathways and uses thereof
US9200061B2 (en) 2004-02-06 2015-12-01 Morpho Sys AG Generation and profiling of fully human HuCAL gold®-derived therapeutic antibodies specific for human CD3i
WO2015193452A1 (en) 2014-06-18 2015-12-23 Ablynx Nv Kv1.3 binding immunoglobulins
US20150368343A1 (en) * 2013-01-25 2015-12-24 Amgen Research (Munich) Gmbh Antibody constructs for cdh19 and cd3
US20150376279A1 (en) * 2014-06-25 2015-12-31 Yale University Cell penetrating nucleolytic antibody based cancer therapy
US9243065B2 (en) 2002-11-08 2016-01-26 Ablynx N.V. Polypeptide constructs including VHH directed against EGFR for intracellular delivery
US20160032019A1 (en) * 2014-07-31 2016-02-04 Amgen Inc. Antibody constructs for CDH19 and CD3
US20160031984A1 (en) * 2014-04-04 2016-02-04 Bionomics Inc. Humanized antibodies that bind lgr5
US20160053025A1 (en) * 2014-08-25 2016-02-25 Samsung Electronics Co., Ltd. Anti-c-met/anti-ang2 bispecific antibody
US9283276B2 (en) 2007-08-14 2016-03-15 Ludwig Institute For Cancer Research Ltd. Monoclonal antibody 175 targeting the EGF receptor and derivatives and uses thereof
CN102164965B (en) * 2008-09-26 2016-03-30 Ucb医药有限公司 Biological product
US9303086B2 (en) 2012-10-03 2016-04-05 Livtech, Inc. Anti-hDlk-1 antibody having an antitumor activity in vivo
EP3009454A2 (en) 2009-04-20 2016-04-20 Oxford Bio Therapeutics Limited Antibodies specific to cadherin-17
US9320792B2 (en) 2002-11-08 2016-04-26 Ablynx N.V. Pulmonary administration of immunoglobulin single variable domains and constructs thereof
US9321844B2 (en) 2011-08-05 2016-04-26 Genentech, Inc. Anti-polyubiquitin antibodies
US20160115241A1 (en) * 2013-03-15 2016-04-28 Amgen Inc. Heterodimeric bispecific antibodies
US9328167B2 (en) 2008-03-04 2016-05-03 Labrys Biologics, Inc. Methods of treating chronic pain
US20160122432A1 (en) * 2013-06-11 2016-05-05 INSERM (Institut National de la Santé et de la Recherche Médicale) Anti-her2 single domain antibodies, polypeptides comprising thereof and their use for treating cancer
WO2016097313A1 (en) 2014-12-19 2016-06-23 Ablynx N.V. Cysteine linked nanobody dimers
US20160176973A1 (en) * 2013-03-15 2016-06-23 Amgen Research (Munich) Gmbh Binding molecules for bcma and cd3
US20160199508A1 (en) * 2012-05-21 2016-07-14 Genentech, Inc. ANTI-Ly6E ANTIBODIES AND IMMUNOCONJUGATES AND METHODS OF USE
US9393304B2 (en) 2008-10-29 2016-07-19 Ablynx N.V. Formulations of single domain antigen binding molecules
US9421372B2 (en) 2011-09-30 2016-08-23 Adi Mashiach Head pain management device having an antenna
US9469689B2 (en) 2010-03-02 2016-10-18 Abbvie Inc. Therapeutic DLL4 binding proteins
US9469688B2 (en) 2009-08-29 2016-10-18 Abbvie Inc. Therapeutic DLL4 binding proteins
US9493558B2 (en) 2010-01-14 2016-11-15 Ucb Pharma, S.A. Bacterial host strain comprising a mutant SPR gene and a wild-type TSP gene
US9493560B2 (en) 2010-08-03 2016-11-15 Abbvie Inc. Dual variable domain immunoglobulins and uses thereof
US9493559B2 (en) 2010-01-14 2016-11-15 Ucb Pharma, S.A. Bacterial host strain expressing recombinant DsbC and having reduced Tsp activity
US9512231B2 (en) * 2010-12-27 2016-12-06 Apo-T B.V. Cross-linking polypeptide that induces apoptosis
US9512228B2 (en) 2011-06-17 2016-12-06 Novo Nordisk A/S Selective elimination of erosive cells
US9533055B2 (en) 2009-03-18 2017-01-03 Armagen Technologies, Inc. Compositions and methods for blood-brain barrier delivery of IgG-decoy receptor fusion proteins
US9534043B2 (en) 2009-02-19 2017-01-03 Glaxo Group Limited Anti-serum albumin binding variants
US9550973B2 (en) 2009-09-24 2017-01-24 Ucb Pharma, S.A. Bacterial host strain
US9556264B2 (en) 2011-12-28 2017-01-31 Chugai Seiyaku Kabushiki Kaisha Humanized anti-epiregulin antibody, and cancer therapeutic agent comprising said antibody as active ingredient
US9562102B2 (en) 2001-05-11 2017-02-07 Ludwig Institute For Cancer Research Specific binding proteins and uses thereof
US20170037130A1 (en) * 2015-07-31 2017-02-09 Amgen Research (Munich) Gmbh Antibody constructs for dll3 and cd3
US9567400B2 (en) 2007-07-27 2017-02-14 Armagen Technologies, Inc. Methods and compositions for increasing α-L-iduronidase activity in the CNS
AU2012296961B2 (en) * 2011-08-17 2017-02-16 Glaxo Group Limited Modified proteins and peptides
US9587227B2 (en) 2010-01-14 2017-03-07 Ucb Pharma, S.A. Bacterial host strain comprising a mutant spr gene and having reduced Tsp activity
US9616120B2 (en) 2010-03-04 2017-04-11 Vet Therapeutics, Inc. Monoclonal antibodies directed to CD20
US9670269B2 (en) 2006-03-31 2017-06-06 Chugai Seiyaku Kabushiki Kaisha Methods of modifying antibodies for purification of bispecific antibodies
US9676845B2 (en) 2009-06-16 2017-06-13 Hoffmann-La Roche, Inc. Bispecific antigen binding proteins
US9688761B2 (en) 2013-12-27 2017-06-27 Merrimack Pharmaceuticals, Inc. Biomarker profiles for predicting outcomes of cancer therapy with ERBB3 inhibitors and/or chemotherapies
US9708393B2 (en) 2011-05-20 2017-07-18 Alderbio Holdings Llc Use of anti-CGRP antibodies and antibody fragments to prevent or inhibit photophobia or light aversion in subjects in need thereof, especially migraine sufferers
US9708412B2 (en) * 2015-05-21 2017-07-18 Harpoon Therapeutics, Inc. Trispecific binding proteins and methods of use
EP3192807A1 (en) 2007-11-27 2017-07-19 The University Of British Columbia 14-3-3 eta antibodies and uses thereof for the diagnosis and treatment of arthritis
US9725516B2 (en) 2011-07-13 2017-08-08 Ucb Pharma, S.A. Bacterial host strain expressing recombinant DSBC
US9745373B2 (en) 2011-05-20 2017-08-29 Alderbio Holdings Llc Anti-CGRP compositions and use thereof
US20170247441A1 (en) * 2014-11-10 2017-08-31 Hoffmann-La Roche Inc. Anti-ang2 antibodies and methods of use
US9751930B2 (en) 2010-07-27 2017-09-05 Ucb Pharma, S.A. Process for purifying proteins
US9777066B2 (en) 2005-06-10 2017-10-03 Chugai Seiyaku Kabushiki Kaisha Pharmaceutical compositions containing sc(Fv)2
WO2017175145A1 (en) 2016-04-05 2017-10-12 Glaxosmithkline Intellectual Property Development Limited Inhibition of tgfbeta in immunotherapy
US9822171B2 (en) 2010-04-15 2017-11-21 AbbVie Deutschland GmbH & Co. KG Amyloid-beta binding proteins
EP3248986A2 (en) 2014-05-16 2017-11-29 Ablynx NV Immunoglobulin variable domains
WO2017210425A1 (en) 2016-06-03 2017-12-07 Janssen Biotech, Inc. Serum albumin-binding fibronectin type iii domains
US9840554B2 (en) 2015-06-15 2017-12-12 Abbvie Inc. Antibodies against platelet-derived growth factor (PDGF)
US9855332B2 (en) 2011-05-20 2018-01-02 Alderbio Holdings Llc Use of anti-CGRP antibodies and antibody fragments to treat diarrhea in subjects with diseases or treatments that result in elevated CGRP levels
WO2018007442A1 (en) 2016-07-06 2018-01-11 Ablynx N.V. Treatment of il-6r related diseases
WO2018020000A1 (en) 2016-07-29 2018-02-01 INSERM (Institut National de la Santé et de la Recherche Médicale) Antibodies targeting tumor associated macrophages and uses thereof
WO2018029182A1 (en) 2016-08-08 2018-02-15 Ablynx N.V. Il-6r single variable domain antibodies for treatment of il-6r related diseases
US9896502B2 (en) 2014-03-21 2018-02-20 Teva Pharmaceuticals International Gmbh Antagonist antibodies directed against calcitonin gene-related peptide and methods using same
US9920115B2 (en) 2016-05-20 2018-03-20 Harpoon Therapeutics, Inc. Single domain serum albumin binding protein
US9944719B2 (en) 2012-12-05 2018-04-17 Hoffman-La Roche Inc. Dual targeting
US9944720B2 (en) 2012-11-01 2018-04-17 Abbvie Inc. Anti-DLL4/VEGF dual variable domain immunoglobulin and uses thereof
US9951125B2 (en) 2006-11-30 2018-04-24 Abbvie Inc. Aβ conformer selective anti-Aβ globulomer monoclonal antibodies
US9951365B2 (en) 2012-05-14 2018-04-24 Ucb Biophara Sprl Recombinant bacterial host cell for protein expression
EP3311837A1 (en) 2011-09-23 2018-04-25 Ablynx NV Prolonged inhibition of interleukin-6 mediated signaling
US9975955B2 (en) 2011-12-02 2018-05-22 Armagen, Inc. Methods and compositions for increasing arylsulfatase A activity in the CNS
US9975966B2 (en) 2014-09-26 2018-05-22 Chugai Seiyaku Kabushiki Kaisha Cytotoxicity-inducing theraputic agent
US9975956B2 (en) 2011-12-22 2018-05-22 I2 Pharmaceuticals, Inc. Surrogate binding proteins which bind DR4 and/or DR5
WO2018091606A1 (en) 2016-11-16 2018-05-24 Ablynx Nv T cell recruiting polypeptides capable of binding cd123 and tcr alpha/beta
US9982036B2 (en) 2011-02-28 2018-05-29 Hoffmann-La Roche Inc. Dual FC antigen binding proteins
WO2018104444A1 (en) 2016-12-07 2018-06-14 Ablynx Nv Improved serum albumin binding immunoglobulin single variable domains
US10005832B2 (en) 2009-05-29 2018-06-26 Chugai Seiyaku Kabushiki Kaisha Method for treating a disease originated from receptor activation by EREG and TGFα
WO2018115231A2 (en) 2016-12-22 2018-06-28 Boehringer Ingelheim International Gmbh Binding molecules for the treatment of cancer
US10011651B2 (en) 2009-10-09 2018-07-03 Armagen, Inc. Methods and compositions for increasing iduronate 2-sulfatase activity in the CNS
US10017561B2 (en) 2009-05-13 2018-07-10 I2 Pharmaceuticals, Inc. Neutralizing molecules to influenza viruses
EP3345926A1 (en) 2010-05-06 2018-07-11 Novartis AG Compositions and methods of use for therapeutic low density lipoprotein-related protein 6 (lrp6) antibodies
WO2018129029A1 (en) 2017-01-04 2018-07-12 Immunogen, Inc. Met antibodies and immunoconjugates and uses thereof
WO2018134234A1 (en) 2017-01-17 2018-07-26 Ablynx Nv Improved serum albumin binders
WO2018134235A1 (en) 2017-01-17 2018-07-26 Ablynx Nv Improved serum albumin binders
US10066016B2 (en) 2016-05-20 2018-09-04 Harpoon Therapeutics, Inc. Single chain variable fragment CD3 binding proteins
WO2018158398A1 (en) 2017-03-02 2018-09-07 INSERM (Institut National de la Santé et de la Recherche Médicale) Antibodies having specificity to nectin-4 and uses thereof
US10072085B2 (en) 2010-01-15 2018-09-11 Kirin-Amgen, Inc. Method of treating psoriasis using an IL-17 receptor antibody formulation
US10093733B2 (en) 2014-12-11 2018-10-09 Abbvie Inc. LRP-8 binding dual variable domain immunoglobulin proteins
US10118962B2 (en) 2008-10-29 2018-11-06 Ablynx N.V. Methods for purification of single domain antigen binding molecules
US10118970B2 (en) 2006-08-30 2018-11-06 Genentech, Inc. Multispecific antibodies
US10138293B2 (en) 2007-12-21 2018-11-27 Hoffmann-La Roche, Inc. Bivalent, bispecific antibodies
US10143748B2 (en) 2005-07-25 2018-12-04 Aptevo Research And Development Llc B-cell reduction using CD37-specific and CD20-specific binding molecules
US20180344868A1 (en) * 2017-05-31 2018-12-06 Boehringer Ingelheim International Gmbh Polypeptides antagonizing wnt signaling in tumor cells
US10184006B2 (en) 2015-06-04 2019-01-22 Merrimack Pharmaceuticals, Inc. Biomarkers for predicting outcomes of cancer therapy with ErbB3 inhibitors
WO2019020480A1 (en) 2017-07-24 2019-01-31 INSERM (Institut National de la Santé et de la Recherche Médicale) Antibodies and peptides to treat hcmv related diseases
US10196442B2 (en) 2011-11-01 2019-02-05 Bionomics Inc. Methods of inhibiting growth of a colon cancer tumor in a subject by administering monoclonal antibodies to G protein-coupled receptor 49 (GPR49)
US10208109B2 (en) 2005-11-30 2019-02-19 Abbvie Inc. Monoclonal antibodies against amyloid beta protein and uses thereof
US10214580B2 (en) 2007-03-27 2019-02-26 I2 Pharmaceuticals, Inc. Constructs and libraries comprising antibody surrogate light chain sequences
US10221245B2 (en) 2013-03-16 2019-03-05 Novartis Ag Treatment of cancer using humanized anti-CD19 chimeric antigen receptor
EP3456743A1 (en) 2013-05-30 2019-03-20 Kiniksa Pharmaceuticals, Ltd. Oncostatin m receptor antigen binding proteins
EP3461844A2 (en) 2009-04-10 2019-04-03 Ablynx N.V. Improved amino acid sequences directed against il-6r and polypeptides comprising the same for the treatment of il-6r related diseases and disorders
US10253086B2 (en) 2015-04-08 2019-04-09 Novartis Ag CD20 therapies, CD22 therapies, and combination therapies with a CD19 chimeric antigen receptor (CAR)-expressing cell
US10253095B2 (en) 2005-01-06 2019-04-09 Innate Pharma S.A.S. Anti-KIR combination treatments and methods
EP3470426A1 (en) 2017-10-10 2019-04-17 Numab Therapeutics AG Multispecific antibody
WO2019072868A1 (en) 2017-10-10 2019-04-18 Numab Therapeutics AG Multispecific antibody
US10300140B2 (en) 2011-07-28 2019-05-28 I2 Pharmaceuticals, Inc. Sur-binding proteins against ERBB3
US10323086B2 (en) 2002-12-24 2019-06-18 Rinat Neuroscience Corp. Methods for treating osteoarthritis pain by administering a nerve growth factor antagonist and compositions containing the same
US10323099B2 (en) 2013-10-11 2019-06-18 Hoffmann-La Roche Inc. Multispecific domain exchanged common variable light chain antibodies
US10344085B2 (en) 2014-11-10 2019-07-09 Hoffman-La Roche, Inc. Anti-IL-1beta antibodies
EP2190875B1 (en) * 2007-04-23 2019-07-10 Sudhir Paul Catalytic Antibodies against HIV gp120
US10377828B2 (en) 2013-03-07 2019-08-13 Boehringer Ingelheim International Gmbh Combination therapy for neoplasia treatment
US10392434B2 (en) 2016-09-23 2019-08-27 Teva Pharmaceuticals International Gmbh Treating refractory migraine
US10464976B2 (en) 2003-01-31 2019-11-05 AbbVie Deutschland GmbH & Co. KG Amyloid β(1-42) oligomers, derivatives thereof and antibodies thereto, methods of preparation thereof and use thereof
EP3569618A1 (en) 2018-05-19 2019-11-20 Boehringer Ingelheim International GmbH Antagonizing cd73 antibody
US10525083B2 (en) 2016-10-07 2020-01-07 Novartis Ag Nucleic acid molecules encoding chimeric antigen receptors comprising a CD20 binding domain
WO2020010250A2 (en) 2018-07-03 2020-01-09 Elstar Therapeutics, Inc. Anti-tcr antibody molecules and uses thereof
WO2020014306A1 (en) 2018-07-10 2020-01-16 Immunogen, Inc. Met antibodies and immunoconjugates and uses thereof
US10538589B2 (en) 2015-01-14 2020-01-21 Armagen Inc. Methods and compositions for increasing N-acetylglucosaminidase (NAGLU) activity in the CNS using a fusion antibody comprising an anti-human insulin receptor antibody and NAGLU
US10538581B2 (en) 2005-11-30 2020-01-21 Abbvie Inc. Anti-Aβ globulomer 4D10 antibodies
US10543271B2 (en) 2017-05-12 2020-01-28 Harpoon Therapeutics, Inc. Mesothelin binding proteins
US10556945B2 (en) 2014-03-21 2020-02-11 Teva Pharmaceuticals International Gmbh Antagonist antibodies directed against calcitonin gene-related peptide and methods using same
US10562966B2 (en) 2010-08-20 2020-02-18 Ucb Biopharma Sprl Antibodies of the class IGG4
WO2020053122A1 (en) 2018-09-10 2020-03-19 INSERM (Institut National de la Santé et de la Recherche Médicale) Combination of her2/neu antibody with heme for treating cancer
US10598653B2 (en) 2011-11-01 2020-03-24 Bionomics Inc. Methods of blocking cancer stem cell growth
US10611825B2 (en) 2011-02-28 2020-04-07 Hoffmann La-Roche Inc. Monovalent antigen binding proteins
WO2020084056A1 (en) 2018-10-25 2020-04-30 Swiss Pharma International Ag Anti-human cd89 antibodies and uses thereof
WO2020099508A1 (en) 2018-11-13 2020-05-22 Emstopa Limited Tissue plasminogen activator antibodies and method of use thereof
US10669337B2 (en) 2014-07-25 2020-06-02 Cytomx Therapeutics, Inc. Bispecific anti-CD3 antibodies, bispecific activatable anti-CD3 antibodies, and methods of using the same
WO2020115283A1 (en) * 2018-12-07 2020-06-11 Baxalta GmbH Bispecific antibodies binding factor ixa and factor x
WO2020114615A1 (en) * 2018-12-07 2020-06-11 Baxalta GmbH Bispecific antibodies binding factor ixa and factor x
US10730954B2 (en) 2017-05-12 2020-08-04 Harpoon Therapeutics, Inc. MSLN targeting trispecific proteins and methods of use
US10730938B2 (en) 2014-11-10 2020-08-04 Hoffmann-La Roche Inc. Bispecific antibodies and methods of use in ophthalmology
EP3693395A1 (en) 2012-05-18 2020-08-12 Amgen Inc. St2 antigen binding proteins
US10745487B2 (en) 2016-03-22 2020-08-18 Bionomics Limited Method of treating cancer by administering an anti-LGR5 monoclonal antibody
WO2020193520A1 (en) 2019-03-25 2020-10-01 INSERM (Institut National de la Santé et de la Recherche Médicale) Treatment of taupathy disorders by targeting new tau species
WO2020198731A2 (en) 2019-03-28 2020-10-01 Danisco Us Inc Engineered antibodies
US10815311B2 (en) 2018-09-25 2020-10-27 Harpoon Therapeutics, Inc. DLL3 binding proteins and methods of use
EP3736293A1 (en) 2013-02-12 2020-11-11 Boehringer Ingelheim International Gmbh Therapeutic and diagnostic target for cancer comprising dll3 binding reagents
US10844134B2 (en) 2016-11-23 2020-11-24 Harpoon Therapeutics, Inc. PSMA targeting trispecific proteins and methods of use
US10849973B2 (en) 2016-11-23 2020-12-01 Harpoon Therapeutics, Inc. Prostate specific membrane antigen binding protein
US10927180B2 (en) 2017-10-13 2021-02-23 Harpoon Therapeutics, Inc. B cell maturation antigen binding proteins
US10946104B2 (en) 2012-01-13 2021-03-16 Apo-Tb.V. Aberrant cell-restricted immunoglobulins provided with a toxic moiety
US10961301B2 (en) 2011-04-01 2021-03-30 Yale University Cell-penetrating anti-DNA antibodies and uses thereof inhibit DNA repair
WO2021058729A1 (en) 2019-09-27 2021-04-01 INSERM (Institut National de la Santé et de la Recherche Médicale) Anti-müllerian inhibiting substance type i receptor antibodies and uses thereof
WO2021058763A1 (en) 2019-09-27 2021-04-01 INSERM (Institut National de la Santé et de la Recherche Médicale) Anti-müllerian inhibiting substance antibodies and uses thereof
US10993420B2 (en) 2013-03-15 2021-05-04 Erasmus University Medical Center Production of heavy chain only antibodies in transgenic mammals
EP3816185A1 (en) 2019-11-04 2021-05-05 Numab Therapeutics AG Multispecific antibody directed against pd-l1 and a tumor-associated antigen
US11028169B2 (en) 2016-05-18 2021-06-08 Boehringer Ingelheim International Gmbh Antibody molecules for cancer treatment
WO2021116119A1 (en) 2019-12-09 2021-06-17 INSERM (Institut National de la Santé et de la Recherche Médicale) Antibodies having specificity to her4 and uses thereof
WO2021123186A1 (en) 2019-12-20 2021-06-24 UCB Biopharma SRL Multi-specific antibody with binding specificity for human il-13 and il-17
WO2021133170A1 (en) 2019-12-24 2021-07-01 Cluster270 Therapeutics B.V. Anti-human hvem (tnfrsf14) antibodies and uses thereof
US11059910B2 (en) 2012-12-03 2021-07-13 Novimmune Sa Anti-CD47 antibodies and methods of use thereof
US11072666B2 (en) 2016-03-14 2021-07-27 Chugai Seiyaku Kabushiki Kaisha Cell injury inducing therapeutic drug for use in cancer therapy
WO2021160133A1 (en) 2020-02-13 2021-08-19 湖南华康恒健生物技术有限公司 Anti-bcma antibody, pharmaceutical composition of same, and applications thereof
US11098115B2 (en) 2011-09-29 2021-08-24 Apo-T B.V. Multi-specific binding molecules targeting aberrant cells
US11136403B2 (en) 2017-10-13 2021-10-05 Harpoon Therapeutics, Inc. Trispecific proteins and methods of use
WO2021205184A1 (en) 2020-04-09 2021-10-14 Autolus Limited Polypeptide
WO2021215919A1 (en) 2020-04-21 2021-10-28 Polpharma Biologics Utrecht B.V. Humanized anti-human cd89 antibodies and uses thereof
WO2021228956A1 (en) 2020-05-12 2021-11-18 INSERM (Institut National de la Santé et de la Recherche Médicale) New method to treat cutaneous t-cell lymphomas and tfh derived lymphomas
US11180563B2 (en) 2020-02-21 2021-11-23 Harpoon Therapeutics, Inc. FLT3 binding proteins and methods of use
WO2021234383A1 (en) 2020-05-19 2021-11-25 Autolus Limited Single domain antibodies against hace2 and their use to prevent sars-cov-2 infection
WO2021236658A1 (en) 2020-05-19 2021-11-25 Boehringer Ingelheim International Gmbh Binding molecules for the treatment of cancer
EP3915580A1 (en) 2020-05-29 2021-12-01 Numab Therapeutics AG Multispecific antibody
US11203636B2 (en) 2017-02-01 2021-12-21 Yale University Treatment of existing left ventricular heart failure
US11234419B2 (en) 2009-12-10 2022-02-01 Regeneran Pharmaceuticals, Inc. Mice that make heavy chain antibodies
US11242395B2 (en) 2017-08-21 2022-02-08 Adagene Inc. Anti-CD137 molecules and use thereof
WO2022042659A1 (en) 2020-08-26 2022-03-03 湖南华康恒健生物技术有限公司 Anti-ox40 antibody, and pharmaceutical composition and application thereof
WO2022044573A1 (en) 2020-08-26 2022-03-03 国立大学法人熊本大学 Human antibody or antigen-binding fragment thereof against coronavirus spike protein
EP3988568A1 (en) 2020-10-21 2022-04-27 Numab Therapeutics AG Combination treatment
WO2022084355A2 (en) 2020-10-21 2022-04-28 Boehringer Ingelheim International Gmbh Agonistic trkb binding molecules for the treatment of eye diseases
WO2022098745A1 (en) 2020-11-03 2022-05-12 Indi Molecular, Inc. Compositions, delivery systems, and methods useful in tumor therapy
WO2022098743A1 (en) 2020-11-03 2022-05-12 Indi Molecular, Inc. Compositions, imaging, and therapeutic methods targeting folate receptor 1 (folr1)
US11345733B2 (en) 2010-06-22 2022-05-31 Precision Biologics, Inc. Colon and pancreas cancer specific antigens and antibodies
US11352426B2 (en) 2015-09-21 2022-06-07 Aptevo Research And Development Llc CD3 binding polypeptides
US11359016B2 (en) 2018-02-02 2022-06-14 Adagene Inc. Anti-CTLA4 antibodies and methods of making and using the same
WO2022122654A1 (en) 2020-12-07 2022-06-16 UCB Biopharma SRL Multi-specific antibodies and antibody combinations
CN114702594A (en) * 2013-12-20 2022-07-05 豪夫迈·罗氏有限公司 Dual specificity antibodies
US11384143B2 (en) 2018-01-05 2022-07-12 Novo Nordisk A/S Methods for treating IL-6 mediated inflammation without immunosuppression
WO2022167460A1 (en) 2021-02-02 2022-08-11 Numab Therapeutics AG Multispecific antibodies having specificity for ror1 and cd3
WO2022178255A2 (en) 2021-02-19 2022-08-25 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Single domain antibodies that neutralize sars-cov-2
WO2022216993A2 (en) 2021-04-08 2022-10-13 Marengo Therapeutics, Inc. Multifuntional molecules binding to tcr and uses thereof
US11472889B2 (en) 2017-10-14 2022-10-18 Cytomx Therapeutics, Inc. Antibodies, activatable antibodies, bispecific antibodies, and bispecific activatable antibodies and methods of use thereof
US11485790B2 (en) 2014-04-07 2022-11-01 Chugai Seiyaku Kabushiki Kaisha Immunoactivating antigen-binding molecule
US11505605B2 (en) 2014-05-13 2022-11-22 Chugai Seiyaku Kabushiki Kaisha T cell-redirected antigen-binding molecule for cells having immunosuppression function
WO2022263507A1 (en) 2021-06-17 2022-12-22 Boehringer Ingelheim International Gmbh Novel tri-specific binding molecules
US11535668B2 (en) 2017-02-28 2022-12-27 Harpoon Therapeutics, Inc. Inducible monovalent antigen binding protein
US11590242B2 (en) 2016-06-15 2023-02-28 Yale University Antibody-mediated autocatalytic, targeted delivery of nanocarriers to tumors
US11623958B2 (en) 2016-05-20 2023-04-11 Harpoon Therapeutics, Inc. Single chain variable fragment CD3 binding proteins
US11639381B2 (en) 2019-01-08 2023-05-02 H. Lundbeck A/S Treatment of headache using anti-CGRP antibodies
US11649262B2 (en) 2015-12-28 2023-05-16 Chugai Seiyaku Kabushiki Kaisha Method for promoting efficiency of purification of Fc region-containing polypeptide
US11649293B2 (en) 2015-11-18 2023-05-16 Chugai Seiyaku Kabushiki Kaisha Method for enhancing humoral immune response
EP4183800A1 (en) 2021-11-19 2023-05-24 Medizinische Hochschule Hannover Novel sars-cov-2 neutralizing antibodies
US11660340B2 (en) 2015-11-18 2023-05-30 Chugai Seiyaku Kabushiki Kaisha Combination therapy using T cell redirection antigen binding molecule against cell having immunosuppressing function
US11661462B2 (en) 2014-07-31 2023-05-30 Amgen Research (Munich) Gmbh Optimized cross-species specific bispecific single chain antibody contructs
WO2023175171A1 (en) 2022-03-18 2023-09-21 Inserm (Institut National De La Sante Et De La Recherche Medicale) Bk polyomavirus antibodies and uses thereof
US11773166B2 (en) 2014-11-04 2023-10-03 Ichnos Sciences SA CD3/CD38 T cell retargeting hetero-dimeric immunoglobulins and methods of their production
WO2024013315A1 (en) 2022-07-15 2024-01-18 Boehringer Ingelheim International Gmbh Binding molecules for the treatment of cancer
US11891433B2 (en) 2012-05-31 2024-02-06 United States Of America As Represented By The Secretary Of The Air Force Camelidae single-domain antibodies against Yersinia pestis and methods of use
WO2024038095A1 (en) 2022-08-16 2024-02-22 Iome Bio NOVEL ANTI-RGMb ANTIBODIES
WO2024052503A1 (en) 2022-09-08 2024-03-14 Institut National de la Santé et de la Recherche Médicale Antibodies having specificity to ltbp2 and uses thereof
WO2024056668A1 (en) 2022-09-12 2024-03-21 Institut National de la Santé et de la Recherche Médicale New anti-itgb8 antibodies and its uses thereof
US11952681B2 (en) 2018-02-02 2024-04-09 Adagene Inc. Masked activatable CD137 antibodies
US11976125B2 (en) 2021-01-15 2024-05-07 Harpoon Therapeutics, Inc. B cell maturation antigen binding proteins

Families Citing this family (62)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
HUP0301002A3 (en) * 2000-06-29 2005-12-28 Abbott Lab Dual specificity antibodies and methods of making and using
US20100081792A1 (en) * 2001-06-28 2010-04-01 Smithkline Beecham Corporation Ligand
US9028822B2 (en) 2002-06-28 2015-05-12 Domantis Limited Antagonists against TNFR1 and methods of use therefor
EP1622942B1 (en) * 2003-05-01 2014-11-19 ImClone LLC Fully human antibodies directed against the human insulin-like growth factor-1 receptor
EP1870458B1 (en) 2005-03-31 2018-05-09 Chugai Seiyaku Kabushiki Kaisha sc(Fv)2 STRUCTURAL ISOMERS
KR101367544B1 (en) 2005-06-10 2014-02-26 추가이 세이야쿠 가부시키가이샤 Stabilizer for protein preparation comprising meglumine and use thereof
SI2100618T1 (en) 2005-06-17 2014-03-31 Imclone Llc An anti-PDGFRalpha antibody for use in the treatment of metastatic bone cancer
EA017265B1 (en) * 2006-02-03 2012-11-30 Имклоун Элэлси Use of imc-a12 antibody being igf-ir inhibitor for treatment of prostate cancer
US20100226920A1 (en) * 2006-03-27 2010-09-09 Ablynx N.V. Medical delivery device for therapeutic proteins based on single domain antibodies
US20070269422A1 (en) * 2006-05-17 2007-11-22 Ablynx N.V. Serum albumin binding proteins with long half-lives
US20070274985A1 (en) * 2006-05-26 2007-11-29 Stefan Dubel Antibody
WO2007146702A2 (en) * 2006-06-12 2007-12-21 Centocor, Inc. Method for detecting il-16 activity and modulation of il-16 activity based on rantes proxy levels
US8324350B2 (en) * 2006-12-29 2012-12-04 Abbott Laboratories Dual-specific IL-1α/IL-1β antibodies
EP2486928A1 (en) 2007-02-27 2012-08-15 Abbott GmbH & Co. KG Method for the treatment of amyloidoses
CA2680854C (en) 2007-03-15 2017-02-14 Ludwig Institute For Cancer Research Treatment method using egfr antibodies and src inhibitors and related formulations
US20100254980A1 (en) 2007-11-02 2010-10-07 Novartis Ag Molecules and methods for modulating low-density-lipoprotein receptor-related protein 6 (lrp6)
US8557242B2 (en) 2008-01-03 2013-10-15 The Scripps Research Institute ERBB2 antibodies comprising modular recognition domains
US8574577B2 (en) 2008-01-03 2013-11-05 The Scripps Research Institute VEGF antibodies comprising modular recognition domains
EP2237797A4 (en) 2008-01-03 2012-11-07 Scripps Research Inst Antibody targeting through a modular recognition domain
US8454960B2 (en) 2008-01-03 2013-06-04 The Scripps Research Institute Multispecific antibody targeting and multivalency through modular recognition domains
US8557243B2 (en) 2008-01-03 2013-10-15 The Scripps Research Institute EFGR antibodies comprising modular recognition domains
AU2010205627B2 (en) 2009-01-14 2015-04-02 Ablynx Nv Pulmonary administration of immunoglobulin single variable domains and constructs thereof
CA2756244A1 (en) * 2009-04-02 2010-10-07 Roche Glycart Ag Multispecific antibodies comprising full length antibodies and single chain fab fragments
CA2757931C (en) 2009-04-07 2019-03-26 Roche Glycart Ag Trivalent, bispecific antibodies
RU2573915C2 (en) 2009-09-16 2016-01-27 Дженентек, Инк. Protein complexes containing superhelix and/or banding, and their use
GB201005063D0 (en) 2010-03-25 2010-05-12 Ucb Pharma Sa Biological products
WO2011044368A1 (en) 2009-10-07 2011-04-14 Macrogenics, Inc. Fc region-containing polypeptides that exhibit improved effector function due to alterations of the extent of fucosylation, and methods for their use
TW201138821A (en) 2010-03-26 2011-11-16 Roche Glycart Ag Bispecific antibodies
WO2012009705A1 (en) 2010-07-15 2012-01-19 Zyngenia, Inc. Ang-2 binding complexes and uses thereof
JP2013536175A (en) 2010-07-16 2013-09-19 アブリンクス エン.ヴェー. Modified single domain antigen binding molecules and uses thereof
EP2601298B1 (en) 2010-08-02 2016-11-30 Regeneron Pharmaceuticals, Inc. Mice that make binding proteins comprising vl domains
EP2609111B1 (en) 2010-08-24 2017-11-01 F. Hoffmann-La Roche AG Bispecific antibodies comprising a disulfide stabilized-fv fragment
US9884909B2 (en) 2010-12-01 2018-02-06 Alderbio Holdings Llc Anti-NGF compositions and use thereof
CN103619879A (en) 2010-12-01 2014-03-05 奥尔德生物控股有限责任公司 Anti-ngf compositions and use thereof
US9067988B2 (en) 2010-12-01 2015-06-30 Alderbio Holdings Llc Methods of preventing or treating pain using anti-NGF antibodies
US9539324B2 (en) 2010-12-01 2017-01-10 Alderbio Holdings, Llc Methods of preventing inflammation and treating pain using anti-NGF compositions
US11214610B2 (en) 2010-12-01 2022-01-04 H. Lundbeck A/S High-purity production of multi-subunit proteins such as antibodies in transformed microbes such as Pichia pastoris
US9078878B2 (en) 2010-12-01 2015-07-14 Alderbio Holdings Llc Anti-NGF antibodies that selectively inhibit the association of NGF with TrkA, without affecting the association of NGF with p75
WO2012085111A1 (en) 2010-12-23 2012-06-28 F. Hoffmann-La Roche Ag Polypeptide-polynucleotide-complex and its use in targeted effector moiety delivery
EP2681242B1 (en) 2011-03-01 2018-01-24 Amgen Inc. Sclerostin and dkk-1 bispecific binding agents
CN103547592A (en) 2011-03-30 2014-01-29 埃博灵克斯股份有限公司 Methods of treating immune disorders with single domain antibodies against TNF-alpha
EP2714738B1 (en) 2011-05-24 2018-10-10 Zyngenia, Inc. Multivalent and monovalent multispecific complexes and their uses
US8951966B2 (en) 2011-07-01 2015-02-10 Ngm Biopharmaceuticals, Inc. Compositions comprising variants and fusions of FGF19 polypeptides, and uses and methods thereof for treatment of metabolic disorders and diseases
ES2732712T3 (en) 2011-10-31 2019-11-25 Chugai Pharmaceutical Co Ltd Antigen binding molecule that has a regulated conjugation between the heavy chain and the light chain
AU2012323987B2 (en) 2011-12-20 2015-07-09 Regeneron Pharmaceuticals, Inc. Humanized light chain mice
EP2867254B1 (en) 2012-06-27 2017-10-25 F. Hoffmann-La Roche AG Method for making antibody fc-region conjugates comprising at least one binding entity that specifically binds to a target and uses thereof
MX354862B (en) 2012-06-27 2018-03-23 Hoffmann La Roche Method for selection and production of tailor-made highly selective and multi-specific targeting entities containing at least two different binding entities and uses thereof.
EP3798228A1 (en) 2012-11-28 2021-03-31 NGM Biopharmaceuticals, Inc. Compositions and methods for treatment of metabolic disorders and diseases
JP6403685B2 (en) 2012-12-27 2018-10-17 エヌジーエム バイオファーマシューティカルス,インコーポレーテッド Methods of regulating bile acid homeostasis and treating bile acid disorders and diseases
MX2015013163A (en) 2013-03-15 2016-04-04 Zyngenia Inc Multivalent and monovalent multispecific complexes and their uses.
RU2714733C2 (en) * 2013-03-15 2020-02-19 Мемориал Слоан-Кеттеринг Кэнсер Сентер Multimerization technologies
PL3039038T3 (en) * 2013-08-30 2021-04-19 Aprilbio Co., Ltd An anti serum albumin fab-effector moiety fusion construct, and the preparing method thereof
JP2017510273A (en) 2014-03-21 2017-04-13 リジェネロン・ファーマシューティカルズ・インコーポレイテッドRegeneron Pharmaceuticals, Inc. VL antigen binding protein showing different binding properties
KR102601491B1 (en) 2014-03-21 2023-11-13 리제너론 파마슈티칼스 인코포레이티드 Non-human animals that make single domain binding proteins
KR102569907B1 (en) 2014-10-23 2023-08-24 엔지엠 바이오파마슈티컬스, 아이엔씨. Pharmaceutical Compositions Comprising Peptide Variants and Methods of Use Thereof
ES2764111T3 (en) 2014-12-03 2020-06-02 Hoffmann La Roche Multispecific antibodies
EP3262068A1 (en) * 2015-02-05 2018-01-03 Ablynx N.V. Nanobody dimers linked via c-terminally engineered cysteins
CN107438622A (en) 2015-03-19 2017-12-05 瑞泽恩制药公司 Non-human animal of the selection with reference to the light chain variable district of antigen
CA3082794A1 (en) 2015-11-09 2017-05-18 Ngm Biopharmaceuticals Inc. Methods for treatment of bile acid-related disorders
IL253015B2 (en) 2017-06-18 2023-07-01 Israel Aerospace Ind Ltd System and method for refueling air vehicles
IL253407B (en) 2017-07-10 2020-08-31 Israel Aerospace Ind Ltd Refueling
WO2023196932A2 (en) * 2022-04-06 2023-10-12 The Regents Of The University Of California Isolating active t-cell cars from bulk transduced human t-cells

Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0368684A1 (en) * 1988-11-11 1990-05-16 Medical Research Council Cloning immunoglobulin variable domain sequences.
WO1991002078A1 (en) * 1989-08-07 1991-02-21 Peptide Technology Ltd Tumour necrosis factor binding ligands
US5644034A (en) * 1989-08-07 1997-07-01 Peptide Technology Ltd. Tumour necrosis factor binding ligands
WO1997030084A1 (en) * 1996-02-15 1997-08-21 Genetics Institute, Inc. Tnf receptor death domain ligand proteins and inhibitors of ligand binding
WO1998040469A1 (en) * 1997-03-13 1998-09-17 Cardiac Crc Nominees Pty. Ltd. Haemocompatible surfaces
WO2000029004A1 (en) * 1998-11-18 2000-05-25 Peptor Ltd. Small functional units of antibody heavy chain variable regions
WO2002072141A2 (en) * 2001-03-09 2002-09-19 William Herman Targeted ligands
WO2003002609A2 (en) * 2001-06-28 2003-01-09 Domantis Limited Dual-specific ligand and its use
WO2003035694A2 (en) * 2001-10-24 2003-05-01 Vlaams Interuniversitair Instituut Voor Biotechnologie Vzw Functional heavy chain antibodies, fragments thereof, library thereof and methods of production thereof

Family Cites Families (120)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CU22545A1 (en) * 1994-11-18 1999-03-31 Centro Inmunologia Molecular OBTAINING A CHEMICAL AND HUMANIZED ANTIBODY AGAINST THE RECEPTOR OF THE EPIDERMAL GROWTH FACTOR FOR DIAGNOSTIC AND THERAPEUTIC USE
US5120712A (en) * 1986-05-05 1992-06-09 The General Hospital Corporation Insulinotropic hormone
WO1989006692A1 (en) * 1988-01-12 1989-07-27 Genentech, Inc. Method of treating tumor cells by inhibiting growth factor receptor function
AU4128089A (en) * 1988-09-15 1990-03-22 Rorer International (Overseas) Inc. Monoclonal antibodies specific to human epidermal growth factor receptor and therapeutic methods employing same
US5530101A (en) * 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
US5116964A (en) * 1989-02-23 1992-05-26 Genentech, Inc. Hybrid immunoglobulins
SE509359C2 (en) * 1989-08-01 1999-01-18 Cemu Bioteknik Ab Use of stabilized protein or peptide conjugates for the preparation of a drug
US6498237B2 (en) * 1989-08-07 2002-12-24 Peptech Limited Tumor necrosis factor antibodies
US5977307A (en) * 1989-09-07 1999-11-02 Alkermes, Inc. Transferrin receptor specific ligand-neuropharmaceutical agent fusion proteins
US5459061A (en) * 1990-01-26 1995-10-17 W. Alton Jones Cell Science Center, Inc. Hybridomas producing monoclonal antibodies which specifically bind to continuous epitope on the human EGF receptor and compete with EGF for binding to the EGF receptor
GB9016299D0 (en) * 1990-07-25 1990-09-12 Brien Caroline J O Binding substances
US5726152A (en) * 1990-09-21 1998-03-10 Merck & Co., Inc. Vascular endothelial cell growth factor II
US5612034A (en) * 1990-10-03 1997-03-18 Redcell, Inc. Super-globuling for in vivo extended lifetimes
US5843440A (en) 1990-10-03 1998-12-01 Redcell Canada, Inc. Cellular and serum protein anchors for modulating pharmacokinetics
WO1992005801A1 (en) 1990-10-04 1992-04-16 University Of Virginia Alumni Patents Foundation Primate erythrocyte bound monoclonal antibody heteropolymers
US5582996A (en) * 1990-12-04 1996-12-10 The Wistar Institute Of Anatomy & Biology Bifunctional antibodies and method of preparing same
HU219537B (en) * 1991-03-06 2001-05-28 Merck Patent Gmbh. Humanized and chimaeric monoclonal antibodies comprising of them pharmaceutical composition and the antibodies coding sequence containing of expression vectors, and process for the praparation of antibodies
OA10149A (en) * 1991-03-29 1996-12-18 Genentech Inc Vascular endothelial cell growth factor antagonists
US6582959B2 (en) * 1991-03-29 2003-06-24 Genentech, Inc. Antibodies to vascular endothelial cell growth factor
WO1993000917A1 (en) * 1991-07-05 1993-01-21 Seragen, Inc. Epidermal growth factor receptor targeted molecules for treatment of inflammatory arthritis
US5474771A (en) * 1991-11-15 1995-12-12 The Trustees Of Columbia University In The City Of New York Murine monoclonal antibody (5c8) recognizes a human glycoprotein on the surface of T-lymphocytes, compositions containing same
DE69233745D1 (en) * 1991-12-02 2008-10-23 Cambridge Antibody Tech Preparation of Autoantibodies on Phage Surfaces Starting from Antibody Segment Libraries
US5965132A (en) * 1992-03-05 1999-10-12 Board Of Regents, The University Of Texas System Methods and compositions for targeting the vasculature of solid tumors
PT1498427E (en) * 1992-08-21 2010-03-22 Univ Bruxelles Immunoglobulins devoid of light chains
US6005079A (en) * 1992-08-21 1999-12-21 Vrije Universiteit Brussels Immunoglobulins devoid of light chains
CA2150262C (en) * 1992-12-04 2008-07-08 Kaspar-Philipp Holliger Multivalent and multispecific binding proteins, their manufacture and use
CN100341896C (en) * 1993-09-02 2007-10-10 达特茅斯学院理事 Anti-GP39 antibodies and uses therefor
US5869049A (en) * 1993-09-02 1999-02-09 Trustees Of Dartmouth College Methods of inducing T cell unresponsiveness to bone marrow with gp39 antagonists
US5922545A (en) * 1993-10-29 1999-07-13 Affymax Technologies N.V. In vitro peptide and antibody display libraries
SE9400088D0 (en) * 1994-01-14 1994-01-14 Kabi Pharmacia Ab Bacterial receptor structures
IL112372A (en) * 1994-02-07 2001-08-26 Res Dev Foundation Non-viral vector for the delivery of genetic information to cells
US5605793A (en) * 1994-02-17 1997-02-25 Affymax Technologies N.V. Methods for in vitro recombination
US5683693A (en) * 1994-04-25 1997-11-04 Trustees Of Dartmouth College Method for inducing T cell unresponsiveness to a tissue or organ graft with anti-CD40 ligand antibody or soluble CD40
GB9410533D0 (en) * 1994-05-26 1994-07-13 Lynxvale Ltd In situ hybridisation and immuno-Chemical localisation of a growth factor
US6010861A (en) * 1994-08-03 2000-01-04 Dgi Biotechnologies, Llc Target specific screens and their use for discovering small organic molecular pharmacophores
ES2167391T3 (en) * 1994-09-16 2002-05-16 Merck Patent Gmbh IMMUNOCONJUGADOS II.
US5876950A (en) * 1995-01-26 1999-03-02 Bristol-Myers Squibb Company Monoclonal antibodies specific for different epitopes of human GP39 and methods for their use in diagnosis and therapy
US5928939A (en) * 1995-03-01 1999-07-27 Ludwig Institute For Cancer Research Vascular endothelial growth factor-b and dna coding therefor
EP0739981A1 (en) 1995-04-25 1996-10-30 Vrije Universiteit Brussel Variable fragments of immunoglobulins - use for therapeutic or veterinary purposes
US7060808B1 (en) * 1995-06-07 2006-06-13 Imclone Systems Incorporated Humanized anti-EGF receptor monoclonal antibody
US6410690B1 (en) * 1995-06-07 2002-06-25 Medarex, Inc. Therapeutic compounds comprised of anti-Fc receptor antibodies
US5833987A (en) * 1995-06-07 1998-11-10 Trustees Of Dartmouth College Treatment of T cell mediated autoimmune disorders
US6020473A (en) * 1995-08-25 2000-02-01 Genentech, Inc. Nucleic acids encoding variants of vascular endothelial cell growth factor
CN1173878A (en) * 1995-10-16 1998-02-18 尤尼利弗公司 Bifunctional or bivalent antibody fragment analogue
US6340459B1 (en) * 1995-12-01 2002-01-22 The Trustees Of Columbia University In The City Of New York Therapeutic applications for the anti-T-BAM (CD40-L) monoclonal antibody 5C8 in the treatment of reperfusion injury in non-transplant recipients
US5664034A (en) * 1996-05-21 1997-09-02 Lucent Technologies Inc. Lightwave communication monitoring switch
US20040248201A1 (en) * 1996-06-27 2004-12-09 Serge Muyldermans Recognition molecules interacting specifically with the active site or cleft of a target molecule
NZ333602A (en) * 1996-07-08 2000-06-23 Biogen Inc Treatment of smooth muscle cell dependent disease using antibodies capable of inhibiting the CD40 - CD40 ligand interaction
US5922845A (en) * 1996-07-11 1999-07-13 Medarex, Inc. Therapeutic multispecific compounds comprised of anti-Fcα receptor antibodies
ATE253593T1 (en) * 1996-07-16 2003-11-15 Plueckthun Andreas Prof Dr IMMUNOGLOBULIN SUPERFAMILY DOMAINS AND FRAGMENTS WITH INCREASED SOLUBILITY
US6013780A (en) * 1996-09-06 2000-01-11 Technion Research & Development Co. Ltd. VEGF145 expression vectors
US6750044B1 (en) * 1996-10-17 2004-06-15 Genentech, Inc. Variants of vascular endothelial cell growth factor having antagonistic properties, nucleic acids encoding the same and host cells comprising those nucleic acids
US20030165467A1 (en) * 1997-01-21 2003-09-04 Technion Research & Development Co., Ltd. Angiogenic factor and use thereof in treating cardiovascular disease
US6294170B1 (en) * 1997-08-08 2001-09-25 Amgen Inc. Composition and method for treating inflammatory diseases
US6485942B1 (en) * 1997-02-14 2002-11-26 Genentech, Inc. Variants of vascular endothelial cell growth factor having altered pharmacological properties, and recombinant methods of production
US20020032315A1 (en) * 1997-08-06 2002-03-14 Manuel Baca Anti-vegf antibodies
US20030207346A1 (en) * 1997-05-02 2003-11-06 William R. Arathoon Method for making multispecific antibodies having heteromultimeric and common components
US20020173629A1 (en) * 1997-05-05 2002-11-21 Aya Jakobovits Human monoclonal antibodies to epidermal growth factor receptor
EP1005372B1 (en) * 1997-06-20 2002-01-02 Tanox Pharma B.V. Anti-cd40l immunotoxins for the treatment of diseases
GB9722131D0 (en) * 1997-10-20 1997-12-17 Medical Res Council Method
DE69841562D1 (en) 1997-10-27 2010-04-29 Bac Ip Bv MULTIVALENT ANTIGENBINDING PROTEINS
US6777534B1 (en) * 1997-12-09 2004-08-17 Children's Medical Center Corporation Peptide antagonists of vascular endothelial growth factor
US20020192211A1 (en) * 1998-03-17 2002-12-19 Hudziak Robert M. Method of treating tumor cells by inhibiting growth factor receptor function
US20030224001A1 (en) * 1998-03-19 2003-12-04 Goldstein Neil I. Antibody and antibody fragments for inhibiting the growth of tumors
ZA200007412B (en) * 1998-05-15 2002-03-12 Imclone Systems Inc Treatment of human tumors with radiation and inhibitors of growth factor receptor tyrosine kinases.
US20030175271A1 (en) * 1998-05-20 2003-09-18 Kyowa Hakko Kogyo Co., Ltd. VEGF activity inhibitor
US7264801B2 (en) * 1998-08-11 2007-09-04 Genentech, Inc. EG-VEGF nucleic acids and polypeptides and method of use
US20020172678A1 (en) * 2000-06-23 2002-11-21 Napoleone Ferrara EG-VEGF nucleic acids and polypeptides and methods of use
US7488590B2 (en) * 1998-10-23 2009-02-10 Amgen Inc. Modified peptides as therapeutic agents
EP1002861A1 (en) * 1998-10-26 2000-05-24 Unilever Plc Antigen-binding proteins comprising a linker which confers restricted conformational flexibility
JP4731016B2 (en) * 1998-12-22 2011-07-20 ジェネンテック, インコーポレイテッド Vascular endothelial growth factor antagonists and their uses
SE9901379D0 (en) * 1999-04-19 1999-04-19 Pharmacia & Upjohn Ab Receptor structures
ES2223705T3 (en) * 1999-04-28 2005-03-01 Board Of Regents, The University Of Texas System COMPOSITIONS AND METHODS FOR THE TREATMENT OF CANCER THROUGH INHIBITION SELECTIVE OF VEGF.
US6703020B1 (en) * 1999-04-28 2004-03-09 Board Of Regents, The University Of Texas System Antibody conjugate methods for selectively inhibiting VEGF
US7049410B2 (en) * 1999-05-14 2006-05-23 Majumdar Adhip P N Antibodies to a novel EGF-receptor related protein (ERRP)
KR20020000223A (en) * 1999-05-14 2002-01-05 존 비. 랜디스 Treatment of Refractory Human Tumors with Epidermal Growth Factor Receptor Antagonists
JP2003502024A (en) 1999-05-14 2003-01-21 メディカル リサーチ カウンシル Protein scaffolds and their use for multimerizing monomeric polypeptides
JP2003507006A (en) * 1999-05-20 2003-02-25 サイオス,インコーポレーテッド Vascular endothelial growth factor variant
US6419758B1 (en) * 1999-09-10 2002-07-16 General Electric Company Cathode wire filament for x-ray tube applications
CA2921260A1 (en) * 1999-12-24 2001-06-28 Genentech, Inc. Methods and compositions for prolonging elimination half-times of bioactive compounds
EP1259626B1 (en) * 2000-02-25 2007-10-31 Ludwig Institute For Cancer Research Materials and methods involving hybrid vascular endothelial growth factor dnas and proteins and screening methods for modulators
US6627785B1 (en) * 2000-02-29 2003-09-30 Virginia Commwealth University Wound dressings with protease-lowering activity
CA2404528A1 (en) * 2000-03-31 2001-10-04 Institut Pasteur Peptides blocking vascular endothelial growth factor (vegf)-mediated angiogenesis, polynucleotides encoding said peptides and methods of use thereof
IL151853A0 (en) * 2000-04-11 2003-04-10 Genentech Inc Multivalent antibodies and uses therefor
WO2001089549A2 (en) * 2000-05-22 2001-11-29 Hyseq, Inc. Therapeutic uses of il-1 receptor antagonist
WO2001090192A2 (en) * 2000-05-24 2001-11-29 Imclone Systems Incorporated Bispecific immunoglobulin-like antigen binding proteins and method of production
DE10038624C2 (en) * 2000-08-03 2002-11-21 Broekelmann Aluminium F W Heat transfer tube with twisted inner fins
UA81743C2 (en) * 2000-08-07 2008-02-11 Центокор, Инк. HUMAN MONOCLONAL ANTIBODY WHICH SPECIFICALLY BINDS TUMOR NECROSIS FACTOR ALFA (TNFα), PHARMACEUTICAL MIXTURE CONTAINING THEREOF, AND METHOD FOR TREATING ARTHRITIS
WO2002030984A1 (en) * 2000-10-13 2002-04-18 Uab Research Foundation Human anti-epidermal growth factor receptor single-chain antibodies
WO2002032925A2 (en) * 2000-10-16 2002-04-25 Phylos, Inc. Protein scaffolds for antibody mimics and other binding proteins
WO2002048193A2 (en) 2000-12-13 2002-06-20 Unilever N.V. Camelidae antibody arrays
US20030133939A1 (en) * 2001-01-17 2003-07-17 Genecraft, Inc. Binding domain-immunoglobulin fusion proteins
US7667004B2 (en) * 2001-04-17 2010-02-23 Abmaxis, Inc. Humanized antibodies against vascular endothelial growth factor
US20050089932A1 (en) * 2001-04-26 2005-04-28 Avidia Research Institute Novel proteins with targeted binding
CN1507355A (en) * 2001-05-08 2004-06-23 Ĭ��ר�����޹�˾ Combination therapy using anti-EGFR antibodies and anti-hormonal agents
EP1417232B1 (en) * 2001-06-13 2014-12-03 Genmab A/S Human monoclonal antibodies to epidermal growth factor receptor (egfr)
US20050271663A1 (en) * 2001-06-28 2005-12-08 Domantis Limited Compositions and methods for treating inflammatory disorders
EP1421113A4 (en) * 2001-08-03 2005-04-13 Commw Scient Ind Res Org Methods of screening based on the egf receptor crystal structure
EP2281837B1 (en) * 2001-08-10 2016-10-05 Aberdeen University Antigen binding domains from fish
IL161418A0 (en) * 2001-10-15 2004-09-27 Immunomedics Inc Direct targeting binding proteins
DE10163459A1 (en) * 2001-12-21 2003-07-03 Merck Patent Gmbh Lyophilized preparation containing antibodies to EGF receptor
CA2478011C (en) * 2002-03-01 2013-05-21 Immunomedics, Inc. Bispecific antibody point mutations for enhancing rate of clearance
JP2006512895A (en) 2002-06-28 2006-04-20 ドマンティス リミテッド Ligand
CN100471871C (en) * 2002-09-06 2009-03-25 安姆根有限公司 Therapeutic human anti-IL-1R1 monoclonal antibody
TW200501960A (en) * 2002-10-02 2005-01-16 Bristol Myers Squibb Co Synergistic kits and compositions for treating cancer
AU2003286004A1 (en) * 2002-11-08 2004-06-07 Ablynx N.V. Single domain antibodies directed against interferon- gamma and uses therefor
MXPA05004955A (en) 2002-11-08 2005-11-17 Ablynx Nv Single domain antibodies directed against tumour necrosis factor-alpha and uses therefor.
GB0230201D0 (en) * 2002-12-27 2003-02-05 Domantis Ltd Retargeting
JP3983183B2 (en) * 2003-02-18 2007-09-26 トヨタ車体株式会社 Transport equipment
US20050043233A1 (en) * 2003-04-29 2005-02-24 Boehringer Ingelheim International Gmbh Combinations for the treatment of diseases involving cell proliferation, migration or apoptosis of myeloma cells or angiogenesis
ME00425B (en) * 2003-05-30 2011-10-10 Genentech Inc Treatment with anti-vegf antibodies
US7354578B2 (en) * 2003-06-06 2008-04-08 Regeneron Pharmaceuticals, Inc. Method of tumor regression with VEGF inhibitors
KR20120098932A (en) * 2003-06-27 2012-09-05 암젠 프레몬트 인코포레이티드 Antibodies directed to the deletion mutants of epidermal growth factor receptor and uses thereof
AR046510A1 (en) * 2003-07-25 2005-12-14 Regeneron Pharma COMPOSITION OF A VEGF ANTAGONIST AND AN ANTI-PROLIFERATIVE AGENT
US20050196340A1 (en) * 2003-08-06 2005-09-08 Jocelyn Holash Use of a VEGF antagonist in combination with radiation therapy
US20050079184A1 (en) * 2003-08-08 2005-04-14 Immunomedics, Inc. Bispecific antibodies for inducing apoptosis of tumor and diseased cells
US7534604B2 (en) * 2004-01-16 2009-05-19 Regeneron Pharmaceuticals, Inc. Fusion polypeptides capable of activating receptors
US7767792B2 (en) * 2004-02-20 2010-08-03 Ludwig Institute For Cancer Research Ltd. Antibodies to EGF receptor epitope peptides
BRPI0511755A (en) * 2004-06-01 2008-01-02 Domantis Ltd drug compositions, fusions and conjugates and methods of production, treatment and use
US7563443B2 (en) * 2004-09-17 2009-07-21 Domantis Limited Monovalent anti-CD40L antibody polypeptides and compositions thereof

Patent Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0368684A1 (en) * 1988-11-11 1990-05-16 Medical Research Council Cloning immunoglobulin variable domain sequences.
WO1991002078A1 (en) * 1989-08-07 1991-02-21 Peptide Technology Ltd Tumour necrosis factor binding ligands
US5644034A (en) * 1989-08-07 1997-07-01 Peptide Technology Ltd. Tumour necrosis factor binding ligands
WO1997030084A1 (en) * 1996-02-15 1997-08-21 Genetics Institute, Inc. Tnf receptor death domain ligand proteins and inhibitors of ligand binding
WO1998040469A1 (en) * 1997-03-13 1998-09-17 Cardiac Crc Nominees Pty. Ltd. Haemocompatible surfaces
WO2000029004A1 (en) * 1998-11-18 2000-05-25 Peptor Ltd. Small functional units of antibody heavy chain variable regions
WO2002072141A2 (en) * 2001-03-09 2002-09-19 William Herman Targeted ligands
WO2003002609A2 (en) * 2001-06-28 2003-01-09 Domantis Limited Dual-specific ligand and its use
WO2003035694A2 (en) * 2001-10-24 2003-05-01 Vlaams Interuniversitair Instituut Voor Biotechnologie Vzw Functional heavy chain antibodies, fragments thereof, library thereof and methods of production thereof

Non-Patent Citations (9)

* Cited by examiner, † Cited by third party
Title
ELS CONRATH K ET AL: "Camel single-domain antibodies as modular building units in bispecific and bivalent antibody constructs" JOURNAL OF BIOLOGICAL CHEMISTRY, AMERICAN SOCIETY OF BIOLOGICAL CHEMISTS, BALTIMORE, MD, US, vol. 276, no. 10, 9 March 2001 (2001-03-09), pages 7346-7350, XP002248402 ISSN: 0021-9258 cited in the application *
HOLLIGER P ET AL: "RETARGETING SERUM IMMUNOGLOBULIN WITH BISPECIFIC DIABODIES" NATURE BIOTECHNOLOGY, NATURE PUBLISHING, US, vol. 15, July 1997 (1997-07), pages 632-636, XP002921893 ISSN: 1087-0156 *
MUYLDERMANS S ET AL: "UNIQUE SINGLE-DOMAIN ANTIGEN BINDING FRAGMENTS DERIVES FROM NATURALLY OCCURRING CAMEL HEAVY-CHAIN ANTIBODIES" JOURNAL OF MOLECULAR RECOGNITION, HEYDEN & SON LTD., LONDON, GB, vol. 12, no. 2, March 1999 (1999-03), pages 131-140, XP009012180 ISSN: 0952-3499 *
REITER Y ET AL: "An antibody single-domain phage display library of a native heavy chain variable region: isolation of functional single-domain VH molecules with a unique interface" JOURNAL OF MOLECULAR BIOLOGY, LONDON, GB, vol. 290, no. 3, 1 January 1999 (1999-01-01), pages 685-698, XP004461990 ISSN: 0022-2836 *
RIECHMANN L ET AL: "Single domain antibodies: Comparison of camel VH and camelised human VH domains" BIOSIS, XP004187632 *
See also references of EP1517921A2 *
SMITH BRYAN J ET AL: "Prolonged in vivo residence times of antibody fragments associated with albumin" BIOCONJUGATE CHEMISTRY, vol. 12, no. 5, September 2001 (2001-09), pages 750-756, XP002270731 ISSN: 1043-1802 *
TANHA J ET AL: "Optimal design features of camelized human single-domain antibody libraries." THE JOURNAL OF BIOLOGICAL CHEMISTRY. UNITED STATES 6 JUL 2001, vol. 276, no. 27, 6 July 2001 (2001-07-06), pages 24774-24780, XP002283749 ISSN: 0021-9258 *
VAN DEN BEUCKEN T ET AL: "Building novel binding ligands to B7.1 and B7.2 based on human antibody single variable light chain domains" JOURNAL OF MOLECULAR BIOLOGY, LONDON, GB, vol. 310, no. 3, 13 July 2001 (2001-07-13), pages 591-601, XP004464206 ISSN: 0022-2836 *

Cited By (812)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7892543B2 (en) * 1996-10-04 2011-02-22 Chugai Seiyako Kabushiki Kaisha Reshaped human anti-HM 1.24 antibody
US8106162B2 (en) * 1997-06-12 2012-01-31 Novartis Ag Artificial antibody polypeptides
US8486391B2 (en) * 1998-07-13 2013-07-16 Board Of Regents, University Of Texas System Cancer treatment kits using antibodies to aminophospholipids
US8703133B2 (en) 1998-11-18 2014-04-22 Genentech, Inc. Antibody variants
US8722859B2 (en) 2000-04-11 2014-05-13 Genentech, Inc. Multivalent antibodies and uses therefor
US9493579B2 (en) 2000-04-11 2016-11-15 Genentech, Inc. Multivalent antibodies and uses therefor
US7829084B2 (en) * 2001-01-17 2010-11-09 Trubion Pharmaceuticals, Inc. Binding constructs and methods for use thereof
US8106161B2 (en) 2001-01-17 2012-01-31 Emergent Product Development Seattle, Llc Binding domain-immunoglobulin fusion proteins
US9005612B2 (en) 2001-01-17 2015-04-14 Emergent Product Development Seattle, Llc Binding domain-immunoglobulin fusion proteins
US8147835B2 (en) 2001-01-17 2012-04-03 Emergent Product Development Seattle, Llc Binding domain-immunoglobulin fusion proteins
US8188237B2 (en) 2001-01-17 2012-05-29 Emergent Product Development Seattle, Llc Binding domain-immunoglobulin fusion proteins
US8197810B2 (en) 2001-01-17 2012-06-12 Emergent Product Development Seattle, Llc Binding domain-immunoglobulin fusion proteins
US8853366B2 (en) 2001-01-17 2014-10-07 Emergent Product Development Seattle, Llc Binding domain-immunoglobulin fusion proteins
US9562102B2 (en) 2001-05-11 2017-02-07 Ludwig Institute For Cancer Research Specific binding proteins and uses thereof
US8097251B2 (en) 2001-10-24 2012-01-17 Vlaams Interuniversitair Instituut Voor Biotechnologie Vzw Functional heavy chain antibodies, fragments thereof, library thereof and methods of production thereof
US9156905B2 (en) 2001-10-24 2015-10-13 Vib Vzw Functional heavy chain antibodies, fragments thereof, library thereof and methods of production thereof
EP1517921A2 (en) 2002-06-28 2005-03-30 Domantis Limited Immunoglobulin single variable antigen-binding domains and dual-specific constructs thereof
US7763244B2 (en) 2002-07-01 2010-07-27 Human Genome Sciences, Inc. Antibodies that specifically bind to Reg IV
US7771724B2 (en) 2002-08-07 2010-08-10 Ablynx N.V. Modulation of platelet adhesion based on the surface-exposed beta-switch loop of platelet glycoprotein IB-alpha
US20120202977A1 (en) * 2002-11-08 2012-08-09 Ablynx N.V. Single domain antibodies directed against tumor necrosis factor-alpha and uses therefor
US9243065B2 (en) 2002-11-08 2016-01-26 Ablynx N.V. Polypeptide constructs including VHH directed against EGFR for intracellular delivery
US20170096481A1 (en) * 2002-11-08 2017-04-06 Ablynx N.V. Single domain antibodies directed against tumor necrosis factor-alpha and uses therefor
US9725522B2 (en) 2002-11-08 2017-08-08 Ablynx N.V. Pulmonary administration of immunoglobulin single variable domains and constructs thereof
EP1558650A2 (en) * 2002-11-08 2005-08-03 Ablynx N.V. Camelidae antibodies against immunoglobulin e and use thereof for the treatment of allergic disorders
US9371381B2 (en) 2002-11-08 2016-06-21 Ablynx, N.V. Single domain antibodies directed against tumor necrosis factor-alpha and uses therefor
US9320792B2 (en) 2002-11-08 2016-04-26 Ablynx N.V. Pulmonary administration of immunoglobulin single variable domains and constructs thereof
US10703818B2 (en) 2002-11-15 2020-07-07 Genmab A/S Human monoclonal antibodies against CD25
US20150259424A1 (en) * 2002-11-15 2015-09-17 Genmab A/S Human monoclonal antibodies against cd25
US11008386B2 (en) 2002-12-24 2021-05-18 Rinat Neuroscience Corp. Anti-NGF antibodies and methods using same
US8088384B2 (en) * 2002-12-24 2012-01-03 Rinat Neuroscience Corp. Anti-NGF antibodies and methods using same
US10323086B2 (en) 2002-12-24 2019-06-18 Rinat Neuroscience Corp. Methods for treating osteoarthritis pain by administering a nerve growth factor antagonist and compositions containing the same
US11034755B2 (en) 2003-01-10 2021-06-15 Ablynx N.V. Polypeptides and polypeptide constructs comprising single domain antibodies directed against von willebrand factor
US10112989B2 (en) 2003-01-10 2018-10-30 Ablynx, N.V. Polypeptides and polypeptide constructs comprising single domain antibodies directed against von Willebrand factor
US9028816B2 (en) 2003-01-10 2015-05-12 Ablynx N.V. Polypeptides and polypeptide constructs comprising single domain antibodies directed against von Willebrand factor
US10464976B2 (en) 2003-01-31 2019-11-05 AbbVie Deutschland GmbH & Co. KG Amyloid β(1-42) oligomers, derivatives thereof and antibodies thereto, methods of preparation thereof and use thereof
US8603473B2 (en) 2003-06-27 2013-12-10 Biogen Idec Ma Inc. Modified binding molecules comprising connecting peptides
WO2004081026A3 (en) * 2003-06-30 2005-01-27 Domantis Ltd Polypeptides
WO2005035572A2 (en) * 2003-10-08 2005-04-21 Domantis Limited Antibody compositions and methods
WO2005035572A3 (en) * 2003-10-08 2007-01-04 Domantis Ltd Antibody compositions and methods
US8163552B2 (en) * 2003-11-14 2012-04-24 Patrys Limited Adenocarcinoma specific antibody SAM-6, and uses thereof
US8263746B2 (en) 2004-02-06 2012-09-11 Morphosys Ag Anti-CD38 human antibodies and uses thereof
US9758590B2 (en) 2004-02-06 2017-09-12 Morphosys Ag Anti-CD38 human antibodies and uses thereof
US9200061B2 (en) 2004-02-06 2015-12-01 Morpho Sys AG Generation and profiling of fully human HuCAL gold®-derived therapeutic antibodies specific for human CD3i
US20110237779A1 (en) * 2004-02-19 2011-09-29 Genentech, Inc. Cdr-repaired antibodies
US8580928B2 (en) * 2004-02-19 2013-11-12 Genetech, Inc. CDR-repaired antibodies
EP2740743A2 (en) 2004-06-01 2014-06-11 Domantis Limited Bispecific fusion antibodies with enhanced serum half-life
JP2012135311A (en) * 2004-06-01 2012-07-19 Domantis Ltd Bispecific domain antibody with increased serum half life
WO2005118642A3 (en) * 2004-06-01 2006-03-23 Domantis Ltd Bispecific fusion antibodies with enhanced serum half-life
AU2005250216B2 (en) * 2004-06-01 2009-12-10 Domantis Limited Bispecific fusion antibodies with enhanced serum half-life
EP2740743A3 (en) * 2004-06-01 2015-08-19 Domantis Limited Bispecific fusion antibodies with enhanced serum half-life
WO2005118642A2 (en) * 2004-06-01 2005-12-15 Domantis Limited Bispecific fusion antibodies with enhanced serum half-life
EA012622B1 (en) * 2004-06-01 2009-10-30 Домэнтис Лимитед Bispecific fusion antibodies with enhanced serum half-life
US8921528B2 (en) 2004-06-01 2014-12-30 Domantis Limited Bispecific fusion antibodies with enhanced serum half-life
JP2008500830A (en) * 2004-06-01 2008-01-17 ドマンティス リミテッド Bispecific fusion antibodies with increased serum half-life
US8029790B2 (en) * 2004-06-29 2011-10-04 B.R.A.H.M.S. Gmbh Monoclonal thyroid stimulating or blocking antibodies, peptide sequences corresponding to their variable regions, and their uses in diagnostic, preventive and therapeutic medicine
EP1777235A1 (en) * 2004-06-30 2007-04-25 Domantis Limited Compositions and methods for treating inflammatory disorders
JP2008504356A (en) * 2004-06-30 2008-02-14 ドマンティス リミテッド Compositions and methods for treating inflammatory diseases
AU2010201856B2 (en) * 2004-06-30 2012-10-18 Cephalon Australia Pty Limited Compositions and methods for treating inflammatory disorders
EP2322554A1 (en) * 2004-06-30 2011-05-18 Domantis Limited Composition comprising an anti-TNF-alpha domain antibody for the treatment of rheumatoid arthritis
JP2013018785A (en) * 2004-06-30 2013-01-31 Domantis Ltd Compositions and methods for treating inflammatory disorders
WO2006003388A3 (en) * 2004-06-30 2006-04-27 Domantis Ltd Compositions and methods for treating inflammatory disorders
JP2013056890A (en) * 2004-06-30 2013-03-28 Domantis Ltd Composition and method for treating inflammatory disorder
EP2261672B1 (en) * 2004-07-14 2014-09-03 The Regents of The University of California Biomarkers for early detection of ovarian cancer
US8921524B2 (en) 2004-07-22 2014-12-30 Erasmus University Medical Centre Binding molecules
US9353179B2 (en) 2004-07-22 2016-05-31 Erasmus University Medical Centre Binding molecules
US8921522B2 (en) 2004-07-22 2014-12-30 Erasmus University Medical Centre Binding molecules
US10906970B2 (en) 2004-07-22 2021-02-02 Erasmus University Medical Centre Methods of making heavy chain only antibodies using transgenic animals
US20100216974A1 (en) * 2004-07-22 2010-08-26 Franklin Gerardus Grosveld Binding molecules
US20100197897A1 (en) * 2004-07-22 2010-08-05 Franklin Gerardus Grosveld Binding molecules
US9346877B2 (en) 2004-07-22 2016-05-24 Erasmus University Medical Centre Binding molecules
US8647625B2 (en) 2004-07-26 2014-02-11 Biogen Idec Ma Inc. Anti-CD154 antibodies
US8961976B2 (en) 2004-07-26 2015-02-24 Biogen Idec Ma Inc. Anti-CD154 antibodies
EP2371862A2 (en) 2004-09-17 2011-10-05 Domantis Limited Compositions monovalent for CD40L binding and methods of use
EP2371862A3 (en) * 2004-09-17 2012-10-24 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
JP2008513425A (en) * 2004-09-17 2008-05-01 ドマンティス リミテッド Monovalent composition 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
WO2006030220A1 (en) * 2004-09-17 2006-03-23 Domantis Limited Compositions monovalent for cd40l binding and methods of use
US7563443B2 (en) 2004-09-17 2009-07-21 Domantis Limited Monovalent anti-CD40L antibody polypeptides and compositions thereof
US7829096B2 (en) 2004-09-17 2010-11-09 Domantis Ltd. CD40L-specific monovalent polypeptides
EP2371390A2 (en) 2004-10-08 2011-10-05 Domantis Limited Antagonists and methods of use therefor
EP2371390A3 (en) * 2004-10-08 2013-03-13 Domantis Limited Antagonists and methods of use therefor
WO2006038027A3 (en) * 2004-10-08 2007-01-04 Domantis Ltd SINGLE DOMAIN ANTIBODIES AGAINST TNFRl AND METHODS OF USE THEREFOR
WO2006038027A2 (en) 2004-10-08 2006-04-13 Domantis Limited SINGLE DOMAIN ANTIBODIES AGAINST TNFRl AND METHODS OF USE THEREFOR
AU2005291017B2 (en) * 2004-10-08 2011-09-15 Domantis Limited Single domain antibodies against TNFRl and methods of use therefor
US8551488B2 (en) * 2004-10-12 2013-10-08 Crucell Holland B.V. Binding molecules for treatment and detection of cancer
US20100297153A1 (en) * 2004-10-12 2010-11-25 Crucell Holland B.V. Binding molecules for treatment and detection of cancer
EP2420251A3 (en) * 2004-11-10 2013-03-13 Domantis Limited Ligands that enhance endogenous compounds
EP2420251A2 (en) 2004-11-10 2012-02-22 Domantis Limited Ligands that enhance endogenous compounds
WO2006051288A3 (en) * 2004-11-10 2006-08-17 Domantis Ltd Ligands that enhance endogenous compounds
WO2006051288A2 (en) * 2004-11-10 2006-05-18 Domantis Limited Ligands that enhance endogenous compounds
US8105592B2 (en) * 2004-11-25 2012-01-31 Vhsquared Limited Heavy chain and single domain antibodies
JP2008521869A (en) * 2004-12-02 2008-06-26 ドマンティス リミテッド Bispecific antibodies targeting serum albumin and GLP-1 or PYY
AU2005311099B2 (en) * 2004-12-02 2012-02-02 Domantis Limited Bispecific domain antibodies targeting serum albumin and GLP-1 or PYY
WO2006059110A2 (en) * 2004-12-02 2006-06-08 Domantis Limited Plad domain peptides with increased serum half life due to conjugation to domain antibodies
JP2008521426A (en) * 2004-12-02 2008-06-26 ドマンティス リミテッド PLAD domain peptide with increased serum half-life by conjugation to domain antibody
WO2006059106A2 (en) * 2004-12-02 2006-06-08 Domantis Limited Bispecific domain antibodies targeting serum albumin and glp-1 or pyy
WO2006059108A2 (en) * 2004-12-02 2006-06-08 Domantis Limited ANTI-IL-IRl SINGLE DOMAIN ANTIBODIES AND THERAPEUTIC USES
EP2769990A2 (en) 2004-12-02 2014-08-27 Domantis Limited Bispecific domain antibodies targeting serum albumin and GLP-1 or PYY
WO2006059110A3 (en) * 2004-12-02 2007-03-15 Domantis Ltd Plad domain peptides with increased serum half life due to conjugation to domain antibodies
EP2769990A3 (en) * 2004-12-02 2015-02-25 Domantis Limited Bispecific domain antibodies targeting serum albumin and GLP-1 or PYY
WO2006059108A3 (en) * 2004-12-02 2007-02-22 Domantis Ltd ANTI-IL-IRl SINGLE DOMAIN ANTIBODIES AND THERAPEUTIC USES
WO2006059106A3 (en) * 2004-12-02 2007-01-04 Domantis Ltd Bispecific domain antibodies targeting serum albumin and glp-1 or pyy
EP1824884B1 (en) 2004-12-16 2015-05-13 Centre National De La Recherche Scientifique (Cnrs) Production of antibody formats and immunological applications of said formats
US8993319B2 (en) 2004-12-28 2015-03-31 Innate Pharma S.A. Monoclonal antibodies against NKG2A
US10160810B2 (en) 2004-12-28 2018-12-25 Innate Pharma, S.A. Monoclonal antibodies against NKG2A
US8021661B2 (en) * 2005-01-05 2011-09-20 Biogen Idec Ma Inc. Cripto binding molecules
US10253095B2 (en) 2005-01-06 2019-04-09 Innate Pharma S.A.S. Anti-KIR combination treatments and methods
US7939277B2 (en) 2005-01-14 2011-05-10 Umc Utrecht Holding Bv Methods and assays for distinguishing between different forms of diseases and disorders characterized by thrombocytopenia and/or by spontaneous interaction between Von Willebrand Factor (vWF) and platelets
US20100278842A1 (en) * 2005-01-31 2010-11-04 Genentech, Inc. Anti-ephb2 antibodies and methods using same
WO2006089133A2 (en) 2005-02-15 2006-08-24 Duke University Anti-cd19 antibodies and uses in oncology
EP2548575A1 (en) 2005-02-15 2013-01-23 Duke University Anti-CD19 antibodies that mediate ADCC for use in treating autoimmune diseases
US7521425B2 (en) 2005-03-03 2009-04-21 Covx Technologies Ireland Limited Anti-angiogenic compounds
EP2292248A2 (en) 2005-03-03 2011-03-09 CovX Technologies Ireland Limited Anti-angiogenic compounds
US8092800B2 (en) 2005-03-18 2012-01-10 Istituto Superiore Di Sanita Antibodies against Candida antigens
WO2006097689A2 (en) * 2005-03-18 2006-09-21 Domantis Limited Antibodies against candida antigens
WO2006097689A3 (en) * 2005-03-18 2007-03-15 Domantis Ltd Antibodies against candida antigens
JP2008533115A (en) * 2005-03-18 2008-08-21 ドマンティス リミテッド Antibodies against Candida antigen
US8759490B2 (en) 2005-03-24 2014-06-24 Millennium Pharamaceuticals, Inc. Antibodies that bind OV064 and methods of use therefor
US8323645B2 (en) * 2005-03-24 2012-12-04 Millennium Pharmaceuticals, Inc. Antibodies that bind OV064 and methods of use therefor
US9421277B2 (en) 2005-03-24 2016-08-23 Millennium Pharmaceuticals, Inc. Antibodies that bind OV064 and methods of use therefor
US9926378B2 (en) 2005-03-24 2018-03-27 Millennium Pharmaceuticals, Inc. Antibodies that bind OV064 and methods of use therefor
US10150807B2 (en) 2005-03-25 2018-12-11 National Research Council Of Canada Method for isolation of soluble polypeptides
US8293233B2 (en) * 2005-03-25 2012-10-23 National Research Council Of Canada Method for isolation of soluble polypeptides
US11091536B2 (en) 2005-03-25 2021-08-17 National Research Council Of Canada Method for isolation of soluble polypeptides
US20110158902A1 (en) * 2005-04-15 2011-06-30 Neogenix Oncology, Inc. Recombinant monoclonal antibodies and corresponding antigens for colon and pancreatic cancers
US10364295B2 (en) 2005-04-15 2019-07-30 Precision Biologics, Inc. Recombinant monoclonal antibodies and corresponding antigens for colon and pancreatic cancers
US11572413B2 (en) 2005-04-15 2023-02-07 Precision Biologics, Inc. Recombinant monoclonal antibodies and corresponding antigens for colon and pancreatic cancers
US9169326B2 (en) * 2005-04-15 2015-10-27 Precision Biologics, Inc. Recombinant monoclonal antibodies and corresponding antigens for colon and pancreatic cancers
US9605077B2 (en) 2005-04-15 2017-03-28 Precision Biologics, Inc. Recombinant monoclonal antibodies and corresponding antigens for colon and pancreatic cancers
US7928214B2 (en) * 2005-04-26 2011-04-19 Agouron Pharmaceuticals, Inc. P-cadherin antibodies
EP2221316A1 (en) 2005-05-05 2010-08-25 Duke University Anti-CD19 antibody therapy for autoimmune disease
EP2365000A2 (en) 2005-05-18 2011-09-14 Ablynx N.V. Improved nanobodiesTM against tumor necrosis factor-alpha
EP2949668A1 (en) 2005-05-18 2015-12-02 Ablynx N.V. Improved nanobodiestm against tumor necrosis factor-alpha
CN103254309A (en) * 2005-05-18 2013-08-21 埃博灵克斯股份有限公司 Improved nanobodies TM against tumor necrosis factor-alpha
NO345340B1 (en) * 2005-05-18 2020-12-21 Ablynx Nv Amino acid sequence, fusion protein or construct containing this amino acid sequence, as well as a method for producing the amino acid sequence.
EP3613767A1 (en) 2005-05-18 2020-02-26 Ablynx N.V. Improved nanobodiestm against tumor cecrosis factor-alpha
JP2017186318A (en) * 2005-05-18 2017-10-12 アブリンクス エン.ヴェー. Improved nanobodies (tm) against tumor necrosis factor-alpha
JP2015157850A (en) * 2005-05-18 2015-09-03 アブリンクス エン.ヴェー. Improved nanobodies (tm) against tumor necrosis factor-alpha
US8188223B2 (en) 2005-05-18 2012-05-29 Ablynx N.V. Serum albumin binding proteins
US9067991B2 (en) 2005-05-18 2015-06-30 Ablynx N.V. Nanobodies against tumor necrosis factor-alpha
EP2479191A2 (en) 2005-05-18 2012-07-25 Ablynx N.V. Improved nanobodiesTM against tumor necrosis factor-alpha
US11472871B2 (en) 2005-05-18 2022-10-18 Ablynx N.V. Nanobodies against tumor necrosis factor-alpha
WO2006122787A1 (en) * 2005-05-18 2006-11-23 Ablynx Nv Serum albumin binding proteins
JP2012167112A (en) * 2005-05-18 2012-09-06 Ablynx Nv IMPROVED NANOBODY (R) AGAINST TUMOR NECROSIS FACTOR-α
EP2365000A3 (en) * 2005-05-18 2013-01-16 Ablynx N.V. Improved nanobodiesTM against tumor necrosis factor-alpha
EP2172484A3 (en) * 2005-05-18 2010-05-19 Ablynx N.V. Serum albumin binding proteins
CN103254309B (en) * 2005-05-18 2017-09-26 埃博灵克斯股份有限公司 For the improved nanometer body of tumor necrosis factor αTM
US8372398B2 (en) 2005-05-20 2013-02-12 Ablynx N.V. Single domain VHH antibodies against Von Willebrand Factor
US7807162B2 (en) 2005-05-20 2010-10-05 Ablynx N.V. Single domain VHH antibodies against von Willebrand factor
US8703131B2 (en) 2005-05-21 2014-04-22 Ablynx N.V. Nanobodies against tumor necrosis factor-alpha
US7875465B2 (en) 2005-05-31 2011-01-25 Canon Kabushiki Kaisha Target substance capturing molecule
WO2006129843A3 (en) * 2005-05-31 2007-04-19 Canon Kk Bispecific capturing molecule
JP2007008925A (en) * 2005-05-31 2007-01-18 Canon Inc Target substance-catching molecule
WO2006129843A2 (en) * 2005-05-31 2006-12-07 Canon Kabushiki Kaisha Bispecific capturing molecule
US7989219B2 (en) 2005-05-31 2011-08-02 Canon Kabushiki Kaisha Bispecific capturing molecule
US9777066B2 (en) 2005-06-10 2017-10-03 Chugai Seiyaku Kabushiki Kaisha Pharmaceutical compositions containing sc(Fv)2
US10307481B2 (en) 2005-07-25 2019-06-04 Aptevo Research And Development Llc CD37 immunotherapeutics and uses thereof
US10143748B2 (en) 2005-07-25 2018-12-04 Aptevo Research And Development Llc B-cell reduction using CD37-specific and CD20-specific binding molecules
EP1945669A4 (en) * 2005-08-15 2009-07-22 Arana Therapeutics Ltd Chimeric antibodies with new world primate regions
EP1945669A1 (en) * 2005-08-15 2008-07-23 Arana Therapeutics Limited Chimeric antibodies with new world primate regions
US8361462B2 (en) * 2005-09-01 2013-01-29 National Research Council Of Canada Anti-apoptotic protein antibodies
US20090324579A1 (en) * 2005-09-01 2009-12-31 Siyaram Pandey Anti-apoptotic protein antibodies
US11939395B2 (en) 2005-10-12 2024-03-26 Morphosys Ag Generation and profiling of fully human HuCAL gold-derived therapeutic antibodies specific for human CD38
US8486394B2 (en) 2005-10-12 2013-07-16 Morphosys Ag Generation and Profiling of fully human hucal gold-derived therapeutic antibodies specific for human CD38
US9193799B2 (en) 2005-10-12 2015-11-24 Morphosys Ag Fully human therapeutic antibodies specific for human CD38
US8088896B2 (en) * 2005-10-12 2012-01-03 Morphosys Ag Generation and profiling of fully human gold-derived therapeutic antibodies specific for human CD38
US11059902B2 (en) 2005-10-12 2021-07-13 Morphosys Ag Generation and profiling of fully human HuCAL GOLD-derived therapeutic antibodies specific for human CD38
US10184005B2 (en) 2005-10-12 2019-01-22 Morphosys Ag Generation and profiling of fully human HuCAL GOLD-derived therapeutic antibodies specific for human CD38
US8129503B2 (en) 2005-10-24 2012-03-06 Domantis Limited Agents that bind a target in pulmonary tissue for targeting respiratory diseases
AU2006307733B2 (en) * 2005-10-24 2012-11-22 Domantis Limited Agents that bind a target in pulmonary tissue for treating respiratory diseases
WO2007049017A3 (en) * 2005-10-24 2008-01-31 Domantis Ltd Agents that bind a target in pulmonary tissue for treating respiratory diseases
US9629909B2 (en) 2005-10-24 2017-04-25 Domantis Limited Methods for targeting pulmonary diseases with agents that bind a target in pulmonary tissue
WO2007049017A2 (en) * 2005-10-24 2007-05-03 Domantis Limited Agents that bind a target in pulmonary tissue for treating respiratory diseases
JP2009512672A (en) * 2005-10-24 2009-03-26 ドマンティス リミテッド Drugs that bind to targets in lung tissue to treat respiratory diseases
US8497244B2 (en) 2005-10-24 2013-07-30 Domantis Limited Methods for targeting pulmonary diseases with agents that bind a target in pulmonary tissue
EA014106B1 (en) * 2005-10-24 2010-10-29 Домантис Лимитед Agents that bind a target in pulmonary tissue for treating respiratory diseases
US8679490B2 (en) * 2005-11-07 2014-03-25 Genentech, Inc. Binding polypeptides with diversified and consensus VH/VL hypervariable sequences
US8734802B1 (en) 2005-11-14 2014-05-27 Labrys Biologics, Inc. Methods of treating vasomotor symptoms using antibodies
EP2366715A2 (en) 2005-11-14 2011-09-21 Amgen Inc. Rankl Antibody-PTH/PTHRP Chimeric Molecules
US8007794B2 (en) * 2005-11-14 2011-08-30 Rinat Neuroscience Corporation Antagonist antibodies directed against calcitonin gene-related peptide and methods using same
US8597649B2 (en) 2005-11-14 2013-12-03 Labrys Biologics, Inc. Antagonist antibodies directed against calcitonin gene-related peptide and methods using same
US9890211B2 (en) 2005-11-14 2018-02-13 Teva Pharmaceuticals International Gmbh Antagonist antibodies directed against calcitonin gene-related peptide
US10329343B2 (en) 2005-11-14 2019-06-25 Teva Pharmaceuticals International Gmbh Methods for treating headache using antagonist antibodies directed against calcitonin gene-related peptide
US20150361173A1 (en) 2005-11-14 2015-12-17 Labrys Biologics, Inc. Antagonist antibodies directed against calcitonin gene-related peptide and methods using same
US9365648B1 (en) 2005-11-14 2016-06-14 Labrys Biologics, Inc. Methods of using anti-CGRP antagonist antibodies
US9884908B2 (en) 2005-11-14 2018-02-06 Teva Pharmaceuticals International Gmbh Methods for treating headache using antagonist antibodies directed against calcitonin gene-related peptide
EP2816060A1 (en) 2005-11-14 2014-12-24 Amgen Inc. Rankl antibody-PTH/PTHRP chimeric molecules
US9340614B2 (en) 2005-11-14 2016-05-17 Labrys Biologics, Inc. Antagonist antibodies directed against calcitonin gene-related peptide and methods using same
US9266951B2 (en) 2005-11-14 2016-02-23 Labrys Biologics, Inc. Antagonist antibodies directed against calcitonin gene-related peptide and methods using same
US9346881B2 (en) 2005-11-14 2016-05-24 Labrys Biologics, Inc. Antagonist antibodies directed against calcitonin gene-related peptide and methods using same
US9328168B2 (en) 2005-11-14 2016-05-03 Labrys Biologics, Inc. Methods of using anti-CGRP antagonist antibodies
US9890210B2 (en) 2005-11-14 2018-02-13 Teva Pharmaceuticals International Gmbh Antagonist antibodies directed against calcitonin gene-related peptide
US9884907B2 (en) 2005-11-14 2018-02-06 Teva Pharmaceuticals International Gmbh Methods for treating headache using antagonist antibodies directed against calcitonin gene-related peptide
US8586045B2 (en) 2005-11-14 2013-11-19 Labrys Biologics, Inc. Methods of using anti-CGRP antagonist antibodies
US9115194B2 (en) 2005-11-14 2015-08-25 Labrys Biologics, Inc. Antagonist antibodies directed against calcitonin gene-related peptide and methods using same
US10208109B2 (en) 2005-11-30 2019-02-19 Abbvie Inc. Monoclonal antibodies against amyloid beta protein and uses thereof
US10323084B2 (en) 2005-11-30 2019-06-18 Abbvie Inc. Monoclonal antibodies against amyloid beta protein and uses thereof
US10538581B2 (en) 2005-11-30 2020-01-21 Abbvie Inc. Anti-Aβ globulomer 4D10 antibodies
WO2007063308A3 (en) * 2005-12-01 2007-09-20 Domantis Ltd Noncompetitive domain antibody formats that bind interleukin 1 receptor type 1
WO2007063311A3 (en) * 2005-12-01 2007-09-20 Domantis Ltd Competitive domain antibody formats that bind interleukin 1 receptor type 1
JP2009518024A (en) * 2005-12-06 2009-05-07 ドマンティス リミテッド Ligands having binding specificity for EGFR and / or VEGF and methods of use thereof
JP2009518025A (en) * 2005-12-06 2009-05-07 ドマンティス リミテッド Bispecific ligands having binding specificity for cell surface targets and methods of use thereof
US9487578B2 (en) 2005-12-15 2016-11-08 Genentech, Inc. Methods and compositions for targeting polyubiquitin
US8603475B2 (en) 2005-12-15 2013-12-10 Genentech, Inc. Methods and compositions for targeting polyubiquitin
WO2007077001A2 (en) 2005-12-30 2007-07-12 Universidad Publica De Navarra Method and system of control of the converter of an electricity generation facility connected to an electricity network in the presence of voltage sags in said network
EP1806365A1 (en) 2006-01-05 2007-07-11 Boehringer Ingelheim International GmbH Antibody molecules specific for fibroblast activation protein and immunoconjugates containing them
JP2009523162A (en) * 2006-01-11 2009-06-18 ドマンティス リミテッド Ligands having binding specificity for VEGF and / or EGFR and methods of use thereof
JP2009523460A (en) * 2006-01-24 2009-06-25 ドマンティス リミテッド Ligand binding to IL-4 and / or IL-13
WO2007085815A2 (en) * 2006-01-24 2007-08-02 Domantis Limited Ligands that bind il-4 and/or il-13
WO2007085815A3 (en) * 2006-01-24 2007-11-15 Domantis Ltd Ligands that bind il-4 and/or il-13
US7846439B2 (en) 2006-02-01 2010-12-07 Cephalon Australia Pty Ltd Domain antibody construct
EP2540741A1 (en) 2006-03-06 2013-01-02 Aeres Biomedical Limited Humanized anti-CD22 antibodies and their use in treatment of oncology, transplantation and autoimmune disease
US10934344B2 (en) 2006-03-31 2021-03-02 Chugai Seiyaku Kabushiki Kaisha Methods of modifying antibodies for purification of bispecific antibodies
US9670269B2 (en) 2006-03-31 2017-06-06 Chugai Seiyaku Kabushiki Kaisha Methods of modifying antibodies for purification of bispecific antibodies
EP2375255A1 (en) 2006-06-06 2011-10-12 Oxford Biotherapeutics Ltd. Proteins
WO2007141280A2 (en) 2006-06-06 2007-12-13 Oxford Genome Sciences (Uk) Ltd Proteins
US8409577B2 (en) 2006-06-12 2013-04-02 Emergent Product Development Seattle, Llc Single chain multivalent binding proteins with effector function
US8206709B2 (en) * 2006-06-30 2012-06-26 Novo Nordisk A/S Anti-NKG2A antibodies and uses thereof
US9683041B2 (en) 2006-06-30 2017-06-20 Novo Nordisk A/S Anti-NKG2A antibodies and uses thereof
US8901283B2 (en) 2006-06-30 2014-12-02 Novo Nordisk A/S Anti-NKG2A antibodies and uses thereof
US8221754B2 (en) * 2006-07-12 2012-07-17 Gene Techno Science Co., Ltd. Antihuman α9 integrin antibody and use of the same
US7867493B2 (en) * 2006-08-18 2011-01-11 Novartis Ag PRLR-specific antibody and uses thereof
US11155631B2 (en) * 2006-08-18 2021-10-26 Armagen, Inc. Macromolecular compositions that cross the blood-brain barrier and methods of use thereof
US20150203586A1 (en) * 2006-08-18 2015-07-23 Armagen Technologies, Inc. Macromolecular compositions that cross the blood-brain barrier and methods of use thereof
US10144783B2 (en) 2006-08-18 2018-12-04 Armagen, Inc. Macromolecular compositions that cross the blood-brain barrier and methods of use thereof
US11008401B2 (en) 2006-08-30 2021-05-18 Genentech, Inc. Multispecific antibodies
US10118970B2 (en) 2006-08-30 2018-11-06 Genentech, Inc. Multispecific antibodies
US11851501B2 (en) 2006-08-30 2023-12-26 Genentech, Inc. Multispecific antibodies
WO2008030611A2 (en) 2006-09-05 2008-03-13 Medarex, Inc. Antibodies to bone morphogenic proteins and receptors therefor and methods for their use
US8388972B2 (en) * 2006-09-15 2013-03-05 Advanced Accelerator Applications S.A. Human anti-folate receptor alpha antibodies and antibody fragments for the radioimmunotherapy of ovarian carcinoma
US20100093977A1 (en) * 2006-09-29 2010-04-15 Ucb Pharma S.A. Altered Antibodies
US8507654B2 (en) * 2006-09-29 2013-08-13 Ucb Pharma S.A. Altered antibodies
EP2486941A1 (en) 2006-10-02 2012-08-15 Medarex, Inc. Human antibodies that bind CXCR4 and uses thereof
US11180564B2 (en) 2006-10-02 2021-11-23 Amgen K-A, Inc. IL-17 Receptor A antigen binding proteins
US11858999B2 (en) 2006-10-02 2024-01-02 Amgen K-A, Inc. IL-17 receptor A antigen binding proteins
WO2008054603A2 (en) 2006-10-02 2008-05-08 Amgen Inc. Il-17 receptor a antigen binding proteins
US10208122B2 (en) 2006-10-02 2019-02-19 Amgen K-A, Inc. IL-17 receptor A antigen binding proteins
US20140356355A1 (en) * 2006-10-02 2014-12-04 Amgen Inc. Il-17 receptor a antigen binding proteins
EP3165539A1 (en) 2006-10-02 2017-05-10 Kirin-Amgen, Inc. Il-17 receptor a antigen binding proteins
WO2008043821A1 (en) * 2006-10-11 2008-04-17 Ablynx N. V. Amino acid sequences that bind to serum proteins in a manner that is essentially independent of the ph, compounds comprising the same, and use thereof
US9017684B2 (en) * 2006-10-12 2015-04-28 Chugai Seiyaku Kabushiki Kaisha Diagnosis and treatment of cancer using anti-EREG antibody
US20090324491A1 (en) * 2006-10-12 2009-12-31 Forerunner Pharma Research Co., Ltd. Diagnosis and Treatment of Cancer Using Anti-EREG Antibody
US8236931B2 (en) 2006-10-30 2012-08-07 Glaxo Group Limited Prevention of aggregation of immunoglobulin light or heavy chains
US8227578B2 (en) * 2006-11-10 2012-07-24 Abe, Ikubo & Katayama Anti-human dlk-1 antibody showing anti-tumor activity in vivo
EP2368908A2 (en) 2006-11-10 2011-09-28 CovX Technologies Ireland Limited Anti- angiogenic compounds
US9951125B2 (en) 2006-11-30 2018-04-24 Abbvie Inc. Aβ conformer selective anti-Aβ globulomer monoclonal antibodies
WO2008070569A2 (en) 2006-12-01 2008-06-12 Medarex, Inc. Human antibodies that bind cd22 and uses thereof
WO2009054863A2 (en) 2006-12-13 2009-04-30 Medarex, Inc. Human antibodies that bind cd19 and uses thereof
WO2008074004A2 (en) 2006-12-14 2008-06-19 Medarex, Inc. Human antibodies that bind cd70 and uses thereof
US20110150905A1 (en) * 2006-12-14 2011-06-23 Regeneron Pharmaceuticals, Inc. Human antibodies to human delta like ligand 4
US11820815B2 (en) 2006-12-14 2023-11-21 Regeneron Pharmaceuticals, Inc. Human antibodies to human delta like ligand 4
US20140377781A1 (en) * 2006-12-14 2014-12-25 Chugai Seiyaku Kabushiki Kaisha Anti-Claudin 3 Monoclonal Antibody and Treatment and Diagnosis of Cancer Using the Same
EP2557090A2 (en) 2006-12-19 2013-02-13 Ablynx N.V. Amino acid sequences directed against GPCRs and polypeptides comprising the same for the treatment of GPCR-related diseases and disorders
EP2514767A1 (en) 2006-12-19 2012-10-24 Ablynx N.V. Amino acid sequences directed against a metalloproteinase from the ADAM family and polypeptides comprising the same for the treatment of ADAM-related diseases and disorders
US9090693B2 (en) 2007-01-25 2015-07-28 Dana-Farber Cancer Institute Use of anti-EGFR antibodies in treatment of EGFR mutant mediated disease
EP2559704A1 (en) * 2007-02-08 2013-02-20 Domantis Limited Antibody single variable domains against serum albumin
EA020464B9 (en) * 2007-02-08 2015-10-30 Домантис Лимитед Antibody single variable domains against serum albumin
EA020464B1 (en) * 2007-02-08 2014-11-28 Домантис Лимитед Antibody single variable domains against serum albumin
WO2008096158A3 (en) * 2007-02-08 2008-12-18 Domantis Ltd Antibody single variable domains against serum albumin
EP2559702A1 (en) * 2007-02-08 2013-02-20 Domantis Limited Antibody single variable domains against serum albumin
US8961966B2 (en) 2007-02-16 2015-02-24 Merrimack Pharmaceuticals, Inc. Antibodies against ERBB3 and uses thereof
US8691225B2 (en) * 2007-02-16 2014-04-08 Merrimack Pharmaceuticals, Inc. Antibodies against the ectodomain of ErbB3 and uses thereof
EP2441775A1 (en) 2007-02-26 2012-04-18 Oxford Biotherapeutics Ltd. Protein
EP2447719A1 (en) 2007-02-26 2012-05-02 Oxford Biotherapeutics Ltd. Proteins
US8283294B2 (en) * 2007-03-01 2012-10-09 Symphogen A/S Method for cloning cognate antibodies
US20100104573A1 (en) * 2007-03-22 2010-04-29 Ucb Parma S.A. Binding proteins, including antibodies, antibody derivatives and antibody fragments, that specifically bind cd154 and uses thereof
US8784823B2 (en) 2007-03-22 2014-07-22 Biogen Idec Ma Inc. Binding proteins, including antibodies, antibody derivatives and antibody fragments, that specifically bind CD154 and uses thereof
US9321840B2 (en) 2007-03-22 2016-04-26 Ucb Biopharma Sprl Binding proteins, including antibodies, antibody derivatives and antibody fragments, that specifically bind CD154 and uses thereof
US8293237B2 (en) * 2007-03-22 2012-10-23 Ucb Pharma, S.A. Binding proteins, including antibodies, antibody derivatives and antibody fragments, that specifically bind CD154 and uses thereof
US10214580B2 (en) 2007-03-27 2019-02-26 I2 Pharmaceuticals, Inc. Constructs and libraries comprising antibody surrogate light chain sequences
US8916159B2 (en) * 2007-04-02 2014-12-23 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Selenocysteine mediated hybrid antibody molecules
US20100104510A1 (en) * 2007-04-02 2010-04-29 The United States Of America, As Represented By The Secretary, Department Of Health And Human Selenocysteine mediated hybrid antibody molecules
EP2190875B1 (en) * 2007-04-23 2019-07-10 Sudhir Paul Catalytic Antibodies against HIV gp120
US10280211B2 (en) * 2007-05-04 2019-05-07 Technophage, Investigação E Desenvolvimento Em Biotecnologia, Sa Engineered rabbit antibody variable domains and uses thereof
US20100136627A1 (en) * 2007-05-04 2010-06-03 Technophage,Investigacao E Desenvolvimento Em Biotechnologia, Sa Engineered Rabbit Antibody Variable Domains and Uses Thereof
EP2703011A2 (en) 2007-05-07 2014-03-05 MedImmune, LLC Anti-icos antibodies and their use in treatment of oncology, transplantation and autoimmune disease
EP2737907A2 (en) 2007-05-07 2014-06-04 MedImmune, LLC Anti-icos antibodies and their use in treatment of oncology, transplantation and autoimmune disease
US8158124B2 (en) * 2007-05-30 2012-04-17 Lpath, Inc. Compositions and methods for binding lysophosphatidic acid
US9163091B2 (en) 2007-05-30 2015-10-20 Lpath, Inc. Compositions and methods for binding lysophosphatidic acid
WO2008153926A2 (en) 2007-06-05 2008-12-18 Yale University Inhibitors of receptor tyrosine kinases and methods of use thereof
US10072089B2 (en) 2007-06-06 2018-09-11 Domantis Limited Polypeptides, antibody variable domains and antagonists
JP2010528646A (en) * 2007-06-06 2010-08-26 ドマンティス リミテッド Polypeptides, antibody variable domains, and antagonists
JP2010529841A (en) * 2007-06-06 2010-09-02 ドマンティス リミテッド Polypeptides, antibody variable domains and antagonists
US8398979B2 (en) 2007-06-06 2013-03-19 Domantis Limited Polypeptides, antibody variable domains and antagonists
EP2746291A2 (en) 2007-06-06 2014-06-25 Domantis Limited Pulmonary formulation comprising an immunoglobulin single variable domain which binds to TNFR1
EP2746290A2 (en) 2007-06-06 2014-06-25 Domantis Limited Polypeptides, antibody variable domains and antagonists
JP2010530220A (en) * 2007-06-06 2010-09-09 ドマンティス リミテッド Polypeptides, antibody variable domains and antagonists
US8580254B2 (en) 2007-06-19 2013-11-12 Boehringer Ingelheim International Gmbh Anti-IGF antibodies
US9567400B2 (en) 2007-07-27 2017-02-14 Armagen Technologies, Inc. Methods and compositions for increasing α-L-iduronidase activity in the CNS
US11512145B2 (en) 2007-07-27 2022-11-29 Armagen, Inc. Methods and compositions for increasing alpha-L-iduronidase activity in the CNS
US10202467B2 (en) 2007-07-27 2019-02-12 Armagen, Inc. Methods and compositions for increasing α-L-iduronidase activity in the CNS
US9283276B2 (en) 2007-08-14 2016-03-15 Ludwig Institute For Cancer Research Ltd. Monoclonal antibody 175 targeting the EGF receptor and derivatives and uses thereof
EP2535350A1 (en) * 2007-09-26 2012-12-19 UCB Pharma S.A. Dual specificity antibody fusions
US8629246B2 (en) 2007-09-26 2014-01-14 Ucb Pharma S.A. Dual specificity antibody fusions
WO2009040562A1 (en) * 2007-09-26 2009-04-02 Ucb Pharma S.A. Dual specificity antibody fusions
US9309327B2 (en) 2007-09-26 2016-04-12 Ucb Pharma S.A. Dual specificity antibody fusions
EP2535349A1 (en) * 2007-09-26 2012-12-19 UCB Pharma S.A. Dual specificity antibody fusions
US9828438B2 (en) 2007-09-26 2017-11-28 Ucb Pharma S.A. Dual specificity antibody fusions
US10100130B2 (en) 2007-09-26 2018-10-16 Ucb Biopharma Sprl Dual specificity antibody fusions
US11427650B2 (en) 2007-09-26 2022-08-30 UCB Biopharma SRL Dual specificity antibody fusions
EP3192808A1 (en) 2007-11-27 2017-07-19 The University Of British Columbia 14-3-3 antagonists for the prevention and treatment of arthritis
EP3192807A1 (en) 2007-11-27 2017-07-19 The University Of British Columbia 14-3-3 eta antibodies and uses thereof for the diagnosis and treatment of arthritis
EP2215125A1 (en) * 2007-11-27 2010-08-11 Ablynx N.V. Method for obtaining polypeptide constructs comprising two or more single domain antibodies
US8975382B2 (en) 2007-11-27 2015-03-10 Ablynx N.V. Amino acid sequences directed against HER2 and polypeptides comprising the same for the treatment of cancers and/or tumors
EP2650311A2 (en) 2007-11-27 2013-10-16 Ablynx N.V. Amino acid sequences directed against heterodimeric cytokines and/or their receptors and polypeptides comprising the same
US9969805B2 (en) 2007-11-27 2018-05-15 Ablynx N.V. Amino acid sequences directed against HER2 and polypeptides comprising the same for the treatment of cancers and/or tumors
US9562090B2 (en) 2007-12-13 2017-02-07 Domantis Limited Polypeptides, antibody variable domains and antagonists
EP3239178A1 (en) 2007-12-14 2017-11-01 Bristol-Myers Squibb Company Binding molecules to the human ox40 receptor
EP2851374A1 (en) 2007-12-14 2015-03-25 Bristol-Myers Squibb Company Binding molecules to the human OX40 receptor
EP2594590A1 (en) 2007-12-14 2013-05-22 Bristol-Myers Squibb Company Binding molecules to the human OX40 receptor
US10927163B2 (en) 2007-12-21 2021-02-23 Hoffmann-La Roche, Inc. Bivalent, bispecific antibodies
US10138293B2 (en) 2007-12-21 2018-11-27 Hoffmann-La Roche, Inc. Bivalent, bispecific antibodies
US10287340B2 (en) 2008-01-07 2019-05-14 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Anti-HIV domain antibodies and method of making and using same
US20110104163A1 (en) * 2008-01-07 2011-05-05 The United States Of America, As Represented By Th Anti-hiv domain antibodies and method of making and using same
US9181327B2 (en) * 2008-01-07 2015-11-10 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Anti-HIV domain antibodies and method of making and using same
US9274119B2 (en) * 2008-01-11 2016-03-01 The University Of Tokyo Anti-CLDN6 antibody
US20110059469A1 (en) * 2008-01-11 2011-03-10 Hiroyuki Aburatani Anti-cldn6 antibody
US8642035B2 (en) * 2008-01-17 2014-02-04 Irm Llc Anti-TrkB antibodies
US9365642B2 (en) 2008-01-18 2016-06-14 Genentech, Inc. Methods and compositions for targeting polyubiquitin
US9081015B2 (en) 2008-01-18 2015-07-14 Genentech, Inc. Methods and compositions for targeting polyubiquitin
US8133488B2 (en) * 2008-01-18 2012-03-13 Genentech, Inc. Methods and compositions for targeting polyubiquitin
US10035849B2 (en) 2008-01-18 2018-07-31 Genentech, Inc. Methods and compositions for targeting polyubiquitin
US11753464B2 (en) 2008-01-18 2023-09-12 Genentech, Inc. Methods and compositions for targeting polyubiquitin
US10808028B2 (en) 2008-01-18 2020-10-20 Genentech, Inc. Methods and compositions for targeting polyubiquitin
US8796427B2 (en) 2008-01-24 2014-08-05 Novo Nordisk A/S Humanized anti-human NKG2A monoclonal antibody
US9422368B2 (en) 2008-01-24 2016-08-23 Novo Nordisk A/S Humanized anti-human NKG2A monoclonal antibody
US10323085B2 (en) 2008-03-04 2019-06-18 Teva Pharmaceuticals International Gmbh Methods of treating fibromyalgia
US9328167B2 (en) 2008-03-04 2016-05-03 Labrys Biologics, Inc. Methods of treating chronic pain
DE112009000507T5 (en) 2008-03-05 2011-02-10 Ablynx Nv Novel antigen-binding dimer complexes, process for their preparation and their use
US8017118B2 (en) * 2008-03-17 2011-09-13 LivTech Inc. — Teikyo University Biotechnology Research Center Anti-hDlk-1 antibody having an antitumor activity in vivo
US20110104256A1 (en) * 2008-03-25 2011-05-05 Yaolin Wang Methods for treating or preventing colorectal cancer
US9169318B2 (en) 2008-03-28 2015-10-27 Sea Lane Biotechnologies, Inc. Neutralizing molecules to viral antigens
US20140328836A1 (en) * 2008-04-02 2014-11-06 Macrogenics, Inc. HER2/neu-Specific Antibodies and Methods of Using Same
US10131713B2 (en) 2008-04-02 2018-11-20 Macrogenics, Inc. HER2/neu-specific antibodies and methods of using same
US11028183B2 (en) 2008-04-02 2021-06-08 Macrogenics, Inc. HER2/neu-specific antibodies and methods of using same
US9243069B2 (en) * 2008-04-02 2016-01-26 Macrogenics, Inc. HER2/neu-specific antibodies and methods of using the same
EP2947097A1 (en) 2008-04-07 2015-11-25 Ablynx N.V. Amino acid sequences directed against the Notch pathways and uses thereof
US9101609B2 (en) 2008-04-11 2015-08-11 Emergent Product Development Seattle, Llc CD37 immunotherapeutic and combination with bifunctional chemotherapeutic thereof
US8333966B2 (en) 2008-04-11 2012-12-18 Emergent Product Development Seattle, Llc CD37 immunotherapeutics and uses thereof
US8217140B2 (en) 2008-04-17 2012-07-10 Ablynx N.V. Peptides capable of binding to serum proteins and compounds, constructs and polypeptides comprising the same
US8168183B2 (en) * 2008-04-28 2012-05-01 Kalobios Pharmaceuticals, Inc. Antibodies to granulocyte-macrophage colony-stimulating factor
US9017674B2 (en) 2008-04-28 2015-04-28 Kalobios Pharmaceuticals, Inc. Antibodies to granulocyte-macrophage colony-stimulating factor
US9193780B2 (en) 2008-06-05 2015-11-24 Ablynx N.V. Amino acid sequences directed against envelope proteins of a virus and polypeptides comprising the same for the treatment of viral diseases
US11518799B2 (en) 2008-06-05 2022-12-06 Ablynx N.V. Amino acid sequences directed against envelope proteins of a virus and polypeptides comprising the same for the treatment of viral diseases
US9834595B2 (en) 2008-06-05 2017-12-05 Ablynx N.V. Amino acid sequences directed against envelope proteins of a virus and polypeptides comprising the same for the treatment of viral diseases
US10550174B2 (en) 2008-06-05 2020-02-04 Ablynx N.V. Amino acid sequences directed against envelope proteins of a virus and polypeptides comprising the same for the treatment of viral diseases
US20190077847A1 (en) 2008-06-05 2019-03-14 Ablynx N.V. Amino acid sequences directed against envelope proteins of a virus and polypeptides comprising the same for the treatment of viral diseases
US8906830B2 (en) 2008-06-18 2014-12-09 California Institute Of Technology Capture agents and related compositions, methods and systems
AU2009259987B2 (en) * 2008-06-18 2015-08-27 California Institute Of Technology Multi-ligand capture agents and related compositions, methods and systems
WO2009155420A1 (en) * 2008-06-18 2009-12-23 California Institute Of Technology Multi-ligand capture agents and related compositions, methods and systems
US9188584B2 (en) 2008-06-18 2015-11-17 California Institute Of Technology Capture agents and related compositions, methods and systems
US9315576B2 (en) 2008-07-02 2016-04-19 Argen-X N.V. Antigen binding polypeptides
US8444976B2 (en) * 2008-07-02 2013-05-21 Argen-X B.V. Antigen binding polypeptides
US20100330080A1 (en) * 2008-07-02 2010-12-30 Torsten Dreier Antigen binding polypeptides
US9428580B2 (en) 2008-07-02 2016-08-30 Argen-X B.V. Antigen binding polypeptides
US9346891B2 (en) 2008-07-02 2016-05-24 Argen-X.N.V. Antigen binding polypeptides
US8524231B2 (en) 2008-07-02 2013-09-03 Argen-X B.V. Antigen binding polypeptides
US9221918B2 (en) 2008-07-02 2015-12-29 Argen-X B.V. Antigen binding polypeptides
EP2700651A1 (en) 2008-07-18 2014-02-26 Bristol-Myers Squibb Company Compositions monovalent for CD28 binding and methods of use
EP3629022A1 (en) 2008-07-25 2020-04-01 Richard W. Wagner Protein screening methods
WO2010011944A2 (en) 2008-07-25 2010-01-28 Wagner Richard W Protein screeing methods
US8623592B2 (en) 2008-08-15 2014-01-07 Merrimack Pharmaceuticals, Inc. Methods and systems for predicting response of cells to a therapeutic agent
US20110158992A1 (en) * 2008-08-27 2011-06-30 Schering Corporation Engineered Anti-IL-23R Antibodies
US9017686B2 (en) 2008-09-03 2015-04-28 Genentech, Inc. Multispecific antibodies
US9522960B2 (en) 2008-09-03 2016-12-20 Genentech, Inc. Multispecific antibodies
US8193321B2 (en) 2008-09-03 2012-06-05 Genentech, Inc. Multispecific antibodies
US8337842B2 (en) * 2008-09-04 2012-12-25 Vet Therapeutics, Inc. Monoclonal antibodies
US20100061988A1 (en) * 2008-09-04 2010-03-11 Hansen Genevieve Monoclonal antibodies
US10407513B2 (en) 2008-09-26 2019-09-10 Ucb Biopharma Sprl Biological products
CN102164965B (en) * 2008-09-26 2016-03-30 Ucb医药有限公司 Biological product
US10981998B2 (en) * 2008-10-01 2021-04-20 Amgen Research (Munich) Gmbh Cross-species-specific single domain bispecific single chain antibody
US20110275787A1 (en) * 2008-10-01 2011-11-10 Microment AG Cross-species-specific single domain bispecific single chain antibody
US9073981B2 (en) 2008-10-13 2015-07-07 Institute For Research In Biomedicine Dengue virus neutralizing antibodies and use thereof
US20120020957A1 (en) * 2008-10-13 2012-01-26 Institute For Research In Biomedicine Dengue virus neutralizing antibodies and use thereof
US11370835B2 (en) 2008-10-29 2022-06-28 Ablynx N.V. Methods for purification of single domain antigen binding molecules
US9393304B2 (en) 2008-10-29 2016-07-19 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
US9993552B2 (en) 2008-10-29 2018-06-12 Ablynx N.V. Formulations of single domain antigen binding molecules
WO2010056804A1 (en) 2008-11-12 2010-05-20 Medimmune, Llc Antibody formulation
US11299538B2 (en) 2008-12-12 2022-04-12 Boehringer Ingelheim International Gmbh Anti-IGF antibodies
US10179810B2 (en) 2008-12-12 2019-01-15 Boehringer Ingelheim International Gmbh Anti-IGF antibodies
EP3064219A2 (en) 2008-12-12 2016-09-07 Boehringer Ingelheim International GmbH Anti-igf antibodies
US8318159B2 (en) 2008-12-12 2012-11-27 Boehringer Ingelheim International Gmbh Anti-IGF antibodies
WO2010084408A2 (en) 2009-01-21 2010-07-29 Oxford Biotherapeutics Ltd. Pta089 protein
WO2010085590A1 (en) 2009-01-23 2010-07-29 Biosynexus Incorporated Opsonic and protective antibodies specific for lipoteichoic acid gram positive bacteria
US20120034212A1 (en) * 2009-01-29 2012-02-09 Michael Bowen Human Anti-IL-6 Antibodies With Extended In Vivo Half-Life And Their Use In Treatment Of Oncology, Autoimmune Diseases And Inflammatory Diseases
US20110207917A1 (en) * 2009-02-09 2011-08-25 Patrys Limited Sam-6 variants, target and methods of use
US9072798B2 (en) 2009-02-18 2015-07-07 Ludwig Institute For Cancer Research Ltd. Specific binding proteins and uses thereof
EP3656788A2 (en) 2009-02-19 2020-05-27 Glaxo Group Limited Improved anti-serum albumin binding variants
US10696738B2 (en) 2009-02-19 2020-06-30 Glaxon Group Limited Anti-serum albumin binding variants
US9534043B2 (en) 2009-02-19 2017-01-03 Glaxo Group Limited Anti-serum albumin binding variants
US9751935B2 (en) 2009-02-19 2017-09-05 Glaxo Group Limited Anti-serum albumin binding variants
WO2010094720A2 (en) 2009-02-19 2010-08-26 Glaxo Group Limited Improved anti-tnfr1 polypeptides, antibody variable domains & antagonists
WO2010094722A2 (en) 2009-02-19 2010-08-26 Glaxo Group Limited Improved anti-serum albumin binding variants
WO2010097385A1 (en) 2009-02-24 2010-09-02 Glaxo Group Limited Antigen-binding constructs
WO2010100135A1 (en) 2009-03-05 2010-09-10 Ablynx N.V. Novel antigen binding dimer-complexes, methods of making/avoiding and uses thereof
WO2010102175A1 (en) 2009-03-05 2010-09-10 Medarex, Inc. Fully human antibodies specific to cadm1
US9533055B2 (en) 2009-03-18 2017-01-03 Armagen Technologies, Inc. Compositions and methods for blood-brain barrier delivery of IgG-decoy receptor fusion proteins
US8597652B2 (en) * 2009-03-20 2013-12-03 Genentech, Inc. Multispecific anti-HER antibodies
US9327035B2 (en) 2009-03-20 2016-05-03 Genentech, Inc. Anti-HER antibodies
US20100255010A1 (en) * 2009-03-20 2010-10-07 Genentech, Inc. Anti-her antibodies
US8569460B2 (en) 2009-03-25 2013-10-29 Vet Therapeutics, Inc. Antibody constant domain regions and uses thereof
EP3461844A2 (en) 2009-04-10 2019-04-03 Ablynx N.V. Improved amino acid sequences directed against il-6r and polypeptides comprising the same for the treatment of il-6r related diseases and disorders
EP3009454A2 (en) 2009-04-20 2016-04-20 Oxford Bio Therapeutics Limited Antibodies specific to cadherin-17
WO2010122090A1 (en) 2009-04-24 2010-10-28 Glaxo Group Limited Fgfr1c antibody combinations
US10017561B2 (en) 2009-05-13 2018-07-10 I2 Pharmaceuticals, Inc. Neutralizing molecules to influenza viruses
US10005832B2 (en) 2009-05-29 2018-06-26 Chugai Seiyaku Kabushiki Kaisha Method for treating a disease originated from receptor activation by EREG and TGFα
EP3205670A1 (en) 2009-06-05 2017-08-16 Ablynx N.V. Improved amino acid sequences directed against human respiratory syncytial virus (hrsv) and polypeptides comprising the same for the prevention and/or treatment of respiratory tract infections
US8945567B2 (en) 2009-06-05 2015-02-03 Ablynx N.V. Monovalent, bivalent and trivalent anti human respiratory syncytial virus (HRSV) nanobody constructs for the prevention and/or treatment of respiratory tract infections
WO2010139808A3 (en) * 2009-06-05 2011-02-24 Ablynx Nv Monovalent, bivalent and trivalent anti human respiratory syncytial virus (hrsv) nanobody constructs for the prevention and/or treatment of respiratory tract infections
US9803001B2 (en) 2009-06-05 2017-10-31 Ablynx N.V. Monovalent, bivalent and trivalent anti human respiratory syncytial virus (hRSV) nanobody constructs for the prevention and/or treatment of respiratory tract infections
US11028151B2 (en) 2009-06-05 2021-06-08 Ablynx N.V. Monovalent, bivalent and trivalent anti human respiratory syncytial virus (HRSV) nanobody constructs for the prevention and/or treatment of respiratory tract infections
US10640555B2 (en) 2009-06-16 2020-05-05 Hoffmann-La Roche Inc. Bispecific antigen binding proteins
US9676845B2 (en) 2009-06-16 2017-06-13 Hoffmann-La Roche, Inc. Bispecific antigen binding proteins
US11673945B2 (en) 2009-06-16 2023-06-13 Hoffmann-La Roche Inc. Bispecific antigen binding proteins
US20120164068A1 (en) * 2009-07-03 2012-06-28 Peter John Hudson Immuno-conjugates and methods for producing them
US9290577B2 (en) * 2009-07-03 2016-03-22 Avipep Pty Limited Immuno-conjugates and methods for producing them
WO2011006915A2 (en) 2009-07-16 2011-01-20 Glaxo Group Limited Improved anti-serum albumin binding single variable domains
WO2011006914A2 (en) 2009-07-16 2011-01-20 Glaxo Group Limited Antagonists, uses & methods for partially inhibiting tnfr1
WO2011012609A2 (en) 2009-07-29 2011-02-03 Glaxo Group Limited Ligands that bind tgf-beta receptor rii
US10597448B2 (en) 2009-08-28 2020-03-24 Teva Pharmaceuticals International Gmbh Methods for treating visceral pain associated with interstitial cystitis by administering antagonist antibodies directed against calcitonin gene-related peptide
US8623366B2 (en) 2009-08-28 2014-01-07 Labrys Biologics, Inc. Methods for treating visceral pain by administering antagonist antibodies directed against calcitonin gene-related peptide
US9469688B2 (en) 2009-08-29 2016-10-18 Abbvie Inc. Therapeutic DLL4 binding proteins
US9550973B2 (en) 2009-09-24 2017-01-24 Ucb Pharma, S.A. Bacterial host strain
US11028156B2 (en) 2009-10-09 2021-06-08 Armagen, Inc. Methods and compositions for increasing iduronate 2-sulfatase activity in the CNS
US10011651B2 (en) 2009-10-09 2018-07-03 Armagen, Inc. Methods and compositions for increasing iduronate 2-sulfatase activity in the CNS
WO2011046958A1 (en) 2009-10-12 2011-04-21 Amgen Inc. Use of il-17 receptor a antigen binding proteins
WO2011047083A1 (en) 2009-10-13 2011-04-21 Oxford Biotherapeutics Ltd. Antibodies against epha10
WO2011051217A1 (en) 2009-10-27 2011-05-05 Glaxo Group Limited Stable anti-tnfr1 polypeptides, antibody variable domains & antagonists
WO2011054007A1 (en) 2009-11-02 2011-05-05 Oxford Biotherapeutics Ltd. Ror1 as therapeutic and diagnostic target
WO2011054893A2 (en) 2009-11-05 2011-05-12 Novartis Ag Biomarkers predictive of progression of fibrosis
WO2011064382A1 (en) 2009-11-30 2011-06-03 Ablynx N.V. Improved amino acid sequences directed against human respiratory syncytial virus (hrsv) and polypeptides comprising the same for the prevention and/or treatment of respiratory tract infections
US9644022B2 (en) 2009-11-30 2017-05-09 Ablynx N.V. Amino acid sequences directed against human respiratory syncytial virus (HRSV) and polypeptides comprising the same for the prevention and/or treatment of respiratory tract infections
US11234419B2 (en) 2009-12-10 2022-02-01 Regeneran Pharmaceuticals, Inc. Mice that make heavy chain antibodies
EP3309176A1 (en) 2009-12-14 2018-04-18 Ablynx N.V. Immunoglobulin single variable domain antibodies against ox40l, constructs and therapeutic use
WO2011073180A1 (en) 2009-12-14 2011-06-23 Ablynx N.V. Single variable domain antibodies against ox40l, constructs and therapeutic use
US20120171115A1 (en) * 2009-12-23 2012-07-05 Avipep Pty Limited Immuno-conjugates and methods for producing them
US9315581B2 (en) * 2009-12-23 2016-04-19 A Vipep Pty Limited Immuno-conjugates and methods for producing them
WO2011083140A1 (en) 2010-01-08 2011-07-14 Ablynx Nv Immunoglobulin single variable domain directed against human cxcr4
US9493559B2 (en) 2010-01-14 2016-11-15 Ucb Pharma, S.A. Bacterial host strain expressing recombinant DsbC and having reduced Tsp activity
US9587227B2 (en) 2010-01-14 2017-03-07 Ucb Pharma, S.A. Bacterial host strain comprising a mutant spr gene and having reduced Tsp activity
US9493558B2 (en) 2010-01-14 2016-11-15 Ucb Pharma, S.A. Bacterial host strain comprising a mutant SPR gene and a wild-type TSP gene
US10072085B2 (en) 2010-01-15 2018-09-11 Kirin-Amgen, Inc. Method of treating psoriasis using an IL-17 receptor antibody formulation
US11505612B2 (en) 2010-01-15 2022-11-22 Amgen K-A, Inc. Method of treating diseases using an IL-17 receptor antibody formulation
US10808033B2 (en) 2010-01-15 2020-10-20 Amgen K-A, Inc. IL-17 receptor antibody formulation
WO2011089183A2 (en) 2010-01-20 2011-07-28 Boehringer Ingelheim International Gmbh Anticoagulant antidotes
EP3195876A1 (en) 2010-01-20 2017-07-26 Boehringer Ingelheim International Gmbh Anticoagulant antidotes
WO2011095545A1 (en) 2010-02-05 2011-08-11 Ablynx Nv Peptides capable of binding to serum albumin and compounds, constructs and polypeptides comprising the same
WO2011098520A1 (en) 2010-02-10 2011-08-18 Novartis Ag Agonist dr5 binding polypeptides
WO2011098449A1 (en) 2010-02-10 2011-08-18 Novartis Ag Methods and compounds for muscle growth
WO2011100403A1 (en) 2010-02-10 2011-08-18 Immunogen, Inc Cd20 antibodies and uses thereof
WO2011098518A2 (en) 2010-02-11 2011-08-18 Ablynx Nv Delivery of immunoglobulin variable domains and constructs thereof
US9469689B2 (en) 2010-03-02 2016-10-18 Abbvie Inc. Therapeutic DLL4 binding proteins
US9616120B2 (en) 2010-03-04 2017-04-11 Vet Therapeutics, Inc. Monoclonal antibodies directed to CD20
US8652470B2 (en) 2010-03-04 2014-02-18 Vet Therapeutics, Inc. Monoclonal antibodies directed to CD52
US8895001B2 (en) 2010-03-11 2014-11-25 Merrimack Pharmaceuticals, Inc. Use of ErbB3 inhibitors in the treatment of triple negative and basal-like breast cancers
US8992919B2 (en) 2010-04-15 2015-03-31 Genentech, Inc. Anti-polyubiquitin antibodies and methods of use
US11066463B2 (en) 2010-04-15 2021-07-20 Genentech, Inc. Anti-polyubiquitin antibodies and methods of use
US9822171B2 (en) 2010-04-15 2017-11-21 AbbVie Deutschland GmbH & Co. KG Amyloid-beta binding proteins
US9556262B2 (en) 2010-04-15 2017-01-31 Genentech, Inc. Anti-polyubiquitin antibodies and methods of use
US10100105B2 (en) 2010-04-15 2018-10-16 Genentech, Inc. Anti-polyubiquitin antibodies and methods of use
WO2011131659A2 (en) 2010-04-21 2011-10-27 Glaxo Group Limited Binding domains
EP3345926A1 (en) 2010-05-06 2018-07-11 Novartis AG Compositions and methods of use for therapeutic low density lipoprotein-related protein 6 (lrp6) antibodies
WO2011138391A1 (en) 2010-05-06 2011-11-10 Novartis Ag Compositions and methods of use for therapeutic low density lipoprotein - related protein 6 (lrp6) multivalent antibodies
EP4234698A2 (en) 2010-05-06 2023-08-30 Novartis AG Compositions and methods of use for therapeutic low density lipoprotein-related protein 6 (lrp6) antibodies
EP3546483A1 (en) 2010-05-20 2019-10-02 Ablynx N.V. Biological materials related to her3
WO2011144751A1 (en) 2010-05-20 2011-11-24 Glaxo Group Limited Improved anti-serum albumin binding variants
WO2011144749A1 (en) 2010-05-20 2011-11-24 Ablynx Nv Biological materials related to her3
US11345733B2 (en) 2010-06-22 2022-05-31 Precision Biologics, Inc. Colon and pancreas cancer specific antigens and antibodies
WO2011161263A1 (en) 2010-06-25 2011-12-29 Ablynx Nv Pharmaceutical compositions for cutaneous administration
US9751930B2 (en) 2010-07-27 2017-09-05 Ucb Pharma, S.A. Process for purifying proteins
US9493560B2 (en) 2010-08-03 2016-11-15 Abbvie Inc. Dual variable domain immunoglobulins and uses thereof
WO2012020096A1 (en) 2010-08-13 2012-02-16 Medimmune Limited Monomeric polypeptides comprising variant fc regions and methods of use
WO2012020143A1 (en) 2010-08-13 2012-02-16 Glaxo Group Limited Improved anti-serum albumin binding variants
US9062101B2 (en) 2010-08-14 2015-06-23 AbbVie Deutschland GmbH & Co. KG Amyloid-beta binding proteins
US10047121B2 (en) 2010-08-14 2018-08-14 AbbVie Deutschland GmbH & Co. KG Amyloid-beta binding proteins
WO2012022734A2 (en) 2010-08-16 2012-02-23 Medimmune Limited Anti-icam-1 antibodies and methods of use
US10562966B2 (en) 2010-08-20 2020-02-18 Ucb Biopharma Sprl Antibodies of the class IGG4
WO2012022814A1 (en) 2010-08-20 2012-02-23 Novartis Ag Antibodies for epidermal growth factor receptor 3 (her3)
WO2012022703A2 (en) 2010-08-20 2012-02-23 Glaxo Group Limited Improved anti-serum albumin binding variants
US9468678B2 (en) 2010-09-09 2016-10-18 Pfizer Inc. Method of producing 4-1BB binding molecules and associated nucleic acids
WO2012032433A1 (en) 2010-09-09 2012-03-15 Pfizer Inc. 4-1bb binding molecules
US8821867B2 (en) 2010-09-09 2014-09-02 Pfizer Inc 4-1BB binding molecules
US10640568B2 (en) 2010-09-09 2020-05-05 Pfizer Inc. 4-1BB binding molecules
US8337850B2 (en) 2010-09-09 2012-12-25 Pfizer Inc. 4-1BB binding molecules
EP3441404A1 (en) 2010-09-09 2019-02-13 Pfizer Inc 4-1bb binding molecules
EP3575321A1 (en) 2010-11-08 2019-12-04 Ablynx N.V. Cxcr2 binding polypeptides
WO2012062713A1 (en) 2010-11-08 2012-05-18 Novartis Ag Cxcr2 binding polypeptides
EP3578568A2 (en) 2010-11-08 2019-12-11 Ablynx N.V. Cxcr2 binding polypeptides
EP3514169A1 (en) 2010-11-26 2019-07-24 Molecular Partners AG Designed repeat proteins binding to serum albumin
WO2012069654A1 (en) 2010-11-26 2012-05-31 Molecular Partners Ag Designed repeat proteins binding to serum albumin
US20140112914A1 (en) * 2010-11-30 2014-04-24 Chugai Seiyaku Kabushiki Kaisha Cytotoxicity-inducing therapeutic agent
US11066483B2 (en) * 2010-11-30 2021-07-20 Chugai Seiyaku Kabushiki Kaisha Cytotoxicity-inducing therapeutic agent
WO2012072731A2 (en) 2010-12-01 2012-06-07 Glaxo Group Limited Improved anti-serum albumin binding single variable domains
US9512231B2 (en) * 2010-12-27 2016-12-06 Apo-T B.V. Cross-linking polypeptide that induces apoptosis
WO2012093125A1 (en) 2011-01-06 2012-07-12 Glaxo Group Limited Ligands that bind tgf-beta receptor ii
WO2012104227A1 (en) 2011-02-02 2012-08-09 Glaxo Group Limited Novel antigen binding proteins
WO2012109624A2 (en) 2011-02-11 2012-08-16 Zyngenia, Inc. Monovalent and multivalent multispecific complexes and uses thereof
US10793621B2 (en) 2011-02-28 2020-10-06 Hoffmann-La Roche Inc. Nucleic acid encoding dual Fc antigen binding proteins
US10611825B2 (en) 2011-02-28 2020-04-07 Hoffmann La-Roche Inc. Monovalent antigen binding proteins
US9982036B2 (en) 2011-02-28 2018-05-29 Hoffmann-La Roche Inc. Dual FC antigen binding proteins
US20140066599A2 (en) * 2011-03-25 2014-03-06 Glenmark Pharmaceuticals S.A. Hetero-Dimeric Immunoglobulins
US9683052B2 (en) 2011-03-25 2017-06-20 Glenmark Pharmaceuticals S.A. Hetero-dimeric immunoglobulins
US9683053B2 (en) 2011-03-25 2017-06-20 Glenmark Pharmaceuticals S.A. Hetero-dimeric immunoglobulins
WO2012130834A1 (en) 2011-03-30 2012-10-04 Boehringer Ingelheim International Gmbh Anticoagulant antidotes
US10961301B2 (en) 2011-04-01 2021-03-30 Yale University Cell-penetrating anti-DNA antibodies and uses thereof inhibit DNA repair
EP3363815A1 (en) 2011-05-05 2018-08-22 Merck Patent GmbH Amino acid sequences directed against il-17a, il-17f and/or il17-a/f and polypeptides comprising the same
EP4105231A1 (en) 2011-05-05 2022-12-21 Merck Patent GmbH Amino acid sequences directed against il-17a, il-17f and/or il17-a/f and polypeptides comprising the same
WO2012156219A1 (en) 2011-05-05 2012-11-22 Ablynx Nv Amino acid sequences directed against il-17a, il-17f and/or il17-a/f and polypeptides comprising the same
US11027018B2 (en) 2011-05-20 2021-06-08 The University Of Iowa Research Foundation Use of anti-CGRP antibodies and antibody fragments to treat diarrhea in subjects with diseases or treatments that result in elevated CGRP levels
US10208112B2 (en) 2011-05-20 2019-02-19 Alderbio Holdings Llc Anti-CGRP compositions and use thereof
US9745373B2 (en) 2011-05-20 2017-08-29 Alderbio Holdings Llc Anti-CGRP compositions and use thereof
US10189895B2 (en) 2011-05-20 2019-01-29 Alderbio Holdings Llc Anti-CGRP compositions and use thereof
US10214582B2 (en) 2011-05-20 2019-02-26 Alderbio Holdings Llc Anti-CGRP compositions and use thereof
US11325967B2 (en) 2011-05-20 2022-05-10 H. Lundbeck A/S Use of anti-CGRP antibodies and antibody fragments to prevent or inhibit photophobia or light a version in subjects in need thereof, especially migraine sufferers
US10066009B2 (en) 2011-05-20 2018-09-04 Alderbio Holdings Llc Anti-CGRP compositions and use thereof
US9855332B2 (en) 2011-05-20 2018-01-02 Alderbio Holdings Llc Use of anti-CGRP antibodies and antibody fragments to treat diarrhea in subjects with diseases or treatments that result in elevated CGRP levels
US9708393B2 (en) 2011-05-20 2017-07-18 Alderbio Holdings Llc Use of anti-CGRP antibodies and antibody fragments to prevent or inhibit photophobia or light aversion in subjects in need thereof, especially migraine sufferers
US10266587B2 (en) 2011-05-20 2019-04-23 Alderbio Holdings Llc Use of anti-CGRP antibodies and antibody fragments to prevent or inhibit photophobia or light aversion in subjects in need thereof, especially migraine sufferers
US10533048B2 (en) 2011-05-20 2020-01-14 Alderbio Holdings Llc Anti-CGRP compositions and use thereof
US10765746B2 (en) 2011-05-20 2020-09-08 Alderbio Holdings Llc Use of anti-CGRP antibodies and antibody fragments to treat diarrhea in subjects with diseases or treatments that result in elevated CGRP levels
US10179809B2 (en) 2011-05-20 2019-01-15 Alderbio Holdings Llc Anti-CGRP compositions and use thereof
US11111289B2 (en) 2011-05-20 2021-09-07 H. Lundbeck A/S Anti-CGRP compositions and use thereof
WO2012163887A1 (en) 2011-05-27 2012-12-06 Ablynx Nv Inhibition of bone resorption with rankl binding peptides
WO2012166906A1 (en) 2011-05-31 2012-12-06 Massachusetts Institute Of Technology Cell-directed synthesis of multifunctional nanopatterns and nanomaterials
US11697687B2 (en) 2011-06-17 2023-07-11 Novo Nordisk A/S Selective elimination of erosive cells
US9512228B2 (en) 2011-06-17 2016-12-06 Novo Nordisk A/S Selective elimination of erosive cells
WO2012175740A1 (en) 2011-06-23 2012-12-27 Ablynx Nv Immunoglobulin single variable domains directed against ige
WO2013001369A2 (en) 2011-06-28 2013-01-03 Oxford Biotherapeutics Ltd. Therapeutic and diagnostic target
WO2013003625A2 (en) 2011-06-28 2013-01-03 Oxford Biotherapeutics Ltd. Antibodies
US9725516B2 (en) 2011-07-13 2017-08-08 Ucb Pharma, S.A. Bacterial host strain expressing recombinant DSBC
US9957328B2 (en) 2011-07-13 2018-05-01 Ucb Pharma, S.A. Bacterial host strain expressing recombinant DSBC
WO2013016220A1 (en) 2011-07-22 2013-01-31 Amgen Inc. Il-17 receptor a is required for il-17c biology
US10300140B2 (en) 2011-07-28 2019-05-28 I2 Pharmaceuticals, Inc. Sur-binding proteins against ERBB3
US9321844B2 (en) 2011-08-05 2016-04-26 Genentech, Inc. Anti-polyubiquitin antibodies
US10738106B2 (en) 2011-08-05 2020-08-11 Genentech, Inc. Nucleic acids encoding anti-polyubiquitin antibodies
US11597761B2 (en) 2011-08-05 2023-03-07 Genentech Inc. Methods of detecting polyubiquitin using anti-polyubiquitin antibodies
AU2012296961B2 (en) * 2011-08-17 2017-02-16 Glaxo Group Limited Modified proteins and peptides
US10808040B2 (en) 2011-08-17 2020-10-20 Glaxo Group Limited Modified proteins and peptides
US20150132288A1 (en) * 2011-09-16 2015-05-14 Biocerox Products B.V. Anti-cd134 (ox40) antibodies and uses thereof
US9475880B2 (en) 2011-09-16 2016-10-25 Biocerox Products, B.V. Anti-CD134 (OX40) antibodies and uses thereof
WO2013038191A2 (en) 2011-09-16 2013-03-21 Bioceros B.V. Anti-cd134 (ox40) antibodies and uses thereof
EP3409689A1 (en) 2011-09-16 2018-12-05 BiocerOX Products B.V. Anti-cd134 (ox40) antibodies and uses thereof
WO2013043933A2 (en) 2011-09-22 2013-03-28 Amgen Inc. Cd27l antigen binding proteins
EP2758434A2 (en) * 2011-09-23 2014-07-30 Technophage, Investigação E Desenvolvimento Em Biotecnologia, SA Anti-tumor necrosis factor-alpha agents and uses thereof
EP3311837A1 (en) 2011-09-23 2018-04-25 Ablynx NV Prolonged inhibition of interleukin-6 mediated signaling
US11098115B2 (en) 2011-09-29 2021-08-24 Apo-T B.V. Multi-specific binding molecules targeting aberrant cells
US9421372B2 (en) 2011-09-30 2016-08-23 Adi Mashiach Head pain management device having an antenna
US9127056B2 (en) 2011-10-17 2015-09-08 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Monospecific and bispecific human monoclonal antibodies targeting insulin-like growth factor II (IGF-II)
WO2013060872A1 (en) 2011-10-27 2013-05-02 Boehringer Ingelheim International Gmbh Anticancer combination therapy
US10196442B2 (en) 2011-11-01 2019-02-05 Bionomics Inc. Methods of inhibiting growth of a colon cancer tumor in a subject by administering monoclonal antibodies to G protein-coupled receptor 49 (GPR49)
US10598653B2 (en) 2011-11-01 2020-03-24 Bionomics Inc. Methods of blocking cancer stem cell growth
EP3290442A1 (en) 2011-11-04 2018-03-07 Novartis AG Low density lipoprotein-related protein 6 (lrp6) half-life extender constructs
EP3252075A1 (en) 2011-11-04 2017-12-06 Novartis AG Low density lipoprotein-related protein 6 (lrp6) - half life extender constructs
WO2013067355A1 (en) 2011-11-04 2013-05-10 Novartis Ag Low density lipoprotein-related protein 6 (lrp6) - half life extender constructs
US9803004B2 (en) 2011-11-11 2017-10-31 Ucb Biopharma Sprl Albumin binding antibodies and binding fragments thereof
US10023631B2 (en) 2011-11-11 2018-07-17 Ucb Biopharma Sprl Albumin binding antibodies and binding fragments thereof
WO2013068571A1 (en) 2011-11-11 2013-05-16 Ucb Pharma S.A. Albumin binding antibodies and binding fragments thereof
US9975955B2 (en) 2011-12-02 2018-05-22 Armagen, Inc. Methods and compositions for increasing arylsulfatase A activity in the CNS
EP3590538A1 (en) 2011-12-05 2020-01-08 Novartis AG Antibodies for epidermal growth factor receptor 3 (her3)
WO2013084148A2 (en) 2011-12-05 2013-06-13 Novartis Ag Antibodies for epidermal growth factor receptor 3 (her3) directed to domain ii of her3
WO2013084147A2 (en) 2011-12-05 2013-06-13 Novartis Ag Antibodies for epidermal growth factor receptor 3 (her3)
US9975956B2 (en) 2011-12-22 2018-05-22 I2 Pharmaceuticals, Inc. Surrogate binding proteins which bind DR4 and/or DR5
US9556264B2 (en) 2011-12-28 2017-01-31 Chugai Seiyaku Kabushiki Kaisha Humanized anti-epiregulin antibody, and cancer therapeutic agent comprising said antibody as active ingredient
US10946104B2 (en) 2012-01-13 2021-03-16 Apo-Tb.V. Aberrant cell-restricted immunoglobulins provided with a toxic moiety
US10633451B2 (en) * 2012-02-03 2020-04-28 Hoffmann-La Roche Inc. Bispecific antibody molecules with antigen-transfected T-cells and their use in medicine
US20150010567A1 (en) * 2012-02-03 2015-01-08 Hoffmann-La Roche Inc. Bispecific antibody molecules with antigen-transfected t-cells and their use in medicine
US9688758B2 (en) * 2012-02-10 2017-06-27 Genentech, Inc. Single-chain antibodies and other heteromultimers
US20150133638A1 (en) * 2012-02-10 2015-05-14 Genentech, Inc. Single-chain antibodies and other heteromultimers
US20150018529A1 (en) * 2012-02-22 2015-01-15 Ucb Pharma S.A. Sequence Symmetric Modified IgG4 Bispecific Antibodies
US11059911B2 (en) 2012-02-22 2021-07-13 UCB Biopharma SRL Sequence symmetric modified IgG4 bispecific antibodies
US20150017169A1 (en) * 2012-02-22 2015-01-15 Ucb Pharma S.A. Sequence Asymmetric Modified IgG4 Bispecific Antibodies
US10221251B2 (en) 2012-02-22 2019-03-05 Ucb Biopharma Sprl Sequence symmetric modified IGG4 bispecific antibodies
US9902768B2 (en) * 2012-02-22 2018-02-27 Ucb Biopharma Sprl Sequence asymmetric modified IgG4 bispecific antibodies
US20150104443A1 (en) * 2012-05-08 2015-04-16 Chong Kun Dang Pharmaceutical Corp. Anti-erbb2 antibody variants
US9403912B2 (en) * 2012-05-08 2016-08-02 Chong Kun Dang Pharmaceutical Corp. Anti-ErbB2 antibody variants
WO2013168108A2 (en) 2012-05-09 2013-11-14 Novartis Ag Chemokine receptor binding polypeptides
US11261470B2 (en) 2012-05-14 2022-03-01 UCB Biopharma SRL Recombinant bacterial host cell for protein expression
US9951365B2 (en) 2012-05-14 2018-04-24 Ucb Biophara Sprl Recombinant bacterial host cell for protein expression
EP3693395A1 (en) 2012-05-18 2020-08-12 Amgen Inc. St2 antigen binding proteins
US10653792B2 (en) 2012-05-21 2020-05-19 Genentech, Inc. Anti-Ly6E antibodies and immunoconjugates and methods of use
US9724427B2 (en) * 2012-05-21 2017-08-08 Genentech, Inc. Anti-Ly6E antibodies and immunoconjugates and methods of use
US20160199508A1 (en) * 2012-05-21 2016-07-14 Genentech, Inc. ANTI-Ly6E ANTIBODIES AND IMMUNOCONJUGATES AND METHODS OF USE
US20150166670A1 (en) * 2012-05-24 2015-06-18 Hoffmann-La Roche Inc. Multispecific antibodies
US11891433B2 (en) 2012-05-31 2024-02-06 United States Of America As Represented By The Secretary Of The Air Force Camelidae single-domain antibodies against Yersinia pestis and methods of use
WO2014020331A1 (en) 2012-08-01 2014-02-06 Oxford Biotherapeutics Ltd. Therapeutic and diagnostic target
US20150203591A1 (en) * 2012-08-02 2015-07-23 Regeneron Pharmaceuticals, Inc. Mutivalent antigen-binding proteins
WO2014029752A1 (en) 2012-08-22 2014-02-27 Glaxo Group Limited Anti lrp6 antibodies
US9303086B2 (en) 2012-10-03 2016-04-05 Livtech, Inc. Anti-hDlk-1 antibody having an antitumor activity in vivo
US9944720B2 (en) 2012-11-01 2018-04-17 Abbvie Inc. Anti-DLL4/VEGF dual variable domain immunoglobulin and uses thereof
US11840553B2 (en) 2012-12-03 2023-12-12 Novimmune Sa Anti-CD47 antibodies and methods of use thereof
US11059910B2 (en) 2012-12-03 2021-07-13 Novimmune Sa Anti-CD47 antibodies and methods of use thereof
US9944719B2 (en) 2012-12-05 2018-04-17 Hoffman-La Roche Inc. Dual targeting
WO2014089335A2 (en) 2012-12-07 2014-06-12 Amgen Inc. Bcma antigen binding proteins
WO2014111550A1 (en) 2013-01-17 2014-07-24 Glaxosmithkline Intellectual Property Development Limited Modified anti-serum albumin binding proteins
US10059766B2 (en) * 2013-01-25 2018-08-28 Amgen Inc. Antibody constructs for CDH19 and CD3
US20150368343A1 (en) * 2013-01-25 2015-12-24 Amgen Research (Munich) Gmbh Antibody constructs for cdh19 and cd3
WO2014114801A1 (en) 2013-01-25 2014-07-31 Amgen Inc. Antibodies targeting cdh19 for melanoma
US11498964B2 (en) 2013-01-25 2022-11-15 Amgen Research (Munich) Gmbh Antibody constructs for CDH19 and CD3
WO2014120916A1 (en) 2013-02-01 2014-08-07 Bristol-Myers Squibb Company Pegylated domain antibodies monovalent for cd28 binding and methods of use
EP3736293A1 (en) 2013-02-12 2020-11-11 Boehringer Ingelheim International Gmbh Therapeutic and diagnostic target for cancer comprising dll3 binding reagents
WO2014133855A1 (en) 2013-02-28 2014-09-04 Caprion Proteomics Inc. Tuberculosis biomarkers and uses thereof
US10377828B2 (en) 2013-03-07 2019-08-13 Boehringer Ingelheim International Gmbh Combination therapy for neoplasia treatment
EP3611189A1 (en) 2013-03-14 2020-02-19 Novartis AG Antibodies against notch 3
WO2014159239A2 (en) 2013-03-14 2014-10-02 Novartis Ag Antibodies against notch 3
US10752694B2 (en) * 2013-03-15 2020-08-25 Amgen Inc. Binding molecules for BCMA and CD3
US20160176973A1 (en) * 2013-03-15 2016-06-23 Amgen Research (Munich) Gmbh Binding molecules for bcma and cd3
US20160115241A1 (en) * 2013-03-15 2016-04-28 Amgen Inc. Heterodimeric bispecific antibodies
US10993420B2 (en) 2013-03-15 2021-05-04 Erasmus University Medical Center Production of heavy chain only antibodies in transgenic mammals
US11634502B2 (en) * 2013-03-15 2023-04-25 Amgen Inc. Heterodimeric bispecific antibodies
US10221245B2 (en) 2013-03-16 2019-03-05 Novartis Ag Treatment of cancer using humanized anti-CD19 chimeric antigen receptor
US10927184B2 (en) 2013-03-16 2021-02-23 Novartis Ag Treatment of cancer using humanized anti-CD19 chimeric antigen receptor
WO2014148895A1 (en) 2013-03-18 2014-09-25 Biocerox Products B.V. Humanized anti-cd134 (ox40) antibodies and uses thereof
EP3409690A1 (en) 2013-03-18 2018-12-05 BiocerOX Products B.V. Humanized anti-cd134 (ox40) antibodies and uses thereof
US10273307B2 (en) 2013-03-18 2019-04-30 Biocerox Products B.V. Humanized anti-CD134 (OX40) antibodies and uses thereof
US9790281B2 (en) 2013-03-18 2017-10-17 Biocerox Products, B.V. Humanized anti-CD134 (OX40) antibodies and uses thereof
EP3971212A1 (en) 2013-05-30 2022-03-23 Kiniksa Pharmaceuticals, Ltd. Oncostatin m receptor antigen binding proteins
EP3456743A1 (en) 2013-05-30 2019-03-20 Kiniksa Pharmaceuticals, Ltd. Oncostatin m receptor antigen binding proteins
EP4349864A2 (en) 2013-05-30 2024-04-10 Kiniksa Pharmaceuticals, Ltd. Oncostatin m receptor antigen binding proteins
US10174117B2 (en) * 2013-06-11 2019-01-08 Inserm (Institut National De La Sante Et De La Recherche Medicale) Anti-HER2 single domain antibodies, polypeptides comprising thereof and their use for treating cancer
US20160122432A1 (en) * 2013-06-11 2016-05-05 INSERM (Institut National de la Santé et de la Recherche Médicale) Anti-her2 single domain antibodies, polypeptides comprising thereof and their use for treating cancer
US20150079088A1 (en) * 2013-07-25 2015-03-19 Cytomx Therapeutics, Inc. Multispecific antibodies, multispecific activatable antibodies and methods of using the same
US11161906B2 (en) * 2013-07-25 2021-11-02 Cytomx Therapeutics, Inc. Multispecific antibodies, multispecific activatable antibodies and methods of using the same
US10144781B2 (en) 2013-08-02 2018-12-04 Pfizer Inc. Anti-CXCR4 antibodies and antibody-drug conjugates
EP4050033A1 (en) 2013-08-02 2022-08-31 Pfizer Inc. Anti-cxcr4 antibodies and antibody-drug conjugates
US9708405B2 (en) 2013-08-02 2017-07-18 Pfizer Inc. Anti-CXCR4 antibodies and antibody-drug conjugates
WO2015015401A2 (en) 2013-08-02 2015-02-05 Pfizer Inc. Anti-cxcr4 antibodies and antibody-drug conjugates
WO2015050959A1 (en) 2013-10-01 2015-04-09 Yale University Anti-kit antibodies and methods of use thereof
WO2015051010A1 (en) 2013-10-02 2015-04-09 Medimmune, Llc Neutralizing anti-influenza a antibodies and uses thereof
EP3733244A1 (en) 2013-10-02 2020-11-04 Medlmmune, LLC Neutralizing anti-influenza a antibodies and uses thereof
US10323099B2 (en) 2013-10-11 2019-06-18 Hoffmann-La Roche Inc. Multispecific domain exchanged common variable light chain antibodies
US11155627B2 (en) 2013-11-29 2021-10-26 Samsung Electronics Co., Ltd. Anti-c-Met/anti-Ang2 bispecific antibody
US20150152192A1 (en) * 2013-11-29 2015-06-04 Samsung Electronics Co., Ltd. Anti-c-met/anti-ang2 bispecific antibody
US9637541B2 (en) * 2013-11-29 2017-05-02 Samsung Electronics Co., Ltd. Anti-C-Met/anti-Ang2 bispecific antibody
CN114702594A (en) * 2013-12-20 2022-07-05 豪夫迈·罗氏有限公司 Dual specificity antibodies
US9688761B2 (en) 2013-12-27 2017-06-27 Merrimack Pharmaceuticals, Inc. Biomarker profiles for predicting outcomes of cancer therapy with ERBB3 inhibitors and/or chemotherapies
US10273304B2 (en) 2013-12-27 2019-04-30 Merrimack Pharmaceuticals, Inc. Biomarker profiles for predicting outcomes of cancer therapy with ERBB3 inhibitors and/or chemotherapies
EP4008726A1 (en) 2014-02-20 2022-06-08 Allergan, Inc. Complement component c5 antibodies
WO2015127134A2 (en) 2014-02-20 2015-08-27 Allergan, Inc. Complement component c5 antibodies
US10752678B2 (en) 2014-02-20 2020-08-25 Allergan, Inc. Complement component C5 antibodies
US9701743B2 (en) 2014-02-20 2017-07-11 Allergan, Inc. Complement component C5 antibodies
US9932395B2 (en) 2014-02-20 2018-04-03 Allergan, Inc. Nucleic acids encoding complement component C5 antibodies
EP3653643A1 (en) 2014-02-20 2020-05-20 Allergan, Inc. Complement component c5 antibodies
WO2015130826A1 (en) 2014-02-27 2015-09-03 Allergan, Inc. COMPLEMENT FACTOR Bb ANTIBODIES
US9896502B2 (en) 2014-03-21 2018-02-20 Teva Pharmaceuticals International Gmbh Antagonist antibodies directed against calcitonin gene-related peptide and methods using same
US10556945B2 (en) 2014-03-21 2020-02-11 Teva Pharmaceuticals International Gmbh Antagonist antibodies directed against calcitonin gene-related peptide and methods using same
US10519224B2 (en) 2014-03-21 2019-12-31 Teva Pharmaceuticals International Gmbh Treating headache comprising administering an antibody to calcitonin gene-related peptide
US11555064B2 (en) 2014-03-21 2023-01-17 Teva Pharmaceuticals International Gmbh Treating headache comprising administering an antibody to calcitonin gene-related peptide
US20160031984A1 (en) * 2014-04-04 2016-02-04 Bionomics Inc. Humanized antibodies that bind lgr5
US9546214B2 (en) * 2014-04-04 2017-01-17 Bionomics, Inc. Humanized antibodies that bind LGR5
US10358500B2 (en) 2014-04-04 2019-07-23 Bionomics Inc. Humanized antibodies that bind LGR5
US20150283178A1 (en) * 2014-04-07 2015-10-08 Carl H. June Treatment of cancer using anti-cd19 chimeric antigen receptor
US10357514B2 (en) 2014-04-07 2019-07-23 The Trustees Of The University Of Pennsylvania Treatment of cancer using anti-CD19 Chimeric Antigen Receptor
US11485790B2 (en) 2014-04-07 2022-11-01 Chugai Seiyaku Kabushiki Kaisha Immunoactivating antigen-binding molecule
US11505605B2 (en) 2014-05-13 2022-11-22 Chugai Seiyaku Kabushiki Kaisha T cell-redirected antigen-binding molecule for cells having immunosuppression function
EP3702369A1 (en) 2014-05-16 2020-09-02 Ablynx NV Immunoglobulin variable domains
EP3693386A1 (en) 2014-05-16 2020-08-12 Ablynx NV Immunoglobulin variable domains
EP3248986A2 (en) 2014-05-16 2017-11-29 Ablynx NV Immunoglobulin variable domains
WO2015193452A1 (en) 2014-06-18 2015-12-23 Ablynx Nv Kv1.3 binding immunoglobulins
US10238742B2 (en) * 2014-06-25 2019-03-26 Yale University Cell penetrating nucleolytic antibody based cancer therapy
US20150376279A1 (en) * 2014-06-25 2015-12-31 Yale University Cell penetrating nucleolytic antibody based cancer therapy
US10669337B2 (en) 2014-07-25 2020-06-02 Cytomx Therapeutics, Inc. Bispecific anti-CD3 antibodies, bispecific activatable anti-CD3 antibodies, and methods of using the same
US11802158B2 (en) 2014-07-25 2023-10-31 Cytomx Therapeutics, Inc. Bispecific anti-CD3 antibodies, bispecific activatable anti-CD3 antibodies, and methods of using the same
US11661462B2 (en) 2014-07-31 2023-05-30 Amgen Research (Munich) Gmbh Optimized cross-species specific bispecific single chain antibody contructs
US20160032019A1 (en) * 2014-07-31 2016-02-04 Amgen Inc. Antibody constructs for CDH19 and CD3
US9765157B2 (en) * 2014-07-31 2017-09-19 Amgen Research (Munich) Gmbh Antibody constructs for CDH19 and CD3
US20160053025A1 (en) * 2014-08-25 2016-02-25 Samsung Electronics Co., Ltd. Anti-c-met/anti-ang2 bispecific antibody
US9808507B2 (en) * 2014-08-25 2017-11-07 Samsung Electronics Co., Ltd. Anti-c-Met/anti-Ang2 bispecific antibody
US11001643B2 (en) 2014-09-26 2021-05-11 Chugai Seiyaku Kabushiki Kaisha Cytotoxicity-inducing therapeutic agent
US9975966B2 (en) 2014-09-26 2018-05-22 Chugai Seiyaku Kabushiki Kaisha Cytotoxicity-inducing theraputic agent
US11773166B2 (en) 2014-11-04 2023-10-03 Ichnos Sciences SA CD3/CD38 T cell retargeting hetero-dimeric immunoglobulins and methods of their production
US10730938B2 (en) 2014-11-10 2020-08-04 Hoffmann-La Roche Inc. Bispecific antibodies and methods of use in ophthalmology
US10538585B2 (en) * 2014-11-10 2020-01-21 Hoffmann-La Roche Inc. Anti-ANG2 antibodies and methods of use
US20170247441A1 (en) * 2014-11-10 2017-08-31 Hoffmann-La Roche Inc. Anti-ang2 antibodies and methods of use
US10344085B2 (en) 2014-11-10 2019-07-09 Hoffman-La Roche, Inc. Anti-IL-1beta antibodies
US10093733B2 (en) 2014-12-11 2018-10-09 Abbvie Inc. LRP-8 binding dual variable domain immunoglobulin proteins
US11426468B2 (en) 2014-12-19 2022-08-30 Ablynx N.V. Cysteine linked nanobody dimers
US20170360955A1 (en) * 2014-12-19 2017-12-21 Ablynx N.V. Cysteine linked nanobody dimers
WO2016097313A1 (en) 2014-12-19 2016-06-23 Ablynx N.V. Cysteine linked nanobody dimers
AU2015366284B2 (en) * 2014-12-19 2021-07-22 Ablynx N.V. Cysteine linked nanobody dimers
US10538589B2 (en) 2015-01-14 2020-01-21 Armagen Inc. Methods and compositions for increasing N-acetylglucosaminidase (NAGLU) activity in the CNS using a fusion antibody comprising an anti-human insulin receptor antibody and NAGLU
US11149076B2 (en) 2015-04-08 2021-10-19 Novartis Ag CD20 therapies, CD22 therapies, and combination therapies with a CD19 chimeric antigen receptor (CAR)-expressing cell
US10253086B2 (en) 2015-04-08 2019-04-09 Novartis Ag CD20 therapies, CD22 therapies, and combination therapies with a CD19 chimeric antigen receptor (CAR)-expressing cell
US10954311B2 (en) 2015-05-21 2021-03-23 Harpoon Therapeutics, Inc. Trispecific binding proteins and methods of use
US9708412B2 (en) * 2015-05-21 2017-07-18 Harpoon Therapeutics, Inc. Trispecific binding proteins and methods of use
US10184006B2 (en) 2015-06-04 2019-01-22 Merrimack Pharmaceuticals, Inc. Biomarkers for predicting outcomes of cancer therapy with ErbB3 inhibitors
US9840554B2 (en) 2015-06-15 2017-12-12 Abbvie Inc. Antibodies against platelet-derived growth factor (PDGF)
US20170037130A1 (en) * 2015-07-31 2017-02-09 Amgen Research (Munich) Gmbh Antibody constructs for dll3 and cd3
US10294300B2 (en) * 2015-07-31 2019-05-21 Amgen Research (Munich) Gmbh Antibody constructs for DLL3 and CD3
US10683351B2 (en) 2015-07-31 2020-06-16 Amgen Research (Munich) Gmbh Antibody constructs for DLL3 and CD3
US11591396B2 (en) 2015-07-31 2023-02-28 Amgen Research (Munich) Gmbh Antibody constructs for DLL3 and CD3
US11352426B2 (en) 2015-09-21 2022-06-07 Aptevo Research And Development Llc CD3 binding polypeptides
US11660340B2 (en) 2015-11-18 2023-05-30 Chugai Seiyaku Kabushiki Kaisha Combination therapy using T cell redirection antigen binding molecule against cell having immunosuppressing function
US11649293B2 (en) 2015-11-18 2023-05-16 Chugai Seiyaku Kabushiki Kaisha Method for enhancing humoral immune response
US11649262B2 (en) 2015-12-28 2023-05-16 Chugai Seiyaku Kabushiki Kaisha Method for promoting efficiency of purification of Fc region-containing polypeptide
US11072666B2 (en) 2016-03-14 2021-07-27 Chugai Seiyaku Kabushiki Kaisha Cell injury inducing therapeutic drug for use in cancer therapy
US10745487B2 (en) 2016-03-22 2020-08-18 Bionomics Limited Method of treating cancer by administering an anti-LGR5 monoclonal antibody
WO2017175145A1 (en) 2016-04-05 2017-10-12 Glaxosmithkline Intellectual Property Development Limited Inhibition of tgfbeta in immunotherapy
US11028169B2 (en) 2016-05-18 2021-06-08 Boehringer Ingelheim International Gmbh Antibody molecules for cancer treatment
US11795219B2 (en) 2016-05-18 2023-10-24 Boehringer Ingelheim International Gmbh Antibody molecules for cancer treatment
EP3848393A1 (en) 2016-05-18 2021-07-14 Boehringer Ingelheim International GmbH Antibody molecules for cancer treatment
US10544221B2 (en) 2016-05-20 2020-01-28 Harpoon Therapeutics, Inc. Single chain variable fragment CD3 binding proteins
US10066016B2 (en) 2016-05-20 2018-09-04 Harpoon Therapeutics, Inc. Single chain variable fragment CD3 binding proteins
US11453716B2 (en) 2016-05-20 2022-09-27 Harpoon Therapeutics, Inc. Single domain serum albumin binding protein
US10100106B2 (en) 2016-05-20 2018-10-16 Harpoon Therapeutics, Inc. Single domain serum albumin binding protein
US9920115B2 (en) 2016-05-20 2018-03-20 Harpoon Therapeutics, Inc. Single domain serum albumin binding protein
US11623958B2 (en) 2016-05-20 2023-04-11 Harpoon Therapeutics, Inc. Single chain variable fragment CD3 binding proteins
WO2017210425A1 (en) 2016-06-03 2017-12-07 Janssen Biotech, Inc. Serum albumin-binding fibronectin type iii domains
US11833190B2 (en) 2016-06-03 2023-12-05 Janssen Biotech, Inc. Serum albumin-binding fibronectin type III domains
US20170348397A1 (en) * 2016-06-03 2017-12-07 Janssen Biotech, Inc. Serum Albumin-Binding Fibronectin Type III Domains
US10925932B2 (en) 2016-06-03 2021-02-23 Janssen Biotech, Inc. Serum albumin-binding fibronectin type III domains
US11590242B2 (en) 2016-06-15 2023-02-28 Yale University Antibody-mediated autocatalytic, targeted delivery of nanocarriers to tumors
WO2018007442A1 (en) 2016-07-06 2018-01-11 Ablynx N.V. Treatment of il-6r related diseases
WO2018020000A1 (en) 2016-07-29 2018-02-01 INSERM (Institut National de la Santé et de la Recherche Médicale) Antibodies targeting tumor associated macrophages and uses thereof
WO2018029182A1 (en) 2016-08-08 2018-02-15 Ablynx N.V. Il-6r single variable domain antibodies for treatment of il-6r related diseases
US10392434B2 (en) 2016-09-23 2019-08-27 Teva Pharmaceuticals International Gmbh Treating refractory migraine
US11028160B2 (en) 2016-09-23 2021-06-08 Teva Pharmaceuticals International Gmbh Treating refractory migraine
US11028161B2 (en) 2016-09-23 2021-06-08 Teva Pharmaceuticals International Gmbh Treating refractory migraine
US11872249B2 (en) 2016-10-07 2024-01-16 Novartis Ag Method of treating cancer by administering immune effector cells expressing a chimeric antigen receptor comprising a CD20 binding domain
US11026976B2 (en) 2016-10-07 2021-06-08 Novartis Ag Nucleic acid molecules encoding chimeric antigen receptors comprising a CD20 binding domain
US10525083B2 (en) 2016-10-07 2020-01-07 Novartis Ag Nucleic acid molecules encoding chimeric antigen receptors comprising a CD20 binding domain
USRE49847E1 (en) 2016-10-07 2024-02-27 Novartis Ag Nucleic acid molecules encoding chimeric antigen receptors comprising a CD20 binding domain
WO2018091606A1 (en) 2016-11-16 2018-05-24 Ablynx Nv T cell recruiting polypeptides capable of binding cd123 and tcr alpha/beta
US10844134B2 (en) 2016-11-23 2020-11-24 Harpoon Therapeutics, Inc. PSMA targeting trispecific proteins and methods of use
US10849973B2 (en) 2016-11-23 2020-12-01 Harpoon Therapeutics, Inc. Prostate specific membrane antigen binding protein
WO2018104444A1 (en) 2016-12-07 2018-06-14 Ablynx Nv Improved serum albumin binding immunoglobulin single variable domains
WO2018115231A2 (en) 2016-12-22 2018-06-28 Boehringer Ingelheim International Gmbh Binding molecules for the treatment of cancer
WO2018129029A1 (en) 2017-01-04 2018-07-12 Immunogen, Inc. Met antibodies and immunoconjugates and uses thereof
WO2018134234A1 (en) 2017-01-17 2018-07-26 Ablynx Nv Improved serum albumin binders
WO2018134235A1 (en) 2017-01-17 2018-07-26 Ablynx Nv Improved serum albumin binders
US11203636B2 (en) 2017-02-01 2021-12-21 Yale University Treatment of existing left ventricular heart failure
EP3360898A1 (en) 2017-02-14 2018-08-15 Boehringer Ingelheim International GmbH Bispecific anti-tnf-related apoptosis-inducing ligand receptor 2 and anti-cadherin 17 binding molecules for the treatment of cancer
US11535668B2 (en) 2017-02-28 2022-12-27 Harpoon Therapeutics, Inc. Inducible monovalent antigen binding protein
WO2018158398A1 (en) 2017-03-02 2018-09-07 INSERM (Institut National de la Santé et de la Recherche Médicale) Antibodies having specificity to nectin-4 and uses thereof
US10543271B2 (en) 2017-05-12 2020-01-28 Harpoon Therapeutics, Inc. Mesothelin binding proteins
US10730954B2 (en) 2017-05-12 2020-08-04 Harpoon Therapeutics, Inc. MSLN targeting trispecific proteins and methods of use
US11607453B2 (en) 2017-05-12 2023-03-21 Harpoon Therapeutics, Inc. Mesothelin binding proteins
US11033636B2 (en) * 2017-05-31 2021-06-15 Boehringer Ingelheim International Gmbh Polypeptides antagonizing Wnt signaling in tumor cells
US20180344868A1 (en) * 2017-05-31 2018-12-06 Boehringer Ingelheim International Gmbh Polypeptides antagonizing wnt signaling in tumor cells
WO2019020480A1 (en) 2017-07-24 2019-01-31 INSERM (Institut National de la Santé et de la Recherche Médicale) Antibodies and peptides to treat hcmv related diseases
US11859003B2 (en) 2017-08-21 2024-01-02 Adagene Inc. Method for treating cancer using anti-CD137 antibody
US11242395B2 (en) 2017-08-21 2022-02-08 Adagene Inc. Anti-CD137 molecules and use thereof
EP3470426A1 (en) 2017-10-10 2019-04-17 Numab Therapeutics AG Multispecific antibody
WO2019072868A1 (en) 2017-10-10 2019-04-18 Numab Therapeutics AG Multispecific antibody
US10927180B2 (en) 2017-10-13 2021-02-23 Harpoon Therapeutics, Inc. B cell maturation antigen binding proteins
US11136403B2 (en) 2017-10-13 2021-10-05 Harpoon Therapeutics, Inc. Trispecific proteins and methods of use
US11859010B2 (en) 2017-10-14 2024-01-02 Cytomx Therapeutics, Inc. Antibodies, activatable antibodies, bispecific antibodies, and bispecific activatable antibodies and methods of use thereof
US11472889B2 (en) 2017-10-14 2022-10-18 Cytomx Therapeutics, Inc. Antibodies, activatable antibodies, bispecific antibodies, and bispecific activatable antibodies and methods of use thereof
US11384143B2 (en) 2018-01-05 2022-07-12 Novo Nordisk A/S Methods for treating IL-6 mediated inflammation without immunosuppression
US11359016B2 (en) 2018-02-02 2022-06-14 Adagene Inc. Anti-CTLA4 antibodies and methods of making and using the same
US11952681B2 (en) 2018-02-02 2024-04-09 Adagene Inc. Masked activatable CD137 antibodies
US11692036B2 (en) 2018-02-02 2023-07-04 Adagene Inc. Anti-CTLA4 antibodies and methods of making and using the same
EP3569618A1 (en) 2018-05-19 2019-11-20 Boehringer Ingelheim International GmbH Antagonizing cd73 antibody
US11312785B2 (en) 2018-05-19 2022-04-26 Boehringer Ingelheim International Gmbh Antagonizing CD73 antibody
WO2019224025A2 (en) 2018-05-19 2019-11-28 Boehringer Ingelheim International Gmbh Antagonizing cd73 antibody
US11845797B2 (en) 2018-07-03 2023-12-19 Marengo Therapeutics, Inc. Anti-TCR antibody molecules and uses thereof
DE202019005887U1 (en) 2018-07-03 2023-06-14 Marengo Therapeutics, Inc. Anti-TCR antibody molecules and uses thereof
US11965025B2 (en) 2018-07-03 2024-04-23 Marengo Therapeutics, Inc. Method of treating solid cancers with bispecific interleukin-anti-TCRß molecules
WO2020010250A2 (en) 2018-07-03 2020-01-09 Elstar Therapeutics, Inc. Anti-tcr antibody molecules and uses thereof
WO2020014306A1 (en) 2018-07-10 2020-01-16 Immunogen, Inc. Met antibodies and immunoconjugates and uses thereof
WO2020053122A1 (en) 2018-09-10 2020-03-19 INSERM (Institut National de la Santé et de la Recherche Médicale) Combination of her2/neu antibody with heme for treating cancer
US11807692B2 (en) 2018-09-25 2023-11-07 Harpoon Therapeutics, Inc. DLL3 binding proteins and methods of use
US10815311B2 (en) 2018-09-25 2020-10-27 Harpoon Therapeutics, Inc. DLL3 binding proteins and methods of use
WO2020084056A1 (en) 2018-10-25 2020-04-30 Swiss Pharma International Ag Anti-human cd89 antibodies and uses thereof
WO2020099508A1 (en) 2018-11-13 2020-05-22 Emstopa Limited Tissue plasminogen activator antibodies and method of use thereof
WO2020115283A1 (en) * 2018-12-07 2020-06-11 Baxalta GmbH Bispecific antibodies binding factor ixa and factor x
WO2020114615A1 (en) * 2018-12-07 2020-06-11 Baxalta GmbH Bispecific antibodies binding factor ixa and factor x
US11639381B2 (en) 2019-01-08 2023-05-02 H. Lundbeck A/S Treatment of headache using anti-CGRP antibodies
US11639380B2 (en) 2019-01-08 2023-05-02 H. Lundbeck A/S Acute treatment and rapid treatment of headache using anti-CGRP antibodies
WO2020193520A1 (en) 2019-03-25 2020-10-01 INSERM (Institut National de la Santé et de la Recherche Médicale) Treatment of taupathy disorders by targeting new tau species
WO2020198731A2 (en) 2019-03-28 2020-10-01 Danisco Us Inc Engineered antibodies
WO2021058729A1 (en) 2019-09-27 2021-04-01 INSERM (Institut National de la Santé et de la Recherche Médicale) Anti-müllerian inhibiting substance type i receptor antibodies and uses thereof
WO2021058763A1 (en) 2019-09-27 2021-04-01 INSERM (Institut National de la Santé et de la Recherche Médicale) Anti-müllerian inhibiting substance antibodies and uses thereof
EP3816185A1 (en) 2019-11-04 2021-05-05 Numab Therapeutics AG Multispecific antibody directed against pd-l1 and a tumor-associated antigen
WO2021089609A1 (en) 2019-11-04 2021-05-14 Numab Therapeutics AG Multispecific antibody
WO2021116119A1 (en) 2019-12-09 2021-06-17 INSERM (Institut National de la Santé et de la Recherche Médicale) Antibodies having specificity to her4 and uses thereof
WO2021123186A1 (en) 2019-12-20 2021-06-24 UCB Biopharma SRL Multi-specific antibody with binding specificity for human il-13 and il-17
WO2021133170A1 (en) 2019-12-24 2021-07-01 Cluster270 Therapeutics B.V. Anti-human hvem (tnfrsf14) antibodies and uses thereof
WO2021160133A1 (en) 2020-02-13 2021-08-19 湖南华康恒健生物技术有限公司 Anti-bcma antibody, pharmaceutical composition of same, and applications thereof
US11180563B2 (en) 2020-02-21 2021-11-23 Harpoon Therapeutics, Inc. FLT3 binding proteins and methods of use
WO2021205183A1 (en) 2020-04-09 2021-10-14 Autolus Limited Polypeptide
WO2021205184A1 (en) 2020-04-09 2021-10-14 Autolus Limited Polypeptide
WO2021215919A1 (en) 2020-04-21 2021-10-28 Polpharma Biologics Utrecht B.V. Humanized anti-human cd89 antibodies and uses thereof
WO2021228956A1 (en) 2020-05-12 2021-11-18 INSERM (Institut National de la Santé et de la Recherche Médicale) New method to treat cutaneous t-cell lymphomas and tfh derived lymphomas
WO2021234383A1 (en) 2020-05-19 2021-11-25 Autolus Limited Single domain antibodies against hace2 and their use to prevent sars-cov-2 infection
WO2021236658A1 (en) 2020-05-19 2021-11-25 Boehringer Ingelheim International Gmbh Binding molecules for the treatment of cancer
EP3915580A1 (en) 2020-05-29 2021-12-01 Numab Therapeutics AG Multispecific antibody
WO2021239987A1 (en) 2020-05-29 2021-12-02 Numab Therapeutics AG Multispecific antibody
WO2022042659A1 (en) 2020-08-26 2022-03-03 湖南华康恒健生物技术有限公司 Anti-ox40 antibody, and pharmaceutical composition and application thereof
WO2022044573A1 (en) 2020-08-26 2022-03-03 国立大学法人熊本大学 Human antibody or antigen-binding fragment thereof against coronavirus spike protein
WO2022084354A1 (en) 2020-10-21 2022-04-28 Boehringer Ingelheim International Gmbh Bispecific anti-vegf and anti-trkb binding molecules for the treatment of eye diseases
WO2022084355A2 (en) 2020-10-21 2022-04-28 Boehringer Ingelheim International Gmbh Agonistic trkb binding molecules for the treatment of eye diseases
EP3988568A1 (en) 2020-10-21 2022-04-27 Numab Therapeutics AG Combination treatment
WO2022084440A2 (en) 2020-10-21 2022-04-28 Numab Therapeutics AG Combination treatment
WO2022098745A1 (en) 2020-11-03 2022-05-12 Indi Molecular, Inc. Compositions, delivery systems, and methods useful in tumor therapy
WO2022098743A1 (en) 2020-11-03 2022-05-12 Indi Molecular, Inc. Compositions, imaging, and therapeutic methods targeting folate receptor 1 (folr1)
US11766414B2 (en) 2020-11-03 2023-09-26 Indi Molecular, Inc. Compositions, delivery systems, and methods useful in tumor therapy
US11733246B2 (en) 2020-11-03 2023-08-22 Indi Molecular, Inc. Compositions, imaging, and therapeutic methods targeting folate receptor 1 (FOLR1)
WO2022122654A1 (en) 2020-12-07 2022-06-16 UCB Biopharma SRL Multi-specific antibodies and antibody combinations
US11976125B2 (en) 2021-01-15 2024-05-07 Harpoon Therapeutics, Inc. B cell maturation antigen binding proteins
WO2022167460A1 (en) 2021-02-02 2022-08-11 Numab Therapeutics AG Multispecific antibodies having specificity for ror1 and cd3
WO2022178255A2 (en) 2021-02-19 2022-08-25 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Single domain antibodies that neutralize sars-cov-2
WO2022216993A2 (en) 2021-04-08 2022-10-13 Marengo Therapeutics, Inc. Multifuntional molecules binding to tcr and uses thereof
WO2022263507A1 (en) 2021-06-17 2022-12-22 Boehringer Ingelheim International Gmbh Novel tri-specific binding molecules
EP4183800A1 (en) 2021-11-19 2023-05-24 Medizinische Hochschule Hannover Novel sars-cov-2 neutralizing antibodies
WO2023089107A1 (en) 2021-11-19 2023-05-25 Medizinische Hochschule Hannover Novel sars-cov-2 neutralizing antibodies
WO2023175171A1 (en) 2022-03-18 2023-09-21 Inserm (Institut National De La Sante Et De La Recherche Medicale) Bk polyomavirus antibodies and uses thereof
WO2024013315A1 (en) 2022-07-15 2024-01-18 Boehringer Ingelheim International Gmbh Binding molecules for the treatment of cancer
WO2024038095A1 (en) 2022-08-16 2024-02-22 Iome Bio NOVEL ANTI-RGMb ANTIBODIES
WO2024052503A1 (en) 2022-09-08 2024-03-14 Institut National de la Santé et de la Recherche Médicale Antibodies having specificity to ltbp2 and uses thereof
WO2024056668A1 (en) 2022-09-12 2024-03-21 Institut National de la Santé et de la Recherche Médicale New anti-itgb8 antibodies and its uses thereof

Also Published As

Publication number Publication date
HK1070081A1 (en) 2005-06-10
DE60305919T2 (en) 2007-01-18
JP2011125342A (en) 2011-06-30
SI1517921T1 (en) 2006-10-31
US20150087813A1 (en) 2015-03-26
US20070093651A1 (en) 2007-04-26
CA2492092A1 (en) 2004-01-08
JP2006512895A (en) 2006-04-20
AU2003244817B2 (en) 2010-08-26
EP2366718A2 (en) 2011-09-21
CN1678634A (en) 2005-10-05
EP2135879A3 (en) 2010-06-23
ATE328906T1 (en) 2006-06-15
US20100234570A1 (en) 2010-09-16
EP1600459A3 (en) 2005-12-07
DK1517921T3 (en) 2006-10-09
EP2366718A3 (en) 2012-05-02
DE60305919D1 (en) 2006-07-20
AU2003244817A2 (en) 2004-01-19
GB0501752D0 (en) 2005-03-02
CY1105191T1 (en) 2010-03-03
PT1517921E (en) 2006-09-29
EP2135879A2 (en) 2009-12-23
EP1600459A2 (en) 2005-11-30
ES2263984T3 (en) 2006-12-16
AU2003244817A1 (en) 2004-01-19
EP1517921B1 (en) 2006-06-07
EP1517921A2 (en) 2005-03-30
US20060106203A1 (en) 2006-05-18
WO2004003019A3 (en) 2004-09-10
US20060063921A1 (en) 2006-03-23

Similar Documents

Publication Publication Date Title
AU2003244817B2 (en) Antigen-binding immunoglobulin single variable domains and dual-specific constructs
EP1578801A2 (en) Dual specific single domain antibodies specific for a ligand and for the receptor of the ligand
CA2583417C (en) Antagonists and methods of use therefor
WO2004003019A9 (en) Immunoglobin single variant antigen-binding domains and dual-specific constructs
EP1777235A1 (en) Compositions and methods for treating inflammatory disorders
US20080233129A1 (en) Ligand that has binding specificity for IL-4 and/or lL-13
US20080008713A1 (en) Single domain antibodies against tnfr1 and methods of use therefor
US9028822B2 (en) Antagonists against TNFR1 and methods of use therefor
US20110223168A1 (en) Ligand that has binding specificity for il-4 and/or il-13
JP2008504356A6 (en) Compositions and methods for treating inflammatory diseases
JP2008504356A (en) Compositions and methods for treating inflammatory diseases
KR20050024397A (en) Ligand

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IT LU MC NL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
WWE Wipo information: entry into national phase

Ref document number: 2492092

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2004516979

Country of ref document: JP

Ref document number: 1020047021231

Country of ref document: KR

WWE Wipo information: entry into national phase

Ref document number: 11023959

Country of ref document: US

WWE Wipo information: entry into national phase

Ref document number: 2003738294

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2003244817

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: GB0501752.0

Country of ref document: GB

WWE Wipo information: entry into national phase

Ref document number: 20038204584

Country of ref document: CN

WWP Wipo information: published in national office

Ref document number: 1020047021231

Country of ref document: KR

WWP Wipo information: published in national office

Ref document number: 2003738294

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 11098758

Country of ref document: US

WWP Wipo information: published in national office

Ref document number: 11098758

Country of ref document: US

COP Corrected version of pamphlet

Free format text: PAGE 106, DESCRIPTION, REPLACED BY CORRECT PAGE 106

WWP Wipo information: published in national office

Ref document number: 11023959

Country of ref document: US

WWG Wipo information: grant in national office

Ref document number: 2003738294

Country of ref document: EP