US20080057063A1 - Antibodies Directed to AlphaVBeta6 and Uses Thereof - Google Patents

Antibodies Directed to AlphaVBeta6 and Uses Thereof Download PDF

Info

Publication number
US20080057063A1
US20080057063A1 US11/833,486 US83348607A US2008057063A1 US 20080057063 A1 US20080057063 A1 US 20080057063A1 US 83348607 A US83348607 A US 83348607A US 2008057063 A1 US2008057063 A1 US 2008057063A1
Authority
US
United States
Prior art keywords
seq
binding agent
antibody
αvβ6
targeted binding
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US11/833,486
Other languages
English (en)
Inventor
Julie Rinkenberger
Ian Foltz
Avril Alfred
Simon Barry
Vahe Bedian
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
MedImmune Ltd
Original Assignee
AstraZeneca AB
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by AstraZeneca AB filed Critical AstraZeneca AB
Priority to US11/833,486 priority Critical patent/US20080057063A1/en
Publication of US20080057063A1 publication Critical patent/US20080057063A1/en
Assigned to ASTRAZENECA AB reassignment ASTRAZENECA AB ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: AMGEN FREMONT INC
Assigned to ASTRAZENECA AB reassignment ASTRAZENECA AB ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: BEDIAN, VAHE, FOLTZ, IAN, ALFRED, AVRIL, RINKENBERGER, JULIE, BARRY, SIMON THOMAS
Priority to US12/782,335 priority patent/US8398975B2/en
Assigned to MEDIMMUNE LIMITED reassignment MEDIMMUNE LIMITED ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: ASTRAZENECA AB
Priority to US13/760,426 priority patent/US8894998B2/en
Priority to US14/535,894 priority patent/US20150166663A1/en
Abandoned legal-status Critical Current

Links

Images

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
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2839Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the integrin superfamily
    • 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
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • 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
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/08Drugs for disorders of the urinary system of the prostate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • 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
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/02Drugs for disorders of the nervous system for peripheral neuropathies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • 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
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/14Drugs for disorders of the endocrine system of the thyroid hormones, e.g. T3, T4
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/14Vasoprotectives; Antihaemorrhoidals; Drugs for varicose therapy; Capillary stabilisers
    • 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
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0684Cells of the urinary tract or kidneys
    • C12N5/0686Kidney cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins 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
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/565Complementarity determining region [CDR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/567Framework region [FR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/998Proteins not provided for elsewhere

Definitions

  • the invention relates to monoclonal antibodies against alphaVbeta6 integrin ( ⁇ V ⁇ 6) and uses of such antibodies.
  • the invention relates to fully human monoclonal antibodies directed to ⁇ V ⁇ 6.
  • the described antibodies are useful as diagnostics and for the treatment of diseases associated with the activity and/or overproduction of ⁇ V ⁇ 6.
  • the integrin superfamily includes at least 24 family members consisting of heterodimers that utilize 18 alpha and 8 beta chains (Hynes, (2002) Cell 110: 673-87). This family of receptors is expressed on the cell surface and mediates cell-cell and cell-extracellular matrix interactions that regulate cell survival, proliferation, migration, and differentiation as well as tumor invasion and metastasis (ffrench-Constant and Colognato, (2004) Trends Cell Biol. 14: 678-86). Integrins bind to other cellular receptors, growth factors and extracellular matrix proteins, with many family members having overlapping binding specificity for particular proteins. This redundancy may ensure that important functions continue in the absence of a particular integrin (Koivisto et al., (2000) Exp. Cell Res.
  • the integrin family can be divided into several sub-families based on ligand specificity of the heterodimers.
  • One subfamily consists of all of the integrins that recognize and bind the RGD tripeptide. These receptors include the ⁇ IIb/ ⁇ 3 and all of the ⁇ V heterodimers (Thomas et al., (2006) J. Oral Pathol. Med. 35: 1-10). While the ⁇ V chain can pair with 5 known beta chains, several of these beta chains can only pair with ⁇ V. The ⁇ 6 chain is selective for heterodimerization to ⁇ V and this pair binds extracellular matrix and cytokine proteins with either high or low affinity.
  • ⁇ V ⁇ 6 binds to the RGD motifs on both TGF ⁇ 1LAP and TGF ⁇ 3LAP latent complexes and activates them (Munger et al, (1999) Cell 96: 319-328; Annes et al., (2002) FEBS Letters 511: 65-68). However, it does not bind to or activate TGF ⁇ 2LAP, which does not have the tri-peptide (Ludbrook et al., (2003) Biochem. J. 369: 311-18). ⁇ V ⁇ 6-mediated activation of TGF ⁇ requires the latent TGF ⁇ binding protein 1 (LTBP1), which tethers the latent TGF ⁇ complex to the extracellular matrix.
  • LTBP1 latent TGF ⁇ binding protein 1
  • Activation is proposed to result from a conformation change induced as the TGF ⁇ LAP is held between the cell and the matrix by ⁇ V ⁇ 6 and LTBP1, respectively (Keski-Oja et al., (2004) Trends Cell Biol. 14: 657-659; Annes et al., (2004) J. Cell Biol. 165: 723-34).
  • the picoMolar binding affinity of ⁇ V ⁇ 6 for the TGF ⁇ LAP complexes is the highest for any of its known ligands.
  • Other ligands for ⁇ V ⁇ 6 include fibronectin, tenascin, vitronectin and osteopontin (Busk et al., (1992) J. Biol. Chem.
  • ⁇ V ⁇ 6 integrin is restricted to areas of active tissue remodeling in the adult, specifically on the epithelia of healing wounds and at the edge of invading tumors (Breuss et al., (1995) J. Cell Sci. 108: 2241-51). Keratinocytes at the wound edge upregulate the expression of ⁇ V ⁇ 6 during their migration into the wound, but expression remains high after the edges of the wound epithelium have joined (Breuss et al., (1995) J. Cell Sci. 108: 2241-51; Haapasalmi et al., (1996) J. Invest. Dermatol. 106: 42-48).
  • the wound extracellular matrix contains fibronectin, tenascin and vitronectin, all of which are ligands for ⁇ V ⁇ 6 (Busk et al., (1992) J. Biol. Chem. 267: 5790-6; Koivisto et al., (1999) Cell Adhes. Commun. 7: 245-57; Hakkinen et al., (2000) J. Histochem. Cytochem. 48: 985-98).
  • ⁇ V ⁇ 6 upregulates the expression of the matrix metalloproteinase, MMP-9, that can degrade Type IV collagen and promote cell movement (Niu et al., (1998) Biochem. Biophys. Res. Com.
  • ⁇ V ⁇ 6 may have dual roles to promote keratinocyte migration during wound closure and later to resolve the wound through the activation of TGF ⁇ .
  • the activation of TGF ⁇ by ⁇ V ⁇ 6 would contribute to wound resolution through the regulation of re-epithelialization, suppression of inflammation and promotion of connective tissue regeneration and scar formation (Thomas et al., (2006) J. Oral Pathol. Med.
  • ⁇ V ⁇ 6 integrin In addition to its expression in wound healing, the ⁇ V ⁇ 6 integrin is upregulated at the periphery of many human tumors. ⁇ V ⁇ 6 expression has been reported in oral (Breuss et al., (1995) J. Cell Sci. 108: 2241-51; Jones et al., (1997) J. Oral Pathol. Med. 26: 63-8; Hamidi et al., (2000) Br. J. Cancer 82: 1433-40; Regezi et al., (2002) Oral Oncology 38: 332-6; Impola et al., (2004) J. Pathol.
  • EMT epithelial-to-mesenchymal transition
  • binding to fibronectin resulted in the recruitment and activation of the Fyn kinase by the beta 6 subunit.
  • Downstream signal transduction resulted in the production of MMP-3, promoted cell proliferation in vitro, tumor invasion in an orthotopic model, and metastasis in a tail vein injection model (Li et al., (2003) J. Biol. Chem. 278: 41646-53).
  • ⁇ V ⁇ 6 Suppression of apoptosis, like cell proliferation, is another way that ⁇ V ⁇ 6 may promote tumor growth.
  • Normal stratified squamous epithelia express the ⁇ V ⁇ 5 integrin but down-regulate it and upregulate ⁇ V ⁇ 6 expression upon transformation to carcinomas.
  • carcinoma cell lines that over-expressed ⁇ V ⁇ 5 Using carcinoma cell lines that over-expressed ⁇ V ⁇ 5, ⁇ V ⁇ 6 expression was shown to prevent suspension-induced cell death (anoikis) in vitro (Janes and Watt, (2004) J. Cell Biol. 166: 419-31).
  • Apoptosis inhibition has also been observed in vitro in ovarian cancer cell lines treated with cisplatin, which may represent a mechanism for drug resistance of these tumors in vivo (Wu et al., (2004) Zhonghua Fu Chan Ke Za Zhi 39: 112-14).
  • a number of investigators have developed therapeutics to target ⁇ V ⁇ 6 activity in fibrosis and cancer.
  • a murine antibody with specificity for the ⁇ V ⁇ 6, ⁇ V ⁇ 3 and ⁇ V ⁇ 5 integrins was shown to prevent adhesion of HT29 colon carcinoma cells to vitronectin and fibronectin in vitro (Lehmann et al., (1994) Can. Res. 54: 2102-07).
  • Another murine antibody therapeutic specific for the human ⁇ V ⁇ 6 protein was demonstrated to inhibit the invasive growth of HSC-3 oral carcinoma cells in a transoral xenograft tumor model in mice (Xue et al., (2001) Biochem. Biophys. Res. Com. 288: 610-18).
  • ⁇ V ⁇ 6 Another recently described role for ⁇ V ⁇ 6 is as a cellular receptor for viral pathogens. It mediates the binding of the viral capsid for foot-and-mouth disease virus and the Coxsackievirus 9 to enable viral entry in vitro (Miller et al., (2001) J. Virol. 75: 4158-64; Williams et al., (2004) J. Virol. 78: 6967-73). Both foot-and-mouth disease virus and Coxsackievirus 9 capsid proteins contain an RGD sequence that is recognized by multiple integrin family members. Viral entry of both pathogens is blocked by antibody to ⁇ V ⁇ 6 integrin (Williams et al., (2004) J. Virol. 78: 6967-73).
  • the invention is generally directed to targeted binding agents that bind to ⁇ V ⁇ 6.
  • Embodiments of the invention relate to fully human targeted binding agents that specifically bind to ⁇ V ⁇ 6 and thereby inhibit binding of ligands to ⁇ V ⁇ 6.
  • the targeted binding agents also inhibit tumor cell adhesion.
  • the targeted binding agents are useful for reducing tumor growth. Mechanisms by which this can be achieved can include and are not limited to either inhibiting binding of a ligand to its receptor ⁇ V ⁇ 6, abrogation of intereactions with ligands such as TGF ⁇ -LAP, thereby reducing the effective concentration of ⁇ V ⁇ 6.
  • the targeted binding agent is a fully human antibody that binds to ⁇ V ⁇ 6 and prevents ⁇ V ⁇ 6 binding to ligands of ⁇ V ⁇ 6.
  • ligands of ⁇ V ⁇ 6 include TGF ⁇ LAP, fibronectin, tenascin, vitronectin and osteopontin.
  • the antibody may bind ⁇ V ⁇ 6 with a K d of less than 35 nM, 25 nM, 10 nM, or 60 pM.
  • Yet another embodiment is a fully human antibody that binds to ⁇ V ⁇ 6 and inhibits greater than 80%, 85%, 90% or 99% of TGF ⁇ -LAP mediated adhesion of HT29 cells at antibody concentrations as low as 1 ⁇ g/ml or less.
  • Yet another embodiment is a fully human antibody that binds to ⁇ V ⁇ 6 and inhibits TGF ⁇ -LAP mediated adhesion of HT29 cells with an IC 50 of less than 0.070 ⁇ g/ml.
  • the targeted binding agent may comprise a heavy chain amino acid sequence having a complementarity determining region (CDR) with one of the sequences shown in Table 8 or Table 29.
  • CDR complementarity determining region
  • the targeted binding agent may comprise a sequence comprising any one of a CDR1, CDR2 or CDR3 sequence as shown in Table 8 or Table 29.
  • the targeted binding agent may comprise a sequence comprising any two of a CDR1, CDR2 or CDR3 sequence as shown in Table 8 or Table 29 (that is either a CDR1 and CDR2, a CDR1 and CDR3 or a CDR2 and CDR3).
  • the targeted binding agent may comprise a sequence comprising a CDR1, CDR2 and CDR3 sequence as shown in Table 8 or Table 29. It is noted that those of ordinary skill in the art can readily accomplish CDR determinations. See for example, Kabat et al., Sequences of Proteins of Immunological Interest , Fifth Edition, NIH Publication 91-3242, Bethesda Md. (1991), vols. 1-3.
  • the targeted binding agent i.e. an antibody
  • the targeted binding agent may comprise a light chain amino acid sequence having a complementarity determining region (CDR), CDR1, CDR2 or CDR3 sequences as shown in Table 9 or Table 30.
  • the targeted binding agent may comprise a sequence comprising any two of a CDR1, CDR2 or CDR3 sequence as shown in Table 9 or Table 30 (that is either a CDR1 and CDR2, a CDR1 and CDR3, or a CDR2 and CDR3).
  • the targeted binding agent may comprise a sequence comprising a CDR1, CDR2 and CDR3 sequence as shown in Table 9 or Table 30.
  • the targeted binding agent of the invention may comprise an antigen-binding site within a non-antibody molecule, normally provided by one or more CDRs e.g. a set of CDRs in a non-antibody protein scaffold, as discussed further below.
  • the targeted binding agent comprises a sequence comprising any one of a CDR1, CDR2 or CDR3 sequence of fully human monoclonal antibodies sc 264 RAD, sc 264 RAD/ADY, sc 188 SDM, sc 133, sc 133 TMT, sc 133 WDS, sc 133 TMT/WDS, sc 188, sc 254, sc 264 or sc 298.
  • the targeted binding agent comprises any two of a CDR1, CDR2 or CDR3 sequence of fully human monoclonal antibodies sc 264 RAD, sc 264 RAD/ADY, sc 188 SDM, sc 133, sc 133 TMT, sc 133 WDS, sc 133 TMT/WDS, sc 188, sc 254, sc 264 or sc 298 (that is either a CDR1 and CDR2, a CDR1 and CDR3 or a CDR2 and CDR3).
  • the targeted binding agent comprises a CDR1, CDR2 and CDR3 sequence of fully human monoclonal antibodies sc 264 RAD, sc 264 RAD/ADY, sc 188 SDM, sc 133, sc 133 TMT, sc 133 WDS, sc 133 TMT/WDS, sc 188, sc 254, sc 264 or sc 298.
  • the targeted binding agent is an antibody. In one embodiment of the invention, the targeted binding agent is a monoclonal antibody. In one embodiment of the invention, the targeted binding agent is a fully human monoclonal antibody.
  • Another embodiment of the invention comprises an antibody that binds to ⁇ V ⁇ 6 and comprises a light chain amino acid sequence comprising any one of a CDR1, CDR2 or CDR3 sequence as shown in Table 9 or Table 30.
  • Another embodiment of the invention comprises an antibody that binds to ⁇ V ⁇ 6 and comprises a light chain amino acid sequence comprising any two of a CDR1, CDR2 or CDR3 sequence as shown in Table 9 or Table 30 (that is either a CDR1 and CDR2, a CDR1 and CDR3 or a CDR2 and CDR3).
  • Another embodiment of the invention comprises an antibody that binds to ⁇ V ⁇ 6 and comprises a light chain amino acid sequence comprising a CDR1, a CDR2 and a CDR3 sequence as shown in Table 9 or Table 30.
  • the antibody is a fully human monoclonal antibody.
  • Yet another embodiment of the invention comprises an antibody that binds to ⁇ V ⁇ 6 and comprises a heavy chain amino acid sequence comprising any one of a CDR1, CDR2 or CDR3 sequence as shown in Table 8 or Table 29.
  • Another embodiment of the invention comprises an antibody that binds to ⁇ V ⁇ 6 and comprises a heavy chain amino acid sequence comprising any two of a CDR1, CDR2 or CDR3 sequence as shown in Table 8 or Table 29 (that is either a CDR1 and CDR2, a CDR1 and CDR3 or a CDR2 and CDR3).
  • Another embodiment of the invention comprises an antibody that binds to ⁇ V ⁇ 6 and comprises a heavy chain amino acid sequence comprising a CDR1, a CDR2 and a CDR3 sequence as shown in Table 8 or Table 29.
  • the antibody is a fully human monoclonal antibody.
  • One embodiment of the invention comprises one or more of fully human monoclonal antibodies sc 264 RAD, sc 264 RAD/ADY, sc 188 SDM, sc 133, sc 133 TMT, sc 133 WDS, sc 133 TMT/WDS, sc 188, sc 254, sc 264 or sc 298 which specifically bind to ⁇ V ⁇ 6, as discussed in more detail below.
  • Yet another embodiment is an antibody that binds to ⁇ V ⁇ 6 and comprises a light chain amino acid sequence having a CDR comprising one of the sequences shown in Table 9 or Table 30.
  • Another embodiment is an antibody that binds to ⁇ V ⁇ 6 and comprises a heavy chain amino acid sequence having a CDR comprising one of the sequences shown in Table 8 or Table 29.
  • the antibody is a fully human monoclonal antibody.
  • a further embodiment is an antibody that binds to ⁇ V ⁇ 6 and comprises a heavy chain amino acid sequence having one of the CDR sequences shown in Table 8 or Table 29 and a light chain amino acid sequence having one of the CDR sequences shown in Table 9 or Table 30.
  • the antibody is a fully human monoclonal antibody.
  • a further embodiment of the invention is a targeted binding agent (i.e. an antibody) that competes for binding to ⁇ V ⁇ 6 with the antibodies of the invention.
  • said targeted binding agent comprises a heavy chain amino acid sequence having at least one of the CDR sequences shown in Table 8 or Table 29 and a light chain amino acid sequence having at least one of the CDR sequences shown in Table 9 or Table 30.
  • a further embodiment of the invention is a targeted binding agent that binds to the same epitope on ⁇ V ⁇ 6 as the antibodies of the invention.
  • said targeted binding agent comprised a heavy chain amino acid sequence having at least one of the CDR sequences shown in Table 8 or Table 29 and a light chain amino acid sequence having at least one of the CDR sequences shown in Table 9 or Table 30.
  • the targeted binding agent comprises a sequence comprising any one of a CDR1, CDR2 or CDR3 sequence as shown in Table 8 or Table 29 and any one of a CDR1, CDR2 or CDR3 sequence as shown in Table 9 or Table 30.
  • the targeted binding agent comprises any two of a CDR1, CDR2 or CDR3 sequence shown in Table 8 or Table 29 and any two of a CDR1, CDR2 or CDR3 sequence as shown in Table 9 or Table 30 (that is either a CDR1 and CDR2, a CDR1 and CDR3 or a CDR2 and CDR3).
  • the targeted binding agent comprises a CDR1, CDR2 and CDR3 sequence as shown in Table 8 or Table 29 and a CDR1, CDR2 and CDR3 sequence as shown in Table 9 or Table 30.
  • a binding agent of the invention may comprise an antigen-binding site within a non-antibody molecule, normally provided by one or more CDRs e.g. a set of CDRs in a non-antibody protein scaffold, as discussed further below.
  • a still further embodiment is an antibody that binds to ⁇ V ⁇ 6 and comprises an amino acid sequence having one or more corrective mutations where the antibody sequence is mutated back to its respective germline sequence.
  • the antibody can have a sequence as shown in any of Tables 10-13.
  • the invention further provides methods for assaying the level of ⁇ V ⁇ 6 in a patient or patient sample, comprising contacting an anti- ⁇ V ⁇ 6 antibody with a biological sample from a patient, and detecting the level of binding between said antibody and ⁇ V ⁇ 6 in said sample.
  • the biological sample is blood, or plasma.
  • compositions comprising targeted binding agent, including an antibody or functional fragment thereof, and a pharmaceutically acceptable carrier.
  • Still further embodiments of the invention include methods of effectively treating an animal suffering from an ⁇ V ⁇ 6-related disease or disorder, including selecting an animal in need of treatment for a neoplastic or non-neoplastic disease, and administering to the animal a therapeutically effective dose of a fully human monoclonal antibody that specifically binds to ⁇ V ⁇ 6.
  • the antibodies of the invention can be used to treat an ⁇ V ⁇ 6-related disease or disorder.
  • An ⁇ V ⁇ 6-related disease or disorder can be any condition arising due to the aberrant activation or expression of ⁇ V ⁇ 6. Examples of such diseases include where ⁇ V ⁇ 6 aberrantly interacts with its ligands thereby altering cell-adhesion or cell signaling properties. This alteration in cell adhesion or cell signaling properties can result in neoplastic diseases.
  • Other ⁇ V ⁇ 6-related diseases or disorders include inflammatory disorders, lung disease, diseases associated with fibrosis and any disease associated with dysregulated TGF- ⁇ .
  • the ⁇ V ⁇ 6-related disease is a neoplastic disease such as melanoma, small cell lung cancer, non-small cell lung cancer, glioma, hepatocellular (liver) carcinoma, thyroid tumor, gastric (stomach) cancer, prostate cancer, breast cancer, ovarian cancer, bladder cancer, lung cancer, glioblastoma, endometrial cancer, kidney cancer, colon cancer, pancreatic cancer, esophageal carcinoma, head and neck cancers, mesothelioma, sarcomas, biliary (cholangiocarcinoma), small bowel adenocarcinoma, pediatric malignancies and epidermoid carcinoma.
  • a neoplastic disease such as melanoma, small cell lung cancer, non-small cell lung cancer, glioma, hepatocellular (liver) carcinoma, thyroid tumor, gastric (stomach) cancer, prostate cancer, breast cancer, ovarian cancer, bladder cancer, lung cancer, glio
  • the ⁇ V ⁇ 6-related disease is an inflammatory disorder such as inflammatory bowel disease; systemic lupus erythematosus; rheumatoid arthritis; juvenile chronic arthritis; spondyloarthropathies; systemic sclerosis, for example, scleroderma; idiopathic inflammatory myopathies for example, dermatomyositis, polymyositis; Sjogren's syndrome; systemic vaculitis; sarcoidosis; thyroiditis, for example, Grave's disease, Hashimoto's thyroiditis, juvenile lymphocytic thyroiditis, atrophic thyroiditis; immune-mediated renal disease, for example, glomerulonephritis, tubulointerstitial nephritis; demyelinating diseases of the central and peripheral nervous systems such as multiple sclerosis, idiopathic polyneuropathy; hepatobiliary diseases such as infectious hepatitis such as hepatitis
  • the ⁇ V ⁇ 6-related disease is fibrosis such as kidney or lung fibrosis.
  • the ⁇ V ⁇ 6-related disease is associated with dysregulated TGF- ⁇ include cancer and connective tissue (fibrotic) disorders.
  • Additional embodiments of the invention include methods of inhibiting ⁇ V ⁇ 6 induced cell adhesion in an animal. These methods include selecting an animal in need of treatment for ⁇ V ⁇ 6 induced cell adhesion, and administering to said animal a therapeutically effective dose of a fully human monoclonal antibody wherein said antibody specifically binds to ⁇ V ⁇ 6.
  • inventions include the use of an antibody in the preparation of medicament for the treatment of an ⁇ V ⁇ 6 related disease or disorder in an animal, wherein said monoclonal antibody specifically binds to ⁇ V ⁇ 6.
  • the targeted binding agents described herein can be used for the preparation of a medicament for the effective treatment of ⁇ V ⁇ 6 induced cell adhesion in an animal, wherein said monoclonal antibody specifically binds to ⁇ V ⁇ 6.
  • Embodiments of the invention described herein relate to monoclonal antibodies that bind ⁇ V ⁇ 6 and affect ⁇ V ⁇ 6 function.
  • Other embodiments relate to fully human anti- ⁇ V ⁇ 6 antibodies and anti- ⁇ V ⁇ 6 antibody preparations with desirable properties from a therapeutic perspective, including high binding affinity for ⁇ V ⁇ 6, the ability to neutralize ⁇ V ⁇ 6 in vitro and in vivo, and the ability to inhibit ⁇ V ⁇ 6 induced cell adhesion and tumor growth.
  • the invention includes antibodies that bind to ⁇ V ⁇ 6 with very high affinities (KD).
  • KD very high affinities
  • a human, rabbit, mouse, chimeric or humanized antibody that is capable of binding ⁇ V ⁇ 6 with a Kd less than, but not limited to, about 10 ⁇ 5 , 10 ⁇ 6 , 10 ⁇ 7 , 10 ⁇ 8 , 10 ⁇ 9 , 10 ⁇ 10 or about 10 ⁇ 11 M, or any range or value therein.
  • Affinity and/or avidity measurements can be measured by KinExA® and/or BIACORE®, as described herein.
  • One embodiment of the invention includes isolated antibodies, or fragments of those antibodies, that bind to ⁇ V ⁇ 6.
  • the antibodies can be, for example, polyclonal, oligoclonal, monoclonal, chimeric, humanized, and/or fully human antibodies.
  • Embodiments of the invention described herein also provide cells for producing these antibodies.
  • the anti- ⁇ V ⁇ 6 antibody may be a full-length antibody (e.g., having an intact human Fc region) or an antibody fragment (e.g., a Fab, Fab′ or F(ab′) 2 , FV or Dab (Dabs are the smallest functional binding units of human antibodies).
  • the antibody may be manufactured from a hybridoma that secretes the antibody, or from a recombinantly produced cell that has been transformed or transfected with a gene or genes encoding the antibody.
  • embodiments of the invention include isolated nucleic acid molecules encoding any of the antibodies described herein, vectors having isolated nucleic acid molecules encoding anti- ⁇ V ⁇ 6 antibodies or a host cell transformed with any of such nucleic acid molecules.
  • one embodiment of the invention is a method of producing an anti- ⁇ V ⁇ 6 antibody by culturing host cells under conditions wherein a nucleic acid molecule is expressed to produce the antibody followed by recovering the antibody.
  • embodiments of the invention also include any nucleic acid molecule which encodes an antibody or fragment of an antibody of the invention including nucleic acid sequences optimized for increasing yields of antibodies or fragments thereof when transfected into host cells for antibody production.
  • a further embodiment includes a method of producing high affinity antibodies to ⁇ V ⁇ 6 by immunizing a mammal with human ⁇ V ⁇ 6, or a fragment thereof, and one or more orthologous sequences or fragments thereof.
  • inventions are based upon the generation and identification of isolated antibodies that bind specifically to ⁇ V ⁇ 6. Inhibition of the biological activity of ⁇ V ⁇ 6 can prevent ⁇ V ⁇ 6 induced cell adhesion and other desired effects.
  • Another embodiment of the invention includes a method of diagnosing diseases or conditions in which an antibody prepared as described herein is utilized to detect the level of ⁇ V ⁇ 6 in a patient sample.
  • the patient sample is blood or blood serum.
  • methods for the identification of risk factors, diagnosis of disease, and staging of disease is presented which involves the identification of the overexpression of ⁇ V ⁇ 6 using anti- ⁇ V ⁇ 6 antibodies.
  • Another embodiment of the invention includes a method for diagnosing a condition associated with the expression of ⁇ V ⁇ 6 in a cell by contacting the serum or a cell with an anti- ⁇ V ⁇ 6 antibody, and thereafter detecting the presence of ⁇ V ⁇ 6.
  • Preferred conditions include an ⁇ V ⁇ 6 related disease or disorder including, but not limited to, neoplastic diseases, such as, melanoma, small cell lung cancer, non-small cell lung cancer, glioma, hepatocellular (liver) carcinoma, glioblastoma, and carcinoma of the thyroid, stomach, prostate, breast, ovary, bladder, lung, uterus, kidney, colon, and pancreas, salivary gland, and colorectum.
  • neoplastic diseases such as, melanoma, small cell lung cancer, non-small cell lung cancer, glioma, hepatocellular (liver) carcinoma, glioblastoma, and carcinoma of the thyroid, stomach, prostate, breast, ovary, bladder, lung
  • the invention includes an assay kit for detecting ⁇ V ⁇ 6 in mammalian tissues, cells, or body fluids to screen for ⁇ V ⁇ 6-related diseases.
  • the kit includes an antibody that binds to ⁇ V ⁇ 6 and a means for indicating the reaction of the antibody with ⁇ V ⁇ 6, if present.
  • the antibody is a monoclonal antibody.
  • the antibody that binds ⁇ V ⁇ 6 is labeled.
  • the antibody is an unlabeled primary antibody and the kit further includes a means for detecting the primary antibody.
  • the means for detecting includes a labeled second antibody that is an anti-immunoglobulin.
  • the antibody may be labeled with a marker selected from the group consisting of a fluorochrome, an enzyme, a radionuclide and a radiopaque material.
  • compositions having an effective amount of an anti- ⁇ V ⁇ 6 antibody in admixture with a pharmaceutically acceptable carrier or diluent.
  • the anti- ⁇ V ⁇ 6 antibody, or a fragment thereof is conjugated to a therapeutic agent.
  • the therapeutic agent can be, for example, a toxin or a radioisotope.
  • Yet another embodiment includes methods for treating diseases or conditions associated with the expression of ⁇ V ⁇ 6 in a patient, by administering to the patient an effective amount of an anti- ⁇ V ⁇ 6 antibody.
  • the anti- ⁇ V ⁇ 6 antibody can be administered alone, or can be administered in combination with additional antibodies or chemotherapeutic drug or radiation therapy.
  • a monoclonal, oligoclonal or polyclonal mixture of ⁇ V ⁇ 6 antibodies that block cell adhesion can be administered in combination with a drug shown to inhibit tumor cell proliferation directly.
  • the method can be performed in vivo and the patient is preferably a human patient.
  • the method concerns the treatment of an ⁇ V ⁇ 6 related disease or disorder including, but not limited to, neoplastic diseases, such as, melanoma, small cell lung cancer, non-small cell lung cancer, glioma, hepatocellular (liver) carcinoma, glioblastoma, and carcinoma of the thyroid, stomach, prostate, breast, ovary, bladder, lung, uterus, kidney, colon, and pancreas, salivary gland, and colorectum.
  • neoplastic diseases such as, melanoma, small cell lung cancer, non-small cell lung cancer, glioma, hepatocellular (liver) carcinoma, glioblastoma, and carcinoma of the thyroid, stomach, prostate, breast, ovary, bladder, lung, uterus, kidney, colon, and pancreas, salivary gland, and colorectum.
  • the invention provides an article of manufacture including a container.
  • the container includes a composition containing an anti- ⁇ V ⁇ 6 antibody, and a package insert or label indicating that the composition can be used to treat an ⁇ V ⁇ 6 related disease or disorder characterized by the overexpression of ⁇ V ⁇ 6.
  • the anti- ⁇ V ⁇ 6 antibody is administered to a patient, followed by administration of a clearing agent to remove excess circulating antibody from the blood.
  • Yet another embodiment is the use of an anti- ⁇ V ⁇ 6 antibody in the preparation of a medicament for the treatment of ⁇ V ⁇ 6-related diseases or disorders such as neoplastic diseases, inflammatory disorders, fibrosis, lung disease or diseases associated with dysregulated TGF- ⁇ .
  • neoplastic diseases include carcinoma, such as breast, ovarian, stomach, endometrial, salivary gland, lung, kidney, colon, colorectum, esophageal, thyroid, pancreatic, prostate and bladder cancer.
  • the ⁇ V ⁇ 6 related diseases or disorders include, but are not limited to, neoplastic diseases, such as, melanoma, small cell lung cancer, non-small cell lung cancer, glioma, hepatocellular (liver) carcinoma, sarcoma, head and neck cancers, mesothelioma, biliary (cholangiocarcinoma), small bowel adenocarcinoma, pediatric malignancies and glioblastoma.
  • neoplastic diseases such as, melanoma, small cell lung cancer, non-small cell lung cancer, glioma, hepatocellular (liver) carcinoma, sarcoma, head and neck cancers, mesothelioma, biliary (cholangiocarcinoma), small bowel adenocarcinoma, pediatric malignancies and glioblastoma.
  • Yet another embodiment of the invention is the use of an anti- ⁇ V ⁇ 6 antibody in the preparation of a medicament for the treatment of inflammatory, or hyperprolifearative diseases including but not limited to arthritis, atherosclerosis, allergic conjunctivitis.
  • FIG. 1 is a line graph showing the ability of the purified monoclonal antibodies to bind to ⁇ V ⁇ 6 and block its binding to a GST-LAP peptide.
  • FIGS. 2A and 2B are line graphs showing a plot of the averaged Geometric Mean Fluorescence (GMF) as a function of molecular mAb concentration, which was used to estimate the binding affinity of one of the antibodies to K562 cells that stably express the human ⁇ V ⁇ 6 antigen. Shown in FIG. 2A is affinity data for mAb 188.
  • GMF Geometric Mean Fluorescence
  • FIG. 2B shows affinity data for mAb 264 RAD.
  • FIGS. 3A-3E are bar graphs showing the ability of the purified monoclonal antibodies to mediate complement-dependent cytotoxicity in 293 cells stably expressing ⁇ V ⁇ 6 integrin.
  • FIG. 4 is a bar graph showing the ability of antibodies 264RAD, 133 and 188 SDM to inhibit tumour growth using the Detroit-562 nasophayngeal cell line.
  • FIG. 5 is a bar chart showing comparison of the activity of 264 RAD with 264 RAD/ADY.
  • Embodiments of the invention relate to targeted binding agents that bind to ⁇ V ⁇ 6 integrin ( ⁇ V ⁇ 6).
  • the binding agents bind to ⁇ V ⁇ 6 and inhibit the binding of ligands to ⁇ V ⁇ 6.
  • the targeted binding agents are monoclonal antibodies, or binding fragments thereof.
  • the antibodies bind only to the ⁇ 6 chain yet are able to inhibit binding of ligands to ⁇ V ⁇ 6.
  • inventions include fully human anti- ⁇ V ⁇ 6 antibodies, and antibody preparations that are therapeutically useful.
  • the anti- ⁇ V ⁇ 6 antibody preparations have desirable therapeutic properties, including strong binding affinity for ⁇ V ⁇ 6, and the ability to inhibit TGF ⁇ LAP mediated cell adhesion in vitro.
  • Embodiments of the invention also include fully human isolated binding fragments of anti- ⁇ V ⁇ 6 antibodies.
  • the binding fragments are derived from fully human anti- ⁇ V ⁇ 6 antibodies.
  • Exemplary fragments include Fv, Fab′ or other well-known antibody fragments, as described in more detail below.
  • Embodiments of the invention also include cells that express fully human antibodies against ⁇ V ⁇ 6. Examples of cells include hybridomas, or recombinantly created cells, such as Chinese hamster ovary (CHO) cells, variants of CHO cells (for example DG44) and NSO cells that produce antibodies against ⁇ V ⁇ 6. Additional information about variants of CHO cells can be found in Andersen and Reilly (2004) Current Opinion in Biotechnology 15, 456-462 which is incorporated herein in its entirety by reference.
  • embodiments of the invention include methods of using these antibodies for treating an ⁇ V ⁇ 6-related disease or disorder.
  • An ⁇ V ⁇ 6-related disease or disorder can be any condition arising due to the aberrant activation or expression of ⁇ V ⁇ 6. Examples of such diseases include where ⁇ V ⁇ 6 aberrantly interacts with its ligands thereby altering cell-adhesion or cell signaling properties. This alteration in cell adhesion or cell signaling properties can result in neoplastic diseases.
  • Other ⁇ V ⁇ 6-related diseases or disorders include inflammatory disorders, lung disease, diseases associated with fibrosis and any disease associated with dysregulated TGF- ⁇ .
  • the ⁇ V ⁇ 6-related disease is a neoplastic disease such as melanoma, small cell lung cancer, non-small cell lung cancer, glioma, hepatocellular (liver) carcinoma, thyroid tumor, gastric (stomach) cancer, prostate cancer, breast cancer, ovarian cancer, bladder cancer, lung cancer, glioblastoma, endometrial cancer, kidney cancer, colon cancer, pancreatic cancer, esophageal carcinoma, head and neck cancers, mesothelioma, sarcomas, biliary (cholangiocarcinoma), small bowel adenocarcinoma, pediatric malignancies and epidermoid carcinoma.
  • a neoplastic disease such as melanoma, small cell lung cancer, non-small cell lung cancer, glioma, hepatocellular (liver) carcinoma, thyroid tumor, gastric (stomach) cancer, prostate cancer, breast cancer, ovarian cancer, bladder cancer, lung cancer, glio
  • the ⁇ V ⁇ 6-related disease is an inflammatory disorder such as inflammatory bowel disease; systemic lupus erythematosus; rheumatoid arthritis; juvenile chronic arthritis; spondyloarthropathies; systemic sclerosis, for example, scleroderma; idiopathic inflammatory myopathies for example, dermatomyositis, polymyositis; Sjogren's syndrome; systemic vaculitis; sarcoidosis; thyroiditis, for example, Grave's disease, Hashimoto's thyroiditis, juvenile lymphocytic thyroiditis, atrophic thyroiditis; immune-mediated renal disease, for example, glomerulonephritis, tubulointerstitial nephritis; demyelinating diseases of the central and peripheral nervous systems such as multiple sclerosis, idiopathic polyneuropathy; hepatobiliary diseases such as infectious hepatitis such as hepatitis
  • the ⁇ V ⁇ 6-related disease is fibrosis such as kidney or lung fibrosis.
  • the ⁇ V ⁇ 6-related disease is associated with dysregulated TGF- ⁇ include cancer and connective tissue (fibrotic) disorders.
  • kits for specifically determining the quantity of ⁇ V ⁇ 6 in a biological sample.
  • the assay kit can include anti- ⁇ V ⁇ 6 antibodies along with the necessary labels for detecting such antibodies.
  • These diagnostic assays are useful to screen for ⁇ V related diseases or ⁇ 6 disorders including, but not limited to, neoplastic diseases, such as, melanoma, small cell lung cancer, non-small cell lung cancer, glioma, hepatocellular (liver) carcinoma, glioblastoma, and carcinoma of the thyroid, stomach, prostate, breast, ovary, bladder, lung, uterus, kidney, colon, and pancreas, salivary gland, and colorectum.
  • neoplastic diseases such as, melanoma, small cell lung cancer, non-small cell lung cancer, glioma, hepatocellular (liver) carcinoma, glioblastoma, and carcinoma of the thyroid, stomach, prostate, breast, ovary, bladder, lung, uterus, kidney, colon, and
  • Another aspect of the invention is an antagonist of the biological activity of ⁇ V ⁇ 6 wherein the antagonist binds to ⁇ V ⁇ 6.
  • the antagonist is a targeted binding agent, such as an antibody.
  • the antagonist may bind to:
  • the antagonist of the biological activity of ⁇ V ⁇ 6 may bind to ⁇ V ⁇ 6 and thereby prevent TGF ⁇ LAP mediated cell adhesion.
  • One embodiment is an antibody which binds to the same epitope or epitopes as fully human monoclonal antibody c 264 RAD, sc 264 RAD/ADY, sc 188 SDM, sc 133, sc 133 TMT, sc 133 WDS, sc 133 TMT/WDS, sc 188, sc 254, sc 264 or sc 298.
  • the targeted binding agent binds ⁇ V ⁇ 6 with a K d of less than 100 nanomolar (nM).
  • the targeted binding agent may bind with a K d less than about 35 nanomolar (nM).
  • the targeted binding agent may bind with a K d less than about 25 nanomolar (nM).
  • the targeted binding agent may bind with a K d less than about 10 nanomolar (nM).
  • the targeted binding agent binds with a K d of less than about 60 picomolar (pM).
  • One embodiment is an antibody-secreting plasma cell that produces the light chain and/or the heavy chain of antibody as described hereinabove.
  • the plasma cell produces the light chain and/or the heavy chain of a fully human monoclonal antibody.
  • the plasma cell produces the light chain and/or the heavy chain of the fully human monoclonal antibody c 264 RAD, sc 264 RAD/ADY, sc 188 SDM, sc 133, sc 133 TMT, sc 133 WDS, sc 133 TMT/WDS, sc 188, sc 254, sc 264 or sc 298.
  • the plasma cell may produce an antibody which binds to the same epitope or epitopes as fully human monoclonal antibody sc c 264 RAD, sc 264 RAD/ADY, sc 188 SDM, sc 133, sc 133 TMT, sc 133 WDS, sc 133 TMT/WDS, sc 188, sc 254, sc 264 or sc 298.
  • fully human monoclonal antibody sc c 264 RAD, sc 264 RAD/ADY, sc 188 SDM, sc 133, sc 133 TMT, sc 133 WDS, sc 133 TMT/WDS, sc 188, sc 254, sc 264 or sc 298.
  • nucleic acid molecule encoding the light chain or the heavy chain of an antibody as described hereinabove.
  • the nucleic acid molecule encodes the light chain or the heavy chain of a fully human monoclonal antibody.
  • nucleic acid molecule encoding the light chain or the heavy chain of a fully human monoclonal antibody selected from antibodies c 264 RAD, sc 264 RAD/ADY, sc 188 SDM, sc 133, sc 133 TMT, sc 133 WDS, sc 133 TMT/WDS, sc 188, sc 254, sc 264 or sc 298.
  • Another embodiment of the invention is a vector comprising a nucleic acid molecule or molecules as described hereinabove, wherein the vector encodes a light chain and/or a heavy chain of an antibody as defined hereinabove.
  • Yet another embodiment of the invention is a host cell comprising a vector as described hereinabove.
  • the host cell may comprise more than one vector.
  • one embodiment of the invention is a method of producing an antibody by culturing host cells under conditions wherein a nucleic acid molecule is expressed to produce the antibody, followed by recovery of the antibody.
  • the invention includes a method of making an antibody by transfecting at least one host cell with at least one nucleic acid molecule encoding the antibody as described hereinabove, expressing the nucleic acid molecule in the host cell and isolating the antibody.
  • the invention includes a method of antagonising the biological activity of ⁇ V ⁇ 6 comprising administering an antagonist as described herein.
  • the method may include selecting an animal in need of treatment for an ⁇ V ⁇ 6 related disease or disorder, and administering to the animal a therapeutically effective dose of an antagonist of the biological activity of ⁇ V ⁇ 6.
  • Another aspect of the invention includes a method of antagonising the biological activity of ⁇ V ⁇ 6 comprising administering an antibody as described hereinabove.
  • the method may include selecting an animal in need of treatment for an ⁇ V ⁇ 6 related disease or disorder, and administering to said animal a therapeutically effective dose of an antibody which antagonises the biological activity of ⁇ V ⁇ 6.
  • a method of treating an ⁇ V ⁇ 6 related disease or disorder in a mammal comprising administering a therapeutically effective amount of an antagonist of the biological activity of ⁇ V ⁇ 6.
  • the method may include selecting an animal in need of treatment for an ⁇ V ⁇ 6 related disease or disorder, and administering to said animal a therapeutically effective dose of an antagonist of the biological activity of ⁇ V ⁇ 6.
  • a method of treating an ⁇ V ⁇ 6 disease or disorder in a mammal comprising administering a therapeutically effective amount of an antibody which antagonizes the biological activity of ⁇ V ⁇ 6.
  • the method may include selecting an animal in need of treatment for an ⁇ V ⁇ 6 related disease or disorder, and administering to said animal a therapeutically effective dose of an antibody which antagonises the biological activity of ⁇ V ⁇ 6.
  • the antibody can be administered alone, or can be administered in combination with additional antibodies or chemotherapeutic drug or radiation therapy.
  • a method of treating cancer in a mammal comprising administering a therapeutically effective amount of an antagonist of the biological activity of ⁇ V ⁇ 6.
  • the method may include selecting an animal in need of treatment for cancer, and administering to said animal a therapeutically effective dose of an antagonist which antagonises the biological activity of ⁇ V ⁇ 6.
  • the antagonist can be administered alone, or can be administered in combination with additional antibodies or chemotherapeutic drug or radiation therapy.
  • a method of treating cancer in a mammal comprising administering a therapeutically effective amount of an antibody which antagonizes the biological activity of ⁇ V ⁇ 6.
  • the method may include selecting an animal in need of treatment for cancer, and administering to said animal a therapeutically effective dose of an antibody which antagonises the biological activity of ⁇ V ⁇ 6.
  • the antibody can be administered alone, or can be administered in combination with additional antibodies or chemotherapeutic drug or radiation therapy.
  • an antagonist of the biological activity of ⁇ V ⁇ 6 for the manufacture of a medicament for the treatment of an ⁇ V ⁇ 6 related disease or disorder.
  • an antibody which antagonizes the biological activity of ⁇ V ⁇ 6 for the manufacture of a medicament for the treatment of an ⁇ V ⁇ 6 related disease or disorder.
  • the present invention is particularly suitable for use in antagonizing ⁇ V ⁇ 6, in patients with a tumor which is dependent alone, or in part, on ⁇ V ⁇ 6 integrin.
  • kits for detecting ⁇ V ⁇ 6 in mammalian tissues, cells, or body fluids to screen for an ⁇ V ⁇ 6 related disease or disorder.
  • the kit includes an antibody that binds to ⁇ V ⁇ 6 and a means for indicating the reaction of the antibody with ⁇ V ⁇ 6, if present.
  • the antibody may be a monoclonal antibody.
  • the antibody that binds ⁇ V ⁇ 6 is labeled.
  • the antibody is an unlabeled primary antibody and the kit further includes a means for detecting the primary antibody.
  • the means includes a labeled second antibody that is an anti-immunoglobulin.
  • the antibody is labeled with a marker selected from the group consisting of a fluorochrome, an enzyme, a radionuclide and a radio-opaque material.
  • Embodiments of the invention include the specific anti- ⁇ V ⁇ 6 antibodies listed below in Table 1. This table reports the identification number of each anti- ⁇ V ⁇ 6 antibody, along with the SEQ ID number of the variable domain of the corresponding heavy chain and light chain genes. Each antibody has been given an identification number that includes the letters “sc” followed by a number.
  • Standard techniques are used for recombinant DNA, oligonucleotide synthesis, and tissue culture and transformation (e.g., electroporation, lipofection). Enzymatic reactions and purification techniques are performed according to manufacturer's specifications or as commonly accomplished in the art or as described herein. The foregoing techniques and procedures are generally performed according to conventional methods well known in the art and as described in various general and more specific references that are cited and discussed throughout the present specification. See e.g., Sambrook et al., Molecular Cloning: A Laboratory Manual (3rd ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. (2001)), which is incorporated herein by reference.
  • An antagonist may be a polypeptide, nucleic acid, carbohydrate, lipid, small molecular weight compound, an oligonucleotide, an oligopeptide, RNA interference (RNAi), antisense, a recombinant protein, an antibody, or conjugates or fusion proteins thereof.
  • RNAi RNA interference
  • antisense see Opalinska J B, Gewirtz A M. (Sci STKE. 2003 Oct. 28; 2003 (206):pe47.)
  • Cell adhesion-related diseases include, but are not limited to, non-solid tumors such as leukemia, multiple myeloma or lymphoma, and also solid tumors such as melanoma, small cell lung cancer, non-small cell lung cancer, glioma, hepatocellular (liver) carcinoma, glioblastoma, carcinoma of the thyroid, bile duct, bone, gastric, brain/CNS, head and neck, hepatic system, stomach, prostate, breast, renal, testicle, ovary, skin, cervix, lung, muscle, neuron, oesophageal, bladder, lung, uterus, vulva, endometrium, kidney, colorectum, pancreas, pleural/peritoneal membranes, salivary gland, and epidermous.
  • non-solid tumors such as leukemia, multiple myeloma or lymphoma
  • solid tumors such as melanoma, small cell lung cancer, non-small cell
  • a compound refers to any small molecular weight compound with a molecular weight of less than about 2000 Daltons.
  • ⁇ V ⁇ 6 refers to the heterodimer integrin molecule consisting of an ⁇ V chain and a ⁇ 6 chain.
  • neutralizing when referring to a targeted binding agent, such as an antibody, relates to the ability of said targeted binding agent to eliminate, or significantly reduce, the activity of a target antigen. Accordingly, a “neutralizing” anti- ⁇ V ⁇ 6 antibody is capable of eliminating or significantly reducing the activity of ⁇ V ⁇ 6.
  • a neutralizing ⁇ V ⁇ 6 antibody may, for example, act by blocking the binding of TGF ⁇ LAP to the integrin ⁇ V ⁇ 6. By blocking this binding, ⁇ V ⁇ 6 mediated cell adhesion is significantly, or completely, eliminated. Ideally, a neutralizing antibody against ⁇ V ⁇ 6 inhibits cell adhesion.
  • isolated polynucleotide shall mean a polynucleotide that has been isolated from its naturally occurring environment. Such polynucleotides may be genomic, cDNA, or synthetic. Isolated polynucleotides preferably are not associated with all or a portion of the polynucleotides they associate with in nature. The isolated polynucleotides may be operably linked to another polynucleotide that it is not linked to in nature. In addition, isolated polynucleotides preferably do not occur in nature as part of a larger sequence.
  • isolated protein means a protein that has been isolated from its naturally occurring environment. Such proteins may be derived from genomic DNA, cDNA, recombinant DNA, recombinant RNA, or synthetic origin or some combination thereof, which by virtue of its origin, or source of derivation, the “isolated protein” (1) is not associated with proteins found in nature, (2) is free of other proteins from the same source, e.g. free of murine proteins, (3) is expressed by a cell from a different species, or (4) does not occur in nature.
  • polypeptide is used herein as a generic term to refer to native protein, fragments, or analogs of a polypeptide sequence. Hence, native protein, fragments, and analogs are species of the polypeptide genus.
  • Preferred polypeptides in accordance with the invention comprise the human heavy chain immunoglobulin molecules and the human kappa light chain immunoglobulin molecules, as well as antibody molecules formed by combinations comprising the heavy chain immunoglobulin molecules with light chain immunoglobulin molecules, such as the kappa or lambda light chain immunoglobulin molecules, and vice versa, as well as fragments and analogs thereof.
  • Preferred polypeptides in accordance with the invention may also comprise solely the human heavy chain immunoglobulin molecules or fragments thereof.
  • naturally-occurring refers to the fact that an object can be found in nature.
  • a polypeptide or polynucleotide sequence that is present in an organism (including viruses) that can be isolated from a source in nature and which has not been intentionally modified by man in the laboratory or otherwise is naturally-occurring.
  • operably linked refers to positions of components so described that are in a relationship permitting them to function in their intended manner.
  • a control sequence “operably linked” to a coding sequence is connected in such a way that expression of the coding sequence is achieved under conditions compatible with the control sequences.
  • polynucleotide as referred to herein means a polymeric form of nucleotides of at least 10 bases in length, either ribonucleotides or deoxynucleotides or a modified form of either type of nucleotide, or RNA-DNA hetero-duplexes.
  • the term includes single and double stranded forms of DNA.
  • oligonucleotide includes naturally occurring, and modified nucleotides linked together by naturally occurring, and non-naturally occurring linkages.
  • Oligonucleotides are a polynucleotide subset generally comprising a length of 200 bases or fewer.
  • oligonucleotides are 10 to 60 bases in length and most preferably 12, 13, 14, 15, 16, 17, 18, 19, or 20 to 40 bases in length.
  • Oligonucleotides are usually single stranded, e.g. for probes; although oligonucleotides may be double stranded, e.g. for use in the construction of a gene mutant.
  • Oligonucleotides can be either sense or antisense oligonucleotides.
  • nucleotides includes deoxyribonucleotides and ribonucleotides.
  • modified nucleotides referred to herein includes nucleotides with modified or substituted sugar groups and the like.
  • oligonucleotide linkages includes oligonucleotides linkages such as phosphorothioate, phosphorodithioate, phosphoroselenoate, phosphorodiselenoate, phosphoroanilothioate, phosphoraniladate, phosphoroamidate, and the like. See e.g., LaPlanche et al., Nucl. Acids Res.
  • oligonucleotide can include a label for detection, if desired.
  • the term “selectively hybridize” referred to herein means to detectably and specifically bind.
  • Polynucleotides, oligonucleotides and fragments thereof selectively hybridize to nucleic acid strands under hybridization and wash conditions that minimize appreciable amounts of detectable binding to nonspecific nucleic acids.
  • High stringency conditions can be used to achieve selective hybridization conditions as known in the art and discussed herein.
  • the nucleic acid sequence homology between the polynucleotides, oligonucleotides, or antibody fragments and a nucleic acid sequence of interest will be at least 80%, and more typically with preferably increasing homologies of at least 85%, 90%, 95%, 99%, and 100%.
  • CDR region or “CDR” is intended to indicate the hypervariable regions of the heavy and light chains of the immunoglobulin as defined by Kabat et al., 1991 (Kabat, E. A. et al., (1991) Sequences of Proteins of Immunological Interest, 5th Edition. US Department of Health and Human Services, Public Service, NIH, Washington), and later editions.
  • An antibody typically contains 3 heavy chain CDRs and 3 light chain CDRs.
  • the term CDR or CDRs is used here in order to indicate, according to the case, one of these regions or several, or even the whole, of these regions which contain the majority of the amino acid residues responsible for the binding by affinity of the antibody for the antigen or the epitope which it recognizes.
  • the third CDR of the heavy chain (HCDR3) has a greater size variability (greater diversity essentially due to the mechanisms of arrangement of the genes which give rise to it). It may be as short as 2 amino acids although the longest size known is 26. CDR length may also vary according to the length that can be accommodated by the particular underlying framework. Functionally, HCDR3 plays a role in part in the determination of the specificity of the antibody (Segal et al., PNAS, 71:4298-4302, 1974, Amit et al., Science, 233:747-753, 1986, Chothia et al., J. Mol.
  • a “set of CDRs” referred to herein comprises CDR1, CDR2 and CDR3.
  • a set of HCDRs refers to HCDR1, HCDR2 and HCDR3 (HCDR refers to a variable heavy chain CDR)
  • a set of LCDRs refers to LCDR1, LCDR2 and LCDR3 (LCDR refers to a variable light chain CDR).
  • a “set of CDRs” includes HCDRs and LCDRs.
  • Two amino acid sequences are “homologous” if there is a partial or complete identity between their sequences. For example, 85% homology means that 85% of the amino acids are identical when the two sequences are aligned for maximum matching. Gaps (in either of the two sequences being matched) are allowed in maximizing matching; gap lengths of 5 or less are preferred with 2 or less being more preferred. Alternatively and preferably, two protein sequences (or polypeptide sequences derived from them of at least about 30 amino acids in length) are homologous, as this term is used herein, if they have an alignment score of at more than 5 (in standard deviation units) using the program ALIGN with the mutation data matrix and a gap penalty of 6 or greater. See Dayhoff, M.
  • the two sequences or parts thereof are more preferably homologous if their amino acids are greater than or equal to 50% identical when optimally aligned using the ALIGN program. It should be appreciated that there can be differing regions of homology within two orthologous sequences. For example, the functional sites of mouse and human orthologues may have a higher degree of homology than non-functional regions.
  • polynucleotide sequence is homologous (i.e., is identical, not strictly evolutionarily related) to all or a portion of a reference polynucleotide sequence, or that a polypeptide sequence is identical to a reference polypeptide sequence.
  • the term “complementary to” is used herein to mean that the complementary sequence is homologous to all or a portion of a reference polynucleotide sequence.
  • the nucleotide sequence “TATAC” corresponds to a reference sequence “TATAC” and is complementary to a reference sequence “GTATA.”
  • sequence identity means that two polynucleotide or amino acid sequences are identical (i.e., on a nucleotide-by-nucleotide or residue-by-residue basis) over the comparison window.
  • percentage of sequence identity is calculated by comparing two optimally aligned sequences over the window of comparison, determining the number of positions at which the identical nucleic acid base (e.g., A, T, C, G, U, or I) or amino acid residue occurs in both sequences to yield the number of matched positions, dividing the number of matched positions by the total number of positions in the comparison window (i.e., the window size), and multiplying the result by 100 to yield the percentage of sequence identity.
  • substantially identical denotes a characteristic of a polynucleotide or amino acid sequence, wherein the polynucleotide or amino acid comprises a sequence that has at least 85 percent sequence identity, preferably at least 90 to 95 percent sequence identity, more preferably at least 99 percent sequence identity, as compared to a reference sequence over a comparison window of at least 18 nucleotide (6 amino acid) positions, frequently over a window of at least 24-48 nucleotide (8-16 amino acid) positions, wherein the percentage of sequence identity is calculated by comparing the reference sequence to the sequence which may include deletions or additions which total 20 percent or less of the reference sequence over the comparison window.
  • the reference sequence may be a subset of a larger sequence.
  • Examples of unconventional amino acids include: 4-hydroxyproline, ⁇ -carboxyglutamate, ⁇ -N,N,N-trimethyllysine, ⁇ -N-acetyllysine, O-phosphoserine, N-acetylserine, N-formylmethionine, 3-methylhistidine, 5-hydroxylysine, ⁇ -N-methylarginine, and other similar amino acids and imino acids (e.g., 4-hydroxyproline).
  • the left-hand direction is the amino terminal direction and the right-hand direction is the carboxy-terminal direction, in accordance with standard usage and convention.
  • the left-hand end of single-stranded polynucleotide sequences is the 5′ end; the left-hand direction of double-stranded polynucleotide sequences is referred to as the 5′ direction.
  • the direction of 5′ to 3′ addition of nascent RNA transcripts is referred to as the transcription direction; sequence regions on the DNA strand having the same sequence as the RNA and which are 5′ to the 5′ end of the RNA transcript are referred to as “upstream sequences”; sequence regions on the DNA strand having the same sequence as the RNA and which are 3′ to the 3′ end of the RNA transcript are referred to as “downstream sequences”.
  • the term “substantial identity” means that two peptide sequences, when optimally aligned, such as by the programs GAP or BESTFIT using default gap weights, share at least 80 percent sequence identity, preferably at least 90 percent sequence identity, more preferably at least 95 percent sequence identity, and most preferably at least 99 percent sequence identity.
  • residue positions that are not identical differ by conservative amino acid substitutions.
  • Conservative amino acid substitutions refer to the interchangeability of residues having similar side chains.
  • a group of amino acids having aliphatic side chains is glycine, alanine, valine, leucine, and isoleucine; a group of amino acids having aliphatic-hydroxyl side chains is serine and threonine; a group of amino acids having amide-containing side chains is asparagine and glutamine; a group of amino acids having aromatic side chains is phenylalanine, tyrosine, and tryptophan; a group of amino acids having basic side chains is lysine, arginine, and histidine; and a group of amino acids having sulfur-containing side chains is cysteine and methionine.
  • Preferred conservative amino acids substitution groups are: valine-leucine-isoleucine, phenylalanine-tyrosine, lysine-arginine, alanine-valine, glutamic-aspartic, and asparagine-glutamine.
  • amino acid sequences of antibodies or immunoglobulin molecules are contemplated as being encompassed by the present invention, providing that the variations in the amino acid sequence maintain at least about 75%, more preferably at least 80%, 90%, 95%, and most preferably about 99% sequence identity to the antibodies or immunoglobulin molecules described herein.
  • conservative amino acid replacements are contemplated. Conservative replacements are those that take place within a family of amino acids that have related side chains.
  • More preferred families are: serine and threonine are an aliphatic-hydroxy family; asparagine and glutamine are an amide-containing family; alanine, valine, leucine and isoleucine are an aliphatic family; and phenylalanine, tryptophan, and tyrosine are an aromatic family.
  • Whether an amino acid change results in a functional peptide can readily be determined by assaying the specific activity of the polypeptide derivative. Assays are described in detail herein. Fragments or analogs of antibodies or immunoglobulin molecules can be readily prepared by those of ordinary skill in the art. Preferred amino- and carboxy-termini of fragments or analogs occur near boundaries of functional domains. Structural and functional domains can be identified by comparison of the nucleotide and/or amino acid sequence data to public or proprietary sequence databases. Preferably, computerized comparison methods are used to identify sequence motifs or predicted protein conformation domains that occur in other proteins of known structure and/or function. Methods to identify protein sequences that fold into a known three-dimensional structure are known. Bowie et al., (1991) Science 253:164. Thus, the foregoing examples demonstrate that those of skill in the art can recognize sequence motifs and structural conformations that may be used to define structural and functional domains in accordance with the antibodies described herein.
  • a further aspect of the invention is a targeting binding agent or an antibody molecule comprising a VH domain that has at least about 60, 70, 80, 85, 90, 95, 98 or about 99% amino acid sequence identity with a VH domain of any of antibodies shown in Table 1, the appended sequence listing, an antibody described herein, or with an HCDR (e.g., HCDR1, HCDR2, or HCDR3) shown in Table 8 or Table 29.
  • HCDR e.g., HCDR1, HCDR2, or HCDR3
  • the targeting binding agent or antibody molecule may optionally also comprise a VL domain that has at least about 60, 70, 80, 85, 90, 95, 98 or about 99% amino acid sequence identity with a VL domain any of antibodies shown in Table 1, the appended sequence listing, an antibody described herein, or with an LCDR (e.g., LCDR1, LCDR2, or LCDR3) shown in Table 9 or Table 30.
  • Algorithms that can be used to calculate % identity of two amino acid sequences comprise e.g. BLAST (Altschul et al., (1990) J. Mol. Biol.
  • the targeting binding agent or antibody that shares amino acid sequence identity as describes above exhibits substantially the same activity as the antibodies referenced.
  • substantially the same activity comprises at least one activity that differed from the activity of the references antibodies by no more that about 50%, 40%, 30%, 20%, 10%, 5%, 2%, 1% or less.
  • An antigen binding site is generally formed by the variable heavy (VH) and variable light (VL) immunoglobulin domains, with the antigen-binding interface formed by six surface polypeptide loops, termed complimentarily determining regions (CDRs).
  • VH variable heavy
  • VL variable light
  • CDRs complimentarily determining regions
  • a VH domain is paired with a VL domain to provide an antibody antigen-binding site, although a VH or VL domain alone may be used to bind antigen.
  • the VH domain (e.g. from Table 1) may be paired with the VL domain (e.g. from Table 1), so that an antibody antigen-binding site is formed comprising both the VH and VL domains.
  • Analogous embodiments are provided for the other VH and VL domains disclosed herein.
  • VH chains in Table 8 or Table 29 are paired with a heterologous VL domain. Light-chain promiscuity is well established in the art. Again, analogous embodiments are provided by the invention for the other VH and VL domains disclosed herein.
  • the VH of the parent or of any of antibodies chain on Table 9 or Table 30 may be paired with the VL of the parent or of any of antibodies on Table 1 or other antibody.
  • An antigen binding site may comprise a set of H and/or L CDRs of the parent antibody or any of antibodies in Table 1 with as many as twenty, sixteen, ten, nine or fewer, e.g. one, two, three, four or five, amino acid additions, substitutions, deletions, and/or insertions within the disclosed set of H and/or L CDRs.
  • an antigen binding site may comprise a set of H and/or L CDRs of the parent antibody or any of antibodies Table 1 with as many as twenty, sixteen, ten, nine or fewer, e.g. one, two, three, four or five, amino acid substitutions within the disclosed set of H and/or L CDRs. Such modifications may potentially be made at any residue within the set of CDRs.
  • Preferred amino acid substitutions are those which: (1) reduce susceptibility to proteolysis, (2) reduce susceptibility to oxidation, (3) alter binding affinity for forming protein complexes, (4) alter binding affinities, and (4) confer or modify other physicochemical or functional properties of such analogs.
  • Analogs can include various muteins of a sequence other than the naturally-occurring peptide sequence. For example, single or multiple amino acid substitutions (preferably conservative amino acid substitutions) may be made in the naturally-occurring sequence (preferably in the portion of the polypeptide outside the domain(s) forming intermolecular contacts.
  • a conservative amino acid substitution should not substantially change the structural characteristics of the parent sequence (e.g., a replacement amino acid should not tend to break a helix that occurs in the parent sequence, or disrupt other types of secondary structure that characterizes the parent sequence).
  • Examples of art-recognized polypeptide secondary and tertiary structures are described in Proteins, Structures and Molecular Principles (Creighton, Ed., W. H. Freeman and Company, New York (1984)); Introduction to Protein Structure (C. Branden and J. Tooze, eds., Garland Publishing, New York, N.Y. (1991)); and Thornton et at. Nature 354:105 (1991), which are each incorporated herein by reference.
  • a further aspect of the invention is an antibody molecule comprising a VH domain that has at least about 60, 70, 80, 85, 90, 95, 98 or about 99% amino acid sequence identity with a VH domain of any of antibodies listed in Table 1, the appended sequence listing or described herein, or with an HCDR (e.g., HCDR1, HCDR2, or HCDR3) shown in Table 8 or Table 29.
  • HCDR e.g., HCDR1, HCDR2, or HCDR3
  • the antibody molecule may optionally also comprise a VL domain that has at least 60, 70, 80, 85, 90, 95, 98 or 99% amino acid sequence identity with a VL domain of any of the antibodies shown in Table 1, the appended sequence listing or described herein, or with an LCDR (e.g., LCDR1, LCDR2, or LCDR3) shown in Table 9 or Table 30.
  • Algorithms that can be used to calculate % identity of two amino acid sequences comprise e.g. BLAST (Altschul et al., (1990) J. Mol. Biol. 215: 405-410), FASTA (Pearson and Lipman (1988) PNAS USA 85: 2444-2448), or the Smith-Waterman algorithm (Smith and Waterman (1981) J. Mol. Biol. 147: 195-197), e.g. employing default parameters.
  • Variants of the VH and VL domains and CDRs of the present invention including those for which amino acid sequences are set out herein, and which can be employed in targeting agents and antibodies for ⁇ V ⁇ 6 can be obtained by means of methods of sequence alteration or mutation and screening for antigen targeting with desired characteristics.
  • desired characteristics include but are not limited to: increased binding affinity for antigen relative to known antibodies which are specific for the antigen; increased neutralization of an antigen activity relative to known antibodies which are specific for the antigen if the activity is known; specified competitive ability with a known antibody or ligand to the antigen at a specific molar ratio; ability to immunoprecipitate complex; ability to bind to a specified epitope; linear epitope, e.g.
  • peptide sequence identified using peptide-binding scan as described herein e.g. using peptides screened in linear and/or constrained conformation; conformational epitope, formed by non-continuous residues; ability to modulate a new biological activity of ⁇ V ⁇ 6, or downstream molecule.
  • peptide-binding scan as described herein, e.g. using peptides screened in linear and/or constrained conformation; conformational epitope, formed by non-continuous residues; ability to modulate a new biological activity of ⁇ V ⁇ 6, or downstream molecule.
  • Variants of antibody molecules disclosed herein may be produced and used in the present invention.
  • quantitative activity-property relationships of antibodies can be derived using well-known mathematical techniques, such as statistical regression, pattern recognition and classification (Norman et al., Applied Regression Analysis. Wiley-Interscience; 3rd edition (April 1998); Kandel, Abraham & Backer, Eric. Computer-Assisted Reasoning in Cluster Analysis.
  • the properties of antibodies can be derived from empirical and theoretical models (for example, analysis of likely contact residues or calculated physicochemical property) of antibody sequence, functional and three-dimensional structures and these properties can be considered singly and in combination.
  • An antibody antigen-binding site composed of a VH domain and a VL domain is typically formed by six loops of polypeptide: three from the light chain variable domain (VL) and three from the heavy chain variable domain (VH).
  • VL light chain variable domain
  • VH heavy chain variable domain
  • Analysis of antibodies of known atomic structure has elucidated relationships between the sequence and three-dimensional structure of antibody combining sites. These relationships imply that, except for the third region (loop) in VH domains, binding site loops have one of a small number of main-chain conformations: canonical structures.
  • the canonical structure formed in a particular loop has been shown to be determined by its size and the presence of certain residues at key sites in both the loop and in framework regions.
  • sequence-structure relationship can be used for prediction of those residues in an antibody of known sequence, but of an unknown three-dimensional structure, which are important in maintaining the three-dimensional structure of its CDR loops and hence maintain binding specificity. These predictions can be backed up by comparison of the predictions to the output from lead optimization experiments.
  • a model can be created of the antibody molecule using any freely available or commercial package, such as WAM.
  • a protein visualisation and analysis software package such as Insight II (Accelrys, Inc.) or Deep View may then be used to evaluate possible substitutions at each position in the CDR. This information may then be used to make substitutions likely to have a minimal or beneficial effect on activity.
  • Variant sequences may be made, with substitutions that may or may not be predicted to have a minimal or beneficial effect on activity, and tested for ability to bind and/or neutralize and/or for any other desired property.
  • VH and VL domains whose sequences are specifically disclosed herein may be employed in accordance with the present invention, as discussed.
  • polypeptide fragment refers to a polypeptide that has an amino-terminal and/or carboxy-terminal deletion, but where the remaining amino acid sequence is identical to the corresponding positions in the naturally-occurring sequence deduced, for example, from a full-length cDNA sequence. Fragments typically are at least about 5, 6, 8 or 10 amino acids long, preferably at least about 14 amino acids long, more preferably at least about 20 amino acids long, usually at least about 50 amino acids long, and even more preferably at least about 70 amino acids long.
  • analog refers to polypeptides which are comprised of a segment of at least about 25 amino acids that has substantial identity to a portion of a deduced amino acid sequence and which has at least one of the following properties: (1) specific binding to ⁇ V ⁇ 6, under suitable binding conditions, (2) ability to block appropriate ligand/ ⁇ V ⁇ 6 binding, or (3) ability to inhibit ⁇ V ⁇ 6 activity.
  • polypeptide analogs comprise a conservative amino acid substitution (or addition or deletion) with respect to the naturally-occurring sequence.
  • Analogs typically are at least 20 amino acids long, preferably at least 50 amino acids long or longer, and can often be as long as a full-length naturally-occurring polypeptide.
  • Peptide analogs are commonly used in the pharmaceutical industry as non-peptide drugs with properties analogous to those of the template peptide. These types of non-peptide compound are termed “peptide mimetics” or “peptidomimetics.” Fauchere, J. Adv. Drug Res. 15:29 (1986); Veber and Freidinger TINS p. 392 (1985); and Evans et al., J. Med. Chem. 30:1229 (1987), which are incorporated herein by reference. Such compounds are often developed with the aid of computerized molecular modeling. Peptide mimetics that are structurally similar to therapeutically useful peptides may be used to produce an equivalent therapeutic or prophylactic effect.
  • peptidomimetics are structurally similar to a paradigm polypeptide (i.e., a polypeptide that has a biochemical property or pharmacological activity), such as human antibody, but have one or more peptide linkages optionally replaced by a linkage selected from the group consisting of: —CH 2 NH—, —CH 2 S—, —CH 2 —CH 2 —, —CH ⁇ CH—(cis and trans), —COCH 2 —, —CH(OH)CH 2 —, and —CH 2 SO—, by methods well known in the art.
  • a paradigm polypeptide i.e., a polypeptide that has a biochemical property or pharmacological activity
  • linkages optionally replaced by a linkage selected from the group consisting of: —CH 2 NH—, —CH 2 S—, —CH 2 —CH 2 —, —CH ⁇ CH—(cis and trans), —COCH 2 —, —CH(OH)CH 2 —
  • Systematic substitution of one or more amino acids of a consensus sequence with a D-amino acid of the same type may be used to generate more stable peptides.
  • constrained peptides comprising a consensus sequence or a substantially identical consensus sequence variation may be generated by methods known in the art (Rizo and Gierasch Ann. Rev. Biochem. 61:387 (1992), incorporated herein by reference); for example, by adding internal cysteine residues capable of forming intramolecular disulfide bridges which cyclize the peptide.
  • antibody refers to a polypeptide or group of polypeptides that are comprised of at least one binding domain that is formed from the folding of polypeptide chains having three-dimensional binding spaces with internal surface shapes and charge distributions complementary to the features of an antigenic determinant of an antigen.
  • An antibody typically has a tetrameric form, comprising two identical pairs of polypeptide chains, each pair having one “light” and one “heavy” chain. The variable regions of each light/heavy chain pair form an antibody binding site.
  • a “targeted binding agent” is an agent, e.g. antibody, or binding fragment thereof, that preferentially binds to a target site.
  • the targeted binding agent is specific for only one target site. In other embodiments, the targeted binding agent is specific for more than one target site.
  • the targeted binding agent may be a monoclonal antibody and the target site may be an epitope.
  • a targeted binding agent may comprise at least one antigen binding domain of an antibody, wherein said domain is fused or contained within a heterologous protein.
  • Binding fragments of an antibody are produced by recombinant DNA techniques, or by enzymatic or chemical cleavage of intact antibodies. Binding fragments include Fab, Fab′, F(ab′) 2 , Fv, and single-chain antibodies. An antibody other than a “bispecific” or “bifunctional” antibody is understood to have each of its binding sites identical. An antibody substantially inhibits adhesion of a receptor to a counter-receptor when an excess of antibody reduces the quantity of receptor bound to counter-receptor by at least about 20%, 40%, 60% or 80%, and more usually greater than about 85% (as measured in an in vitro competitive binding assay).
  • An antibody may be oligoclonal, a polyclonal antibody, a monoclonal antibody, a chimeric antibody, a CDR-grafted antibody, a multi-specific antibody, a bi-specific antibody, a catalytic antibody, a chimeric antibody, a humanized antibody, a fully human antibody, an anti-idiotypic antibody and antibodies that can be labeled in soluble or bound form as well as fragments, variants or derivatives thereof, either alone or in combination with other amino acid sequences provided by known techniques.
  • An antibody may be from any species.
  • the term antibody also includes binding fragments of the antibodies of the invention; exemplary fragments include Fv, Fab, Fab′, single stranded antibody (svFC), dimeric variable region (Diabody) and disulphide stabilized variable region (dsFv).
  • binding fragments are (Ward, E. S. et al., (1989) Nature 341, 544-546) the Fab fragment consisting of VL, VH, CL and CH1 domains; (McCafferty et al., (1990) Nature, 348, 552-554) the Fd fragment consisting of the VH and CH1 domains; (Holt et al., (2003) Trends in Biotechnology 21, 484-490) the Fv fragment consisting of the VL and VH domains of a single antibody; (iv) the dAb fragment (Ward, E. S.
  • Fv, scFv or diabody molecules may be stabilized by the incorporation of disulphide bridges linking the VH and VL domains (Reiter, Y. et al., Nature Biotech, 14, 1239-1245, 1996).
  • Minibodies comprising a scFv joined to a CH3 domain may also be made (Hu, S. et al., (1996) Cancer Res., 56, 3055-3061).
  • binding fragments are Fab′, which differs from Fab fragments by the addition of a few residues at the carboxyl terminus of the heavy chain CH1 domain, including one or more cysteines from the antibody hinge region, and Fab′-SH, which is a Fab′ fragment in which the cysteine residue(s) of the constant domains bear a free thiol group.
  • epitopic determinants includes any protein determinant capable of specific binding to an immunoglobulin or T-cell receptor.
  • Epitopic determinants usually consist of chemically active surface groupings of molecules such as amino acids or sugar side chains and may, but not always, have specific three-dimensional structural characteristics, as well as specific charge characteristics.
  • An antibody is said to specifically bind an antigen when the dissociation constant is ⁇ 1 ⁇ M, preferably ⁇ 100 nM and most preferably ⁇ 10 nM.
  • agent is used herein to denote a chemical compound, a mixture of chemical compounds, a biological macromolecule, or an extract made from biological materials.
  • ⁇ V ⁇ 6 heterodimeric polypeptide refers to a portion of an ⁇ V ⁇ 6 heterodimeric polypeptide that has a biological or an immunological activity of a native ⁇ V ⁇ 6 polypeptide.
  • Biological when used herein refers to a biological function that results from the activity of the native ⁇ V ⁇ 6 polypeptide.
  • a preferred ⁇ V ⁇ 6 biological activity includes, for example, ⁇ V ⁇ 6 induced cell adhesion.
  • mammal when used herein refers to any animal that is considered a mammal. Preferably, the mammal is human.
  • Digestion of antibodies with the enzyme, papain results in two identical antigen-binding fragments, known also as “Fab” fragments, and a “Fc” fragment, having no antigen-binding activity but having the ability to crystallize.
  • Digestion of antibodies with the enzyme, pepsin results in the a F(ab′) 2 fragment in which the two arms of the antibody molecule remain linked and comprise two-antigen binding sites.
  • the F(ab′) 2 fragment has the ability to crosslink antigen.
  • Fv when used herein refers to the minimum fragment of an antibody that retains both antigen-recognition and antigen-binding sites.
  • Fab when used herein refers to a fragment of an antibody that comprises the constant domain of the light chain and the CH1 domain of the heavy chain.
  • mAb refers to monoclonal antibody.
  • “Liposome” when used herein refers to a small vesicle that may be useful for delivery of drugs that may include the ⁇ V ⁇ 6 polypeptide of the invention or antibodies to such an ⁇ V ⁇ 6 polypeptide to a mammal.
  • Label refers to the addition of a detectable moiety to a polypeptide, for example, a radiolabel, fluorescent label, enzymatic label chemiluminescent labeled or a biotinyl group.
  • Radioisotopes or radionuclides may include 3 H, 14 C, 15 N, 35 S 90 Y, 99 Tc, 111 In, 125 I, 131 I, fluorescent labels may include rhodamine, lanthanide phosphors or FITC and enzymatic labels may include horseradish peroxidase, ⁇ -galactosidase, luciferase, alkaline phosphatase.
  • Additional labels include, by way of illustration and not limitation: enzymes, such as glucose-6-phosphate dehydrogenase (“G6PDH”), alpha-D-galactosidase, glucose oxydase, glucose amylase, carbonic anhydrase, acetylcholinesterase, lysozyme, malate dehydrogenase and peroxidase; dyes; additional fluorescent labels or fluorescers include, such as fluorescein and its derivatives, fluorochrome, GFP (GFP for “Green Fluorescent Protein”), dansyl, umbelliferone, phycoerythrin, phycocyanin, allophycocyanin, o-phthaldehyde, and fluorescamine; fluorophores such as lanthanide cryptates and chelates e.g.
  • G6PDH glucose-6-phosphate dehydrogenase
  • alpha-D-galactosidase glucose oxydase
  • glucose amylase carbonic
  • chemoluminescent labels or chemiluminescers such as isoluminol, luminol and the dioxetanes; sensitizers; coenzymes; enzyme substrates; particles, such as latex or carbon particles; metal sol; crystallite; liposomes; cells, etc., which may be further labelled with a dye, catalyst or other detectable group; molecules such as biotin, digoxygenin or 5-bromodeoxyuridine; toxin moieties, such as for example a toxin moiety selected from a group of Pseudomonas exotoxin (PE or a cytotoxic fragment or mutant thereof), Diptheria toxin or a cytotoxic fragment or mutant thereof, a botulinum toxin A, B, C, D, E or F, ricin or a cytotoxic fragment thereof e.g. ricin A, abrin or a cytotoxic fragment thereof, saporin
  • PE Pseudomonas exotoxi
  • pharmaceutical agent or drug refers to a chemical compound or composition capable of inducing a desired therapeutic effect when properly administered to a patient.
  • Other chemistry terms herein are used according to conventional usage in the art, as exemplified by The McGraw - Hill Dictionary of Chemical Terms (Parker, S., Ed., McGraw-Hill, San Francisco (1985)), (incorporated herein by reference).
  • substantially pure means an object species is the predominant species present (i.e., on a molar basis it is more abundant than any other individual species in the composition), and preferably a substantially purified fraction is a composition wherein the object species comprises at least about 50 percent (on a molar basis) of all macromolecular species present. Generally, a substantially pure composition will comprise more than about 80 percent of all macromolecular species present in the composition, more preferably more than about 85%, 90%, 95%, and 99%. Most preferably, the object species is purified to essential homogeneity (contaminant species cannot be detected in the composition by conventional detection methods) wherein the composition consists essentially of a single macromolecular species.
  • patient includes human and veterinary subjects.
  • Human antibodies avoid some of the problems associated with antibodies that possess murine or rat variable and/or constant regions.
  • the presence of such murine or rat derived proteins can lead to the rapid clearance of the antibodies or can lead to the generation of an immune response against the antibody by a patient.
  • fully human antibodies can be generated through the introduction of functional human antibody loci into a rodent, other mammal or animal so that the rodent, other mammal or animal produces fully human antibodies.
  • XenoMouse® strains of mice that have been engineered to contain up to but less than 1000 kb-sized germline configured fragments of the human heavy chain locus and kappa light chain locus. See Mendez et al., Nature Genetics 15:146-156 (1997) and Green and Jakobovits J. Exp. Med. 188:483-495 (1998).
  • the XenoMouse® strains are available from Amgen, Inc. (Fremont, Calif.).
  • minilocus In an alternative approach, others, including GenPharm International, Inc., have utilized a “minilocus” approach. In the minilocus approach, an exogenous Ig locus is mimicked through the inclusion of pieces (individual genes) from the Ig locus. Thus, one or more V H genes, one or more D H genes, one or more J H genes, a mu constant region, and usually a second constant region (preferably a gamma constant region) are formed into a construct for insertion into an animal. This approach is described in U.S. Pat. No. 5,545,807 to Surani et al., and U.S. Pat. Nos.
  • Kirin has also demonstrated the generation of human antibodies from mice in which, through microcell fusion, large pieces of chromosomes, or entire chromosomes, have been introduced. See European Patent Application Nos. 773 288 and 843 961, the disclosures of which are hereby incorporated by reference. Additionally, KMTM-mice, which are the result of cross-breeding of Kirin's Tc mice with Medarex's minilocus (Humab) mice have been generated. These mice possess the human IgH transchromosome of the Kirin mice and the kappa chain transgene of the Genpharm mice (Ishida et al., Cloning Stem Cells, (2002) 4:91-102).
  • Human antibodies can also be derived by in vitro methods. Suitable examples include but are not limited to phage display (CAT, Morphosys, Dyax, Biosite/Medarex, Xoma, Symphogen, Alexion (formerly Proliferon), Affimed) ribosome display (CAT), yeast display, and the like.
  • mice were prepared through the utilization of the XenoMouse® technology, as described below. Such mice, then, are capable of producing human immunoglobulin molecules and antibodies and are deficient in the production of murine immunoglobulin molecules and antibodies. Technologies utilized for achieving the same are disclosed in the patents, applications, and references disclosed in the background section herein. In particular, however, a preferred embodiment of transgenic production of mice and antibodies therefrom is disclosed in U.S. patent application Ser. No. 08/759,620, filed Dec. 3, 1996 and International Patent Application Nos. WO 98/24893, published Jun. 11, 1998 and WO 00/76310, published Dec. 21, 2000, the disclosures of which are hereby incorporated by reference. See also Mendez et al., Nature Genetics 15:146-156 (1997), the disclosure of which is hereby incorporated by reference.
  • XenoMouse® lines of mice are immunized with an antigen of interest (e.g. ⁇ V ⁇ 6), lymphatic cells (such as B-cells) are recovered from the hyper-immunized mice, and the recovered lymphocytes are fused with a myeloid-type cell line to prepare immortal hybridoma cell lines.
  • lymphatic cells such as B-cells
  • myeloid-type cell line to prepare immortal hybridoma cell lines.
  • These hybridoma cell lines are screened and selected to identify hybridoma cell lines that produced antibodies specific to the antigen of interest.
  • Provided herein are methods for the production of multiple hybridoma cell lines that produce antibodies specific to ⁇ V ⁇ 6.
  • characterization of the antibodies produced by such cell lines including nucleotide and amino acid sequence analyses of the heavy and light chains of such antibodies.
  • B cells can be directly assayed.
  • CD19+ B cells can be isolated from hyperimmune XenoMouse® mice and allowed to proliferate and differentiate into antibody-secreting plasma cells.
  • Antibodies from the cell supernatants are then screened by ELISA for reactivity against the ⁇ V ⁇ 6 immunogen.
  • the supernatants might also be screened for immunoreactivity against fragments of ⁇ V ⁇ 6 to further map the different antibodies for binding to domains of functional interest on ⁇ V ⁇ 6.
  • the antibodies may also be screened against other related human integrins and against the rat, the mouse, and non-human primate, such as Cynomolgus monkey, orthologues of ⁇ V ⁇ 6, the last to determine species cross-reactivity.
  • B cells from wells containing antibodies of interest may be immortalized by various methods including fusion to make hybridomas either from individual or from pooled wells, or by infection with EBV or transfection by known immortalizing genes and then plating in suitable medium.
  • single plasma cells secreting antibodies with the desired specificities are then isolated using a ⁇ V ⁇ 6-specific hemolytic plaque assay (see for example Babcook et al., Proc. Natl. Acad. Sci. USA 93:7843-48 (1996)).
  • Cells targeted for lysis are preferably sheep red blood cells (SRBCs) coated with the ⁇ V ⁇ 6 antigen.
  • SRBCs sheep red blood cells
  • a plaque In the presence of a B-cell culture containing plasma cells secreting the immunoglobulin of interest and complement, the formation of a plaque indicates specific ⁇ V ⁇ 6-mediated lysis of the sheep red blood cells surrounding the plasma cell of interest.
  • the single antigen-specific plasma cell in the center of the plaque can be isolated and the genetic information that encodes the specificity of the antibody is isolated from the single plasma cell.
  • RT-PCR reverse-transcription followed by PCR
  • Such cloned DNA can then be further inserted into a suitable expression vector, preferably a vector cassette such as a pcDNA, more preferably such a pcDNA vector containing the constant domains of immunglobulin heavy and light chain.
  • a suitable expression vector preferably a vector cassette such as a pcDNA, more preferably such a pcDNA vector containing the constant domains of immunglobulin heavy and light chain.
  • the generated vector can then be transfected into host cells, e.g., HEK293 cells, CHO cells, and cultured in conventional nutrient media modified as appropriate for inducing transcription, selecting transformants, or amplifying the genes encoding the desired sequences.
  • antibodies produced by the fused hybridomas were human IgG2 heavy chains with fully human kappa or lambda light chains.
  • Antibodies described herein possess human IgG4 heavy chains as well as IgG2 heavy chains.
  • Antibodies can also be of other human isotypes, including IgG1.
  • the antibodies possessed high affinities, typically possessing a Kd of from about 10 ⁇ 6 through about 10 ⁇ 12 M or below, when measured by solid phase and solution phase techniques.
  • Antibodies possessing a Kd of at least 10 ⁇ 11 M are preferred to inhibit the activity of ⁇ V ⁇ 6.
  • antibodies can be expressed in cell lines other than hybridoma cell lines. Sequences encoding particular antibodies can be used to transform a suitable mammalian host cell. Transformation can be by any known method for introducing polynucleotides into a host cell, including, for example packaging the polynucleotide in a virus (or into a viral vector) and transducing a host cell with the virus (or vector) or by transfection procedures known in the art, as exemplified by U.S. Pat. Nos. 4,399,216, 4,912,040, 4,740,461, and 4,959,455 (which patents are hereby incorporated herein by reference). The transformation procedure used depends upon the host to be transformed.
  • Methods for introducing heterologous polynucleotides into mammalian cells include dextran-mediated transfection, calcium phosphate precipitation, polybrene mediated transfection, protoplast fusion, electroporation, encapsulation of the polynucleotide(s) in liposomes, and direct microinjection of the DNA into nuclei.
  • Mammalian cell lines available as hosts for expression are well known in the art and include many immortalized cell lines available from the American Type Culture Collection (ATCC), including but not limited to Chinese hamster ovary (CHO) cells, HeLa cells, baby hamster kidney (BHK) cells, monkey kidney cells (COS), human hepatocellular carcinoma cells (e.g., Hep G2), human epithelial kidney 293 cells, and a number of other cell lines. Cell lines of particular preference are selected through determining which cell lines have high expression levels and produce antibodies with constitutive ⁇ V ⁇ 6 binding properties.
  • ATCC American Type Culture Collection
  • the antibodies and methods herein relate to the treatment of symptoms resulting from ⁇ V ⁇ 6 induced cell adhesion or signaling induced as a result of ⁇ V ⁇ 6 interaction with it s ligands
  • a pharmaceutical composition comprising an antagonist of the biological activity of ⁇ V ⁇ 6, and a pharmaceutically acceptable carrier.
  • the antagonist comprises an antibody.
  • a pharmaceutical composition comprising an antagonist of the biological activity of ⁇ V ⁇ 6, and a pharmaceutically acceptable carrier.
  • the antagonist comprises an antibody.
  • Anti- ⁇ V ⁇ 6 antibodies are useful in the detection of ⁇ V ⁇ 6 in patient samples and accordingly are useful as diagnostics for disease states as described herein. In addition, based on their ability to significantly inhibit ⁇ V ⁇ 6 activity (as demonstrated in the Examples below), anti- ⁇ V ⁇ 6 antibodies have therapeutic effects in treating symptoms and conditions resulting from ⁇ V ⁇ 6 expression. In specific embodiments, the antibodies and methods herein relate to the treatment of symptoms resulting from ⁇ V ⁇ 6 induced cell adhesion.
  • ⁇ V ⁇ 6 related disease or disorder including neoplastic diseases, such as, melanoma, small cell lung cancer, non-small cell lung cancer, glioma, hepatocellular (liver) carcinoma, thyroid tumor, gastric (stomach) cancer, prostate cancer, breast cancer, ovarian cancer, bladder cancer, lung cancer, glioblastoma, endometrial cancer, kidney cancer, colon cancer, and pancreatic cancer.
  • neoplastic diseases such as, melanoma, small cell lung cancer, non-small cell lung cancer, glioma, hepatocellular (liver) carcinoma, thyroid tumor, gastric (stomach) cancer, prostate cancer, breast cancer, ovarian cancer, bladder cancer, lung cancer, glioblastoma, endometrial cancer, kidney cancer, colon cancer, and pancreatic cancer.
  • Embodiments of the invention include sterile pharmaceutical formulations of anti- ⁇ V ⁇ 6 antibodies that are useful as treatments for diseases. Such formulations would inhibit the binding of ligands to the ⁇ V ⁇ 6 integrin, thereby effectively treating pathological conditions where, for example, tissue ⁇ V ⁇ 6 is abnormally elevated.
  • Anti- ⁇ V ⁇ 6 antibodies preferably possess adequate affinity to potently inhibit ⁇ V ⁇ 6 activity, and preferably have an adequate duration of action to allow for infrequent dosing in humans. A prolonged duration of action will allow for less frequent and more convenient dosing schedules by alternate parenteral routes such as subcutaneous or intramuscular injection.
  • Sterile formulations can be created, for example, by filtration through sterile filtration membranes, prior to or following lyophilization and reconstitution of the antibody.
  • the antibody ordinarily will be stored in lyophilized form or in solution.
  • Therapeutic antibody compositions generally are placed into a container having a sterile access port, for example, an intravenous solution bag or vial having an adapter that allows retrieval of the formulation, such as a stopper pierceable by a hypodermic injection needle.
  • the route of antibody administration is in accord with known methods, e.g., injection or infusion by intravenous, intraperitoneal, intracerebral, intramuscular, intraocular, intraarterial, intrathecal, inhalation or intralesional routes, direct injection to a tumor site, or by sustained release systems as noted below.
  • the antibody is preferably administered continuously by infusion or by bolus injection.
  • an effective amount of antibody to be employed therapeutically will depend, for example, upon the therapeutic objectives, the route of administration, and the condition of the patient. Accordingly, it is preferred that the therapist titer the dosage and modify the route of administration as required to obtain the optimal therapeutic effect. Typically, the clinician will administer antibody until a dosage is reached that achieves the desired effect. The progress of this therapy is easily monitored by conventional assays or by the assays described herein.
  • Antibodies as described herein, can be prepared in a mixture with a pharmaceutically acceptable carrier.
  • This therapeutic composition can be administered intravenously or through the nose or lung, preferably as a liquid or powder aerosol (lyophilized).
  • the composition may also be administered parenterally or subcutaneously as desired.
  • the therapeutic composition should be sterile, pyrogen-free and in a parenterally acceptable solution having due regard for pH, isotonicity, and stability. These conditions are known to those skilled in the art.
  • dosage formulations of the compounds described herein are prepared for storage or administration by mixing the compound having the desired degree of purity with pharmaceutically acceptable carriers, excipients, or stabilizers.
  • Such materials are non-toxic to the recipients at the dosages and concentrations employed, and include buffers such as TRIS HCl, phosphate, citrate, acetate and other organic acid salts; antioxidants such as ascorbic acid; low molecular weight (less than about ten residues) peptides such as polyarginine, proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidinone; amino acids such as glycine, glutamic acid, aspartic acid, or arginine; monosaccharides, disaccharides, and other carbohydrates including cellulose or its derivatives, glucose, mannose, or dextrins; chelating agents such as EDTA; sugar alcohols such as mannitol or sorbitol; counterions such as sodium and/or nonionic surfactants such as TWEEN, PLURONICS or polyethyleneglycol.
  • buffers such as TRIS HCl, phosphate, citrate, a
  • Sterile compositions for injection can be formulated according to conventional pharmaceutical practice as described in Remington: The Science and Practice of Pharmacy (20th ed, Lippincott Williams & Wilkens Publishers (2003)). For example, dissolution or suspension of the active compound in a pharmaceutically acceptable carrier such as water or naturally occurring vegetable oil like sesame, peanut, or cottonseed oil or a synthetic fatty vehicle like ethyl oleate or the like may be desired. Buffers, preservatives, antioxidants and the like can be incorporated according to accepted pharmaceutical practice.
  • sustained-release preparations include semipermeable matrices of solid hydrophobic polymers containing the polypeptide, which matrices are in the form of shaped articles, films or microcapsules.
  • sustained-release matrices include polyesters, hydrogels (e.g., poly(2-hydroxyethyl-methacrylate) as described by Langer et al., J. Biomed Mater. Res ., (1981) 15:167-277 and Langer, Chem. Tech ., (1982) 12:98-105, or poly(vinylalcohol)), polylactides (U.S. Pat. No.
  • stabilization may be achieved by modifying sulfhydryl residues, lyophilizing from acidic solutions, controlling moisture content, using appropriate additives, and developing specific polymer matrix compositions.
  • the antibodies of the invention also encompass antibodies that have half-lives (e.g., serum half-lives) in a mammal, preferably a human, of greater than that of an unmodified antibody.
  • said antibody anybody half life is greater than 15 days, greater than 20 days, greater than 25 days, greater than 30 days, greater than 35 days, greater than 40 days, greater than 45 days, greater than 2 months, greater than 3 months, greater than 4 months, or greater than 5 months.
  • the increased half-lives of the antibodies of the present invention or fragments thereof in a mammal, preferably a human, result in a higher serum titer of said antibodies or antibody fragments in the mammal, and thus, reduce the frequency of the administration of said antibodies or antibody fragments and/or reduces the concentration of said antibodies or antibody fragments to be administered.
  • Antibodies or fragments thereof having increased in vivo half-lives can be generated by techniques known to those of skill in the art.
  • antibodies or fragments thereof with increased in vivo half-lives can be generated by modifying (e.g., substituting, deleting or adding) amino acid residues identified as involved in the interaction between the Fc domain and the FcRn receptor (see, e.g., International Publication Nos. WO 97/34631 and WO 02/060919, which are incorporated herein by reference in their entireties).
  • Antibodies or fragments thereof with increased in vivo half-lives can be generated by attaching to said antibodies or antibody fragments polymer molecules such as high molecular weight polyethyleneglycol (PEG).
  • PEG high molecular weight polyethyleneglycol
  • PEG can be attached to said antibodies or antibody fragments with or without a multifunctional linker either through site-specific conjugation of the PEG to the N- or C-terminus of said antibodies or antibody fragments or via epsilon-amino groups present on lysine residues.
  • Linear or branched polymer derivatization that results in minimal loss of biological activity will be used.
  • the degree of conjugation will be closely monitored by SDS-PAGE and mass spectrometry to ensure proper conjugation of PEG molecules to the antibodies.
  • Unreacted PEG can be separated from antibody-PEG conjugates by, e.g., size exclusion or ion-exchange chromatography.
  • Sustained-released compositions also include preparations of crystals of the antibody suspended in suitable formulations capable of maintaining crystals in suspension. These preparations when injected subcutaneously or intraperitonealy can produce a sustained release effect.
  • Other compositions also include liposomally entrapped antibodies. Liposomes containing such antibodies are prepared by methods known per se: U.S. Pat. No. DE 3,218,121; Epstein et al., Proc. Natl. Acad. Sci. USA , (1985) 82:3688-3692; Hwang et al., Proc. Natl. Acad. Sci.
  • the dosage of the antibody formulation for a given patient will be determined by the attending physician taking into consideration various factors known to modify the action of drugs including severity and type of disease, body weight, sex, diet, time and route of administration, other medications and other relevant clinical factors.
  • Therapeutically effective dosages may be determined by either in vitro or in vivo methods.
  • an effective amount of the antibodies, described herein, to be employed therapeutically will depend, for example, upon the therapeutic objectives, the route of administration, and the condition of the patient. Accordingly, it is preferred for the therapist to titer the dosage and modify the route of administration as required to obtain the optimal therapeutic effect.
  • a typical daily dosage might range from about 0.001 mg/kg to up to 100 mg/kg or more, depending on the factors mentioned above.
  • the clinician will administer the therapeutic antibody until a dosage is reached that achieves the desired effect. The progress of this therapy is easily monitored by conventional assays or as described herein.
  • compositions and methods herein will be administered with suitable carriers, excipients, and other agents that are incorporated into formulations to provide improved transfer, delivery, tolerance, and the like.
  • suitable carriers, excipients, and other agents that are incorporated into formulations to provide improved transfer, delivery, tolerance, and the like.
  • formulations include, for example, powders, pastes, ointments, jellies, waxes, oils, lipids, lipid (cationic or anionic) containing vesicles (such as LipofectinTM), DNA conjugates, anhydrous absorption pastes, oil-in-water and water-in-oil emulsions, emulsions carbowax (polyethylene glycols of various molecular weights), semi-solid gels, and semi-solid mixtures containing carbowax.
  • any of the foregoing mixtures may be appropriate in treatments and therapies in accordance with the present invention, provided that the active ingredient in the formulation is not inactivated by the formulation and the formulation is physiologically compatible and tolerable with the route of administration. See also Baldrick P. “Pharmaceutical excipient development: the need for preclinical guidance.” Regul. Toxicol. Pharmacol. 32(2):210-8 (2000), Wang W. “Lyophilization and development of solid protein pharmaceuticals.” Int. J. Pharm. 203(1-2):1-60 (2000), Charman W N “Lipids, lipophilic drugs, and oral drug delivery-some emerging concepts.” J Pharm Sci.
  • Such modalities include, without limitation, advanced antibody therapeutics, such as bispecific antibodies, immunotoxins, and radiolabeled therapeutics, single domain antibodies, generation of peptide therapeutics, ⁇ V ⁇ 6 binding domains in novel scaffolds, gene therapies, particularly intrabodies, antisense therapeutics, and small molecules.
  • Bispecific antibodies can be generated that comprise (i) two antibodies one with a specificity to ⁇ V ⁇ 6 and another to a second molecule that are conjugated together, (ii) a single antibody that has one chain specific to ⁇ V ⁇ 6 and a second chain specific to a second molecule, or (iii) a single chain antibody that has specificity to ⁇ V ⁇ 6 and the other molecule.
  • Such bispecific antibodies can be generated using techniques that are well known; for example, in connection with (i) and (ii) see e.g., Fanger et al., Immunol Methods 4:72-81 (1994) and Wright and Harris, supra. and in connection with (iii) see e.g., Traunecker et al., Int. J.
  • the second specificity can be made to the heavy chain activation receptors, including, without limitation, CD16 or CD64 (see e.g., Deo et al., Immunol. Today 18:127 (1997)) or CD89 (see e.g., Valerius et al., Blood 90:4485-4492 (1997)).
  • antibodies can be modified to act as immunotoxins utilizing techniques that are well known in the art. See e.g., Vitetta Immunol Today 14:252 (1993). See also U.S. Pat. No. 5,194,594.
  • modified antibodies can also be readily prepared utilizing techniques that are well known in the art. See e.g., Junghans et al., in Cancer Chemotherapy and Biotherapy 655-686 (2d edition, Chafner and Longo, eds., Lippincott Raven (1996)). See also U.S. Pat. Nos. 4,681,581, 4,735,210, 5,101,827, 5,102,990 (RE 35,500), 5,648,471, and 5,697,902.
  • An antigen binding site may be provided by means of arrangement of CDRs on non-antibody protein scaffolds, such as fibronectin or cytochrome B etc. (Haan & Ma ⁇ gos (2004) BioCentury, 12(5): A1-A6; Koide et al., (1998) Journal of Molecular Biology, 284: 1141-1151; Nygren et al., (1997) Current Opinion in Structural Biology, 7: 463-469) or by randomising or mutating amino acid residues of a loop within a protein scaffold to confer binding specificity for a desired target.
  • non-antibody protein scaffolds such as fibronectin or cytochrome B etc.
  • Protein scaffolds for antibody mimics are disclosed in WO/0034784, which is herein incorporated by reference in its entirety, in which the inventors describe proteins (antibody mimics) that include a fibronectin type III domain having at least one randomised loop.
  • a suitable scaffold into which to graft one or more CDRs, e.g. a set of HCDRs, may be provided by any domain member of the immunoglobulin gene superfamily.
  • the scaffold may be a human or non-human protein.
  • non-antibody protein scaffold may provide an antigen-binding site in a scaffold molecule that is smaller and/or easier to manufacture than at least some antibody molecules.
  • Small size of a binding agent may confer useful physiological properties, such as an ability to enter cells, penetrate deep into tissues or reach targets within other structures, or to bind within protein cavities of the target antigen.
  • Use of antigen binding sites in non-antibody protein scaffolds is reviewed in Wess, 2004 (Wess, L. In: BioCentury, The Bernstein Report on BioBusiness, 12(42), A1-A7, 2004).
  • Typical are proteins having a stable backbone and one or more variable loops, in which the amino acid sequence of the loop or loops is specifically or randomly mutated to create an antigen-binding site that binds the target antigen.
  • proteins include the IgG-binding domains of protein A from S. aureus , transferrin, albumin, tetranectin, fibronectin (e.g. 10th fibronectin type III domain), lipocalins as well as gamma-crystalline and other AffilinTM scaffolds (Scil Proteins).
  • Examples of other approaches include synthetic “Microbodies” based on cyclotides—small proteins having intra-molecular disulphide bonds, Microproteins (VersabodiesTM, Amunix) and ankyrin repeat proteins (DARPins, Molecular Partners).
  • a binding agent according to the present invention may comprise other amino acids, e.g. forming a peptide or polypeptide, such as a folded domain, or to impart to the molecule another functional characteristic in addition to ability to bind antigen.
  • Binding agents of the invention may carry a detectable label, or may be conjugated to a toxin or a targeting moiety or enzyme (e.g. via a peptidyl bond or linker).
  • a binding agent may comprise a catalytic site (e.g. in an enzyme domain) as well as an antigen binding site, wherein the antigen binding site binds to the antigen and thus targets the catalytic site to the antigen.
  • the catalytic site may inhibit biological function of the antigen, e.g. by cleavage.
  • anti-tumor treatment may be applied as a sole therapy or may involve, in addition to the compounds of the invention, conventional surgery or radiotherapy or chemotherapy.
  • chemotherapy may include one or more of the following categories of anti tumor agents:
  • antiproliferative/antineoplastic drugs and combinations thereof as used in medical oncology, such as alkylating agents (for example cis-platin, oxaliplatin, carboplatin, cyclophosphamide, nitrogen mustard, melphalan, chlorambucil, busulphan, temozolamide and nitrosoureas); antimetabolites (for example gemcitabine and antifolates such as fluoropyrimidines like 5-fluorouracil and tegafur, raltitrexed, methotrexate, cytosine arabinoside, and hydroxyurea); antitumour antibiotics (for example anthracyclines like adriamycin, bleomycin, doxorubicin, daunomycin, epirubicin, idarubicin, mitomycin-C, dactinomycin and mithramycin); antimitotic agents (for example vinca alkaloids like vincristine, vinblast
  • cytostatic agents such as antioestrogens (for example tamoxifen, fulvestrant, toremifene, raloxifene, droloxifene and iodoxyfene), antiandrogens (for example bicalutamide, flutamide, nilutamide and cyproterone acetate), LHRH antagonists or LHRH agonists (for example goserelin, leuprorelin and buserelin), progestogens (for example megestrol acetate), aromatase inhibitors (for example as anastrozole, letrozole, vorazole and exemestane) and inhibitors of 5 ⁇ -reductase such as finasteride;
  • antioestrogens for example tamoxifen, fulvestrant, toremifene, raloxifene, droloxifene and iodoxyfene
  • antiandrogens for example
  • anti-invasion agents for example c-Src kinase family inhibitors like 4-(6-chloro-2,3-methylenedioxyanilino)-7-[2-(4-methylpiperazin-1-yl)ethoxy]-5-tetrahydropyran-4-yloxyquinazoline (AZDO530; International Patent Application WO 01/94341) and N-(2-chloro-6-methylphenyl)-2- ⁇ 6-[4-(2-hydroxyethyl)piperazin-1-yl]-2-methylpyrimidin-4-ylamino ⁇ thiazole-5-carboxamide (dasatinib, BMS-354825 ; J. Med. Chem., 2004, 47, 6658-6661), and metalloproteinase inhibitors like marimastat, inhibitors of urokinase plasminogen activator receptor function or antibodies to Heparanase);
  • c-Src kinase family inhibitors like 4-(6-
  • inhibitors of growth factor function include growth factor antibodies and growth factor receptor antibodies (for example the anti-erbB2 antibody trastuzumab [HerceptinTM], the anti-EGFR antibody panitumumab, the anti-EGFR inhibitor Bevacizumab (AvastinTM), the anti-erbB1 antibody cetuximab [Erbitux, C225] and any growth factor or growth factor receptor antibodies disclosed by Stern et al., Critical reviews in oncology/haematology, 2005, Vol.
  • growth factor antibodies and growth factor receptor antibodies for example the anti-erbB2 antibody trastuzumab [HerceptinTM], the anti-EGFR antibody panitumumab, the anti-EGFR inhibitor Bevacizumab (AvastinTM), the anti-erbB1 antibody cetuximab [Erbitux, C225] and any growth factor or growth factor receptor antibodies disclosed by Stern et al., Critical reviews in oncology/haematology, 2005, Vol.
  • inhibitors also include tyrosine kinase inhibitors, for example inhibitors of the epidermal growth factor family (for example EGFR family tyrosine kinase inhibitors such as N-(3-chloro-4-fluorophenyl)-7-methoxy-6-(3-morpholinopropoxy)quinazolin-4-amine (gefitinib, ZD1839), N-(3-ethynylphenyl)-6,7-bis(2-methoxyethoxy)quinazolin-4-amine (erlotinib, OSI-774) and 6-acrylamido-N-(3-chloro-4-fluorophenyl)-7-(3-morpholinopropoxy)-quinazolin-4-amine (CI 1033), erbB2 tyrosine kinase inhibitors such as lapatinib, inhibitors of the hepatocyte growth factor family, inhibitors of the platelet-derived
  • antiangiogenic agents such as those which inhibit the effects of vascular endothelial growth factor, [for example the anti-vascular endothelial cell growth factor antibody bevacizumab (AvastinTM) and VEGF receptor tyrosine kinase inhibitors such as 4-(4-bromo-2-fluoroanilino)-6-methoxy-7-(1-methylpiperidin-4-ylmethoxy)quinazoline (ZD6474; Example 2 within WO 01/32651), 4-(4-fluoro-2-methylindol-5-yloxy)-6-methoxy-7-(3-pyrrolidin-1-ylpropoxy)quinazoline (AZD2171; Example 240 within WO 00/47212), vatalanib (PTK787; WO 98/35985) and SU11248 (sunitinib; WO 01/60814), compounds such as those disclosed in International Patent Applications WO97/22596, WO 97/
  • vascular damaging agents such as Combretastatin A4 and compounds disclosed in International Patent Applications WO 99/02166, WO 00/40529, WO 00/41669, WO 01/92224, WO 02/04434 and WO 02/08213;
  • antisense therapies for example those which are directed to the targets listed above, such as ISIS 2503, an anti-ras antisense;
  • gene therapy approaches including for example approaches to replace aberrant genes such as aberrant p53 or aberrant BRCA1 or BRCA2, GDEPT (gene-directed enzyme pro-drug therapy) approaches such as those using cytosine deaminase, thymidine kinase or a bacterial nitroreductase enzyme and approaches to increase patient tolerance to chemotherapy or radiotherapy such as multi-drug resistance gene therapy; and
  • GDEPT gene-directed enzyme pro-drug therapy
  • immunotherapy approaches including for example ex-vivo and in-vivo approaches to increase the immunogenicity of patient tumour cells, such as transfection with cytokines such as interleukin 2, interleukin 4 or granulocyte-macrophage colony stimulating factor, approaches to decrease T-cell anergy, approaches using transfected immune cells such as cytokine-transfected dendritic cells, approaches using cytokine-transfected tumor cell lines and approaches using anti-idiotypic antibodies.
  • cytokines such as interleukin 2, interleukin 4 or granulocyte-macrophage colony stimulating factor
  • Such conjoint treatment may be achieved by way of the simultaneous, sequential or separate dosing of the individual components of the treatment.
  • Such combination products employ the compounds of this invention, or pharmaceutically acceptable salts thereof, within the dosage range described hereinbefore and the other pharmaceutically active agent within its approved dosage range.
  • Immunizations were conducted using soluble ⁇ V ⁇ 6 and cell-bound ⁇ V ⁇ 36 (CHO transfectants expressing human ⁇ V ⁇ 6 at the cell surface), respectively.
  • human full length ⁇ V ⁇ 36 cDNA was inserted into the pcDNA 3 expression vector.
  • CHO cells were transiently transfected via electroporation. Expression of human ⁇ V ⁇ 6 on the cell surface at the level suitable for immunogen purpose was confirmed by Fluorescene-Activated Cell Sorter (FACS) analysis.
  • FACS Fluorescene-Activated Cell Sorter
  • mice of the antibody against human ⁇ V ⁇ 6 were tested by ELISA assay for mice immunized with soluble antigen. Titers of the antibody for mice immunized with native (cell-bound) antigen were tested by FACS. The ELISA and FACS analyses showed that there were some mice which appeared to be specific for ⁇ V ⁇ 6. Therefore, at the end of the immunization program, twenty mice were selected for harvest, and lymphocytes were isolated from the spleens and lymph nodes of the immunized mice, as described in Example 2.
  • lymphoid cells were dissociated by grinding in DMEM to release the cells from the tissues and the cells were suspended in DMEM.
  • B cells were enriched by negative selection in IgM and positive selection on IgG. The cells were cultured to allow B cell expansion and differentiation into antibody-secreting plasma cells.
  • Antibody-secreting plasma cells were grown as routine in the selective medium. Exhaustive supernatants collected from the cells that potentially produce anti-human ⁇ V ⁇ 6 antibodies were subjected to subsequent screening assays as detailed in the examples below.
  • binding of secreted antibodies to ⁇ V ⁇ 6 was assessed. Binding to cell-bound ⁇ V ⁇ 6 was assessed using an FMAT macroconfocal scanner, and binding to soluble ⁇ V ⁇ 6 was analyzed by ELISA, as described below.
  • Supernatants collected from harvested cells were tested to assess the binding of secreted antibodies to HEK 293 cells stably overexpressing ⁇ V ⁇ 6.
  • a parental 293F cell line was used as a negative control.
  • Cells in Freestyle media (Invitrogen) were seeded into 384-well FMAT plates in a volume of 50 ⁇ L/well at a density of 2500 cells/well for the stable transfectants, and at a density of 22,500 cells/well for the parental cells, and cells were incubated overnight at 37° C.
  • ⁇ V ⁇ 6 antibody binding to soluble ⁇ V ⁇ 6 was analyzed by ELISA.
  • Costar medium binding 96-well plates (Costar catalog #3368) were coated by incubating overnight at 4° C. with ⁇ V ⁇ 6 at a concentration of 5 ⁇ g/ml in TBS/1 mM MgCl 2 buffer for a total volume of 50 ⁇ L/well. Plates were then washed with TBS/1 mM MgCl 2 buffer, and blocked with 250 ⁇ L of 1 ⁇ PBS/1% milk for thirty minutes at room temperature. Ten ⁇ L of supernatant was then added to 40 ⁇ L TBS/1 mM MgCl 2 /1% milk and incubated for one hour at room temperature.
  • the antibodies were assessed for their ability to inhibit TGF ⁇ LAP-mediated adhesion of ⁇ V ⁇ 6-positive HT29 cells. Plates were coated overnight with 10 ⁇ g/ml TGF ⁇ LAP, and pre-blocked with 3% BSA/PBS for 1 hour prior to the assay. Cells were then pelleted and washed twice in HBBS, after which the cells were then resuspended in HBSS at appropriate concentrations. The cells were incubated in the presence of appropriate antibodies at 4° C. for 30 minutes in a V-bottom plate. The antigen coating solution was removed and the plates were blocked with 100 ⁇ L of 3% BSA for one hour at room temperature.
  • Cross-reactivity to human ⁇ V was also tested.
  • cross-reactivity was tested on the supernatants using FACS analysis on parental A375M cells, which express ⁇ V ⁇ 3 and ⁇ V ⁇ 5, but not ⁇ V ⁇ 6. This screen was designed to show that the antibodies were specifically recognizing either the ⁇ 6 chain or the ⁇ 6 chain in combination with ⁇ V.
  • the human ⁇ V assay was run at the same time as the macaque ⁇ V ⁇ 6 cross-reactivity screen.
  • the assays were performed as follows. A375M cells that were approximately 75% confluent were labeled with CFSE intracellular dye by dissociating and then pelleting cells (equivalent to 250,000 to 300,000 cells per well) in a falcon tube, then resuspending in 0.125 ⁇ M CFSE in FACS buffer to a final volume of 100 ⁇ L for every 250,000 cells, and then by incubating at 37° C. for five minutes. The cells were then pelleted, the supernatant discarded, and resuspended in FACS buffer and incubated for 30 minutes at 37° C. Cells were then washed twice with FACS buffer and resuspended in a final volume of 100 ⁇ L FACS buffer per well.
  • HEK-293 cells were transiently transfected with cynomolgus ⁇ V and cynomolgus ⁇ 6. After 48 hours, the cells were collected and resuspended in FACS buffer to reach a final concentration of approximately 50,000 cells in 100 ⁇ L.
  • Approximately 100,000 cells total comprising a 50:50 mix of CFSE-labeled A375M cells and transfected 293 cells, were used in each reaction as follows. 100 ⁇ L of CFSE-labeled A375M cells and 100 ⁇ L of 293 cells were dispensed into a V-bottom plate. The cells in the plate were pelleted at 1500 rpm for 3 minutes, and then resuspended in 100 ⁇ L FACS buffer. The pelleting step was repeated, and the FACS buffer supernatant was removed. The harvested antibody-containing supernatants, or control primary antibodies were added in a volume of 50 ⁇ L and the cells were resuspended.
  • Primary antibody controls were murine ⁇ V ⁇ 6 (Cat#MAB2077z, Chemicon) and an anti- ⁇ V recombinant.
  • the plate was incubated on ice for 45 minutes, after which 100 ⁇ L FACS buffer was added to dilute the primary antibody.
  • the cells were then pelleted by centrifuging at 1500 rpm for 3 minutes, and the pellet was resuspended in 100 ⁇ L FACS buffer. The pelleting step was repeated, and the FACS buffer supernatant was removed. Cells were then resuspended in the appropriate secondary antibody (5 ⁇ g/ml) with 7AAD dye (10 ⁇ g/ml), and stained on ice for 7 minutes.
  • FACS buffer 150 ⁇ L was added and the cells were pelLeted at 1500 rpm for 3 minutes, after which the cells were washed in 100 ⁇ L FACS buffer, pelleted, and then resuspended in 250 ⁇ L buffer and added to FACS tubes. Samples were analyzed on a high throughput FACS machine and analyzed using Cell Quest Pro software.
  • Antibody-secreting plasma cells were selected from each harvest for the production of recombinant antibodies.
  • a fluorescent plaque assay was used to identify single plasma cells expressing antibodies against ⁇ V ⁇ 6. Then, the single cells were subjected to reverse transcription and polymerase chain reaction to rescue and amplify the variable heavy and variable light chains that encoded the initial antibody specificity, as described in Example 7.
  • the preparation of a number of specialized reagents and materials needed to conduct the ⁇ V ⁇ 6-specific hemolytic plaque assay are described below.
  • SRBC Sheep red blood cells
  • the SRBCs were centrifuged at 3000 g for 5 minutes. The supernatant was drawn off and 25 mL PBS at pH 7.4 was added as a wash. The wash cycle was repeated 3 times, then 4.75 mL immune cell media (RPMI 1640 with 10% FCS) was added to the 250 ⁇ l biotinylated-SRBC (B-SRBC) pellet to gently re-suspend the B-SRBC (5% B-SRBC stock). The stock was stored at 4° C. until needed.
  • the cells were re-suspended in 50 ⁇ l of PBS and incubated with 2 ⁇ g/mL Gt-anti Human IgG Fc antibody conjugated to the photostable fluorescent dye Alexa488 (Molecular Probes, Eugene, Oreg.).
  • the tubes were rotated at room temperature for 25 min, followed by washing with 100 ⁇ l PBS and re-suspension in 10 ⁇ l PBS.
  • 10 ⁇ l of the stained cells were spotted onto a clean glass microscope slide, covered with a glass coverslip, observed under fluorescent light, and scored on an arbitrary scale of 0-4 to assess the quality of the isolated cells.
  • Plasma Cells The Contents of a Single B Cell Culture Well previously identified as neutralizing for ⁇ V ⁇ 6 activity (therefore containing a B cell clone secreting the immunoglobulin of interest), was harvested.
  • the B cell culture present in the well was recovered by addition of RPMI+10% FCS at 37° C.
  • the cells were re-suspended by pipetting and then transferred to a fresh 1.5 mL eppendorf tube (final volume approximately 500-700 ⁇ l).
  • the cells were centrifuged in a microfuge at 1500 rpm (240 rcf) for 2 minutes at room temperature, then the tube was rotated 180 degrees and centrifuged again for 2 minutes at 1500 rpm.
  • the freeze media was drawn off and the immune cells were resuspended in 100 ⁇ l RPMI (10% FCS), then centrifuged. This washing with RPMI (10% FCS) was repeated and the cells re-suspended in 60 ⁇ l RPMI (FCS) and stored on ice until ready to use.
  • Plaque Assay Results Parent Plate ID Plaque Assay Plate Row Column Assay Single Cells 68 B 10 Fluorescent 45-57 296 D 10 Fluorescent 58-59 318 F 1 Hemolytic 60-62 612 G 1 Fluorescent 187-189 752 D 12 Fluorescent 95-100 762 D 8 Fluorescent 277-286 766 B 5 Fluorescent 132-143, 147-150 827 E 12 Fluorescent 159-170 659 F 11 Fluorescent 252-263 761 H 3 Fluorescent 264-276 765 A 8 Fluorescent 287-298 652 D 2 Fluorescent 374-379, 392-397 806 A 6 Fluorescent 312-321
  • mRNA was extracted and reverse transcriptase PCR was conducted to generate cDNA encoding the variable heavy and light chains of the antibody secreted by each cell.
  • the human variable heavy chain cDNA was digested with restriction enzymes that were added during the PCR and the product of this reaction were cloned into an IgG2 expression vector with compatible overhangs for cloning. This vector was generated by cloning the constant domain of human IgG2 into the multiple cloning site of pcDNA3.1+/Hygro (Invitrogen, Burlington, Ontario, Canada).
  • the human variable light chain cDNA was digested with restriction enzymes that were added during the PCR reaction and the products of this reaction were cloned into an IgKappa or IgLamda expression vector with compatible overhangs for cloning.
  • This vector was generated by cloning the constant domain of human IgK or IgL into the multiple cloning site of pcDNA3.1+/Neo (Invitrogen).
  • the heavy chain and the light chain expression vectors were then co-transfected using lipofectamine into a 60 mm dish of 70% confluent human embryonal kidney (HEK) 293 cells.
  • the transfected cells secreted a recombinant antibody with the identical specificity as the original plasma cell for 24 to 72 hours.
  • the supernatant (3 mL) was harvested from the HEK 293 cells and the secretion of an intact antibody was demonstrated with a sandwich ELISA to specifically detect human IgG. Specificity was confirmed through binding of the recombinant antibody to ⁇ V ⁇ 6 using ELISA.
  • the rescued clones secreting antibody that could bind to the target antigen are summarized in Table 7.
  • heavy and light chain expression vectors 2.5 ⁇ g of each chain/dish were lipofected into ten 100 mm dishes that were 70% confluent with HEK 293 cells.
  • the transfected cells were incubated at 37° C. for 4 days, the supernatant (6 mL) was harvested and replaced with 6 mL of fresh media. At day 7, the supernatant was removed and pooled with the initial harvest (120 mL total from 10 plates).
  • the antibodies were purified from the supernatant using Protein-A Sepharose (Amersham Biosciences, Piscataway, N.J.) affinity chromatography (1 mL).
  • the antibodies were eluted from the Protein-A column with 500 ⁇ L of 0.1 M Glycine pH 2.5. The eluate was dialyzed in PBS pH 7.4 and filter sterilized. The antibodies were analyzed by non-reducing SDS-PAGE to assess purity and yield. Protein concentration was determined by determining the optical density at 280 nm.
  • variable heavy chains and the variable light chains of the antibodies were sequenced to determine their DNA sequences.
  • the complete sequence information for the anti- ⁇ V ⁇ 6 antibodies is provided in the sequence listing with nucleotide and amino acid sequences for each gamma and kappa/lambda chain combination.
  • the variable heavy sequences were analyzed to determine the VH family, the D-region sequence and the J-region sequence.
  • the sequences were then translated to determine the primary amino acid sequence and compared to the germline VH, D and J-region sequences to assess somatic hypermutations.
  • Table 8 is a table comparing the antibody heavy chain regions to their cognate germ line heavy chain region.
  • Table 9 is a table comparing the antibody kappa or lambda light chain regions to their cognate germ line light chain region.
  • the variable (V) regions of immunoglobulin chains are encoded by multiple germ line DNA segments, which are joined into functional variable regions (V H DJ H or V K J K ) during B-cell ontogeny.
  • V H DJ H or V K J K functional variable regions
  • the primary structure of the heavy chains of sc 298 and sc 374 are similar, but not identical.
  • sc 254 is structurally different from the other two. It should also be appreciated that where a particular antibody differs from its respective germline sequence at the amino acid level, the antibody sequence can be mutated back to the germline sequence. Such corrective mutations can occur at one, two, three or more positions, or a combination of any of the mutated positions, using standard molecular biological techniques.
  • Table 9 shows that the light chain sequence of sc 298 (SEQ ID NO.: 40) differs from the corresponding germline sequence (SEQ ID NO.:68) by a Val to Ala mutation (mutation 1) in the FR1 region, via a Leu to Ala mutation (mutation 2) in the CDR1 region and an Asn to Ser in the FR3 region.
  • the amino acid or nucleotide sequence encoding the light chain of sc 298 can be modified to change mutation 1 to yield the germline sequence at the site of mutation 1.
  • the amino acid or nucleotide sequence encoding the light chain of mAb sc 298 can be modified to change mutation 2 to yield the germline sequence at the site of mutation 2.
  • amino acid or nucleotide sequence encoding the light chain of mAb sc 298 can be modified to change mutation 3 to yield the germline sequence at the site of mutation 3. Still further again, the amino acid or nucleotide sequence encoding the light chain of sc 298 can be modified to change mutation 1, mutation 2 and mutation 3 to yield the germline sequence at the sites of mutations 1, 2 and 3. Still further again, the amino acid or nucleotide sequence encoding the light chain of sc 298 can be modified to change any combination of mutation 1, mutation 2 and mutation 3.
  • heavy chain of sc 264 (SEQ ID NO: 30) differs from it's germline (SEQ ID NO: 55) at position 61.
  • amino acid or nucleotide sequence encoding the heavy chain of sc 264 can be modified from a N to Y to yield the germline sequence.
  • Tables 10-13 below illustrate the position of such variations from the germline for sc 133, sc 188 and sc 264. Each row represents a unique combination of germline and non-germline residues at the position indicated by bold type.
  • an antibody sequence that can be mutated back to the germline sequence include: sc 133 where the L at amino acid 70 of the heavy chain is mutated back to the germline amino acid of M (referred to herein as sc 133 TMT); sc 133 where the N at amino acid 93 of the light chain is mutated back to the germline amino acid of D (referred to herein as sc 133 WDS); and sc 264 where the A at amino acid 84 of the light chain is mutated back to the germline amino acid of D (referred to herein as sc 264 ADY).
  • the invention features modifying one or more of the amino acids in the CDR regions, i.e., CDR1, CDR2 and/or CDR3.
  • the CDR3 of the heavy chain of an antibody described herein is modified.
  • the amino acid is substituted with an amino acid having a similar side chain (a conservative amino acid substitution) or can be substituted with any appropriate amino acid such as an alanine or a leucine.
  • the sc 264 CDR3, VATGRGDYHFYAMDV (amino acid residues 100-114 of SEQ ID NO: 30), can be modified at one or more amino acids.
  • the CDR3 region can be modified without adversely affecting activity, i.e., see sc 264 RAD where the second G in the CDR3 region is substituted for an A.
  • Other modifications within the CDR3 region are also envisaged.
  • the sc 133 CDR3 region, RLDV can be modified at one or more amino acids including substituting the L for an A and/or the V for an A.
  • Means of substituting amino acids are well known in the art and include site-directed mutagenesis.
  • the invention includes replacing any structural liabilities in the sequence that might affect the heterogeneity or specificity of binding of the antibodies of the invention.
  • the antibody sc 264 has an RGD sequence in the CDR3 region that might cause cross-reactive binding. Therefore the glycine residue in the RGD can be replaced with an alanine (sc 264 RAD).
  • TGF ⁇ LAP TGF Beta1 LAP
  • HT29 cells grown to the optimal density were then pelleted and washed twice in HBBS (with 1% BSA and without Mn 2+ ), after which the cells were then resuspended in HBSS at 30,000 cell per well.
  • the coating liquid was removed from the plates, which were then blocked with 100 ⁇ L 3% BSA at room temperature for 1 hour and thereafter washed twice with PBS.
  • Antibody titrations were prepared in 1:3 serial dilutions in a final volume of 30 ⁇ L and at two times the final concentration. Care was taken to ensure that the PBS concentration in the control wells matched the PBS concentration in the most dilute antibody well. 30 ⁇ L of cells were added to each well, and the cells were incubated in the presence of the antibodies at 4° C. for 40 minutes in a V-bottom plate. The cell-antibody mixtures were transferred to the coated plate and the plate was incubated at 37° C. for 40 minutes. The cells on the coated plates were then washed four times in warm HBSS, and the cells were thereafter frozen at ⁇ 80° C. for 15 minutes.
  • the cells were allowed to thaw at room temperature, and then 100 ⁇ L of CyQuant dye/lysis buffer (Molecular Probes) was added to each well according to the manufacturer's instructions. Fluorescence was read at an excitation wavelength of 485 nm and an emission wavelength of 530 nm. An estimated IC 50 value for each mAb was calculated based on the maximal and minimal amount of cell adhesion possible in the assay, as determined by positive and negative control wells. The results for twelve antibodies are summarized in Table 14.
  • the mAbs (in 1:3 serial dilutions ranging from 10 ⁇ g/ml to 0.01 ⁇ g/ml) were incubated overnight with ⁇ V ⁇ 6 (250 ng/ml in assay diluent containing 0.05% sodium azide). The following day, 50 ⁇ L/well of the pre-incubated primary antibody was transferred to the GST-LAP peptide-coated plate and incubated for one hour at room temperature. The wells were then washed three times using 300 ⁇ L/well of assay diluent.
  • mAb 2075 (Chemicon) was added at a concentration of 1 ⁇ g/ml in assay diluent (50 ⁇ L/well) and incubated for one hour at room temperature. The wells were then washed three times using 300 ⁇ L/well of assay diluent, and incubated with goat anti-mouse IgG Fc-peroxidase at 400 ng/ml in assay diluent (50 ⁇ L/well) for one hour at room temperature.
  • the following assay was performed to test the ability of the antibodies to inhibit the adhesion of A375M cells to an osteopontin peptide.
  • Assay plates were coated with osteopontin peptide. Two fragments of osteopontin were used: OPN 17-168 and OPN 17-314. Assay plates were pre-blocked with 3% BSA/PBS for one hour prior to the assay. The A375M cells were removed from a culture flask, pelleted and washed twice with HBSS containing 1% BSA and 1 mM Ca 2+ and 1 mM Mg 2+ . The cells were then resuspended in HBSS at a concentration of 30,000 cells per well. The coating liquid containing the osteopontin fragments was removed, and the plates were blocked with 100 ⁇ L of 3% BSA for one hour at room temperature.
  • the coated plates were washed twice with HBSS containing 1% BSA.
  • Antibody titrations were prepared in 1:4 serial dilutions in a final volume of 30 ⁇ L and at twice the final concentration.
  • the resuspended cells were added to the wells containing the titrated antibody in a V-bottom plate, and the cells and antibodies were co-incubated at 4° C. for 40 minutes.
  • the cell-antibody mixture was then transferred to the coated plate, which was thereafter incubated at 37° C. for 40 minutes.
  • the cells on the coated plates were next washed four times in warm HBSS, and the cells in the plates were then frozen at ⁇ 80° C. for 15 minutes.
  • the cells were allowed to thaw at room temperature, and then 100 ⁇ L of CyQuant dye/lysis buffer (Molecular Probes) was added to each well according to the manufacturer's instructions. Fluorescence was read at an excitation wavelength of 485 nm and an emission wavelength of 530 nm.
  • an assay was performed to test the ability of the antibodies to inhibit the adhesion of J6.77 cells to the CS-1 peptide of fibronectin.
  • the assay was performed as described in Example 12 above, with the exception that J6.77 cells were used for binding and the CS-1 peptide of fibronectin was used to coat the assay plates.
  • an adhesion assay was performed to test the ability of the antibodies to inhibit the adhesion of K562 cells to fibronectin.
  • Assay plates were coated with the FN9-10 peptide of fibronectin at a concentration of 12.5 ⁇ g/mL. Assay plates were pre-blocked with 3% BSA/PBS for one hour prior to the assay. The K562 cells were removed from a culture flask, pelleted and washed twice with HBSS containing 1% BSA and 1 mM Mn 2+ . The cells were then resuspended in HBSS at a concentration of 30,000 cells per well. The coating liquid containing the osteopontin fragments was removed, and the plates were blocked with 100 ⁇ L of 3% BSA for one hour at room temperature. The coated plates were washed twice with HBSS containing 1% BSA.
  • Antibody titrations were prepared in 1:4 serial dilutions in a final volume of 30 ⁇ L and at twice the final concentration.
  • the resuspended cells were added to the wells containing the titrated antibody in a V-bottom plate, and the cells and antibodies were co-incubated at 4° C. for 60 minutes.
  • the cell-antibody mixture was then transferred to the coated plate, which was thereafter incubated at 37° C. for 40 minutes.
  • the cells on the coated plates were next washed four times in warm HBSS, and the cells in the plates were then frozen at ⁇ 80° C. for 15 minutes.
  • the cells were allowed to thaw at room temperature, and then 100 ⁇ L of CyQuant dye/lysis buffer (Molecular Probes) was added to each well according to the manufacturer's instructions. Fluorescence was read at an excitation wavelength of 485 nm and an emission wavelength of 530 nm.
  • Test antibodies were compared to a commercially available ⁇ 5 ⁇ 1 antibody as a positive control and an ⁇ V ⁇ 6 specific antibody as a negative control. None of the test antibodies were able to block adhesion in the assay at the 5 ⁇ g/ml concentration used in this assay. TABLE 17 Cross-Reactivity to ⁇ 5 ⁇ 1 Integrin Percent Antibody ID Inhibition sc 133 ⁇ 12 sc 188 5 sc 254 ⁇ 9 sc 264 ⁇ 4 sc 298 2 ⁇ V ⁇ 6 Control 7 ⁇ 5 ⁇ 1 Control 78 Human IgG 11 Control
  • the plate was incubated on ice for 45 minutes, after which 100 ⁇ L FACS buffer was added to dilute the primary antibody.
  • the cells were then pelleted by centrifuging at 1500 rpm for 3 minutes, and the pellet was resuspended in 100 ⁇ L FACS buffer. The pelleting step was repeated, and the FACS buffer supernatant was removed. Cells were then resuspended in the appropriate secondary antibody (5 ⁇ g/ml) with 7AAD dye (10 ⁇ g/ml), and stained on ice for 7 minutes.
  • FACS buffer 150 ⁇ L was added and the cells were pelleted at 1500 rpm for 3 minutes, after which the cells were washed in 100 ⁇ L FACS buffer, pelleted, and then resuspended in 250 ⁇ L buffer and added to FACS tubes. Samples were analyzed on a high throughput FACS machine and analyzed using Cell Quest Pro software.
  • the internalization of the antibodies was tested using a K562 cell line that stably expressed human ⁇ V ⁇ 6. Internalization of the purified antibodies was compared to a commercially available ⁇ V ⁇ 6 antibody that was not internalized in this assay.
  • the Biacore analysis was performed as follows. A high-density goat ⁇ human IgG antibody surface over two CM5 Biacore chips was prepared using routine amine coupling. Each mAb was diluted in degassed HBS-P running buffer containing 100 ⁇ g/ml BSA, 1 mM MgCl 2 , and 1 mM CaCl 2 to a concentration of approximately 1 ⁇ g/mL.
  • mAb sc 133 was diluted to 0.98 ⁇ g/mL
  • mAb sc 188 was diluted to 0.96 ⁇ g/mL
  • mAb sc 264 was diluted to 0.94 ⁇ g/mL
  • mAb sc 254.2 was diluted to 0.87 ⁇ g/mL
  • mAb sc 298 was diluted to 1.6 ⁇ g/mL.
  • a capture level protocol was developed for each mAb by capturing each mAb over a separate flow cell at a 10 ⁇ L/min flow rate at the concentrations listed above.
  • mAbs sc 133, sc 298, and sc 254.2 were captured for 30 seconds while mAbs sc 188 and sc 264 were captured for 1 minute.
  • a 4-minute wash step at 50 ⁇ L/min followed to stabilize the mAb baseline.
  • Soluble ⁇ V ⁇ 6 was injected for 4 minutes at a concentration range of 116-3.6 nM for mAbs sc 133, sc 188, sc 264, and sc 298, and 233-3.6 nM for mAb sc 254.2, with a 2 ⁇ serial dilution for each concentration range.
  • a 10-minute dissociation followed each antigen injection.
  • the antigen samples were prepared in the HBS-P running described above. All samples were randomly injected in triplicate with several mAb capture/buffer inject cycles interspersed for double referencing.
  • the high-density goat ⁇ mouse antibody surfaces were regenerated with one 18-second pulse of 146 mM phosphoric acid (pH 1.5) after each cycle at a flow rate of 100 ⁇ L/min. A flow rate of 50 ⁇ L/min was used for all antigen injection cycles.
  • FACS analysis was also used to estimate the binding affinity of one of the antibodies to K562 cells that stably express the human ⁇ V ⁇ 6 antigen.
  • the amount of antigen was titrated to generate a binding curve and estimate the binding affinity to the antigen.
  • K562 cells expressing ⁇ V ⁇ 6 were resuspended in filtered HBS buffer containing 1 mM of MgCl 2 and 1 mM of CaCl 2 at a concentration of approximately 6 million cells/mL. The cells were kept on ice. Purified mAb sc 188 was serially diluted by a factor of 1:2 in HBS across 11 wells in a 96-well plate. The 12 th well in each row contained buffer only. Titrations were done in triplicate. Additional HBS and cells were added to each well so that the final volume was 300 ⁇ L/well and each well contained approximately 120,000 cells. The final molecular concentration range for mAb sc 188 was 4.9-0.019 nM.
  • the plates were placed into a plate shaker for 5 hours at 4° C., after which the plates were spun and washed three times with HBS, following which, 200 ⁇ L of 131 nM Cy5 goat ⁇ -human polyclonal antibody (Jackson Laboratories, #109-175-008) were added to each well. The plates were then shaken for 40 minutes at 4° C., and thereafter were spun and washed once again three times with HBS.
  • the Geometric Mean Fluorescence (GMF) of 20,000 “events” for each mAb concentration was recorded using a FACSCalibur instrument, and the corresponding triplicate titration points were averaged to give one GMF point for each mAb concentration.
  • F geometric mean fluorescence
  • L T total molecular mAb concentration
  • P′ proportionality constant that relates arbitrary fluorescence units to bound mAb
  • M cellular concentration in molarity
  • n number of receptors per cell
  • B background signal
  • K D equilibrium dissociation constant
  • Binding affinity for sc 188 as determined by FACS was significantly tighter than as determined by Biacore (Example 17).
  • Biacore Biacore
  • the first reason is that the two assays used different forms of the antigen for the measurement—Biacore used soluble antigen and the FACs analysis used a cell-bound form of the antigen.
  • the second reason is that the antibodies that were tested were raised against the cell-bound form of the antigen and may not bind with as high an affinity to the soluble antigen as they do to the cell-bound antigen.
  • the purified antibodies described in the examples above are of the IgG1 isotype and can have effector function.
  • the following assay was performed using 293 cells stably expressing ⁇ V ⁇ 6 (293-10A11) and parental 293 cells (293F).
  • calcein staining of cells For calcein staining of cells, aliquots of approximately 25 ⁇ 10e6 each of HT29, 293-10A11, and 293F cells were individually resuspended in 3 ml serum-free RPMI media. 45 ⁇ L of 1 mM calcein was then added to each 3 ml sample of cells, and the samples were incubated at 37° C. for 45 minutes. The cells were centrifuged at 1200 ⁇ RPM for 3 minutes, the supernatant was discarded and the cells were resuspended in each respective cell line's culture media. The centrifugation step was repeated and the cells were resuspended to give a final concentration of about 100,000 cells in 50 ⁇ L media.
  • Serial 1:2 dilutions of each antibody were prepared in a v-bottom 96-well plate, with concentrations ranging from 20 ⁇ g/ml to 0.625 ⁇ g/ml in a volume of 50 ⁇ L. Then, 100,000 of the cells prepared as described above were added in a volume of 50 ⁇ L to the antibody-containing plates, and the resulting mixture was incubated on ice for two hours. Following the incubation, the cells were pelleted, and the supernatant was discarded. The cells were resuspended in 100 ⁇ L of their respective media containing 10% human sera (ABI donor #27), and incubated at 37° C. for 30 minutes. The cells were then centrifuged, and 80 L of the supernatant was transferred to a FMAT plate. The plate was read on a Tecan reader using an excitation wavelength of 485 nm and an emission wavelength of 530 nm.
  • sc 264 One of the antibodies (sc 264) prepared from the immunizations (Example 1) showed strong functional blocking activity in vitro in the TGF ⁇ LAP binding inhibition assay (see Example 4), but exhibited cross-reactive binding to non- ⁇ V ⁇ 6 expressing cell lines (see Example 15).
  • This antibody, sc 264 has an RGD sequence in the CDR3 region, which is presumed to be responsible for the cross-reactive binding. Therefore, site-directed mutagenesis was used to replace the glycine residue in the RGD with an alanine (sc 264 RAD).
  • a second antibody (sc 188) has a glycosylation site within the FR3 region. This site was eliminated through site-directed mutagenesis with a substitution from NLT to KLT (sc 188 SDM). The mutated versions of these two antibodies were then expressed and purified as described in Examples 7 and 8, and the purified antibodies were analyzed as described in the following examples.
  • a binding assay was performed to test whether the cross-reactive binding observed in Example 15 was reduced because of site-directed mutagenesis of sc 264. Binding of the antibodies was analyzed on 293T and 293F cell lines to test whether removing the RGD site from sc 264 would result in decreased binding compared with the original antibody.
  • 293T and 293F cells were spun down after collection and resuspended in HBSS with 1% BSA and 1 mM CaCl 2 and 1 mM MgCl 2 (wash buffer), so that at least 150,000 cells were used in each reaction.
  • Cells were divided between reactions in a V-bottom 96-well plate (Sarstedt), and the cells in the plate were pelleted at 1500 rpm for 3 minutes, after which the HBSS supernatant was removed.
  • the primary antibody was added at the concentration indicated in Table 19 in a volume of 50 ⁇ L, and the cells were resuspended and thereafter incubated on ice for 60 minutes.
  • TGF ⁇ LAP TGF Beta1 LAP
  • HT29 cells grown to the optimal density were then pelleted and washed twice in HBBS (with 1% BSA and with 1 mM Ca and 1 mM Mg 2+ ), after which the cells were then resuspended in HBSS at 30,000 cell per well.
  • the coating liquid was removed from the plates, which were then blocked with 100 ⁇ L 3% BSA at room temperature for 1 hour and thereafter washed twice with PBS.
  • Antibody titrations were prepared in 1:4 serial dilutions in a final volume of 30 ⁇ L and at two times the final concentration. Care was taken to ensure that the PBS concentration in the control wells matched the PBS concentration in the most dilute antibody well. 30 ⁇ L of cells were added to each well, and the cells were incubated in the presence of the antibodies at 4° C. for 40 minutes in a V-bottom plate. The cell-antibody mixtures were transferred to the coated plate and the plate was incubated at 37° C. for 40 minutes. The cells on the coated plates were then washed four times in warm HBSS, and the cells were thereafter frozen at ⁇ 80° C. for 15 minutes.
  • Assay plates were coated with the CS-1 peptide of fibronectin. Assay plates were pre-blocked with 3% BSA/PBS for one hour prior to the assay. The J6.77 cells were grown to confluency, then removed from a culture flask, pelleted and washed three times with HBSS. The cells were then resuspended in HBSS at a concentration of 30,000 cells per well. The coating liquid containing the fibronectin fragments was removed, and the plates were blocked with 100 ⁇ L of 3% BSA for one hour at room temperature. The coated plates were washed three times with HBSS. Antibody titrations were prepared in 1:4 serial dilutions in a final volume of 30 ⁇ L and at twice the final concentration.
  • the resuspended cells were added to the wells containing the titrated antibody in a V-bottom plate, and the cells and antibodies were co-incubated at 4° C. for 40 minutes.
  • the cell-antibody mixture was then transferred to the coated plate, which was thereafter incubated at 37° C. for 40 minutes.
  • the cells on the coated plates were next washed four times in warm HBSS, and the cells in the plates were then frozen at ⁇ 80° C. for 15 minutes.
  • the cells were allowed to thaw at room temperature, and then 100 ⁇ L of CyQuant dye/lysis buffer (Molecular Probes) was added to each well according to the manufacturer's instructions. Fluorescence was read at an excitation wavelength of 485 nm and an emission wavelength of 530 nm.
  • K562 parental cells, or K562 cells expressing Cynomolgus or mouse ⁇ V ⁇ 6 were spun down after collection and resuspended in HBSS with 1% BSA and 1 mM CaCl 2 and 1 mM MgCl 2 (wash buffer), so that at least 150,000 cells were used in each reaction.
  • Cells were divided between reactions in a V-bottom 96-well plate (Sarstedt), and the cells in the plate were pelleted at 1500 rpm for 3 minutes, after which the HBSS supernatant was removed.
  • the primary antibody was added in a volume of 50 ⁇ L, and the cells were resuspended and thereafter incubated on ice for 60 minutes.
  • the internalization of the mutant antibodies was tested using a K562 cell line that stably expressed human ⁇ V ⁇ 6.
  • the assay was performed as described in Example 15. Internalization of the purified antibodies was compared to a commercially available ⁇ V ⁇ 6 antibody that was not internalized in this assay.
  • Plates were coated with 0.5 ⁇ g/ml GST-TGF-b LAP fusion protein at 4° C. overnight and the following morning, washed, and then blocked with 3% BSA/PBS for 1 hour. Detroit-562 cells (25000 cells per well) were then allowed to adhere to the plates for 45 minutes at 37° C. in HBSS containing 2 mM MgCl 2 . After 45 minutes the plates were washed three times in PBS and then fixed in ethanol. Cells were visualized by staining with Hoescht and quantitated by counting the number of cells bound per well on a Cellomics Arrayscan II.
  • the data shown in FIG. 5 indicates that both sc 264 RAD and sc 264 RAD/ADY have similar activity and that the ability to block ⁇ V ⁇ 6 function is maintained in the modified antibody.
  • Detroit 562 cells were cultured in EMEM with Earle's BSS and 2 mM L-Glu+1.0 mM sodium pyruvate, 0.1 mM NEAA+1.5 g/L sodium bicarbonate+10% FBS. Cells were harvested and resuspended in 50% PBS+50% matrigel. The suspension was then implanted at 5 ⁇ 10 ⁇ 6 per mouse in a volume of 0.1 ml within the right flank. Animals were 6-8 week old NCR female nude mice. Dosing was initiated when tumours reached 0.1 cm3 and dosed at 20 mg/kg once weekly for the duration of the study.
  • 264RAD was the most effective, followed by 133, and 188. This data clearly shows that the antibodies 264RAD, 133 and 188 are active in vivo and are able reduce the growth of a tumour dependent on ⁇ V ⁇ 6 signalling for growth.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Immunology (AREA)
  • Genetics & Genomics (AREA)
  • Biochemistry (AREA)
  • Biomedical Technology (AREA)
  • Urology & Nephrology (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biophysics (AREA)
  • Neurology (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Biotechnology (AREA)
  • Microbiology (AREA)
  • Pulmonology (AREA)
  • Endocrinology (AREA)
  • Oncology (AREA)
  • Rheumatology (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Neurosurgery (AREA)
  • Cardiology (AREA)
  • Dermatology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • General Engineering & Computer Science (AREA)
  • Cell Biology (AREA)
US11/833,486 2006-08-03 2007-08-03 Antibodies Directed to AlphaVBeta6 and Uses Thereof Abandoned US20080057063A1 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
US11/833,486 US20080057063A1 (en) 2006-08-03 2007-08-03 Antibodies Directed to AlphaVBeta6 and Uses Thereof
US12/782,335 US8398975B2 (en) 2006-08-03 2010-05-18 Antibodies directed to αVβ6 and uses thereof
US13/760,426 US8894998B2 (en) 2006-08-03 2013-02-06 Antibodies directed to αVβ6 and uses thereof
US14/535,894 US20150166663A1 (en) 2006-08-03 2014-11-07 Antibodies Directed To Alpha V Beta 6 And Uses Thereof

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US83555906P 2006-08-03 2006-08-03
US11/833,486 US20080057063A1 (en) 2006-08-03 2007-08-03 Antibodies Directed to AlphaVBeta6 and Uses Thereof

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US12/782,335 Continuation US8398975B2 (en) 2006-08-03 2010-05-18 Antibodies directed to αVβ6 and uses thereof

Publications (1)

Publication Number Publication Date
US20080057063A1 true US20080057063A1 (en) 2008-03-06

Family

ID=39146822

Family Applications (4)

Application Number Title Priority Date Filing Date
US11/833,486 Abandoned US20080057063A1 (en) 2006-08-03 2007-08-03 Antibodies Directed to AlphaVBeta6 and Uses Thereof
US12/782,335 Active 2028-08-20 US8398975B2 (en) 2006-08-03 2010-05-18 Antibodies directed to αVβ6 and uses thereof
US13/760,426 Active US8894998B2 (en) 2006-08-03 2013-02-06 Antibodies directed to αVβ6 and uses thereof
US14/535,894 Abandoned US20150166663A1 (en) 2006-08-03 2014-11-07 Antibodies Directed To Alpha V Beta 6 And Uses Thereof

Family Applications After (3)

Application Number Title Priority Date Filing Date
US12/782,335 Active 2028-08-20 US8398975B2 (en) 2006-08-03 2010-05-18 Antibodies directed to αVβ6 and uses thereof
US13/760,426 Active US8894998B2 (en) 2006-08-03 2013-02-06 Antibodies directed to αVβ6 and uses thereof
US14/535,894 Abandoned US20150166663A1 (en) 2006-08-03 2014-11-07 Antibodies Directed To Alpha V Beta 6 And Uses Thereof

Country Status (18)

Country Link
US (4) US20080057063A1 (ja)
EP (1) EP2064244B1 (ja)
JP (2) JP5362563B2 (ja)
KR (1) KR20090039739A (ja)
CN (2) CN103524619B (ja)
AR (1) AR062213A1 (ja)
AU (1) AU2007348941B2 (ja)
BR (1) BRPI0715141A2 (ja)
CA (1) CA2658612C (ja)
ES (1) ES2746925T3 (ja)
IL (1) IL196848A0 (ja)
MX (1) MX2009001293A (ja)
NO (1) NO20090877L (ja)
RU (1) RU2009107277A (ja)
TW (1) TW200815474A (ja)
UY (1) UY30524A1 (ja)
WO (1) WO2008112004A2 (ja)
ZA (1) ZA200900395B (ja)

Cited By (27)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100172895A1 (en) * 2008-12-23 2010-07-08 Boone Thomas C Human CGRP Receptor Binding Proteins
US20100330103A1 (en) * 2006-08-03 2010-12-30 Astrazeneca Ab Antibodies Directed to Alpha V Beta 6 And Uses Thereof
CN102858975A (zh) * 2010-02-18 2013-01-02 明治制果药业株式会社 铜绿假单胞菌a血清型脂多糖的抗体
WO2014057074A1 (en) 2012-10-12 2014-04-17 Spirogen Sàrl Pyrrolobenzodiazepines and conjugates thereof
US20140199295A1 (en) * 2011-06-23 2014-07-17 Ablynx N.V. Techniques for predicting, detecting and reducing aspecific protein interference in assays involving immunoglobulin single variable domains
WO2014140174A1 (en) 2013-03-13 2014-09-18 Spirogen Sàrl Pyrrolobenzodiazepines and conjugates thereof
WO2016037644A1 (en) 2014-09-10 2016-03-17 Medimmune Limited Pyrrolobenzodiazepines and conjugates thereof
WO2016057306A1 (en) * 2014-10-06 2016-04-14 University Of Houston Rho associated kinase (rock) inhibitors and their use in treating disease
US9346884B2 (en) 2011-09-30 2016-05-24 Ablynx N.V. Biological materials related to c-Met
WO2016090329A3 (en) * 2014-12-05 2016-07-28 Memorial Sloan-Kettering Cancer Center Antibodies targeting g-protein coupled receptor and methods of use
US9573992B2 (en) 2011-06-23 2017-02-21 Ablynx N.V. Serum albumin binding proteins
EP3042956A4 (en) * 2013-09-05 2017-03-15 University of Miyazaki Antibody which specifically reacts with human integrin a6b4
US9683045B2 (en) 2010-09-30 2017-06-20 Ablynx N.V. Biological materials related to c-Met
WO2018069289A1 (en) 2016-10-11 2018-04-19 Medimmune Limited Antibody-drug conjugates with immune-mediated therapy agents
AU2016244220B2 (en) * 2008-12-23 2018-05-17 Amgen Inc. Human CGRP receptor binding proteins
US10259877B2 (en) 2015-04-24 2019-04-16 Amgen Inc. Methods for treating or preventing migraine headache
CN109843911A (zh) * 2016-08-18 2019-06-04 国家生物技术研究所公司 用于治疗骨相关疾病的组合物和方法
WO2019224275A1 (en) 2018-05-23 2019-11-28 Adc Therapeutics Sa Molecular adjuvant
US20200095317A1 (en) * 2018-05-09 2020-03-26 Legochem Biosciences, Inc. Compositions and methods related to anti-cd19 antibody drug conjugates
US10633426B2 (en) 2014-12-05 2020-04-28 Memorial Sloan Kettering Cancer Center Chimeric antigen receptors targeting G-protein coupled receptor and uses thereof
US10934362B2 (en) 2014-09-15 2021-03-02 Amgen Inc. Bi-specific anti-CGRP receptor/PAC1 receptor antigen binding proteins and uses thereof
CN112533948A (zh) * 2018-05-31 2021-03-19 台湾醣联生技医药股份有限公司 与双触角Lewis B以及Lewis Y抗原结合的治疗性抗体
GB202102396D0 (en) 2021-02-19 2021-04-07 Adc Therapeutics Sa Molecular adjuvant
WO2022079211A1 (en) 2020-10-16 2022-04-21 Adc Therapeutics Sa Glycoconjugates
US11407838B2 (en) 2018-04-02 2022-08-09 Amgen Inc. Erenumab compositions and uses thereof
US11644471B2 (en) * 2010-09-30 2023-05-09 Ablynx N.V. Techniques for predicting, detecting and reducing aspecific protein interference in assays involving immunoglobulin single variable domains
US11975076B2 (en) 2015-11-25 2024-05-07 Legochem Biosciences, Inc. Antibody-drug conjugates comprising branched linkers and methods related thereto

Families Citing this family (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2379596A1 (en) * 2008-12-23 2011-10-26 AstraZeneca AB Targeted binding agents directed to 5 1 and uses therefor
CN106995495A (zh) * 2009-01-12 2017-08-01 希托马克斯医疗有限责任公司 修饰抗体组合物及其制备和使用方法
MX360141B (es) 2012-02-17 2018-10-24 Seattle Genetics Inc Anticuerpos contra integrina alfavhbeta6 y uso de los mismos para tratar cancer.
US8834881B2 (en) 2012-03-08 2014-09-16 Crucell Holland B.V. Human binding molecules capable of binding to and neutralizing influenza B viruses and uses thereof
EP2784511A1 (en) 2013-03-27 2014-10-01 Universität Zürich Integrin alpha-v-beta6 for diagnosis/prognosis of colorectal carcinoma
EP3052523B1 (en) * 2013-10-01 2021-03-10 Medimmune Limited Methods of treating and diagnosing alpha-v-beta-6 overexpressing cancer
WO2015085179A1 (en) * 2013-12-06 2015-06-11 The Regents Of The University Of California Alpha-v beta-6 integrin-binding antibody fragments
WO2015164627A1 (en) * 2014-04-23 2015-10-29 Discovery Genomics, Inc. Chimeric antigen receptors specific to avb6 integrin and methods of use thereof to treat cancer
KR102218714B1 (ko) * 2016-06-14 2021-02-24 머크 샤프 앤드 돔 코포레이션 항응고 인자 xi 항체
CN107991481A (zh) * 2016-10-27 2018-05-04 武汉科前生物股份有限公司 一种检测猪伪狂犬病毒和口蹄疫病毒的二联阻断elisa抗体检测试剂盒及其应用
IL266343B2 (en) * 2016-11-01 2024-01-01 Arrowhead Pharmaceuticals Inc Ligands of alpha-V beta-6 integrin and their uses
EP4252629A3 (en) 2016-12-07 2023-12-27 Biora Therapeutics, Inc. Gastrointestinal tract detection methods, devices and systems
JP7150723B2 (ja) 2016-12-14 2022-10-11 ビオラ・セラピューティクス・インコーポレイテッド 消化管疾病のインテグリン阻害薬による治療
EP3810085A1 (en) 2018-06-20 2021-04-28 Progenity, Inc. Treatment of a disease of the gastrointestinal tract with an integrin inhibitor
EP3883635A1 (en) 2018-11-19 2021-09-29 Progenity, Inc. Methods and devices for treating a disease with biotherapeutics
MX2022006132A (es) 2019-12-05 2022-06-17 Seagen Inc Anticuerpos anti alfa-v beta-6 (avb6) y conjugados de anticuerpo-farmaco.
WO2021119482A1 (en) 2019-12-13 2021-06-17 Progenity, Inc. Ingestible device for delivery of therapeutic agent to the gastrointestinal tract
JP2023509760A (ja) 2020-01-08 2023-03-09 シンシス セラピューティクス,インコーポレイテッド Alk5阻害剤複合体およびその使用
CN114195893A (zh) * 2020-09-17 2022-03-18 百奥泰生物制药股份有限公司 抗整联蛋白抗体或抗原结合片段及其应用

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7084257B2 (en) * 2001-10-05 2006-08-01 Amgen Inc. Fully human antibody Fab fragments with human interferon-gamma neutralizing activity
US20080152587A1 (en) * 2006-04-10 2008-06-26 Amgen Fremont Inc. TARGETED BINDING AGENTS DIRECTED TO uPAR AND USES THEREOF

Family Cites Families (56)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB8823869D0 (en) 1988-10-12 1988-11-16 Medical Res Council Production of antibodies
US5175384A (en) 1988-12-05 1992-12-29 Genpharm International Transgenic mice depleted in mature t-cells and methods for making transgenic mice
US5859205A (en) * 1989-12-21 1999-01-12 Celltech Limited Humanised antibodies
US6673986B1 (en) 1990-01-12 2004-01-06 Abgenix, Inc. Generation of xenogeneic antibodies
EP0463151B1 (en) 1990-01-12 1996-06-12 Cell Genesys, Inc. Generation of xenogeneic antibodies
US6075181A (en) 1990-01-12 2000-06-13 Abgenix, Inc. Human antibodies derived from immunized xenomice
US6150584A (en) 1990-01-12 2000-11-21 Abgenix, Inc. Human antibodies derived from immunized xenomice
US5633425A (en) 1990-08-29 1997-05-27 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
US5877397A (en) 1990-08-29 1999-03-02 Genpharm International Inc. Transgenic non-human animals capable of producing heterologous antibodies of various isotypes
US6255458B1 (en) 1990-08-29 2001-07-03 Genpharm International High affinity human antibodies and human antibodies against digoxin
US5661016A (en) 1990-08-29 1997-08-26 Genpharm International Inc. Transgenic non-human animals capable of producing heterologous antibodies of various isotypes
US5545806A (en) 1990-08-29 1996-08-13 Genpharm International, Inc. Ransgenic non-human animals for producing heterologous antibodies
US6300129B1 (en) 1990-08-29 2001-10-09 Genpharm International Transgenic non-human animals for producing heterologous antibodies
US5814318A (en) 1990-08-29 1998-09-29 Genpharm International Inc. Transgenic non-human animals for producing heterologous antibodies
ES2108048T3 (es) 1990-08-29 1997-12-16 Genpharm Int Produccion y utilizacion de animales inferiores transgenicos capaces de producir anticuerpos heterologos.
US5625126A (en) 1990-08-29 1997-04-29 Genpharm International, Inc. Transgenic non-human animals for producing heterologous antibodies
US5874299A (en) 1990-08-29 1999-02-23 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
CA2090473A1 (en) 1990-08-29 1992-03-01 Robert M. Kay Homologous recombinatin in mammalian cells
US5770429A (en) 1990-08-29 1998-06-23 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
US5789650A (en) 1990-08-29 1998-08-04 Genpharm International, Inc. Transgenic non-human animals for producing heterologous antibodies
WO1992022670A1 (en) 1991-06-12 1992-12-23 Genpharm International, Inc. Early detection of transgenic embryos
WO1992022645A1 (en) 1991-06-14 1992-12-23 Genpharm International, Inc. Transgenic immunodeficient non-human animals
AU2515992A (en) 1991-08-20 1993-03-16 Genpharm International, Inc. Gene targeting in animal cells using isogenic dna constructs
DE69233528T2 (de) 1991-11-25 2006-03-16 Enzon, Inc. Verfahren zur Herstellung von multivalenten antigenbindenden Proteinen
EP0746609A4 (en) 1991-12-17 1997-12-17 Genpharm Int NON-HUMAN TRANSGENIC ANIMALS CAPABLE OF PRODUCING HETEROLOGOUS ANTIBODIES
NZ253943A (en) 1992-06-18 1997-01-29 Genpharm Int Transfering polynucleotides into eukaryotic cells using co-lipofection complexes of a cationic lipid and the polynucleotide
AU675661B2 (en) 1992-07-24 1997-02-13 Abgenix, Inc. Generation of xenogeneic antibodies
AU690528B2 (en) 1992-12-04 1998-04-30 Medical Research Council Multivalent and multispecific binding proteins, their manufacture and use
US5981175A (en) 1993-01-07 1999-11-09 Genpharm Internation, Inc. Methods for producing recombinant mammalian cells harboring a yeast artificial chromosome
JPH08509612A (ja) 1993-04-26 1996-10-15 ジェンファーム インターナショナル インコーポレイテッド 異種抗体を産生することができるトランスジェニック非ヒト動物
US5625825A (en) 1993-10-21 1997-04-29 Lsi Logic Corporation Random number generating apparatus for an interface unit of a carrier sense with multiple access and collision detect (CSMA/CD) ethernet data network
US5643763A (en) 1994-11-04 1997-07-01 Genpharm International, Inc. Method for making recombinant yeast artificial chromosomes by minimizing diploid doubling during mating
WO1996034096A1 (en) 1995-04-28 1996-10-31 Abgenix, Inc. Human antibodies derived from immunized xenomice
AU718138B2 (en) 1995-08-29 2000-04-06 Kyowa Hakko Kirin Co., Ltd. Chimeric animal and method for constructing the same
ATE279947T1 (de) 1996-03-18 2004-11-15 Univ Texas Immunglobulinähnliche domäne mit erhöhten halbwertszeiten
EP1500329B1 (en) 1996-12-03 2012-03-21 Amgen Fremont Inc. Human antibodies that specifically bind human TNF alpha
ATE286742T1 (de) 1997-08-08 2005-01-15 Univ California Behandlung von nierenfibrose durch antikörper gegen integrin alpha-v-beta 6
PL341569A1 (en) 1998-01-23 2001-04-23 Merck Patent Gmbh Monoclonal anti-alphav-integrin antibody and application thereof as an inhibitore of alphavbeta6-integrin to fibronectin bond
CN1335853A (zh) 1998-12-19 2002-02-13 默克专利股份公司 整联蛋白αvβb的抑制剂
US6521593B1 (en) 1999-02-01 2003-02-18 Childrens Hospital Los Angeles Methods for inhibiting brain tumor growth
US6833268B1 (en) 1999-06-10 2004-12-21 Abgenix, Inc. Transgenic animals for producing specific isotypes of human antibodies via non-cognate switch regions
DE19929410A1 (de) 1999-06-26 2000-12-28 Merck Patent Gmbh Inhibitoren des Integrins avß6
DE19933173A1 (de) 1999-07-15 2001-01-18 Merck Patent Gmbh Cyclische Peptidderivate als Inhibitoren des Integrins alpha¶v¶beta¶6¶
US7163681B2 (en) 2000-08-07 2007-01-16 Centocor, Inc. Anti-integrin antibodies, compositions, methods and uses
PT1355919E (pt) 2000-12-12 2011-03-02 Medimmune Llc Moléculas com semivida longa, composições que as contêm e suas utilizações
US20090028853A1 (en) 2006-10-19 2009-01-29 The Regents Of The University Of California, A California Corporation TREATMENT AND PREVENTION OF CHRONIC ASTHMA USING ANTAGONISTS OF INTEGRIN ALPHAvBETA6
CN102659946A (zh) * 2002-03-13 2012-09-12 拜奥根Idec马萨诸塞公司 抗αvβ6抗体
US20040048312A1 (en) 2002-04-12 2004-03-11 Ronghao Li Antibodies that bind to integrin alpha-v-beta-6 and methods of use thereof
MX2007007011A (es) 2004-12-09 2007-09-21 Johnson & Johnson Inmunoconjugados anti-integrina, metodos y usos.
CN104072614B (zh) 2005-07-08 2017-04-26 生物基因Ma公司 抗-αvβ6 抗体及其用途
GB0520068D0 (en) 2005-10-03 2005-11-09 Cancer Res Technology av peptide ligand
BRPI0715141A2 (pt) 2006-08-03 2013-06-04 Astrazeneca Ab agente de ligaÇço alvejante, anticorpo isolado, fragmento de anticorpo isolado, molÉcula de Ácido nucleico, vetor, cÉlula hospedeira, mÉtodos para produzir um agente de ligaÇço alvejado, para produzir um anticorpo, para produzir um fragmento de anticorpo, para tratar um tumor maligno em um animal, para tratar inflamaÇço, e para inibir a interaÇço de alfabeta6 com seu ligando, e, conjugado
US20090196913A1 (en) 2007-05-11 2009-08-06 Ken Shi Kun Huang Anti-Alpha-V Immunoliposome Composition, Methods, and Uses
US20090163698A1 (en) 2007-05-11 2009-06-25 John Joseph Grigsby Method for Preparing Antibody Conjugates
US20090175784A1 (en) 2007-05-11 2009-07-09 Joshua Goldstein Anti-Alpha V Immunoliposome Composition, Methods, and Uses
GB0718843D0 (en) 2007-09-26 2007-11-07 Cancer Rec Tech Ltd Materials and methods relating to modifying the binding of antibodies

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7084257B2 (en) * 2001-10-05 2006-08-01 Amgen Inc. Fully human antibody Fab fragments with human interferon-gamma neutralizing activity
US20080152587A1 (en) * 2006-04-10 2008-06-26 Amgen Fremont Inc. TARGETED BINDING AGENTS DIRECTED TO uPAR AND USES THEREOF

Cited By (51)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8894998B2 (en) 2006-08-03 2014-11-25 Medimmune Limited Antibodies directed to αVβ6 and uses thereof
US8398975B2 (en) 2006-08-03 2013-03-19 Medimmune Limited Antibodies directed to αVβ6 and uses thereof
US20100330103A1 (en) * 2006-08-03 2010-12-30 Astrazeneca Ab Antibodies Directed to Alpha V Beta 6 And Uses Thereof
AU2018203471B2 (en) * 2008-12-23 2020-03-05 Amgen Inc. Human CGRP receptor binding proteins
US20100172895A1 (en) * 2008-12-23 2010-07-08 Boone Thomas C Human CGRP Receptor Binding Proteins
AU2016244220B2 (en) * 2008-12-23 2018-05-17 Amgen Inc. Human CGRP receptor binding proteins
AU2009330175B2 (en) * 2008-12-23 2014-01-16 Amgen Inc. Human CGRP receptor binding proteins
US9862771B2 (en) 2008-12-23 2018-01-09 Amgen Inc. Human CGRP receptor binding proteins
US9102731B2 (en) 2008-12-23 2015-08-11 Amgen Inc. Human CGRP receptor binding proteins
AU2009330175C1 (en) * 2008-12-23 2018-10-04 Amgen Inc. Human CGRP receptor binding proteins
AU2013205271C1 (en) * 2008-12-23 2018-10-04 Amgen Inc. Human CGRP receptor binding proteins
CN102858975A (zh) * 2010-02-18 2013-01-02 明治制果药业株式会社 铜绿假单胞菌a血清型脂多糖的抗体
US20130045207A1 (en) * 2010-02-18 2013-02-21 Symphogen A/S Antibody against serotype g lipopolysaccharide of pseudomonas aeruginosa
US20130004499A1 (en) * 2010-02-18 2013-01-03 Symphogen A/S Antibody against serotype e lipopolysaccharide of pseudomonas aeruginosa
US9683045B2 (en) 2010-09-30 2017-06-20 Ablynx N.V. Biological materials related to c-Met
US11644471B2 (en) * 2010-09-30 2023-05-09 Ablynx N.V. Techniques for predicting, detecting and reducing aspecific protein interference in assays involving immunoglobulin single variable domains
US20140199295A1 (en) * 2011-06-23 2014-07-17 Ablynx N.V. Techniques for predicting, detecting and reducing aspecific protein interference in assays involving immunoglobulin single variable domains
US11192938B2 (en) 2011-06-23 2021-12-07 Ablynx N.V. Serum albumin binding proteins containing immunoglobulin single variable domains
US10858418B2 (en) 2011-06-23 2020-12-08 Ablynx N.V. Techniques for predicting, detecting and reducing aspecific protein interference in assays involving immunoglobulin single variable domains
US9573992B2 (en) 2011-06-23 2017-02-21 Ablynx N.V. Serum albumin binding proteins
US11192937B2 (en) 2011-06-23 2021-12-07 Ablynx N.V. Techniques for predicting, detecting and reducing aspecific protein interference in assays involving immunoglobulin single variable domains
US9346884B2 (en) 2011-09-30 2016-05-24 Ablynx N.V. Biological materials related to c-Met
EP2839860A1 (en) 2012-10-12 2015-02-25 Spirogen Sàrl Pyrrolobenzodiazepines and conjugates thereof
WO2014057074A1 (en) 2012-10-12 2014-04-17 Spirogen Sàrl Pyrrolobenzodiazepines and conjugates thereof
WO2014140174A1 (en) 2013-03-13 2014-09-18 Spirogen Sàrl Pyrrolobenzodiazepines and conjugates thereof
EP3042956A4 (en) * 2013-09-05 2017-03-15 University of Miyazaki Antibody which specifically reacts with human integrin a6b4
US10030071B2 (en) 2013-09-05 2018-07-24 University Of Miyazaki Antibody which specifically reacts with human integrin A6B4
WO2016037644A1 (en) 2014-09-10 2016-03-17 Medimmune Limited Pyrrolobenzodiazepines and conjugates thereof
US11919964B2 (en) 2014-09-15 2024-03-05 Amgen Inc. Bi-specific anti-CGRP receptor/PAC1 receptor antigen binding proteins and uses thereof
US10934362B2 (en) 2014-09-15 2021-03-02 Amgen Inc. Bi-specific anti-CGRP receptor/PAC1 receptor antigen binding proteins and uses thereof
WO2016057306A1 (en) * 2014-10-06 2016-04-14 University Of Houston Rho associated kinase (rock) inhibitors and their use in treating disease
US10335449B2 (en) 2014-10-06 2019-07-02 University Of Houston System Rho associated kinase (ROCK) inhibitors and their use in treating disease
US11566071B2 (en) 2014-12-05 2023-01-31 Memorial Sloan Kettering Cancer Center Nucleic acid molecules encoding anti-GPRC5D antibodies
WO2016090329A3 (en) * 2014-12-05 2016-07-28 Memorial Sloan-Kettering Cancer Center Antibodies targeting g-protein coupled receptor and methods of use
US11820806B2 (en) 2014-12-05 2023-11-21 Memorial Sloan-Kettering Cancer Center Chimeric antigen receptors targeting G-protein coupled receptor and uses thereof
US10906956B2 (en) 2014-12-05 2021-02-02 Memorial Sloan Kettering Cancer Center Methods of treatments using chimeric antigen receptors targeting G-protein coupled receptor
US10590196B2 (en) 2014-12-05 2020-03-17 Memorial Sloan-Kettering Cancer Center Antibodies targeting G-protein coupled receptor and methods of use
US10633426B2 (en) 2014-12-05 2020-04-28 Memorial Sloan Kettering Cancer Center Chimeric antigen receptors targeting G-protein coupled receptor and uses thereof
US11866478B2 (en) 2014-12-05 2024-01-09 Memorial Sloan-Kettering Cancer Center Nucleic acid molecules encoding chimeric antigen receptors targeting G-protein coupled receptor
US11466090B2 (en) 2015-04-24 2022-10-11 Amgen Inc. Methods for treating or preventing migraine headache
US10259877B2 (en) 2015-04-24 2019-04-16 Amgen Inc. Methods for treating or preventing migraine headache
US11975076B2 (en) 2015-11-25 2024-05-07 Legochem Biosciences, Inc. Antibody-drug conjugates comprising branched linkers and methods related thereto
CN109843911A (zh) * 2016-08-18 2019-06-04 国家生物技术研究所公司 用于治疗骨相关疾病的组合物和方法
WO2018069289A1 (en) 2016-10-11 2018-04-19 Medimmune Limited Antibody-drug conjugates with immune-mediated therapy agents
US11407838B2 (en) 2018-04-02 2022-08-09 Amgen Inc. Erenumab compositions and uses thereof
US20200095317A1 (en) * 2018-05-09 2020-03-26 Legochem Biosciences, Inc. Compositions and methods related to anti-cd19 antibody drug conjugates
US11827703B2 (en) * 2018-05-09 2023-11-28 Legochem Biosciences, Inc. Compositions and methods related to anti-CD19 antibody drug conjugates
WO2019224275A1 (en) 2018-05-23 2019-11-28 Adc Therapeutics Sa Molecular adjuvant
CN112533948A (zh) * 2018-05-31 2021-03-19 台湾醣联生技医药股份有限公司 与双触角Lewis B以及Lewis Y抗原结合的治疗性抗体
WO2022079211A1 (en) 2020-10-16 2022-04-21 Adc Therapeutics Sa Glycoconjugates
GB202102396D0 (en) 2021-02-19 2021-04-07 Adc Therapeutics Sa Molecular adjuvant

Also Published As

Publication number Publication date
US20150166663A1 (en) 2015-06-18
AR062213A1 (es) 2008-10-22
AU2007348941A1 (en) 2008-09-18
US20130202621A1 (en) 2013-08-08
CN101553505B (zh) 2013-07-17
CN103524619B (zh) 2016-10-05
RU2009107277A (ru) 2010-09-10
KR20090039739A (ko) 2009-04-22
EP2064244B1 (en) 2019-06-05
TW200815474A (en) 2008-04-01
CA2658612A1 (en) 2008-09-18
WO2008112004A2 (en) 2008-09-18
JP2013256504A (ja) 2013-12-26
CN101553505A (zh) 2009-10-07
JP2009545327A (ja) 2009-12-24
ES2746925T3 (es) 2020-03-09
ZA200900395B (en) 2010-01-27
BRPI0715141A2 (pt) 2013-06-04
US20100330103A1 (en) 2010-12-30
US8398975B2 (en) 2013-03-19
EP2064244A2 (en) 2009-06-03
MX2009001293A (es) 2009-02-11
WO2008112004A3 (en) 2009-04-02
CA2658612C (en) 2015-11-17
UY30524A1 (es) 2008-02-29
AU2007348941B2 (en) 2011-08-04
NO20090877L (no) 2009-03-03
JP5362563B2 (ja) 2013-12-11
US8894998B2 (en) 2014-11-25
IL196848A0 (en) 2011-08-01
CN103524619A (zh) 2014-01-22

Similar Documents

Publication Publication Date Title
CA2658612C (en) Antibodies directed to .alpha.v.beta.6 and uses thereof
US20120107324A1 (en) TARGETED BINDING AGENTS DIRECTED TO a5ß1 AND USES THEREOF
US20190389967A1 (en) Method of treating and diagnosing alpha-v-beta-6 overexpressing cancer
US8119130B2 (en) Targeted binding agents directed to KDR and uses thereof—035
US8697664B2 (en) Targeted binding agents directed to PDGRF-alpha and uses thereof
US20120114667A1 (en) TARGETED BINDING AGENTS DIRECTED TO a5BETA1 AND USES THEREOF
EP2379596A1 (en) Targeted binding agents directed to 5 1 and uses therefor

Legal Events

Date Code Title Description
AS Assignment

Owner name: ASTRAZENECA AB, SWEDEN

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:ALFRED, AVRIL;BARRY, SIMON THOMAS;BEDIAN, VAHE;AND OTHERS;REEL/FRAME:021755/0151;SIGNING DATES FROM 20080212 TO 20080818

Owner name: ASTRAZENECA AB, SWEDEN

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:AMGEN FREMONT INC;REEL/FRAME:021755/0950

Effective date: 20070425

STCB Information on status: application discontinuation

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

AS Assignment

Owner name: MEDIMMUNE LIMITED, UNITED KINGDOM

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:ASTRAZENECA AB;REEL/FRAME:025790/0810

Effective date: 20110208