WO2007066106A1 - Ligands that have binding specificity for egfr and/or vegf and methods of use therefor - Google Patents

Ligands that have binding specificity for egfr and/or vegf and methods of use therefor Download PDF

Info

Publication number
WO2007066106A1
WO2007066106A1 PCT/GB2006/004559 GB2006004559W WO2007066106A1 WO 2007066106 A1 WO2007066106 A1 WO 2007066106A1 GB 2006004559 W GB2006004559 W GB 2006004559W WO 2007066106 A1 WO2007066106 A1 WO 2007066106A1
Authority
WO
WIPO (PCT)
Prior art keywords
seq
dom16
doml
egfr
ligand
Prior art date
Application number
PCT/GB2006/004559
Other languages
French (fr)
Inventor
Olga Ignatovich
Steve Holmes
Roland Beckmann
Haiqun Liu
Rudolf M. T. De Wildt
Laurent S. Jespers
Michael Steward
Armin Sepp
Malgorzata Pupecka
Original Assignee
Domantis Limited
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Domantis Limited filed Critical Domantis Limited
Priority to US12/086,020 priority Critical patent/US20100056439A1/en
Priority to BRPI0619463-0A priority patent/BRPI0619463A2/en
Priority to EA200801172A priority patent/EA013878B1/en
Priority to CA002632417A priority patent/CA2632417A1/en
Priority to EP06820440A priority patent/EP1966242A1/en
Priority to AU2006323412A priority patent/AU2006323412A1/en
Priority to JP2008543892A priority patent/JP2009518024A/en
Priority to PCT/GB2007/000227 priority patent/WO2007085814A1/en
Priority to AU2007209201A priority patent/AU2007209201A1/en
Priority to EP07705002A priority patent/EP1976991A1/en
Priority to CA002640066A priority patent/CA2640066A1/en
Priority to US12/087,924 priority patent/US20100047171A1/en
Priority to JP2008551863A priority patent/JP2009523459A/en
Priority to TW096102778A priority patent/TW200804593A/en
Priority to EP11157601.3A priority patent/EP2441838A3/en
Publication of WO2007066106A1 publication Critical patent/WO2007066106A1/en
Priority to JP2009533859A priority patent/JP2010508016A/en
Priority to CL200703101A priority patent/CL2007003101A1/en
Priority to ARP070104757A priority patent/AR063416A1/en
Priority to EP07821877A priority patent/EP2079761A2/en
Priority to US12/447,908 priority patent/US8236931B2/en
Priority to TW096140428A priority patent/TW200825103A/en
Priority to PCT/EP2007/061514 priority patent/WO2008052933A2/en
Priority to PE2007001465A priority patent/PE20081141A1/en
Priority to CA002667141A priority patent/CA2667141A1/en
Priority to US12/006,933 priority patent/US20080241166A1/en
Priority to NO20082386A priority patent/NO20082386L/en
Priority to US13/644,414 priority patent/US20130041136A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/22Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against growth factors ; against growth regulators
    • 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
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2863Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against receptors for growth factors, growth regulators
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/46Hybrid immunoglobulins
    • 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
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/31Immunoglobulins specific features characterized by aspects of specificity or valency multispecific
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/569Single domain, e.g. dAb, sdAb, VHH, VNAR or nanobody®

Definitions

  • Cancer is a leading cause of mortality and morbidity.
  • Approaches to treating cancer include surgical intervention to remove tumors and chemotherapy. These approaches can successfully cure some patients. However, even patients that appear to have been cured often suffer a recurrence of the cancer necessitating further therapy.
  • Chemotherapeutic agents generally are nonselective agents that are toxic to cells, such as proliferating cells. Accordingly, such agents may effectively kill cancer cells but also kill healthy cells producing several deleterious side effects.
  • Certain cancer cells express or overexpress certain cellular components such as cell surface proteins, or express different cellular components when compared to normal cells.
  • chemotherapeutic approaches to cancer therapy and diagnosis involves targeting cancer cells, for example using antibodies or antibody fragments that bind to proteins that are expressed or overexpressed on cancerous cells.
  • a number of such target proteins have been identified.
  • EGFR epidermal growth factor receptor
  • EGFR is a member of the ErbBl family and transduces signals that lead to cellular proliferation and survival, and the elaboration of growth and angiogenic factors upon binding epidermal growth factor (EGF) and or transforming growth factor alpha (TGF alpha). Accordingly, EGFR has been demonstrated to be involved in tumor growth, metastasis and angiogenesis. Further, many cancers express EGFR, such as bladder cancer, ovarian cancer, colorectal cancer, breast cancer, lung cancer (e.g., non-small cell lung carcinoma), gastric cancer, pancreatic cancer, prostate cancer, head and neck cancer, renal cancer and gall bladder cancer.
  • ERBITUX cetuximab; Imclone Systems Inc
  • ERBITUX cetuximab; Imclone Systems Inc
  • VEGF vascular endothelial growth factor
  • Another approach to treating certain cancers is to inhibit tumor angiogenesis mediated by VEGF, thereby starving the tumor.
  • AVASTIN bevacizumab; Genetech, Inc.
  • An antibody referred to as antibody 2C3 is reported to bind VEGF and inhibit binding of VEGF to epidermal growth factor receptor 2.
  • EGFR or VEGF Targeting EGFR or VEGF with currently available therapeutics is not effective in all patients, or for all cancers (e.g., EGFR-expressing cancers). Thus, a need exists for improved agents for treating cancer and other pathological conditions.
  • the invention relates to ligands that have binding specificity for VEGF (e.g., human VEGF), ligands that have binding specificity for EGFR (e.g., human EGFR), and to ligands that have binding specificity for VEGF and EGFR (e.g., human VEGF and human EGFR).
  • VEGF vascular endothelial growth factor
  • EGFR e.g., human EGFR
  • the ligand can comprise a polypeptide domain having a binding site with binding specificity for VEGF, a polypeptide domain having a binding site with binding specificity EGFR, or comprise a polypeptide domain having a binding site with binding specificity for VEGF and a polypeptide domain having a binding site with binding specificity EGFR.
  • the invention relates to a ligand that has binding specificity for
  • Such ligands comprise at least one protein moiety that has a binding site with binding specificity for VEGF and at least one protein moiety that has a binding site with binding specificity for EGFR.
  • the protein moiety that has a binding site with binding specificity for VEGF and the protein moiety that has a binding site with binding specificity for EGFR can each be any suitable binding moiety.
  • the protein moieties can be a peptide moiety, polypeptide moiety or protein moiety.
  • the protein moieties can be provided by an antibody fragment that has a binding site with binding specificity for VEGF or EGFR, such as an immunoglobulin single variable domain that has binding specificity for VEGF or EGFR.
  • the ligand can comprise a protein moiety that has a binding site with binding specificity for VEGF that competes for binding to VEGF with AVASTIN
  • the ligand can comprise a protein moiety that has a binding site with binding specificity for EGFR that competes for binding to EGFR with ERBITUX (cetuximab; Imclone Systems, Inc.) and/or VECTIBIX (panitumumab; Amgen, Inc.).
  • the ligand comprises a protein moiety that has a binding site with binding specificity for VEGF that competes for binding to VEGF with bevacizumab and/or antibody 2C3 (ATCC Accession No. PTA 1595), and further comprises a protein moiety that has a binding site with binding specificity for EGFR that competes for binding to EGFR with cetuximab.
  • the ligand comprises a protein moiety that has a binding site with binding specificity for VEGF ⁇ e.g., an immunoglobulin single variable domain) that competes for binding to VEGF with an anti-VEGF domain antibody (dAb) selected from the group consisting of TAR15-1 (SEQ ID NO:100), TARl 5-3 (SEQ ID NO: 101), TARl 5-4 (SEQ ID NO: 102), TARl 5-9 (SEQ ID NO:103), TAR15-10 (SEQ ID NO: 104), TAR15-11 (SEQ ID NO:105), TAR15-12 (SEQ ID NO:106), TAR15-13 (SEQ ID NO:107), TAR15-14 (SEQ ID NO:108), TAR15-15 (SEQ ID NO:109), TAR15-16 (SEQ ID NO:110), TAR15-17 (SEQ ID NO:111), TAR15-18 (SEQ ID NO:112), TAR15-19 (SEQ ID NO:113), TAR15-20 (SEQ ID
  • TARl 5-23 SEQ ID NO: 120
  • TARl 5-24 SEQ ID NO: 121
  • TARl 5-25 SEQ ID NO: 122
  • TARl 5-26 SEQ ID NO: 123
  • TARl 5-27 SEQ ID NO: 124
  • TAR15-29 (SEQ ID NO.125), TAR15-30 (SEQ ID NO:126), TAR15-6-500 (SEQ ID NO:127), TAR15-6-501 (SEQ ID NO:128), TAR15-6-502 (SEQ ID NO:129), TAR15-6-503 (SEQ ID NO:130), TAR15-6-504 (SEQ ID NO:131), TAR15-6-505 (SEQ ID NO: 132), TARl 5-6-506 (SEQ ID NO: 133), TARl 5-6-507 (SEQ ID NO:134), TAR15-6-508 (SEQ ID NO:135), TAR15-6-509 (SEQ ID NO:136), TAR15-6-510 (SEQ ID NO:137), TAR15-8-500 (SEQ ID NO:138), TAR15-8-501 (SEQ ID NO:139), TAR15-8-502 (SEQ ID NO:140), TAR15-8-503 (SEQ ID NO:141), TAR15-8-505 (SEQ ID NO:142
  • the ligand comprises a protein moiety that has a binding site with binding specificity for VEGF (e.g., an immunoglobulin single variable domain) that competes for binding to VEGF with TARl 5-26-555 (SEQ ID NO:704).
  • VEGF e.g., an immunoglobulin single variable domain
  • the ligand can comprise a protein moiety that has a binding site with binding specificity for EGFR (e.g., an immunoglobulin single variable domain) that competes for binding to EGFR with an anti-EGFR domain antibody (dAb) selected from the group consisting of DOM 16- 17 (SEQ ID NO:325), DOM16-18 (SEQ ID NO:326), DOM16-19 (SEQ ID NO:325), DOM16- 17 (SEQ ID NO:325), DOM16-18 (SEQ ID NO:326), DOM16-19 (SEQ ID).
  • dAb anti-EGFR domain antibody
  • DOM16-109 SEQ ID NO:405
  • DOM16-110 SEQ ID NO:406
  • DOM16-111 SEQ ID NO:407
  • DOM16-112 SEQ ID NO:408
  • DOM16-113 SEQ ID NO:409
  • DOM16-114 SEQ ID NO:410
  • DOM16-115 SEQ ID NO:
  • the ligand has binding specificity for VEGF and for EGFR and comprises a protein moiety that has a binding site with binding specificity for VEGF that competes for binding to VEGF with an anti-VEGF domain antibody (dAb) selected from the group consisting of TARl 5-6 (SEQ ID NO: 117), TAR15-8 (SEQ ID NO:119), and TAR15-26 (SEQ ID NO:123), and further comprises a protein moiety that has a binding site with binding specificity for EGFR that competes for binding to EGFR with an anti-EGFR domain antibody (dAb) selected from the group consisting of DOM16-39 (SEQ ID NO:345), DOM16-39-87 (SEQ ID NO:420), DOM16-39-100 (SEQ ID NO:423), DOM16-39-107 (SEQ ID NO:430), DOM16-39-109 (SEQ ID NO:432), DOM16-39-115 (SEQ ID NO:438), and
  • the ligand has binding specificity for VEGF and for EGFR and comprises a protein moiety that has a binding site with binding specificity for VEGF that competes for binding to VEGF with an anti-VEGF domain antibody (dAb) selected from the group consisting of TARl 5-6 (SEQ ID NO:117), TAR15-8 (SEQ ID NO:119), and TAR15-26 (SEQ ID NO:123), and further comprises a protein moiety that has a binding site with binding specificity for EGFR that competes for binding to EGFR with an anti-EGFR domain antibody (dAb) selected from the group consisting of DOMl 6-39-521 (SEQ ID NO:577), D0M16- 39-541 (SEQ ID NO:585), DOM16-39-542 (SEQ ID NO:586), DOM16-39-551 (SEQ ID NO:591), DOM16-39-601 (SEQ ID NO:608), DOM16-39-604 (SEQ ID NO:61
  • the ligand has binding specificity for VEGF and for EGFR and comprises at least one immunoglobulin single variable domain with binding specificity for VEGF and at least one immunoglobulin single variable domain with binding specificity for EGFR, wherein an immunoglobulin single variable domain with binding specificity for VEGF competes for binding to VEGF with an anti-VEGF domain antibody (dAb) selected from the group consisting of TAR15-1 (SEQ ID NO:100), TAR15-3 (SEQ ID NOiIOl), TAR15-4 (SEQ ID NO: 102), TARl 5-9 (SEQ ID NO: 103), TARl 5-10 (SEQ ID NO: 104), TARl 5-11 (SEQ ID NO: 105), TARl 5-12 (SEQ ID NO: 106), TARl 5-13 (SEQ ID NO: 107), TAR15-14 (SEQ ID NO:108), TAR15-15 (SEQ ID NO: 109), TAR15-16 (SEQ ID NO-.110),
  • dAb anti
  • TAR15-8 (SEQ ID NO:119), TAR15-23 (SEQ ID NO: 120), TAR15-24 (SEQ ID NO:121), TAR15-25 (SEQ ID NO:122), TAR15-26 (SEQ ID NO:123), TAR15-27 (SEQ ID NO: 124), TAR15-29 (SEQ ID NO:125), TAR15-30 (SEQ ID NO:126), TAR15-6-500 (SEQ ID NO: 127), TAR15-6-501 (SEQ ID NO:128), TAR15-6-502 (SEQ ID NO:129), TAR15-6-503 (SEQ ID NO:130), TAR15-6-504 (SEQ ID NO:131), TAR15-6-505 (SEQ ID NO:132), TAR15-6-506 (SEQ ID NO: 133), TARl 5-6-507 (SEQ ID NO: 134), TARl 5-6-508 (SEQ ID NO: 135), TAR15-6-509 (SEQ ID NO:136), TAR15
  • TARl 5-8-506 (SEQ ID NO: 143), TARl 5-8-507 (SEQ ID NO: 144), TARl 5-8-508 (SEQ ID NO:145), TAR15-8-509 (SEQ ID NO:146), TAR15-8-510 (SEQ ID NO:147), TARl 5-8-511 (SEQ ID NO:148), TAR15-26-500 (SEQ ID NO: 149), TAR15-26-501 (SEQ ID NO:150), TAR15-26-502 (SEQ ID NO:151), TAR15-26- 503 (SEQ ID NO: 152), TARl 5-26-504 (SEQ ID NO: 153), TARl 5-26-505 (SEQ ID NO: 154), TARl 5-26-506 (SEQ ID NO: 155), TARl 5-26-507 (SEQ ID NO: 156), TARl 5-26-508 (SEQ ID NO: 157), TARl 5-26-509 (SEQ ID NO: 158), TARl 5-26
  • the immunoglobulin single variable domain with binding specificity for VEGF can comprise an amino acid sequence that has at least about 85% amino acid sequence identity with the amino acid sequence of a dAb selected from the group consisting of TARl 5-1 (SEQ ID NO: 100), TARl 5-3 (SEQ ID NO:101), TAR15-4 (SEQ ID NO:102), TAR15-9 (SEQ ID NO:103), TAR15-10 (SEQ ID NO:104), TAR15-11 (SEQ ID NO:105), TAR15-12 (SEQ ID NO:106), TAR15-13 (SEQ ID NO:107), TAR15-14 (SEQ ID NO:108), TAR15-15 (SEQ ID NO:109), TAR15-16 (SEQ ID NO:110), TAR15-17 (SEQ ID NO:111), TAR15-18 (SEQ ID NO:112), TAR15-19 (SEQ ID NO:113), TAR15-20 (SEQ ID NO:114), TAR 15-22 (SEQ ID NO: 115), TARl 5-5
  • the ligand has binding specificity for VEGF and for EGFR and comprises at least one immunoglobulin single variable domain with binding specificity for VEGF and at least one immunoglobulin single variable domain with binding specificity for EGFR, wherein an immunoglobulin single variable domain with binding specificity for EGFR competes for binding to EGFR with an anti-EGFR domain antibody (dAb) selected from the group consisting of DOM16-17 (SEQ ID NO:325), DOM16-18 (SEQ ID NO:326), DOM16-19 (SEQ ID NO:327), DOM16-20 (SEQ ID NO:328), DOM16-21 (SEQ ID NO:329), DOM16- 22 (SEQ ID NO:330), DOM16-23 (SEQ ID NO:331), DOM16-24 (SEQ ID
  • dAb anti-EGFR domain antibody
  • the ligand has binding specificity for VEGF and for EGFR and comprises at least one immunoglobulin single variable domain with binding specificity for VEGF and at least one immunoglobulin single variable domain with binding specificity for EGFR, wherein an immunoglobulin single variable domain with binding specificity for EGFR competes for binding to EGFR with an anti-EGFR domain antibody (dAb) selected from the group consisting of DOM16-39-210 (SEQ ID NO:541), DOM16-39-211 (SEQ ID NO:542), D0M16- 39-212 (SEQ ID NO:543), DOM16-39-213 (SEQ ID NO:544), DOM16-39-214 (SEQ ID NO:545), DOM16-39-215 (SEQ ID NO:546), DOM16-39-216 (SEQ ID NO:547), DOM16-39-217 (SEQ ID NO:548), DOM16-39-218 (SEQ ID NO:549), DOM16-39-210
  • the immunoglobulin single variable domain with binding specificity for EGFR can comprise an amino acid sequence that has at least about 85% amino acid sequence identity with the amino acid sequence of a dAb selected from the group consisting of DOM16-17 (SEQ ID NO:325), DOM16-18 (SEQ ID NO:326), DOM16-19 (SEQ ID NO:327), DOM16-20 (SEQ ID NO:328), DOM16- 21 (SEQ ID NO:329), DOM16-22 (SEQ ID NO:330), DOM16-23 (SEQ ID NO:
  • DOMl 6-90 SEQ ID NO:387)
  • DOMl 6-91 SEQ ID NO:388
  • DOMl 6- 92 SEQ ID NO:389
  • DOM16-94 SEQ ID NO:390
  • DOM16-95 SEQ ID NO:
  • the immunoglobulin single variable domain with binding specificity for EGFR can comprise an amino acid sequence that has at least about 85% amino acid sequence identity with the amino acid sequence of a dAb selected from the group consisting of DOM16-39-210 (SEQ ID NO:541), DOM16-39-21I (SEQ ID NO:542), DOM16-39-212 (SEQ ID NO:543), DOM16-39-213 (SEQ ID NO:544), DOM16-39-214 (SEQ ID NO:545), DOM16-39-215 (SEQ ID NO:546), DOM16-39-216 (SEQ ID NO:547), DOM16-39-217 (SEQ ID NO:548), DOM16- 39-218 (SEQ ID NO:549), DOM16-39-219 (SEQ ID NO:550), DOM16-39-220 (SEQ ID NO:551), DOM16-39-221 (SEQ ID NO:552), DOM16-39-222 (SEQ ID NO
  • the ligand has binding specificity for VEGF and for EGFR and comprises at least one immunoglobulin single variable domain with binding specificity for VEGF and at least one immunoglobulin single variable domain with binding specificity for EGFR, wherein an immunoglobulin single variable domain with binding specificity for VEGF competes for binding to VEGF with an anti-VEGF domain antibody (dAb) selected from the group consisting of TARl 5-1 (SEQ ID NO: 100), TARl 5-3 (SEQ ID NO: 101), TARl 5-4 (SEQ ID NO:102), TAR15-9 (SEQ ID NO:103), TAR15-10 (SEQ ID NO: 104), TARl 5-11 (SEQ ID NO:105), TAR15-12 (SEQ ID NO:106), TAR15-13 (SEQ ID NO:107), TARl 5-14 (SEQ ID NO: 108), TARl 5-15 (SEQ ID NO: 109), TARl 5-16 (SEQ ID NO:110), TAR15-17 (SEQ ID NO:
  • DOM16-90 SEQ ID NO:387)
  • DOM16-91 SEQ ID NO:388
  • DOM16-92 SEQ ID NO:389
  • DOM16-94 SEQ ID NO:390
  • DOM16-95 SEQ ID NO:
  • the ligand has binding specificity for VEGF and for EGFR and comprises at least one immunoglobulin single variable domain with binding specificity for VEGF and at least one immunoglobulin single variable domain with binding specificity for EGFR, wherein an immunoglobulin single variable domain with binding specificity for VEGF competes for binding to VEGF with an anti-VEGF domain antibody (dAb) selected from the group consisting of TAR15-1 (SEQ ID NO:100), TAR15-3 (SEQ ID NOrIOl), TAR15-4 (SEQ ID NO:102), TAR15-9 (SEQ ID NOrI 03), TAR15-10 (SEQ ID NOrI 04), TAR15-11 (SEQ ID NO:105), TAR15-12 (SEQ ID NO:106), TAR15-13 (SEQ ID NO: 107), TAR15-14 (SEQ ID NO:108), TAR15-15 (SEQ ID NO:109), TAR15-16 (SEQ ID NOrI lO), TAR15-17 (SEQ ID NO:
  • TARl 5-6-509 SEQ ID NO: 136
  • TARl 5-6-510 SEQ ID NO: 137
  • TARl 5-8-500 SEQ ID NO: 138
  • TARl 5-8-501 SEQ ID NO: 139
  • TARl 5-8-502 SEQ ID NO:140
  • TAR15-8-503 SEQ ID NO:141
  • TAR15-8-505 SEQ ID NO: 142
  • TARl 5-8-506 SEQ ID NO: 143
  • TARl 5-8-507 SEQ ID NO: 144
  • TARl 5-8-508 SEQ ID NO: 145
  • TARl 5-8-509 SEQ ID NO: 146
  • TARl 5-8-510 SEQ ID NO: 147
  • TARl 5-8-511 SEQ ID NO: 148
  • TARl 5-26-500 SEQ ID NO: 149
  • TARl 5-26-501 SEQ ID NO: 150
  • TARl 5-26-502 SEQ ID NO:151
  • the ligand can comprise an immunoglobulin single variable domain with binding specificity for VEGF that comprises an amino acid sequence that has at least about 85% amino acid sequence identity with the amino acid sequence of a dAb selected from the group consisting of TARl 5-1 (SEQ ID NOrIOO), TAR15-3 (SEQ ID NO:101), TAR15-4 (SEQ ID NO:102), TAR15-9 (SEQ ID NO:103), TAR15-10 (SEQ ID NO:104), TAR15-11 (SEQ ID NO: 105), TAR15-12 (SEQ ID NO:106), TAR15-13 (SEQ ID NO:107), TAR15-14 (SEQ ID NO: 108), TAR15-15 (SEQ ID NO: 109), TAR15-16 (SEQ ID NO:110), TAR15-17 (SEQ ID NO:111), TAR15-18 (SEQ ID NO:112), TAR15-19 (SEQ ID NO:113), TARl 5-20 (SEQ ID NO: 114), TAR 15-22 (SEQ ID NO
  • DOM16-109 SEQ ID NO:405
  • DOM16-110 SEQ ID NO:406
  • DOMl 6-111 SEQ ID NO:407
  • DOM16-112 SEQ ID NO:408
  • DOM16-113 SEQ ID NO:409
  • DOM16-114 SEQ ID NO:410
  • DOM16-115 SEQ ID NO:
  • NB20 SEQ ID NO:470
  • NB21 SEQ ID NO:471
  • NB22 SEQ ID NO:472.
  • the ligand can comprise an immunoglobulin single variable domain with binding specificity for VEGF that comprises an amino acid sequence that has at least about 85% amino acid sequence identity with the amino acid sequence of a dAb selected from the group consisting of TARl 5-1 (SEQ ID NO: 1)
  • TAR15-6-510 SEQ ID NO:137
  • TAR15-8-500 SEQ ID NO: 138
  • TAR15-8-501 SEQ ID NO:139
  • TAR15-8-502 SEQ ID NO: 140
  • TAR15-8-503 SEQ ID NO:141
  • TAR15-8-505 SEQ ID NO: 142
  • TAR15-8-506 SEQ ID NO:
  • the ligand has binding specificity for VEGF and for EGFR and comprises at least one immunoglobulin single variable domain with binding specificity for VEGF and at least one immunoglobulin single variable domain with binding specificity for EGFR, wherein an immunoglobulin single variable domain with binding specificity for VEGF competes for binding to VEGF with an anti-VEGF domain antibody (dAb) selected from the group consisting of TAR15-1 (SEQ ID NOrIOO), TAR15-3 (SEQ ID NOrIOl), TAR15-4 (SEQ ID NO:102), TAR15-9 (SEQ ID NO:103), TAR15-10 (SEQ ID NO:104), TAR15-11 (SEQ ID NO:105), TAR15-12 (SEQ ID NO:106), TAR15-13 (SEQ ID NO: 107), TAR15-14 (SEQ ID NO:108), TAR15-15 (SEQ ID NO:109), TAR15-16 (SEQ ID NO:110), TAR15-17 (SEQ ID NO:111),
  • dAb
  • the immunoglobulin single variable domain with binding specificity for VEGF can comprise an amino acid sequence that has at least about 85% amino acid sequence identity with the amino acid sequence of a dAb selected from the group consisting of TAR15-1 (SEQ ID NO:100), TAR15-3 (SEQ ID NO:101), TAR15-4 (SEQ ID NO:102), TAR15-9 (SEQ ID NO:103), TAR15-10 (SEQ ID NO:104), TAR15-11 (SEQ ID NO:105), TAR15-12 (SEQ ID NO:106), TAR15-13 (SEQ ID NO:107), TAR15-14 (SEQ ID NO:108), TAR15-15 (SEQ ID NO:109), TAR15-16 (SEQ ID NO:110), TAR15-17 (SEQ ID NO:111), TAR15-18 (SEQ ID NO:112), TAR15-19 (SEQ ID NO:113), TAR15-20 (SEQ ID NO:114), , TAR 15-22 (SEQ ID NO: 115), TARl 5-5
  • TARl 5-6-506 (SEQ ID NO: 133), TARl 5-6-507 (SEQ ID NO: 134), TARl 5-6-508 (SEQ ID NO: 135), TARl 5-6-509 (SEQ ID NO: 136), TARl 5-6-510 (SEQ ID NO:137), TAR15-8-500 (SEQ ID NO:138), TAR15-8-501 (SEQ ID NO:139), TAR15-8-502 (SEQ ID NO: 140), TAR15-8-503 (SEQ ID NO:141), TAR15-8-505 (SEQ ID NO:142), TAR15-8-506 (SEQ ID NO:143), TAR15-8-507 (SEQ ID NO:144), TAR15-8-508 (SEQ ID NO: 145), TAR15-8-509 (SEQ ID NO: 146), TAR15-8-510 (SEQ ID NO:147), TAR15-8-511 (SEQ ID NO:148), TAR15-26-500 (SEQ ID NO:149), TAR15-
  • the ligand has binding specificity for VEGF and for EGFR and comprises at least one immunoglobulin single variable domain with binding specificity for VEGF and at least one immunoglobulin single variable domain with binding specificity for EGFR, wherein an immunoglobulin single variable domain with binding specificity for VEGF competes for binding to VEGF with bevacizumab and/or antibody 2C3 (ATCC Accession No.
  • an immunoglobulin single variable domain with binding specificity for EGFR competes for binding to EGFR with an anti-EGFR domain antibody (dAb) selected from the group consisting of DOMl 6-17 (SEQ ID NO:325), DOMl 6-18 (SEQ ID NO:326), DOMl 6-19 (SEQ ID NO:327), DOMl 6-20 (SEQ ID NO:328), DOMl 6- 21 (SEQ ID NO:329), DOM16-22 (SEQ ID NO:330), DOM16-23 (SEQ ID NO:331), DOM16-24 (SEQ ID NO:332), DOM16-25 (SEQ ID NO:333), D0M16- 26 (SEQ ID NO:334), DOMl 6-27 (SEQ ID NO:335), DOMl 6-28 (SEQ ID NO:336), DOM16-29 (SEQ ID NO:337), DOM16-30 (SEQ ID NO:338), D0M16- 31 (SEQ ID NO:339),
  • the ligand has binding specificity for VEGF and for EGFR and comprises at least one immunoglobulin single variable domain with binding specificity for VEGF and at least one immunoglobulin single variable domain with binding specificity for EGFR, wherein an immunoglobulin single variable domain with binding specificity for VEGF competes for binding to VEGF with bevacizumab and/or antibody 2C3 (ATCC Accession No.
  • an immunoglobulin single variable domain with binding specificity for EGFR competes for binding to EGFR with an anti-EGFR domain antibody (dAb) selected from the group consisting of DOM16-39-210 (SEQ ID NO:541), DOM16-39-211 (SEQ ID NO:542), DOM16-39-212 (SEQ ID NO:543), DOM16-39-213 (SEQ ID NO:544), DOM16-39-214 (SEQ ID NO:545), DOM16-39-215 (SEQ ID NO:546), DOM16-39-216 (SEQ ID NO:547), DOM16-39-217 (SEQ ID NO:548), DOM16- 39-218 (SEQ ID NO:549), DOM16-39-219 (SEQ ID NO:550), DOM16-39-220 (SEQ ID NO:551), DOM16-39-221 (SEQ ID NO:552), DOM16-39-222 (SEQ ID NO:553)
  • the immunoglobulin single variable domain with binding specificity for EGFR can comprise an amino acid sequence that has at least about 85% amino acid sequence identity with the amino acid sequence of a dAb selected from the group consisting of DOM16-17 (SEQ ID NO:325), DOM16-18 (SEQ ID NO:326), DOM16-19 (SEQ ID NO:327), DOM16-20 (SEQ ID NO:328), DOM16- 21 (SEQ ID NO:329), DOM16-22 (SEQ ID NO:330), DOM16-23 (SEQ ID NO:
  • DOM16-90 SEQ ID NO:387)
  • DOM16-91 SEQ ID NO:388
  • D0M16- 92 SEQ ID NO:389
  • DOM16-94 SEQ ID NO:390
  • DOM16-95 SEQ ID NO:
  • the immunoglobulin single variable domain with binding specificity for EGFR can comprise an amino acid sequence that has at least about 85% amino acid sequence identity with the amino acid sequence of a dAb selected from the group consisting of DOM16-39-210 (SEQ ID NO:541), DOM16-39-211 (SEQ ID NO:542), DOM16-39-212 (SEQ ID NO:543), DOM16-39-213 (SEQ ID NO:544), DOM16-39-214 (SEQ ID NO:545), DOM16-39-215 (SEQ ID NO:546), DOM16-39-216 (SEQ ID NO:547), DOM16-39-217 (SEQ ID NO:548), DOM16- 39-218 (SEQ ID NO:549), DOM16-39-219 (SEQ ID NO:550), DOM16-39-220 (SEQ ID NO:551), DOM16-39-221 (SEQ ID NO:552), DOM16-39-222 (SEQ ID NO
  • the ligand that has binding specificity for VEGF and for EGFR comprises a first immunoglobulin single variable domain with binding specificity for VEGF and a second immunoglobulin single variable domain with binding specificity for EGFR, wherein said first immunoglobulin single variable domain competes for binding to VEGF with bevacizumab and/or antibody 2C3 (ATCC Accession No. PTA 1595); and said second immunoglobulin single variable domain competes for binding to EGFR with cetuximab.
  • the ligand has binding specificity for VEGF and for EGFR and comprises at least one immunoglobulin single variable domain with binding specificity for VEGF and at least one immunoglobulin single variable domain with binding specificity for EGFR, wherein the ligand comprises an immunoglobulin single variable domain with binding specificity for VEGF that comprises an amino acid sequence that has at least 90% amino acid sequence identity with the amino acid sequence of an anti-VEGF dAb selected from the group consisting of TARl 5-6 (SEQ ID NO: 117), TARl 5-8 (SEQ ID NO: 119), and TAR15-26 (SEQ ID NO:123), and further comprises an immunoglobulin single variable domain with binding specificity for EGFR that comprises an amino acid sequence that has at least 90% amino acid sequence identity with an amino acid sequence selected from the group consisting of DOM16-39 (SEQ ID NO:345), DOM16-39-87 (SEQ ID NO:420), DOM16-39-100 (SEQ ID NO:423),
  • the ligand has binding specificity for VEGF and for EGFR and comprises at least one immunoglobulin single variable domain with binding specificity for VEGF and at least one immunoglobulin single variable domain with binding specificity for EGFR, wherein the ligand comprises an immunoglobulin single variable domain with binding specificity for VEGF that comprises an amino acid sequence that has at least 90% amino acid sequence identity with the amino acid sequence of an anti-VEGF dAb selected from the group consisting of TAR15-6 (SEQ ID NO:117), TAR15-8 (SEQ ID NO:119), and TAR15-26 (SEQ ID NO: 123), and further comprises an immunoglobulin single variable domain with binding specificity for EGFR that comprises an amino acid sequence that has at least 90% amino acid sequence identity with an amino acid sequence selected from the group consisting of DOM16-39-521 (SEQ ID NO:577), DOM16-39-541 (SEQ ID NO:585), DOM16-39-542 (SEQ ID NO:586),
  • the ligand that has binding specificity for VEGF and for EGFR can inhibit binding of epidermal growth factor (EGF) and/or transforming growth factor alpha (TGFalpha) to EGFR, inhibit the activity of EGFR, and/or inhibit the activity of EGFR without substantially inhibiting binding of epidermal growth factor (EGF) and/or transforming growth factor alpha (TGFalpha) to EGFR.
  • EGF epidermal growth factor
  • TGFalpha transforming growth factor alpha
  • the ligand that has binding specificity for VEGF and for EGFR can inhibit binding of VEGF to vascular endothelial growth factor receptor 1 (VEGFRl) and/or vascular endothelial growth factor receptor 2 (VEGFR2), inhibit the activity of VEGF and/or inhibit the activity of VEGF without substantially inhibiting binding of VEGF to VEGFRl and/or VEGFR2.
  • VEGFRl vascular endothelial growth factor receptor 1
  • VEGFR2 vascular endothelial growth factor receptor 2
  • the ligand that has binding specificity for VEGF and for EGFR can contain a protein binding moiety (e.g., immunoglobulin single variable domain) with binding specificity for VEGF that binds VEGF with an affinity (KD) that is between about 100 nM and about 1 pM, as determined by surface plasmon resonance.
  • a protein binding moiety e.g., immunoglobulin single variable domain
  • KD affinity
  • the ligand that has binding specificity for VEGF and for EGFR can contain a protein binding moiety (e.g., immunoglobulin single variable domain) with binding specificity for EGFR that binds EGFR with an affinity (KD) that is between about 100 nM and about 1 pM or about 10 nM to about 100 pM, as determined by surface plasmon resonance.
  • a protein binding moiety e.g., immunoglobulin single variable domain
  • KD affinity
  • the ligand that has binding specificity for VEGF and for EGFR can bind VEGF with an affinity (KD) that is between about 100 nM and about 1 pM, as determined by surface plasmon resonance.
  • KD affinity
  • the ligand that has binding specificity for VEGF and for EGFR can bind EGFR with an affinity (KD) that is between about 100 nM and about 1 pM or about 10 nM to about 100 pM, as determined by surface plasmon resonance.
  • KD affinity
  • the ligand that has binding specificity for VEGF and for EGFR can comprise an immunoglobulin single variable domain with binding specificity for VEGF that is a V HH and/or an immunoglobulin single variable domain with bining specificity for EGFR that is a V HH.
  • the ligand that has binding specificity for VEGF and for EGFR can comprise an immunoglobulin single variable domain with binding specificity for VEGF and an immunoglobulin single variable domain with binding specificity for EGFR, wherein the immunoglobuoin single domains are selected from the group consisting of a human V H and a human V L .
  • the ligand that has binding specificity for VEGF and for EGFR can be an IgG-like format comprising two immunoglobulin single variable domains with binding specificity for VEGF, and two immunoglobulin single variable domains with binding specificity for EGFR.
  • the ligand that has binding specificity for VEGF and for EGFR can comprise an antibody Fc region.
  • the invention also relates to a ligand that has binding specificity for VEGF, comprising at least one immunoglobulin single variable domain with binding specificity for VEGF, wherein an immunoglobulin single variable domain with binding specificity for VEGF competes for binding to VEGF with an anti-VEGF domain antibody (dAb) selected from the group consisting of TARl 5-1 (SEQ ID NO:100), TAR15-3 (SEQ ID NO:101), TAR15-4 (SEQ ID NO:102), TAR15-9 (SEQ ID NO: 103), TARl 5-10 (SEQ ID NO: 104), TARl 5-11 (SEQ ID NO: 105), TAR15-12 (SEQ ID NO:106), TAR15-13 (SEQ ID NO:107), TAR15-14 (SEQ ID NO:108), TAR15-15 (SEQ ID NO:109), TAR15-16 (SEQ ID NO:110), TAR15-17 (SEQ ID NO:111), TAR15-18 (SEQ ID NO:112), TAR15-19
  • an immunoglobulin single variable domain with binding specificity for VEGF can comprise an amino acid sequence that has at least about 85% amino acid sequence identity with the amino acid sequence of a dAb selected from the group consisting of TAR15-1 (SEQ ID NO:100), TAR15-3 (SEQ ID NO:101), TAR15-4 (SEQ ID NO: 102), TAR15-9 (SEQ ID NO:103), TAR15-10 (SEQ ID NO: 104), TARl 5-11 (SEQ ID NO:105), TAR15-12 (SEQ ID NO:106), TAR15-13 (SEQ ID NO:107), TAR15-14 (SEQ ID NO: 108), TAR15-15 (SEQ ID NO:109), TAR15-16 (SEQ ID NO:110), TAR15-17 (SEQ ID NO:111), TAR15-18 (SEQ ID NO:112), TARl 5-19 (SEQ ID NO:113), TAR15-20 (SEQ ID NO:114), TAR 15-22 (SEQ ID NO: 115), TAR
  • the ligand that has binding specificity for VEGF can inhibit binding of VEGF to vascular endothelial growth factor receptor 1 (VEGFRl) and/or vascular endothelial growth factor receptor 2 (VEGFR2), inhibit the activity of VEGF and/or inhibit the activity of VEGF without substantially inhibiting binding of VEGF to VEGFRl and/or VEGFR2.
  • VEGFRl vascular endothelial growth factor receptor 1
  • VEGFR2 vascular endothelial growth factor receptor 2
  • the ligand that has binding specificity for VEGF can contain an
  • immunoglobulin single variable domain with binding specificity for VEGF that binds VEGF with an affinity (KD) that is between about 100 nM and about 1 pM, as determined by surface plasmon resonance.
  • the ligand that has binding specificity for VEGF can bind VEGF with an affinity (KD) that is between about 100 nM and about 1 pM, as determined by surface plasmon resonance.
  • KD affinity
  • the ligand that has binding specificity for VEGF can comprise an immunoglobulin single variable domain with binding specificity for VEGF that is a
  • the ligand that has binding specificity for VEGF can comprise an immunoglobulin single variable domain with binding specificity for VEGF that is selected from the group consisting of a human V H and a human V L -
  • the ligand that has binding specificity for VEGF is an IgG-like format comprising at least two immunoglobulin single variable domains with binding specificity for VEGF.
  • the ligand that has binding specificity for VEGF comprises an antibody Fc region.
  • the invention also relates to a ligand that has binding specificity for EGFR comprising at least one immunoglobulin single variable domain with binding specificity for EGFR, wherein an immunoglobulin single variable domain with binding specificity for EGFR competes for binding to EGFR with an anti-EGFR domain antibody (dAb) selected from the group consisting of DOM16-17 (SEQ ID NO:325), DOM16-18 (SEQ ID NO:326), DOM16-19 (SEQ ID NO:327), DOM16- 20 (SEQ ID NO:328), DOM16-21 (SEQ ID NO:329), DOM16-22 (SEQ ID NO:330), DOMl 6-23 (SEQ ID NO:331), DOMl 6-24 (SEQ ID NO:332), DOMl 6- 25 (SEQ ID NO:333), DOMl 6-26 (SEQ ID NO:334), DOMl 6-27 (SEQ ID NO:335), DOM16-28 (SEQ ID NO:336), DOM16-29 (SEQ
  • the invention also relates to a ligand that has binding specificity for EGFR comprising at least one immunoglobulin single variable domain with binding specificity for EGFR, wherein an immunoglobulin single variable domain with binding specificity for EGFR competes for binding to EGFR with an anti-EGFR domain antibody (dAb) selected from the group consisting of DOMl 6-39-210 (SEQ ID NO:541), DOM16-39-211 (SEQ ID NO:542), DOM16-39-212 (SEQ ID NO:543), DOM 16-39-213 (SEQ ID NO:544), DOM16-39-214 (SEQ ID NO:545), DOM16-39-215 (SEQ ID NO:546), DOM16-39-216 (SEQ ID NO:547), DOM16- 39-217 (SEQ ID NO:548), DOM16-39-218 (SEQ ID NO:549), DOM16-39-219 (SEQ ID NO:550), DOM16-39-220 (S
  • the immunoglobulin single variable domain with binding specificity for EGFR can comprise an amino acid sequence that has at least about 85% amino acid sequence identity with the amino acid sequence of a dAb selected from the group consisting of DOM16-17 (SEQ ID NO.325), DOM16-18 (SEQ ID NO:326), DOMl 6-19 (SEQ ID NO:327), DOMl 6-20 (SEQ ID NO:328), DOMl 6- 21 (SEQ ID NO:329), DOMl 6-22 (SEQ ID NO:330), DOMl 6-23 (SEQ ID
  • DOM16-90 SEQ ID NO:387)
  • DOM16-91 SEQ ID NO:388
  • DOM16-92 SEQ ID NO:389
  • DOM16-94 SEQ ID NO:390
  • DOM16-95 SEQ ID NO:
  • the immunoglobulin single variable domain with binding specificity for EGFR can comprise an amino acid sequence that has at least about 85% amino acid sequence identity with the amino acid sequence of a dAb selected from the group consisting of DOM 16-39-210 (SEQ ID NO:541), DOM16-39-211 (SEQ ID NO:542), DOM16-39-212 (SEQ ID NO:543), DOM16-39-213 (SEQ ID NO:544), DOM16-39-214 (SEQ ID NO:545), DOM16-39-215 (SEQ ID NO:546), DOM16-39-216 (SEQ ID NO:547), DOM16-39-217 (SEQ ID NO:548), DOM16- 39-218 (SEQ ID NO:549), DOM16-39-219 (SEQ ID NO:550), DOM16-39-220 (SEQ ID NO:551), DOM16-39-221 (SEQ ID NO:552), DOM16-39-222 (SEQ ID NO
  • the ligand that has binding specificity for EGFR can inhibit binding of epidermal growth factor (EGF) and/or transforming growth factor alpha (TGF alpha) to EGFR, inhibit the activity of EGFR, and/or inhibit the activity of EGFR without substantially inhibiting binding of epidermal growth factor (EGF) and/or transforming growth factor alpha (TGF alpha) to EGFR.
  • EGF epidermal growth factor
  • TGF alpha transforming growth factor alpha
  • the ligand that has binding specificity for EGFR can contain an
  • immunoglobulin single variable domain with binding specificity for EGFR that binds EGFR with an affinity (KD) that is between about 100 nM and about 1 pM or about 10 nM to about 100 pM, as determined by surface plasmon resonance.
  • KD affinity
  • the ligand that has binding specificity for VEGF and for EGFR can bind EGFR with an affinity (KD) that is between about 100 nM and about 1 pM or about 10 nM to about 100 pM, as determined by surface plasmon resonance.
  • KD affinity
  • the ligand that has binding specificity for EGFR can comprise an immunoglobulin single variable domain with binding specificity for EGFR that is a
  • the ligand that has binding specificity for EGFR can comprise an immunoglobulin single variable domain with binding specificity for EGFR that is selected from the group consisting of a human V H and a human V L .
  • the ligand that has binding specificity for EGFR is an IgG-like format comprising at least two immunoglobulin single variable domains with binding specificity for EGFR.
  • the ligand that has binding specificity for EGFR comprises an antibody Fc region.
  • the ligand comprises a single immunoglobulin variable domain polypeptide that antagonizes (inhibits) human EGFR binding to a receptor, wherein said single immunoglobulin variable domain polypeptide comprises a CDR3 sequence that is the same sequence of CDR3 of an anti-EGFR dAb disclosed herein.
  • the ligand comprises a single immunoglobulin variable domain polypeptide that binds EGFR, wherein the polypeptide has an amino acid sequence that is identical to the amino acid sequence of an anti-EGFR dAb disclosed herein, or differs from the amino acid sequence of an anti-EGFR dAb disclosed herein at no more than 25 amino acid positions and has a CDRl sequence that has at least 50% identity to the CDRl sequence of the anti-EGFR dAb.
  • the ligand comprises single immunoglobulin variable domain polypeptide that binds to EGFR, wherein the polypeptide has an amino acid sequence that is identical to the amino acid sequence of anti-EGFR dAb disclosed herein, or differs from the amino acid sequence of anti-EGFR dAb disclosed herein at no more than 25 amino acid positions and has a CDR2 sequence that has at least 50% identity to the CDR2 sequence of the anti-EGFR dAb.
  • the ligand comprises an immunoglobulin single variable domain polypeptide that binds EGFR, wherein the polypeptide has an amino acid sequence that is identical to the amino acid sequence of anti-EGFR dAb disclosed herein, or differs from the amino acid sequence of anti-EGFR dAb disclosed herein at no more than 25 amino acid positions and has a CDR3 sequence that has at least 50% identity to the CDR3 sequence of the anti-EGFR dAb.
  • the ligand comprises an immunoglobulin single variable domain polypeptide that binds EGFR, wherein the polypeptide has an amino acid sequence that is identical to the amino acid sequence of an anti-EGFR dAb disclosed herein, or differs from the amino acid sequence of an anti-EGFR dAb disclosed herein at no more than 25 amino acid positions and has a CDRl sequence that has at least 50% identity to the CDRl sequence of the anti-EGFR dAb and has a CDR2 sequence has at least 50% identity to the CDR2 sequence of the anti-EGFR dAb.
  • the ligand comprises an immunoglobulin single variable domain polypeptide that binds EGFR, wherein the polypeptide has an amino acid sequence that is identical to the amino acid sequence of an anti-EGFR dAb disclosed herein, or differs from the amino acid sequence of an anti-EGFR dAb disclosed herein at no more than 25 amino acid positions and has a CDR2 sequence that has at least 50% identity to the CDR2 sequence of the anti-EGFR dAb and has a CDR3 sequence that has at least 50% identity to the CDR3 sequence of the anti- EGFR dAb.
  • the ligand comprises an immunoglobulin single variable domain polypeptide that binds EGFR, wherein the polypeptide has an amino acid sequence that is identical to the amino acid sequence of an anti-EGFR dAb disclosed herein, or differs from the amino acid sequence of an anti-EGFR dAb disclosed herein at no more than 25 amino acid positions and has a CDRl sequence that has at least 50% identity to the CDRl sequence of the anti-EGFR dAb and has a CDR3 sequence that has at least 50% identity to the CDR3 sequence of the anti- EGFR dAb.
  • the ligand comprises an immunoglobulin single variable domain polypeptide that binds EGFR, wherein the polypeptide has an amino acid sequence that is identical to the amino acid sequence of an anti-EGFR dAb disclosed herein, or differs from the amino acid sequence of an anti-EGFR dAb disclosed herein at no more than 25 amino acid positions and has a CDRl sequence that has at least 50% identity to the CDRl sequence of the anti-EGFR dAb and has a CDR2 sequence that has at least 50% identity to the CDR2 sequence of the anti- EGFR dAb and has a CDR3 sequence that has at least 50% identity to the CDR3 sequence of the anti-EGFR dAb.
  • the invention is an EGFR antagonist having a CDRl sequence that has at least 50% identity to the CDRl sequence of an anti-EGFR dAb disclosed herein. In another embodiment, the invention is an EGFR antagonist having a CDR2 sequence that has at least 50% identity to the CDR2 sequence of an anti-EGFR dAb disclosed herein.
  • the invention is an EGFR antagonist having a CDR3 sequence that has at least 50% identity to the CDR3 sequence of an anti-EGFR dAb disclosed herein.
  • the invention is an EGFR antagonist having a CDRl sequence that has at least 50% identity to the CDRl sequence of an anti-EGFR dAb disclosed herein and a CDR2 sequence that has at least 50% identity to the CDR2 sequence of the anti-EGFR dAb.
  • the invention is an EGFR antagonist having a CDR2 sequence that has at least 50% identity to the CDR2 sequence of an anti-EGFR dAb disclosed herein and a CDR3 sequence that has at least 50% identity to the CDR3 sequence of the anti-EGFR dAb .
  • the invention is an EGFR antagonist having a CDRl sequence that has at least 50% identity to the CDRl sequence of an anti-EGFR dAb disclosed herein and a CDR3 sequence that has at least 50% identity to the CDR3 sequence of the anti-EGFR dAb.
  • the invention is an EGFR antagonist having a CDRl sequence that has at least 50% identity to the CDRl sequence of an anti-EGFR dAb disclosed herein and a CDR2 sequence that has at least 50% identity to the CDR2 sequence of the anti-EGFR dAb and a CDR3 sequence that has at least 50% identity to the CDR3 sequence of the anti-EGFR dAb.
  • the ligand comprises a single immunoglobulin variable domain polypeptide that antagonizes (inhibits) human VEGF binding to a receptor, wherein said single immunoglobulin variable domain polypeptide comprises a CDR3 sequence that is the same sequence of CDR3 of an anti-VEGF dAb disclosed herein.
  • the ligand comprises a single immunoglobulin variable domain polypeptide that binds VEGF, wherein the polypeptide has an amino acid sequence that is identical to the amino acid sequence of an anti-VEGF dAb disclosed herein, or differs from the amino acid sequence of an anti-VEGF dAb disclosed herein at no more than 25 amino acid positions and has a CDRl sequence that has at least 50% identity to the CDRl sequence of the anti-VEGF dAb.
  • the ligand comprises single immunoglobulin variable domain polypeptide that binds to VEGF, wherein the polypeptide has an amino acid sequence that is identical to the amino acid sequence of anti-VEGF dAb disclosed herein, or differs from the amino acid sequence of anti-VEGF dAb disclosed herein at no more than 25 amino acid positions and has a CDR2 sequence that has at least 50% identity to the CDR2 sequence of the anti-VEGF dAb.
  • the ligand comprises an immunoglobulin single variable domain polypeptide that binds VEGF, wherein the polypeptide has an amino acid sequence that is identical to the amino acid sequence of anti-VEGF dAb disclosed herein, or differs from the amino acid sequence of anti-VEGF dAb disclosed herein at no more than 25 amino acid positions and has a CDR3 sequence that has at least 50% identity to the CDR3 sequence of the anti-VEGF dAb.
  • the ligand comprises an immunoglobulin single variable domain polypeptide that binds VEGF, wherein the polypeptide has an amino acid sequence that is identical to the amino acid sequence of an anti-VEGF dAb disclosed herein, or differs from the amino acid sequence of an anti-VEGF dAb disclosed herein at no more than 25 amino acid positions and has a CDRl sequence that has at least 50% identity to the CDRl sequence of the anti-VEGF dAb and has a CDR2 sequence has at least 50% identity to the CDR2 sequence of the anti-VEGF dAb.
  • the ligand comprises an immunoglobulin single variable domain polypeptide that binds VEGF, wherein the polypeptide has an amino acid sequence that is identical to the amino acid sequence of an anti-VEGF dAb disclosed herein, or differs from the amino acid sequence of an anti-VEGF dAb disclosed herein at no more than 25 amino acid positions and has a CDR2 sequence that has at least 50% identity to the CDR2 sequence of the anti-VEGF dAb and has a CDR3 sequence that has at least 50% identity to the CDR3 sequence of the anti- VEGF dAb.
  • the ligand comprises an immunoglobulin single variable domain polypeptide that binds VEGF, wherein the polypeptide has an amino acid sequence that is identical to the amino acid sequence of an anti-VEGF dAb disclosed herein, or differs from the amino acid sequence of an anti-VEGF dAb disclosed herein at no more than 25 amino acid positions and has a CDRl sequence that has at least 50% identity to the CDRl sequence of the anti-VEGF dAb and has a CDR3 sequence that has at least 50% identity to the CDR3 sequence of the anti- VEGF dAb.
  • the ligand comprises an immunoglobulin single variable domain polypeptide that binds VEGF, wherein the polypeptide has an amino acid sequence that is identical to the amino acid sequence of an anti-VEGF dAb disclosed herein, or differs from the amino acid sequence of an anti-VEGF dAb disclosed herein at no more than 25 amino acid positions and has a CDRl sequence that has at least 50% identity to the CDRl sequence of the anti-VEGF dAb and has a CDR2 sequence that has at least 50% identity to the CDR2 sequence of the anti- VEGF dAb and has a CDR3 sequence that has at least 50% identity to the CDR3 sequence of the anti-VEGF dAb .
  • the invention is an VEGF antagonist having a CDRl sequence that has at least 50% identity to the CDRl sequence of an anti-VEGF dAb disclosed herein.
  • the invention is an VEGF antagonist having a CDR2 sequence that has at least 50% identity to the CDR2 sequence of an anti-VEGF dAb disclosed herein.
  • the invention is an VEGF antagonist having a CDR3 sequence that has at least 50% identity to the CDR3 sequence of an anti-VEGF dAb disclosed herein.
  • the invention is an VEGF antagonist having a CDRl sequence that has at least 50% identity to the CDRl sequence of an anti-VEGF dAb disclosed herein and a CDR2 sequence that has at least 50% identity to the CDR2 sequence of the anti-VEGF dAb.
  • the invention is an VEGF antagonist having a CDR2 sequence that has at least 50% identity to the CDR2 sequence of an anti-VEGF dAb disclosed herein and a CDR3 sequence that has at least 50% identity to the CDR3 sequence of the anti-VEGF dAb .
  • the invention is an VEGF antagonist having a CDRl sequence that has at least 50% identity to the CDRl sequence of an anti-VEGF dAb disclosed herein and a CDR3 sequence that has at least 50% identity to the CDR3 sequence of the anti-VEGF dAb.
  • the invention is an VEGF antagonist having a CDRl sequence that has at least 50% identity to the CDRl sequence of an anti-VEGF dAb disclosed herein and a CDR2 sequence that has at least 50% identity to the CDR2 sequence of the anti-VEGF dAb and a CDR3 sequence that has at least 50% identity to the CDR3 sequence of the anti-VEGF dAb.
  • any of the ligands described herein further comprise a toxin, such as a cytotoxin, free radical generator, antimetabolite, protein, polypeptide, peptide, photoactive agent, antisense compound, chemotherapeutic, radionuclide or intrabody.
  • a toxin such as a cytotoxin, free radical generator, antimetabolite, protein, polypeptide, peptide, photoactive agent, antisense compound, chemotherapeutic, radionuclide or intrabody.
  • the toxin is a surface active toxin (e.g., a free radical generator, a radionuclide).
  • the ligand further comprises a half-life extending moiety, such as a polyalkylene glycol moiety, serum albumin or a fragment thereof, transferrin receptor or a transferrin-binding portion thereof, or a moiety comprising a binding site for a polypeptide that enhances half-life in vivo.
  • the half-life extending moiety is a moiety comprising a binding site for a polypeptide that enhances half-life in vivo selected from the group consisting of an affibody, an SpA domain, an LDL receptor class A domain, an EGF domain, and an avimer.
  • the half-life extending moiety is an antibody or antibody fragment (e.g., an immunoglobulin single variable domain) comprising a binding site for serum albumin or neonatal Fc receptor.
  • an antibody or antibody fragment e.g., an immunoglobulin single variable domain
  • the half-life extending moiety is an
  • immunoglobulin single variable domain comprising a binding site for serum albumin that competes for binding to human serum albumin with a dAb selected from the group consisting of DOM7m-16 (SEQ ID NO: 473), DOM7m-12 (SEQ ID NO: 474), DOM7m-26 (SEQ ID NO: 475), DOM7r-l (SEQ ID NO: 476), DOM7r-3 (SEQ ID NO: 477), DOM7r-4 (SEQ ID NO: 478), DOM7r-5 (SEQ ID NO: 479), DOM7r-7 (SEQ ID NO: 480), DOM7r-8 (SEQ ID NO: 481), DOM7h-2 (SEQ ID NO: 482), DOM7h-3 (SEQ ID NO: 483), DOM7h-4 (SEQ ID NO: 484), DOM7h-6 (SEQ ID NO: 485), DOM7h-l (SEQ ID NO: 486), DOM7h-7 (SEQ ID NO: 487),
  • the immunoglobulin single variable domain comprising a binding site for serum albumin can comprise an amino acid sequence that has at least 85% amino acid sequence identity with the amino acid sequence of a dAb selected from the group consisting of DOM7m-16 (SEQ ID NO: 473), DOM7m-12 (SEQ ID NO: 474), DOM7m-26 (SEQ ID NO: 475), DOM7r-l (SEQ ID NO: 476), DOM7r-3 (SEQ ID NO: 477), DOM7r-4 (SEQ ID NO: 478), DOM7r-5 (SEQ ID NO: 479), DOM7r-7 (SEQ ID NO: 480), DOM7r-8 (SEQ ID NO: 481), DOM7h-2 (SEQ ID NO: 482), DOM7h-3 (SEQ ID NO: 483), DOM7h-4 (SEQ ID NO: 484), DOM7h-6 (SEQ ID NO: 485), DOM7h-l (SEQ ID NO: 486),
  • the invention also relates to an isolated or recombinant nucleic acid encoding a ligand described herein, and to a vector (e.g., recombinant vector) comprising the recombinant nucleic acid.
  • a vector e.g., recombinant vector
  • the invention also relates to a host cell
  • the invention also relates to a method for producing a ligand, comprising maintaining a host cell of the invention under conditions suitable for expression of said nucleic acid or vector, whereby a ligand is produced. In some embodiments, the method further comprises isolating the ligand.
  • the invention also relates to a ligand of the invention for use in therapy or diagnosis, and to the use of a ligand of the invention for the manufacture of a medicament for treatment, prevention or suppression of a disease described herein
  • the invention also relates to a pharmaceutical compositon for the treatment, prevention or suppression of a disease described herein (e.g., cancer) comprising as an active ingredient a ligand of the invention.
  • a disease described herein e.g., cancer
  • the invention relates to a ligand for use in treating cancer, or cancer cells that overexpress EGFR and/or VEGF.
  • the invention relates to use of a ligand for the manufacture of a medicament for killing cells (e.g., selectively killing cancer cells over normal cells).
  • the invention relates to use of a ligand for the manufacture of a medicament for treating cancer cells that overexpress EGFR and/or VEGF.
  • the invention also relates to therapeutic methods that comprise
  • the invention relates to a method for treating cancer comprising administering to a subject in need thereof a
  • the method further comprises administering to the subject a chemotherapeutic agent
  • the method for treating cancer comprises
  • antineoplastic composition comprises at least one chemotherapeutic agent.
  • the chemotherapeutic agent can be selected from the group consisting of alkylating agents, antimetabolites, folic acid analogs, pyrimidine analogs, purine analogs and related inhibitors, vinca alkaloids, epipodopyyllotoxins, antibiotics, L-Asparaginase, topoisomerase inhibitor, interferons, platinum cooridnation complexes,
  • anthracenedione substituted urea methyl hydrazine derivatives, adrenocortical suppressant, adrenocorticosteroides, progestins, estrogens, antiestrogen, androgens, antiandrogen, and gonadotropin-releasing hormone analog.
  • the chemotherapeutic agent is selected from the group consisting of cisplatin, dicarbazine, dactinomycin, mechlorethamine, streptozocin, cyclophosphamide, capecitabine, carmustine, lomustine, doxorubicin, daunorubicin, procarbazine, mitomycin, cytarabine, etoposide, methotrexate, 5-fluorouracil, vinblastine, vincristine, bleomycin, paclitaxel, docetaxel, doxetaxe, aldesleukin, asparaginase, busulfan, carboplatin, cladribine, dacarbazine, fioxuridine, fludarabine, hydroxyurea, ifosfamide, interferon alpha, irinotecan, leuprolide, leucovorin, megestrol, melphalan, mercaptopurine, o
  • the method is a method of treating a cancer selected from the group consisting of bladder cancer, ovarian cancer, colorectal cancer
  • cancer colonal carcinoma
  • breast cancer breast cancer
  • lung cancer non-small cell lung carcinoma
  • gastric cancer pancreatic cancer
  • prostate cancer head and neck cancer
  • renal cancer gall bladder cancer
  • the invention also relates to a method of administering to a subject anti- VEGF treatment and anti-EGFR treatment, the method comprising simultaneous administration of an anti-VEGF treatment and an anti-EGFR treatment by administering to said subject a therapeutically effective amount of a ligand that has binding specificity for VEGF and EGFR.
  • the invention also relates to a composition ⁇ e.g., pharmaceutical
  • composition comprising a ligand of the invention and a physiologically or pharmaceutically acceptable carrier.
  • the composition comprises a vehicle for intravenous, intramuscular, intraperitoneal, intraarterial, intrathecal, intraarticular subcutaneous administration, pulmonary, intranasal, vaginal, or rectal administration.
  • the invention also relates to a drug delivery device comprising the composition ⁇ e.g., pharmaceutical composition) of the invention or a ligand of the invention.
  • the drug delivery device is for simultaneously administering to a subject anti-VEGF treatment and anti-EGFR treatment, and the device comprising a ligand that has binding specificity for VEGF and EGFR.
  • the drug device comprises a plurality of therapeutically effective doses of ligand.
  • the drug delivery device is selected from the group consisting of a parenteral delivery device, intravenous delivery device, intramuscular delivery device, intraperitoneal delivery device, transdermal delivery device, pulmonary delivery device, intraarterial delivery device, intrathecal delivery device, intraarticular delivery device, subcutaneous delivery device, intranasal delivery device, vaginal delivery device, rectal delivery device, a syringe, a transdermal delivery device, a capsule, a tablet, a nebulizer, an inhaler, an atomizer, an aerosolizer, a mister, a dry powder inhaler, a metered dose inhaler, a metered dose sprayer, a metered dose mister, a metered dose atomizer, a catheter.
  • a parenteral delivery device intravenous delivery device, intramuscular delivery device, intraperitoneal delivery device, transdermal delivery device, pulmonary delivery device, intraarterial delivery device, intrathecal delivery device, intraarticular delivery device, subcutaneous delivery device,
  • the invention also relates to a ligand that has binding specificity for vascular endothelial growth factor (VEGF) and epidermal growth factor receptor (EGFR), comprising at least one protein moiety that has a binding site with binding specificity for VEGF, at least one protein moiety that has a binding site with binding specificity for EGFR, and an Fc region of an antibody.
  • VEGF vascular endothelial growth factor
  • EGFR epidermal growth factor receptor
  • Such ligands can consist of a single polypeptide.
  • two ligands that contain Fc regions are bonded together, for example through a disulfide bond ⁇ e.g., in the hinge region), to form a dimer.
  • the ligand that has binding specificity for VEGF can comprise a protein moiety that has a binding site with binding specificity for VEGF and an Fc region of an antibody.
  • the protein moiety having binding specificity for VEGF is fused to an Fc region of an antibody.
  • the ligand that has binding specificity for EGFR can comprise a protein moiety that has a binding site with binding specificity for EGFR and an Fc region of an antibody.
  • the protein moiety having binding specificity for EGFR is fused to an Fc region of an antibody.
  • the ligand can comprise two protein moieties that have binding sites with binding specificity for EGFR and an Fc region of an antibody.
  • the ligand that has binding specificity for VEGF and EGFR comprises a single variable domain with binding specificity for VEGF, a single variable domain with binding specificity for EGFR, and optionally a linker.
  • the single variable domain with binding specificity for EGFR can be bonded via the linker to the immunoglobulin single variable domain with binding specificity for VEGF.
  • Suitable linkers include SEQ ID NO:706, SEQ ID NO:707, SEQ ID NO:708, SEQ ID NO:709, SEQ ID NO:710, SEQ ID NO:711, SEQ ID NO:712, SEQ ID NO:713, SEQ ID NO:714, SEQ ID NO:723 and SEQ ID NO:724.
  • the ligand can also comprise an Fc region of an antibody if desired.
  • a linker can bind an immunoglobulin variable domain to the Fc region.
  • two ligands that contain Fc regions are bonded together, for example through a disulfide bond (e.g., in the hinge region), to form a dimer.
  • the ligand that has binding specificity for VEGF and EGFR comprises a single variable domain with binding specificity for VEGF directly fused to a single variable domain with binding specificity for EGFR.
  • the single variable domains can independently be a light chain variable domain or a heavy chain variable domain.
  • the ligand can comprise a) a single variable domain with binding specificity for VEGF that is a heavy chain variable region, and the immunoglobulin single variable domain with binding specificity for EGFR that is a light chain variable region; b) a single variable domain with binding specificity for VEGF that is a light chain variable domain, and a single variable domain with binding specificity for EGFR that is a heavy chain variable domain; c) a single variable domain with binding specificity for VEGF that is a heavy chain variable domain, and a single variable domain with binding specificity for EGFR that is a heavy chain variable domain; or d) a single variable domain with binding specificity for VEGF that is a light chain variable domain, and a single variable domain with binding specificity for EGFR that is a light chain variable domain.
  • the heavy chain variable region is a V H or V HH - In further embodiments, the heavy chain variable region is a V H or V HH - In further embodiments, the heavy chain variable region is a V H or V HH - In further embodiments, the heavy chain variable region is a V H or V HH - In further embodiments, the heavy chain variable region is a V H or V HH - In further embodiments, the heavy chain variable region is a V H or V HH - In further
  • the V H is a human V H .
  • the light chain variable region is a V K -
  • a ligand that has binding specificity for VEGF and EGFR comprises at least one immunoglobulin single variable domain with binding specificity for VEGF and at least one immunoglobulin single variable domain with binding specificity for EGFR, wherein the immunoglobulin single variable domain with binding specificity for EGFR is bonded via a disulfide bond to the immunoglobulin single variable domain with binding specificity for VEGF.
  • a ligand that has binding specificity for VEGF and EGFR can comprises at least one immunoglobulin single variable domain with binding specificity for VEGF and at least one immunoglobulin single variable domain with binding specificity for EGFR, wherein the immunoglobulin single variable domain with binding specificity for EGFR is directly fused to the immunoglobulin single variable domain with binding specificity for VEGF (z. e. , a single polypeptide comprising two dAbs).
  • the ligand is a fusion of a dAb to an antiserum albumin dAb (a DOM7 dAb).
  • the ligand can have the structure, from amino-terminal to carboxy-terminal, DOMl 5-10— DOMl 6-39— anti-serum albumin dAb, DOM16-39— DOM15-10— anti-serum albumin dAb, DOM15-26- 501— DOM16-39— anti-serum albumin dAb, or DOM16-39— DOM15-26-501— anti-serum albumin dAb.
  • the ligand that has a binding site with binding specificity for EGFR can compete for binding to EGFR with cetuximab and/or panitumumab and is fused to an anti-serum albumin dAb.
  • the ligand can comprise two or more dAbs (e.g. anti-EGFR dAbs) fused to an anti-serum albumin dAb.
  • dAbs e.g. anti-EGFR dAbs
  • FIG. IA- IE illustrates twenty-seven nucleotide sequences that encode human (Homo sapiens) domain antibodies (dAbs) that specifically bind human VEGF.
  • the nucleotide sequences presented are SEQ ID NOS:l-27, 535 and 536.
  • FIG. 2A-2C is a alignment of twelve nucleotide sequences that encode human dAbs that bind human VEGF.
  • the nucleotide sequences presented are SEQ ID NO: 18 and SEQ ID NOS:28-38.
  • FIG. 3A-3D is a alignment of twelve nucleotide sequences that encode human dAbs that bind human VEGF.
  • the nucleotide sequences presented are SEQ ID NO:20 and SEQ ID NOS:39-49.
  • FIG. 4A-4J is a alignment of fifty-three nucleotide sequences that encode human dAbs that bind human VEGF.
  • the nucleotide sequences presented are SEQ ID NO:24, 50-99, 537 and 538.
  • FIG. 5A-5C illustrates the amino acid sequences of dAbs encoded by several of the nucleic acid sequences shown in FIG. 1A-1E.
  • the amino acid sequences presented are SEQ ID NOS: 100-126.
  • FIG. 6 is an alignment of the amino acid sequences of the dAbs encoded by the nucleic acid sequences shown in FIG. 2A-2C.
  • the amino acid sequences presented are SEQ ID NO:117 AND SEQ ID NOS:127-137.
  • FIG. 7A-7B is an alignment of the amino acid sequences of the dAbs encoded by the nucleic acid sequences shown in FIG. 3A-2D.
  • the symbol ⁇ has been inserted into the sequence of TARl 5-8-500 to facilitate alignment.
  • the amino acid sequences presented are SEQ ID NO:119 and SEQ ID NOS:138-148.
  • FIG. 8A-8D is an alignment of the amino acid sequences of the dAbs encoded by the nucleic acid sequences shown in FIG. 4A-4J.
  • the amino acid sequences presented are SEQ ID NO:123, 149-198, 539 and 540.
  • FIG. 9A-9O illustrates several nucleotide sequences that encode human (Homo sapiens) domain antibodies (dAbs) that specifically bind human EGFR.
  • the nucleotide sequences presented are SEQ ID NOS: 199-324.
  • FIG. 10A- 101 illustrates the amino acid sequences of the dAbs encoded by the nucleic acid sequences shown in FIG. 9A-9O.
  • the amino acid sequences presented are SEQ ID NOS:325-450.
  • FIG. 1 IA-I IB illustrates the amino acid sequences of several Camelid V HH S that bind EGFR that are disclosed in WO 2005/044858.
  • NBl SEQ ID NO:451
  • NB2 SEQ ID NO:452
  • NB3 SEQ ID NO:453
  • NB4 SEQ ID NO:454
  • NB5 SEQ ID NO:455
  • NB6 SEQ ID NO:456
  • NB7 SEQ ID NO:457
  • NB8 SEQ ID NO:458
  • NB9 SEQ ID NO:459
  • NBlO SEQ ID NO:460
  • NBI l SEQ ID NO:461
  • NB12 SEQ ID NO:462
  • NB13 SEQ ID NO:463
  • NB14 SEQ ID NO:464
  • NB15 SEQ ID NO:465)
  • NB16 SEQ ID NO:466)
  • NB17 SEQ ID NO:467)
  • NB 18 SEQ
  • FIG. 12A is an alignment of the amino acid sequences of three Vies that bind mouse serum albumin (MSA).
  • the aligned amino acid sequences are from VKS designated MSA16, which is also referred to as DOM7m-16 (SEQ ID NO: 473), MSA 12, which is also referred to as DOM7m-12 (SEQ ID NO: 474), and MSA 26, which is also referred to as DOM7m-26 (SEQ ID NO: 475).
  • FIG. 12B is an alignment of the amino acid sequences of six VKS that bind rat serum albumin (RSA).
  • the aligned amino acid sequences are from VKS designated DOM7r-l (SEQ ID NO: 476), DOM7r-3 (SEQ ID NO: 477), DOM7r-4 (SEQ ID NO: 478), DOM7r-5 (SEQ ID NO: 479), DOM7r-7 (SEQ ID NO: 480), and DOM7r-8 (SEQ ID NO: 481).
  • FIG. 12C is an alignment of the amino acid sequences of six VKS that bind human serum albumin (HSA).
  • the aligned amino acid sequences are from VKS designated DOM7h-2 (SEQ ID NO: 482), DOM7h-3 (SEQ ID NO: 483), DOM7h-4 (SEQ ID NO: 484), DOM7h-6 (SEQ ID NO: 485), DOM7h-l (SEQ ID NO: 486), and DOM7h-7 (SEQ ID NO: 487).
  • FIG. 12D is an alignment of the amino acid sequences of seven VHS that bind human serum albumin and a consensus sequence (SEQ ID NO: 488).
  • the aligned sequences are from V H s designated DOM7h-22 (SEQ ID NO: 489), DOM7h-23 (SEQ ID NO: 490), DOM7h-24 (SEQ ID NO: 491), DOM7h-25 (SEQ ID NO: 492), DOM7h-26 (SEQ ID NO: 493), DOM7h-21 (SEQ ID NO: 494), and DOM7h-27 (SEQ ID NO: 495).
  • FIG. 12E is an alignment of the amino acid sequences of three VKS that bind human serum albumin and rat serum albumin.
  • the aligned amino acid sequences are from VKS designated DOM7h-8 (SEQ ID NO: 496), DOM7r-13 (SEQ ID NO: 497), and DOM7r-14 (SEQ ID NO: 498).
  • FIG. 13 is an illustration of the amino acid sequences of Vies that bind rat serum albumin (RSA).
  • the illustrated sequences are from VKS designated DOM7r- 15 (SEQ ID NO: 499), DOM7r-16 (SEQ ID NO: 500), DOM7r-17 (SEQ ID NO: 501), DOM7r-18 (SEQ ID NO: 502), DOM7r-19 (SEQ ID NO: 503).
  • FIG. 14A-14B is an illustration of the amino acid sequences of V H S that bind rat serum albumin (RSA).
  • the illustrated sequences are from V H S designated DOM7r-20 (SEQ ID NO: 504), DOM7r-21 (SEQ ID NO: 505), DOM7r-22 (SEQ ID NO: 506), DOM7r-23 (SEQ ID NO: 507), DOM7r-24 (SEQ ID NO: 508), DOM7r- 25 (SEQ ID NO: 509), DOM7r-26 (SEQ ID NO: 510), DOM7r-27 (SEQ ID NO: 511), DOM7r-28 (SEQ ID NO: 512), DOM7r-29 (SEQ ID NO: 513), DOM7r-30 (SEQ ID NO: 514), DOM7r-31 (SEQ ID NO: 515), DOM7r-32 (SEQ ID NO: 516), and DOM7r-33 (SEQ ID NO: 517).
  • FIG. 15 illustrates the amino acid sequences of several Camelid V HH S that bind mouse serum albumin that are disclosed in WO 2004/041862.
  • Sequence A (SEQ ID NO: 518), Sequence B (SEQ ID NO: 519), Sequence C (SEQ ID NO: 520), Sequence D (SEQ ID NO: 521), Sequence E (SEQ ID NO: 522), Sequence F (SEQ ID NO: 523), Sequence G (SEQ ID NO: 524), Sequence H (SEQ ID NO: 525),
  • Sequence I (SEQ ID NO:526), Sequence J (SEQ ID NO:527), Sequence K (SEQ ID NO: 528), Sequence L (SEQ ID NO:529), Sequence M (SEQ ID NO:530), Sequence N (SEQ ID NO:531), Sequence O (SEQ ID NO: 532), Sequence P (SEQ ID NO:
  • FIG. 16 is a map of a vector used to prepare IgG-like formats.
  • FIG. 17A-17F illustrates the amino acid sequences of human dAbs that bind human EGFR.
  • the amino acid sequences presented are SEQ ID NOS:541-622, 725 and 726.
  • the sequences are continuous with no gaps, the symbols ⁇ , ⁇ and ⁇ have been inserted to show the locations of the CDRs.
  • CDRl is flanked by ⁇
  • CDR2 is flanked by ⁇
  • CDR3 is flanked by ⁇ .
  • FIG. 18A-18L illustrates nucleotide sequences that encode the dAbs shown in FIG. 17A-17F.
  • the nucleotide sequences presented are SEQ ID NOS:623-703, 727 and 728.
  • FIG. 19 illustrates the amino acid sequence (SEQ ID NO:704) of a human dAb that binds VEGF, and a nucleotide sequence (SEQ ID NO:705) that encodes the dAb.
  • the sequences are continuous with no gaps, the symbols ⁇ ,— ⁇ and ⁇ have been inserted to show the locations of the CDRs.
  • CDRl is flanked by ⁇
  • CDR2 is flanked by—
  • CDR3 is flanked by ⁇ .
  • ligand refers to a compound that comprises at least one peptide, polypeptide or protein moiety that has a binding site with binding specificity for a desired endogenous target compound.
  • the ligands according to the invention preferably comprise immunoglobulin variable domains which have different binding specificities, and do not contain variable domain pairs which have the same specificity.
  • each domain which has a binding site that has binding specificity for a cell surface target is an immunoglobulin single variable domain ⁇ e.g., immunoglobulin single heavy chain variable domain ⁇ e.g., V H , V HH ) immunoglobulin single light chain variable domain ⁇ e.g., V L )) that has binding specificity for a desired cell surface target ⁇ e.g., a membrane protein, such as a receptor protein).
  • Each polypeptide domain which has a binding site that has binding specificity for a cell surface target can also comprise one or more complementarity determining regions (CDRs) of an antibody or antibody fragment ⁇ e.g., an immunoglobulin single variable domain) that has binding specificity for a desired cell surface target in a suitable format, such that the binding domain has binding specificity for the cell surface target.
  • CDRs can be grafted onto a suitable protein scaffold or skeleton, such as an affibody, an SpA scaffold, an LDL receptor class A domain, or an EGF domain.
  • the ligand can be bivalent (heterobivalent) or multivalent (heteromultivalent) as described herein.
  • the first and second domains lack domains that share the same specificity.
  • Ligands include polypeptides that comprise two dAbs wherein each dAb binds to a different cell surface target.
  • Ligands also include polypeptides that comprise at least two dAbs that bind different cell surface targets (or the CDRs of a dAbs) in a suitable format, such as an antibody format (e.g., IgG-like format, scFv, Fab, Fab', F(ab') 2 ) or a suitable protein scaffold or skeleton, such as an affibody, an SpA scaffold, an LDL receptor class A domain, an EGF domain, avimer and
  • a suitable format such as an antibody format (e.g., IgG-like format, scFv, Fab, Fab', F(ab') 2 ) or a suitable protein scaffold or skeleton, such as an affibody, an SpA scaffold, an LDL receptor class A domain, an EGF domain, avimer and
  • the polypeptide domain which has a binding site that has binding specificity for a cell surface target can also be a protein domain comprising a binding site for a desired target, e.g., a protein domain is selected from an affibody, an SpA domain, an LDL receptor class A domain, an avimer (see, e.g., U.S. Patent Application Publication Nos. 2005/0053973, 2005/0089932, 2005/0164301).
  • a "ligand” can further comprise one or more additional moieties, that can each independently be a peptide, polypeptide or protein moiety or a non-peptidic moiety (e.g., a polyalkylene glycol, a lipid, a carbohydrate).
  • the ligand can further comprise a half-life extending moiety as described herein (e.g., a
  • polyalkylene glycol moiety a moiety comprising albumin, an albumin fragment or albumin variant, a moiety comprising transferrin, a transferrin fragment or transferrin variant, a moiety that binds albumin, a moiety that binds neonatal Fc receptor).
  • target refers to a biological molecule (e.g., peptide, polypeptide, protein, lipid, carbohydrate) to which a polypeptide domain which has a binding site can bind.
  • the target can be, for example, an intracellular target (e.g., an intracellular protein target) or a cell surface target (e.g., a membrane protein, a receptor protein).
  • the target is VEGF or EGFR.
  • immunoglobulin single variable domain refers to an antibody variable region (V H , V HH , V L ) that specifically binds a target, antigen or epitope independently of other V domains; however, as the term is used herein, an immuno globulin single variable domain can be present in a format (e.g., hetero- multimer) with other variable regions or variable domains where the other regions or domains are not required for antigen binding by the single immunoglobulin variable domain (i.e., where the immunoglobulin single variable domain binds antigen independently of the additional variable domains).
  • immunoglobulin single variable domain encompasses not only an isolated antibody single variable domain polypeptide, but also larger polypeptides that comprise one or more monomers of an antibody single variable domain polypeptide sequence.
  • a "domain antibody” or “dAb” is the same as an "immunoglobulin single variable domain” polypeptide as the term is used herein.
  • An immunoglobulin single variable domain polypeptide, as used herein refers to a mammalian immunoglobulin single variable domain polypeptide, preferably human, but also includes rodent (for example, as disclosed in WO 00/29004, the contents of which are incorporated herein by reference in their entirety) or camelid V HH dAbs.
  • camelid dAbs are immunoglobulin single variable domain polypeptides which are derived from species including camel, llama, alpaca, dromedary, and guanaco, and comprise heavy chain antibodies naturally devoid of light chain (V HH )- Similar dAbs, can be obtained for single chain antibodies from other species, such as nurse shark.
  • Preferred ligands comprises at least two different immunoglobulin single variable domain polypeptides or at least two different dAbs.
  • a "human” immunoglobulin single variable domain (e.g., dAb, V H , V L , V K , V ⁇ ) can be derived from an antibody of human origin or from a library prepared using human antibody variable region genes.
  • human immunoglobulin single variable domains have one or more framework regions that are encoded by a human germline antibody gene segment, or that have up to 5 amino acid differences relative to the amino acid sequence encoded by a human germline antibody gene segment.
  • vascular endothelial growth factor refers to naturally occurring or endogenous mammalian VEGF-A proteins and to proteins having an amino acid sequence which is the same as that of a naturally occurring or endogenous corresponding mammalian VEGF-A protein (e.g., recombinant proteins, synthetic proteins (i.e., produced using the methods of synthetic organic chemistry)).
  • the term includes mature VEGF-A protein, polymorphic or allelic variants, and other isoforms of a VEGF-A (e.g., produced by alternative splicing or other cellular processes), and modified or unmodified forms of the foregoing (e.g., lipidated, glycosylated).
  • Alternative splicing of RNA encoding human (Homo sapiens) VEGF-A yield several isoforms of human VEGF- A that differ in the number of amino acids in the protein sequence. For example, isoforms referred to as VEGF-121, VEGF-165, VEGF-189 and VEGF-206 are produced in humans.
  • Naturally occurring or endogenous VEGF-A include wild type proteins such as mature VEGF-A, polymorphic or allelic variants and other isoforms which occur naturally in mammals (e.g., humans, non-human primates). Such proteins can be recovered or isolated from a source which naturally produces VEGF-A, for example. These proteins and proteins having the same amino acid sequence as a naturally occurring or endogenous corresponding VEGF, are referred to by the name of the corresponding mammal. For example, where the corresponding mammal is a human, the protein is designated as a human VEGF.
  • a ligand e.g., immunoglobulin single variable domain
  • a ligand that inhibits binding of VEGF to VEGFRl or VEGFR2 inhibits binding in the VEGFRl binding assay or VEGFR2 assay described herein by at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 85%, at least about 90%, or at least about 95% when the ligand is assayed at a concentration of about 1 nM, about 10 nM, about 50 nM, about 100 nM, about 1 ⁇ M, about 10 ⁇ M or about 100 ⁇ M.
  • a ligand that inhibits binding of VEGF to VEGFRl or VEGFR2 can also or alternatively, inhibit binding in the VEGFRl binding assay or VEGFR2 assay with an IC50 of about 1 ⁇ M or less, about 500 nM or less, about 100 nM or less, about 75 nM or less, about 50 nM or less, about 10 nM or less or about 1 nM or less.
  • a ligand e.g., immunoglobulin single variable domain
  • a ligand that inhibits activity of VEGF inhibits viability in the VEGF bioassay described herein by at least about 20%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 85%, at least about 90%, or at least about 95%.
  • a ligand e.g., immunoglobulin single variable domain
  • a ligand that does not substantially inhibit binding of VEGF to VEGFRl or VEGFR2 does not
  • such a ligand might inhibit binding of VEGF in the VEGFRl binding assay or VEGFR2 assay described herein with an IC50 of about 1 mM or higher, or inhibit binding by no more than about 20%, no more than about 15%, no more than about 10% or no more than about 5%.
  • EGFR epidermal growth factor receptor
  • the term includes mature EGFR protein, polymorphic or allelic variants, and other isoforms of a EGFR (e.g., produced by alternative splicing or other cellular processes), and modified or unmodified forms of the foregoing (e.g., lipidated, glycosylated).
  • Naturally occurring or endogenous EGFR include wild type proteins such as mature EGFR, polymorphic or allelic variants and other isoforms which occur naturally in mammals (e.g., humans, non-human primates). Such proteins can be recovered or isolated from a source which naturally produces EGFR, for example. These proteins and proteins having the same amino acid sequence as a naturally occurring or endogenous corresponding EGFR, are referred to by the name of the corresponding mammal. For example, where the corresponding mammal is a human, the protein is designated as a human EGFR.
  • a ligand e.g., immunoglobulin single variable domain
  • a ligand that inhibits binding of EGF and/or TGF alpha to EGFR inhibits binding in the EGFR binding assay or EGFR kinase assay described herein with an IC50 of about 1 ⁇ M or less, about 500 nM or less, about 100 nM or less, about 75 nM or less, about 50 nM or less, about 10 nM or less or about 1 nM or less.
  • a ligand ⁇ e.g., immunoglobulin single variable domain) that inhibits activity of EGFR inhibits kinase activity of EGFR in the EGFR kinase assay described herein with an IC50 of about 1 ⁇ M or less, about 500 nM or less, about 100 nM or less, about 75 nM or less, about 50 nM or less, about 10 nM or less or about 1 nM or less.
  • a ligand ⁇ e.g., immunoglobulin single variable domain) that does not substantially inhibit binding of EGF or TGF alpha to EGFR does not significantly inhibit binding of EGF and/or TGF alpha to EGFR in the receptor binding assay or kinase assay described herein.
  • a ligand might inhibit binding of EGF or TGF alpha to EGFR in the receptor binding assay or kinase assay described herein with an IC50 of about 1 mM or higher.
  • avidity refers to the overall strength of binding between the targets ⁇ e.g., first cell surface target and second cell surface target) on the cell and the ligand. Avidity is more than the sum of the individual affinities for the individual targets.
  • toxin moiety refers to a moiety that comprises a toxin.
  • a toxin is an agent that has deleterious effects on or alters cellular physiology ⁇ e.g., causes cellular necrosis, apoptosis or inhibits cellular division).
  • dose refers to the quantity of ligand administered to a subject all at one time (unit dose), or in two or more administrations over a defined time interval.
  • dose can refer to the quantity of ligand ⁇ e.g., ligand comprising an immunoglobulin single variable domain that binds VEGF and an immunoglobulin single variable domain that binds EGFR) administered to a subject over the course of one day (24 hours) (daily dose), two days, one week, two weeks, three weeks or one or more months ⁇ e.g., by a single administration, or by two or more administrations).
  • the interval between doses can be any desired amount of time.
  • complementary refers to when two immunoglobulin domains belong to families of structures which form cognate pairs or groups or are derived from such families and retain this feature. For example, a Vpj domain and a VL domain of an antibody are complementary; two Vg domains are not
  • VL domains are not complementary.
  • Complementary domains may be found in other members of the immunoglobulin superfamily, such as the V ⁇ and V ⁇ (or ⁇ and ⁇ ) domains of the T-cell receptor. Domains which are artificial, such as domains based on protein scaffolds which do not bind epitopes unless engineered to do so, are non- complementary. Likewise, two domains based on (for example) an immunoglobulin domain and a fibronectin domain are not complementary.
  • immunoglobulin refers to a family of polypeptides which retain the immunoglobulin fold characteristic of antibody molecules, which contains two ⁇ sheets and, usually, a conserved disulphide bond.
  • immunoglobulin superfamily are involved in many aspects of cellular and non- cellular interactions in vivo, including widespread roles in the immune system (for example, antibodies, T-cell receptor molecules and the like), involvement in cell adhesion (for example the ICAM molecules) and intracellular signaling (for example, receptor molecules, such as the PDGF receptor).
  • the present invention is applicable to all immunoglobulin superfamily molecules which possess binding domains.
  • the present invention relates to antibodies.
  • domain refers to a folded protein structure which retains its tertiary structure independently of the rest of the protein. Generally, domains are responsible for discrete functional properties of proteins, and in many cases maybe added, removed or transferred to other proteins without loss of function of the remainder of the protein and/or of the domain.
  • single antibody variable domain is meant a folded polypeptide domain comprising sequences characteristic of antibody variable domains.
  • each ligand comprises at least two different domains.
  • "Repertoire" A collection of diverse variants, for example polypeptide variants which differ in their primary sequence.
  • a library used in the present invention will encompass a repertoire of polypeptides comprising at least 1000 members.
  • Library refers to a mixture of heterogeneous polypeptides or nucleic acids.
  • the library is composed of members, each of which have a single polypeptide or nucleic acid sequence. To this extent, library is synonymous with repertoire. Sequence differences between library members are responsible for the diversity present in the library.
  • the library may take the form of a simple mixture of polypeptides or nucleic acids, or may be in the form of organisms or cells, for example bacteria, viruses, animal or plant cells and the like, transformed with a library of nucleic acids.
  • each individual organism or cell contains only one or a limited number of library members.
  • a library may take the form of a population of host organisms, each organism containing one or more copies of an expression vector containing a single member of the library in nucleic acid form which can be expressed to produce its
  • the population of host organisms has the potential to encode a large repertoire of genetically diverse polypeptide variants.
  • an antibody refers to IgG, IgM, IgA, IgD or IgE or a fragment (such as a Fab , F(ab')2, Fv, disulphide linked Fv, scFv, closed conformation multispecific antibody, disulphide-linked scFv, diabody) whether derived from any species naturally producing an antibody, or created by recombinant DNA
  • an "antigen' is a molecule that is bound by a binding domain according to the present invention.
  • antigens are bound by antibody ligands and are capable of raising an antibody response in vivo. It may be a polypeptide, protein, nucleic acid or other molecule.
  • the dual-specific ligands according to the invention are selected for target specificity against two particular targets ⁇ e.g., antigens).
  • the antibody binding site defined by the variable loops (Ll , L2, L3 and Hl, H2, H3) is capable of binding to the antigen.
  • An “epitope” is a unit of structure conventionally bound by an
  • immunoglobulin VJJ/VL pair- Epitopes define the minimum binding site for an antibody, and thus represent the target of specificity of an antibody.
  • an epitope represents the unit of structure bound by a variable domain in isolation.
  • Universal framework refers to a single antibody framework sequence corresponding to the regions of an antibody conserved in sequence as defined by Kabat ("Sequences of Proteins of Immunological Interest", US Department of Health and Human Services) or corresponding to the human germline
  • the invention provides for the use of a single framework, or a set of such frameworks, which has been found to permit the derivation of virtually any binding specificity though variation in the hypervariable regions alone.
  • half-life refers to the time taken for the serum concentration of the ligand to reduce by 50%, in vivo, for example due to degradation of the ligand and/or clearance or sequestration of the dual-specific ligand by natural mechanisms.
  • the ligands of the invention are stabilized in vivo and their half-life increased by binding to molecules which resist degradation and/or clearance or sequestration.
  • such molecules are naturally occurring proteins which themselves have a long half-life in vivo.
  • the half-life of a ligand is increased if its functional activity persists, in vivo, for a longer period than a similar ligand which is not specific for the half-life increasing molecule.
  • a ligand specific for HSA and two target molecules is compared with the same ligand wherein the specificity to HSA is not present, that is does not bind HSA but binds another molecule. For example, it may bind a third target on the cell.
  • the half-life is increased by 10%, 20%, 30%, 40%, 50% or more.
  • the term "competes" means that the binding of a first target to its cognate target binding domain is inhibited when a second target is bound to its cognate target binding domain.
  • binding may be inhibited sterically, for example by physical blocking of a binding domain or by alteration of the structure or environment of a binding domain such that its affinity or avidity for a target is reduced.
  • a protein moiety competes for binding to a target (e.g., EGFR, VEGF, serum albumin) with another agent, when the protein moiety inhibits binding of the other agent to the target in a competitive binding assay (e.g., a competitive ELISA or other suitable binding assay).
  • the protein moiety can inhibit binding of another agent that binds a target (e.g., EGFR, VEGF, serum albumin) in a competitive binding assay by at least about 25%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, or at least about 95%.
  • a target e.g., EGFR, VEGF, serum albumin
  • the terms “low stringency,” “medium stringency,” “high stringency,” or “very high stringency conditions” describe conditions for nucleic acid hybridization and washing. Guidance for performing hybridization reactions can be found in Current Protocols in Molecular Biology, John Wiley & Sons, N.Y. (1989), 6.3.1-6.3.6, which is incorporated herein by reference in its entirety.
  • Aqueous and nonaqueous methods are described in that reference and either can be used.
  • Specific hybridization conditions referred to herein are as follows: (1) low stringency hybridization conditions in 6X sodium chloride/sodium citrate (SSC) at about 45C, followed by two washes in 0.2X SSC, 0.1% SDS at least at 5OC (the temperature of the washes can be increased to 55C for low stringency conditions); (2) medium stringency hybridization conditions in 6X SSC at about 45C, followed by one or more washes in 0.2X SSC, 0.1 % SDS at 6OC; (3) high stringency hybridization conditions in 6X SSC at about 45C, followed by one or more washes in 0.2X SSC, 0.1% SDS at 65C; and preferably (4) very high stringency
  • hybridization conditions are 0.5M sodium phosphate, 7% SDS at 65C, followed by one or more washes at 0.2X SSC, 1% SDS at 65C.
  • Very high stringency conditions (4) are the preferred conditions and the ones that should be used unless otherwise specified. Sequences similar or homologous (e.g., at least about 70% sequence identity) to the sequences disclosed herein are also part of the invention. In some
  • the sequence identity at the amino acid level can be about 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or higher.
  • the sequence identity can be about 70%, 75%, 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or higher.
  • substantial identity exists when the nucleic acid segments will hybridize under selective hybridization conditions (e.g., very high stringency hybridization conditions), to the complement of the strand.
  • the nucleic acids may be present in whole cells, in a cell lysate, or in a partially purified or substantially pure form.
  • sequence identity or “sequence identity” or “similarity” between two sequences (the terms are used interchangeably herein) are performed as follows.
  • the sequences are aligned for optimal comparison purposes (e.g., gaps can be introduced in one or both of a first and a second amino acid or nucleic acid sequence for optimal alignment and non-homologous sequences can be disregarded for comparison purposes).
  • the length of a reference sequence aligned for comparison purposes is at least 30%, preferably at least 40%, more preferably at least 50%, even more preferably at least 60%, and even more preferably at least 70%, 80%, 90%, or 100% of the length of the reference sequence.
  • amino acid residues or nucleotides at corresponding amino acid positions or nucleotide positions are then compared.
  • a position in the first sequence is occupied by the same amino acid residue or nucleotide as the corresponding position in the second sequence, then the molecules are identical at that position (as used herein amino acid or nucleic acid " homology” is equivalent to amino acid or nucleic acid “identity”).
  • the percent identity between the two sequences is a function of the number of identical positions shared by the sequences, taking into account the number of gaps, and the length of each gap, which need to be introduced for optimal alignment of the two sequences.
  • Amino acid and nucleotide sequence alignments and homology, similarity or identity, as defined herein are preferably prepared and determined using the algorithm BLAST 2 Sequences, using default parameters (Tatusova, T. A. et al., FEMS Microbiol Lett, 174:187-188 (1999)).
  • the BLAST algorithm version 2.0 is employed for sequence alignment, with parameters set to default values.
  • BLAST Basic Local Alignment Search Tool
  • blastp, blastn, blastx, tblastn, and tblastx are the heuristic search algorithm employed by the programs blastp, blastn, blastx, tblastn, and tblastx; these programs ascribe significance to their findings using the statistical methods of Karlin and Altschul, 1990, Proc. Natl. Acad. Sd. USA 87(6):2264-8.
  • the invention relates to ligands that have binding specificity for VEGF ⁇ e.g. , human VEGF), ligands that have binding specificity for EGFR ⁇ e.g., human EGFR), and to ligands that have binding specificity for VEGF and EGFR ⁇ e.g., human VEGF and human EGFR).
  • the ligand can comprise a polypeptide domain having a binding site with binding specificity for VEGF, a polypeptide domain having a binding site with binding specificity for EGFR, or comprise a polypeptide domain having a binding site with binding specificity for VEGF and a polypeptide domain having a binding site with binding specificity for EGFR.
  • the ligands of the invention provide several advantages.
  • the ligand can be tailored to have a desired in vivo serum half-life.
  • the ligands can be used to control, reduce, or eliminate general toxicity of therapeutic agents, such as cytotoxin used to treat cancer.
  • dAbs are much smaller than conventional antibodies, and can be administered to achieve better tissue penetration than conventional antibodies.
  • dAbs and ligands that comprise a dAb provide advantages over conventional antibodies when administered to treat cancer, for example by targeting solid tumors.
  • cancers overexpress EGFR, and ligands that have binding specificity for EGFR and VEGF can be administered to target VEGF-inhibitory activity to tumors or the environment of cancer cells.
  • This approach provides two beneficial activities directly at the site of a tumor or cancer, i.e., direct anti-cancer activity by binding to EGFR and inhibiting binding of ligands ⁇ e.g., EGF, TGF alpha) to the receptor, and inhibition of angiogenesis that supports tumor formation and development.
  • ligands that have binding specificity for VEGF and EGFR can be administered to a patient with cancer ⁇ e.g., EGFR-expressing cancer) to provide superior therapy using a single therapeutic agent.
  • signals transduced through EGFR can lead to the production of angiogenic factors, such as VEGF.
  • Cancer cells ⁇ e.g., in a tumor) that express or overexpress EGFR can produce a high level of VEGF that acts locally to induce formation of tumor vasculature.
  • the ligands of the invention that have binding specificity for VEGF and EGFR can be administered to a subject to target delivery of the VEGF inhibitory activity of the ligand to cells that overexpress EGFR.
  • anti-angiogenic therapy can be delivered specifically to sites where VEGF is being produced ⁇ e.g., to cells that overexpress EGFR).
  • the ligand has binding specificity for VEGF and comprises an (at least one) immunoglobulin single variable domain with binding specificity for VEGF. In other embodiments, the ligand has binding specificity for EGFR and comprises an (at least one) immunoglobulin single variable domain with binding specificity for EGFR. In certain embodiments, the ligand has binding specificity for VEGF and EGFR, and comprises an (at least one) immunoglobulin single variable domain with binding specificity for VEGF and an (at least one) immunoglobulin single variable domain with binding specificity for EGFR.
  • the ligand of the invention can be formatted as described herein
  • the ligand of the invention can be formatted to tailor in vivo serum half- life.
  • the ligand can further comprise a toxin or a toxin moiety as described herein.
  • the ligand comprises a surface active toxin, such as a free radical generator (e.g., selenium containing toxin) or a radionuclide.
  • the toxin or toxin moiety is a polypeptide domain (e.g., a dAb) having a binding site with binding specificity for an intracellular target.
  • the ligand is an IgG-like format that has binding specificity for VEGF and EGFR (e.g., human VEGF and human EGFR).
  • the ligand of the invention can be formatted as a monospecific, dual specific or multispecific ligand as described herein. See, also WO 03/002609, the entire teachings of which are incorporated herein by reference, regarding ligand formatting.
  • Such dual specific ligands comprise immunoglobulin single variable domains that have different binding specificities.
  • Such dual specific ligands can comprise combinations of heavy and light chain domains.
  • the dual specific ligand may comprise a V H domain and a V L domain, which may be linked together in the form of an scFv (e.g., using a suitable linker such as GIy 4 S er), or formatted into a bispecif ⁇ c antibody or antigen-binding fragment thereof (e.g.
  • the dual specific ligands do not comprise complementary V J /V L pairs which form a conventional two chain antibody antigen-binding site that binds antigen or epitope co-operatively. Instead, the dual format ligands comprise a V H /V L complementary pair, wherein the V domains have different binding specificities.
  • the dual specific ligands may comprise one or more C H or C L domains if desired.
  • a hinge region may also be included if desired.
  • Such combinations of domains may, for example, mimic natural antibodies, such as IgG or IgM, or fragments thereof, such as Fv, scFv, Fab or F(ab') 2 molecules.
  • Other structures, such as a single arm of an IgG molecule comprising V H , V L , C H I and C L domains, are envisaged.
  • the dual specific ligand of the invention comprises only two variable domains although several such ligands may be incorporated together into the same protein, for example two such ligands can be incorporated into an IgG or a multimeric immunoglobulin, such as IgM.
  • a plurality of dual specific ligands are combined to form a multimer.
  • two different dual specific ligands are combined to create a tetra-specific molecule.
  • the light and heavy variable regions of a dual-specific ligand produced according to the method of the present invention may be on the same polypeptide chain, or alternatively, on different polypeptide chains.
  • the variable regions are on different polypeptide chains, then they may be linked via a linker, generally a flexible linker (such as a polypeptide chain), a chemical linking group, or any other method known in the art.
  • the linker may be a "natural linker" that contains carboxy-terminal amino acids of an antibody variable domain and amino-terminal amino acids of an antibody constant domain.
  • a natural linker can contain the carboxy-terminal amino acids of Vk and amino-terminal amino acids of Ck (e.g. KVEIKRTVAAPS (SEQ ID NO:706)).
  • the linker can contain fewer Lys and Arg residues than the natural linker (e.g., LVTVSSAST (SEQ ID NO:707) or (LVTVS S GGGGS GGGS (SEQ ID NO:708)). If desired, the linker can be mutated to substitute some or all of the positively charged residues
  • linker e.g., in a natural linker
  • Lys and/or Arg with residues that are not positively charged at physiological pH.
  • Lys and/or Arg residues can be replaced with Asn, Leu, GIn or Ser.
  • This type of linker provides the advantage of reducing protease sensitivity (e.g., serine protease, cysteine protease, matrix metalloprotease, pepsin, trypsin, elastase, chymotrypsin, carboxypeptidase, cathepsin (e.g., cathepsin G), proteinase 3).
  • linkers include, GQGTNVEINRTVAAPS (SEQ ID NO:710), GQGTNVEINQTVAAPS (SEQ ID NO:711),
  • GQGTNVEIQRTVAAPS SEQ ID NO:712
  • GQGTLVTVSSTVAAPS SEQ ID NO:713
  • Proteases e.g., a serine protease, cysteine protease, matrix metalloprotease, pepsin, trypsin, elastase, chymotrypsin, carboxypeptidase, cathepsin (e.g., cathepsin G), proteinase 3) function in the normal turn over and metabolism of proteins.
  • proteases present in a tissue, organ or animal (e.g., in the lung, in or adjacent to a tumor) can increase.
  • This increase in proteases can result in accelerated degradation and inactivation of endogenous proteins and of therapeutic or diagnostic peptides, polypeptides and proteins that are administered.
  • agents that have potential for in vivo use e.g., use in treating, diagnosing or preventing disease
  • have only limited efficacy because they are rapidly degraded and inactivated by proteases .
  • the invention relates to ligand comprising a linker that is resistant to protease degradation.
  • the protease resistant ligands of the invention provide several advantages. For example, a protease resistant ligand can be administered to a subject and remain active in vivo longer than protease sensitive agents.
  • protease resistant ligand will remain functional for a period of time that is sufficient to produce biological effects.
  • a ligand or linker that is resistant to protease degradation is not substantially degraded by a protease when incubated with the protease under conditions suitable for protease activity for at least about 2 hours, at least about 3 hours, at least about 4 hours, at least about 5 hours, at least about 6 hours, at least about 7 hours, at least about 8 hours, at least about 9 hours, at least about 10 hours, at least about 11 hours, at least about 12 hours, at least about 24 hours, at least about 36 hours, or at least about 48 hours.
  • a ligand or linker is not substantially degraded when no more than about 25%, no more than about 20%, no more than about 15%, no more than about 14%, no more than about 13%, no more than about 12%, no more than about 11%, no more than about 10%, no more than about 9%, no more than about 8%, no more than about 7% no more than about 6%, no more than about 5%, no more than about 4%, no more than about 3%, no more than about 2%, no more than about 1%, or substantially none of the ligand or linker is degraded by protease after incubation with the protease for at least about 2 hours. Protein degradation can be assessed using any suitable method, for example, by SDS-PAGE.
  • Protease resistance can be assessed using any suitable method.
  • a protease can be added to a solution of ligand or linker in a suitable buffer ⁇ e.g., PBS) to produce a ligand or linker/protease solution, such as a solution of at least about 0.01% (w/w) protease, about 0.01% to about 5% (w/w) protease, about 0.05% to about 5% (w/w) protease, about 0.1% to about 5% (w/w) protease, about 0.5% to about 5% (w/w) protease, about 1% to about 5% (w/w) protease, at least about 0.01% (w/w) protease, at least about 0.02% (w/w) protease, at least about 0.03% (w/w) protease, at least about 0.04% (w/w) protease, at least about 0.05% (w/w) protease,
  • the ligand or linker/protease mixture can be incubated at a suitable temperature for protease activity (e.g., at 37 0 C) and samples can be taken at time intervals (e.g., at 1 hour, 2 hours, 3 hours, etc.) and the protease reaction stopped.
  • the samples can then be analyzed for protein degradation using any suitable method, such as SDS-PAGE analysis. The results can be used to establish a time course of degradation.
  • Ligands can be formatted as bi- or multispecific antibodies or antibody fragments or into bi- or multispecific non-antibody structures.
  • Suitable formats include, any suitable polypeptide structure in which an antibody variable domain or one or more of the CDRs thereof can be incorporated so as to confer binding specificity for antigen on the structure.
  • bispecific IgG-like formats e.g., chimeric antibodies, humanized antibodies, human antibodies, single chain antibodies, heterodimers of antibody heavy chains and/or light chains, antigen-binding fragments of any of the foregoing (e.g., a Fv fragment (e.g., single chain Fv (scFv), a disulfide bonded Fv), a Fab fragment, a Fab' fragment, a F(ab') 2 fragment), a single variable domain (e.g., V H , V L , V HH ), a dAb), and modified versions of any of the foregoing (e.g., modified by the covalent attachment of polyalkylene glycol (e.g., polyethylene glycol, polypropylene glycol, polybutylene glycol) or other suitable polymer).
  • polyalkylene glycol e.g., polyethylene glycol, polypropylene glycol, polybutylene glycol
  • ligands including dAb monomers, dimers and trimers, can be linked to an antibody Fc region, comprising one or both of CH2 and C H 3 domains, and optionally a hinge region.
  • vectors encoding ligands linked as a single nucleotide sequence to an Fc region may be used to prepare such polypeptides.
  • the ligand comprises one, two or more dAbs of the same or different binding specificities, and C H 2, C H 3, C H 2-C H 3, hinge-Cj H [2, hinge-C H 3, hinge-C ⁇ 2-CH3, a portion of hinge-C H 2, a portion of hinge- C H 3, or a portion of hinge-C H 2-C H 3.
  • C H 2, C H 3, C H 2-C H 3, hinge-C ⁇ 2, hinge-C ⁇ 3, hinge-C H 2-C ⁇ 3, the portion of hinge-C ⁇ 2, the portion of hinge-C H 3, and the portion of hinge-C H 2-C H 3 can be from any desired antibody, such as a human IgG, such as a human IgGl or a human IgG4.
  • the ligand of the invention comprises an anti-EGFR dAb or an anti-VEGF dAb that is fused (e.g., directly or through a linker) to an Fc region of an antibody.
  • the ligand is an Fc fusion that comprises an anti-VEGF dAb that is disulfide bonded to an anti-EGFR dAb.
  • the ligand comprises two or more dAbs (e.g., two dAbs that bind EGFR, two dAbs that bind VEGF, a dAb that binds EGFR and a dAb that binds VEGF) and an Fc region, and the ligand has the structure, from amino terminus to carboxy terminus, V H -V H -FC, V L -V L -FC, V H -V L -FC, V L -V H -FC.
  • the ligand can have the structure V H -V K -hinge-CH2-CH3, V K -V H -hinge-CH2-CH3, V ⁇ - V K -hinge-CH2-CH3, or V H -V H -hinge-CH2-CH3.
  • the V H shown in any of the foregoing formulae can be a V HH - TWO ligands that contain Fc regions can be bonded together to form a dimer, for example, through a disulfide bond (e.g., in the hinge region).
  • orientation of the polypeptide domains that have a binding site with binding specificity for a target is a matter of design choice. However, some orientations, with or without linkers, may provide preferred binding characteristics in comparison to other orientations.
  • AU orientations e.g., dAbl -dAb2-Fc; dAb2 -dAb 1-Fc
  • ligands that contain an orientation that provides desired binding characteristics can be easily identified by routine screening.
  • Ligands and dAb monomers can also be combined and/or formatted into non-antibody multi-ligand structures to form multivalent complexes, which bind target molecules with the same antigen, thereby providing superior avidity.
  • natural bacterial receptors such as SpA can be used as scaffolds for the grafting of CDRs to generate ligands which bind specifically to one or more epitopes. Details of this procedure are described in US 5,831,012.
  • Other suitable scaffolds include those based on fibronectin and affibodies. Details of suitable procedures are described in WO 98/58965.
  • Other suitable scaffolds include lipocallin and CTLA4, as described in van den Beuken et at., J. MoI. Biol.
  • Protein scaffolds may be combined; for example, CDRs may be grafted on to a CTLA4 scaffold and used together with immunoglobulin V H or V L domains to form a ligand. Likewise, fibronectin, lipocallin and other scaffolds may be combined
  • antibody chains and formats ⁇ e.g., bispecific IgG-like formats, chimeric antibodies, humanized antibodies, human antibodies, single chain antibodies, homodimers and heterodimers of antibody heavy chains and/or light chains
  • suitable expression constructs and/or culture of suitable cells e.g., hybridomas, heterohybridomas, recombinant host cells containing recombinant constructs encoding the format).
  • formats such as antigen-binding fragments of antibodies or antibody chains ⁇ e.g., bispecific binding fragments, such as a Fv fragment ⁇ e.g., single chain Fv (scFv), a disulfide bonded Fv), a Fab fragment, a Fab' fragment, a F(ab') 2 fragment), can be prepared by expression of suitable expression constructs or by enzymatic digestion of antibodies, for example using papain or pepsin.
  • Fv fragment ⁇ e.g., single chain Fv (scFv), a disulfide bonded Fv), a Fab fragment, a Fab' fragment, a F(ab') 2 fragment
  • the ligand can be formatted as a multispecific ligand, for example as described in WO 03/002609, the entire teachings of which are incorporated herein by reference. Such multispecific ligands possess more than one epitope binding specificity.
  • the multi-specific ligand comprises two or more epitope binding domains, such as dAbs or non-antibody protein domain comprising a binding site for an epitope, e.g., an affibody, an SpA domain, an LDL receptor class A domain, an EGF domain, an avimer.
  • Multispecific ligands can be formatted further as described herein.
  • the ligand is an IgG-like format.
  • Such formats have the conventional four chain structure of an IgG molecule (2 heavy chains and two light chains), in which one or more of the variable regions (V H and or V L ) have been replaced with a dAb or single variable domain of a desired specificity.
  • each of the variable regions (2 V H regions and 2 V L regions) is replaced with a dAb or single variable domain.
  • the dAb(s) or single variable domain(s) that are included in an IgG-like format can have the same specificity or different specificities.
  • the IgG-like format is tetravalent and can have one, two, three or four specificities.
  • the IgG-like format can be monospecific and comprises 4 dAbs that have the same specificity; bispecific and comprises 3 dAbs that have the same specificity and another dAb that has a different specificity;
  • Antigen- binding fragments of IgG-like formats e.g., Fab, F(ab') 2 , Fab', Fv, scFy
  • IgG-like formats e.g., Fab, F(ab') 2 , Fab', Fv, scFy
  • a particular constant region of Fc portion (e.g., of an IgG, such as IgGl), variant or portion thereof can be selected in order to tailor effector function.
  • the ligand can be an IgGl -like format.
  • the IgG-like format can comprise a mutated constant region (variant IgG heavy chain constant region) to minimize binding to Fc receptors and/or ability to fix complement, (see e.g. Winter et al, GB 2,209,757 B; Morrison et al, WO 89/07142;
  • the IgG-like formats can comprise an anti-EGFR dAb (e.g., DOM16-39-542, DOM16-39-618 or DOM16-39-619), an anti-VEGF dAb (e.g., DOMl 5-26-501), or an anti-EGFR dAb and an anti-VEGF dAb.
  • an anti-EGFR dAb e.g., DOM16-39-542, DOM16-39-618 or DOM16-39-619
  • an anti-VEGF dAb e.g., DOMl 5-26-501
  • an anti-EGFR dAb and an anti-VEGF dAb e.g., DOMl 5-26-501
  • the ligands of the invention can be formatted as a fusion protein that contains a first immunoglobulin single variable domain that is fused directly to a second immunoglobulin single variable domain. If desired such a format can further comprise a half-life extending moiety.
  • the ligand can comprise a first immunoglobulin single variable domain that is fused directly to a second immunoglobulin single variable domain.
  • the ligand can be an in line fusion of two or more protein moieties that have a binding site with binding specificity for EGFR that competes for binding to EGFR with an anti-EGFR domain antibody (e.g., any of the DOMl 6 dAbs disclosed herein) and fused to an anti-serum albumin dAb (e.g., any of the D0M7 dAbs disclosed herein).
  • an anti-EGFR domain antibody e.g., any of the DOMl 6 dAbs disclosed herein
  • an anti-serum albumin dAb e.g., any of the D0M7 dAbs disclosed herein.
  • the protein moieties that have a binding site with binding specificity for EGFR have different epitopic specificities.
  • the ligand is an in line fusion protein comprising a protein moiety that has a binding site with binding specificity for EGFR (e.g., anti-EGFR dAb), a protein moiety that has a binding site with binding specificity for VEGF, and an anti-serum albumin dAb.
  • such an in line fusion comprises DOMl 6-39-618 dAb and/or DOM16-39-619 and an anti-serum albumin dAb (e.g., DOM16-39- 618— DOM7h-14, DOM7h-14— DOMl 6-39-618, DOM16-39-619— DOM7h-14, DOM7h-14— DOM16-39-619).
  • an anti-serum albumin dAb e.g., DOM16-39- 618— DOM7h-14, DOM7h-14— DOMl 6-39-618, DOM16-39-619— DOM7h-14, DOM7h-14— DOM16-39-619.
  • the in line fusion comprises a protein moiety (e.g., dAb) that has at least about 80%, 85%, 87%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% amino acid sequence identiy with the amino acid sequence of DOMl 6-39-618, a protein moiety that has at least about 80%, 85%, 87%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% amino acid sequence identity with the amino acid sequence of DOM 16-39-619, and/or a protein moiety (e.g., dAb) that has at least about 80%, 85%, 87%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% amino acid sequence identiy with the amino acid sequence of an anti-serum albumin dAb disclosed herein, such as DOM7h-14.
  • a protein moiety e
  • the ligand comprises an anti-VEGF dAb, an anti-EGFR dAb, and an anti-serum albumin dAb (e.g., DOMl 5-10— DOMl 6-39— anti-serum albumin dAb, DOMl 6-39— DOMl 5-10— anti-serum albumin dAb, DOM15-26-501— DOM16-39— anti-serum albumin dAb, DOM16-39— D0M15- 26-501— anti-serum albumin dAb).
  • an anti-serum albumin dAb e.g., DOMl 5-10— DOMl 6-39— anti-serum albumin dAb, DOMl 6-39— DOMl 5-10— anti-serum albumin dAb, DOM15-26-501— DOM16-39— anti-serum albumin dAb, DOM16-39— D0M15- 26-501— anti-serum albumin dAb
  • the in line fusion comprises a protein moiety (e.g., dAb) that has at least about 80%, 85%, 87%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% amino acid sequence identiy with the amino acid sequence of an anti-VEGF dAb disclosed herein, such as DOMl 5-10 or DOMl 5-25-501, and/or a protein moiety that has at least about 80%, 85%, 87%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% amino acid sequence identity with the amino acid sequence of an anti-EGFR dAb disclosed herein, such as DOM16-39, and/or a protein moiety (e.g., dAb) that has at least about 80%, 85%, 87%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% amino acid sequence identi
  • orientation of the polypeptide domains that have a binding site with binding specificity for a target, and whether the ligand comprises a linker is a matter of design choice. However, some orientations, with or without linkers, may provide better binding characteristics than other orientations.
  • AU orientations e.g., dAbl-linker-dAb2; dAb2-linker-dAbl
  • ligands that contain an orientation that provides desired binding characteristics can be easily identified by screening.
  • the ligands and dAb monomers disclosed herein can be formatted to extend their in vivo serum half-life. Increased in vivo half-life is useful in in vivo applications of immunoglobulins, especially antibodies and most especially antibody fragments of small size such as dAbs. Such fragments (Fvs, disulphide bonded Fvs, Fabs, scFvs, dAbs) are rapidly cleared from the body, which can limit clinical applications.
  • a ligand can be formatted as a larger antigen-binding fragment of an antibody or as an antibody (e.g., formatted as a Fab, Fab', F(ab) 2 , F(ab') 2 , IgG, scFv) that has larger hydrodynamic size.
  • Ligands can also be formatted to have a larger hydrodynamic size, for example, by attachment of a polyalkyleneglycol group (e.g. polyethyleneglycol (PEG) group, polypropylene glycol, polybutylene glycol), serum albumin, transferrin, transferrin receptor or at least the transferrin-binding portion thereof, an antibody Fc region, or by conjugation to an antibody domain.
  • a polyalkyleneglycol group e.g. polyethyleneglycol (PEG) group, polypropylene glycol, polybutylene glycol
  • serum albumin e.g. polyethyleneglycol (PEG) group, polypropylene glycol, polybuty
  • the ligand e.g., dAb monomer
  • the PEGylated ligand e.g., dAb monomer
  • the ligand can be a PEGylated ligand comprising a dAb that binds VEGF or EGFR with an affinity or avidity that differs from the avidity of ligand in unPEGylated form by no more than a factor of about 1000, preferably no more than a factor of about 100, more preferably no more than a factor of about 10, or with affinity or avidity substantially unchanged relative to the unPEGylated form. See,
  • PCT/GB03/002804 filed June 30, 2003, which designated the United States, (WO 2004/081026) regarding PEGylated single variable domains and dAbs, suitable methods for preparing same, increased in vivo half-life of the PEGylated single variable domains and dAb monomers and multimers, suitable PEGs, preferred hydrodynamic sizes of PEGs, and preferred hydrodynamic sizes of PEGylated single variable domains and dAb monomers and multimers.
  • the entire teaching of PCT/GB03/002804 (WO 2004/081026), including the portions referred to above, are incorporated herein by reference.
  • Hydrodynamic size of the ligands (e.g., dAb monomers and multimers) of the invention may be determined using methods which are well known in the art. For example, gel filtration chromatography may be used to determine the hydrodynamic size of a ligand. Suitable gel filtration matrices for determining the hydrodynamic sizes of ligands, such as cross-linked agarose matrices, are well known and readily available.
  • the size of a ligand format (e.g., the size of a PEG moiety attached to a dAb monomer), can be varied depending on the desired application. For example, where ligand is intended to leave the circulation and enter into peripheral tissues, it is desirable to keep the hydrodynamic size of the ligand low to facilitate extravazation from the blood stream.
  • the size of the ligand can be increased, for example by formatting as an Ig like protein or by addition of a 30 to 60 kDa PEG moiety (e.g., linear or branched PEG 30 to 40 IcDa PEG, such as addition of two 20IcDa PEG moieties.)
  • the size of the ligand format can be tailored to achieve a desired in vivo serum half-life, for example to control exposure to a toxin and/or to reduce side effects of toxic agents.
  • the hydrodynamic size of ligand (e.g., dAb monomer) and its serum half-life can also be increased by conjugating or linking the ligand to a binding domain (e.g., antibody or antibody fragment) that binds an antigen or epitope that increases half- life in vivo, as described herein.
  • a binding domain e.g., antibody or antibody fragment
  • the ligand (e.g., dAb monomer) can be conjugated or linked to an anti-serum albumin or anti-neonatal Fc receptor antibody or antibody fragment, e.g., an anti-SA or anti-neonatal Fc receptor dAb, Fab, Fab' or scFv, or to an anti-SA affibody or anti-neonatal Fc receptor affibody.
  • albumin, albumin fragments or albumin variants for use in a ligand according to the invention are described in WO 2005/077042A2, which is incorporated herein by reference in its entirety.
  • albumin, albumin fragments or albumin variants can be used in the present invention:
  • Albumin fragment or variant comprising or consisting of amino acids 1-387 of SEQ ID NO:1 in WO 2005/077042A2;
  • Albumin or fragment or variant thereof, comprising an amino acid sequence selected from the group consisting of: (a) amino acids 54 to 61 of SEQ ID NO:1 in WO 2005/077042 A2; (b) amino acids 76 to 89 of SEQ ID NO:1 in WO 2005/077042A2; (c) amino acids 92 to 100 of SEQ ID NO: 1 in WO
  • albumin fragments and analogs for use in a ligand according to the invention are described in WO 03/076567A2, which is incorporated herein by reference in its entirety.
  • albumin, fragments or variants can be used in the present invention:
  • HA Human serum albumin
  • An albumin fragment or variant as described in EP 322094 e.g., HA(l-373., HA(l-388), HA(l-389), HA(l-369), and HA(1-419) and fragments between 1-369 and 1-419;
  • An albumin fragment or variant as described in EP 399666 e.g., HA(1-177) and HA(I -200) and fragments between HA(I-X), where X is any number from 178 to 199.
  • a (one or more) half-life extending moiety ⁇ e.g., albumin, transferrin and fragments and analogues thereof
  • it can be conjugated to the ligand using any suitable method, such as, by direct fusion to the target-binding moiety (e.g., dAb or antibody fragment), for example by using a single nucleotide construct that encodes a fusion protein, wherein the fusion protein is encoded as a single polypeptide chain with the half-life extending moiety located N- or C-terminally to the cell surface target binding moieties.
  • conjugation can be achieved by using a peptide linker between moieties, e.g., a peptide linker as described in WO 03/076567A2 or WO 2004/003019 (these linker disclosures being incorporated by reference in the present disclosure to provide examples for use in the present invention).
  • a polypeptide that enhances serum half-life in vivo is a polypeptide which occurs naturally in vivo and which resists degradation or removal by endogenous mechanisms which remove unwanted material from the organism ⁇ e.g., human).
  • a polypeptide that enhances seram half-life in vivo can be selected from proteins from the extracellular matrix, proteins found in blood, proteins found at the blood brain barrier or in neural tissue, proteins localized to the kidney, liver, lung, heart, skin or bone, stress proteins, disease-specific proteins, or proteins involved in Fc transport.
  • Suitable polypeptides that enhance serum half-life in vivo include, for example, transferrin receptor specific ligand-neuropharmaceutical agent fusion proteins (see U.S. Patent No. 5,977,307, the teachings of which are incorporated herein by reference), brain capillary endothelial cell receptor, transferrin, transferrin receptor ⁇ e.g., soluble transferrin receptor), insulin, insulin-like growth factor 1 (IGF 1) receptor, insulin-like growth factor 2 (IGF 2) receptor, insulin receptor, blood coagulation factor X, ⁇ l-antitrypsin and HNF l ⁇ .
  • transferrin receptor specific ligand-neuropharmaceutical agent fusion proteins see U.S. Patent No. 5,977,307, the teachings of which are incorporated herein by reference
  • brain capillary endothelial cell receptor transferrin, transferrin receptor ⁇ e.g., soluble transferrin receptor
  • insulin insulin-like growth factor 1
  • Suitable polypeptides that enhance serum half-life also include alpha- 1 glycoprotein (orosomucoid; AAG), alpha-1 antichymotrypsin (ACT), alpha-1 microglobulin (protein HC; AIM), antithrombin III (AT III), apolipoprotein A-I (Apo A-I), apolipoprotein B (Apo B), ceruloplasmin (Cp), complement component C3 (C3), complement component C4 (C4), Cl esterase inhibitor (Cl INH), C-reactive protein (CRP), ferritin (FER), hemopexin (HPX), lipoprotein(a) (Lp(a)), mannose-binding protein (MBP), myoglobin (Myo), prealbumin (transthyretin; PAL), retinol-binding protein (RBP), and rheumatoid factor (RF).
  • alpha- 1 glycoprotein orosomucoid
  • AAG alpha-1 antichymotryp
  • Suitable proteins from the extracellular matrix include, for example, collagens, laminins, integrins and fibronectin.
  • Collagens are the major proteins of the extracellular matrix.
  • about 15 types of collagen molecules are currently known, found in different parts of the body, e.g. type I collagen (accounting for 90% of body collagen) found in bone, skin, tendon, ligaments, cornea, internal organs or type II collagen found in cartilage, vertebral disc, notochord, and vitreous humor of the eye.
  • Suitable proteins from the blood include, for example, plasma proteins ⁇ e.g., fibrin, ⁇ -2 macroglobulin, serum albumin, fibrinogen ⁇ e.g., fibrinogen A, fibrinogen B), serum amyloid protein A, haptoglobin, profilin, ubiquitin, uteroglobulin and ⁇ -2- microglobulin), enzymes and enzyme inhibitors ⁇ e.g., plasminogen, lysozyme, cystatin C, alpha-1 -antitrypsin and pancreatic trypsin inhibitor), proteins of the immune system, such as immunoglobulin proteins ⁇ e.g., IgA, IgD, IgE, IgG, IgM, immunoglobulin light chains (kappa/lambda)), transport proteins ⁇ e.g., retinol binding protein, ⁇ -1 microglobulin), defensins ⁇ e.g., beta-defensin 1, neutrophil defensin 1
  • Suitable proteins found at the blood brain barrier or in neural tissue include, for example, melanocortin receptor, myelin, ascorbate transporter and the like.
  • Suitable polypeptides that enhance serum half-life in vivo also include proteins localized to the kidney ⁇ e.g., polycystin, type IV collagen, organic anion transporter Kl, Heymann's antigen), proteins localized to the liver ⁇ e.g., alcohol dehydrogenase, G250), proteins localized to the lung ⁇ e.g., secretory component, which binds IgA), proteins localized to the heart ⁇ e.g., HSP 27, which is associated with dilated cardiomyopathy), proteins localized to the skin ⁇ e.g., keratin), bone specific proteins such as morphogenic proteins (BMPs), which are a subset of the transforming growth factor ⁇ superfamily of proteins that demonstrate osteogenic activity ⁇ e.g., BMP-2, BMP-4, BMP-5, BMP-6, BMP-7, BMP-8), tumor specific proteins ⁇ e.g., trophoblast antigen, herceptin receptor, oestrogen receptor, catheps
  • Suitable disease-specific proteins include, for example, antigens expressed only on activated T-cells, including LAG-3 (lymphocyte activation gene), osteoprotegerin ligand (OPGL; see Nature 402, 304-309 (1999)), OX40 (a member of the TNF receptor family, expressed on activated T cells and specifically up- regulated in human T cell leukemia virus type-I (HTLV-I)-producing cells; see Immunol. 165 (l):263-70 (2000)).
  • LAG-3 lymphocyte activation gene
  • osteoprotegerin ligand OPGL
  • OX40 a member of the TNF receptor family, expressed on activated T cells and specifically up- regulated in human T cell leukemia virus type-I (HTLV-I)-producing cells; see Immunol. 165 (l):263-70 (2000)).
  • Suitable disease-specific proteins also include, for example, metalloproteases (associated with arthritis/cancers) including CG6512 Drosophila, human paraplegin, human FtsH, human AFG3L2, murine ftsH; and angiogenic growth factors, including acidic fibroblast growth factor (FGF-I), basic fibroblast growth factor (FGF-2), vascular endothelial growth factor/vascular permeability factor (VEGF/VPF), transforming growth factor- ⁇ (TGF ⁇ ), tumor necrosis factor-alpha (TNF- ⁇ ), angiogenin, interleukin-3 (IL-3), interleukin-8 (IL- 8), platelet-derived endothelial growth factor (PD-ECGF), placental growth factor (PlGF), midkine platelet-derived growth factor-BB (PDGF), and fractalkine.
  • metalloproteases associated with arthritis/cancers
  • FGF-I acidic fibroblast growth factor
  • FGF-2 basic fibroblast growth factor
  • Suitable polypeptides that enhance serum half-life in vivo also include stress proteins such as heat shock proteins (HSPs). HSPs are normally found
  • Suitable proteins involved in Fc transport include, for example, Brambell receptor (also known as FcRB).
  • FcRB Brambell receptor
  • This Fc receptor has two functions, both of which are potentially useful for delivery. The functions are (1) transport of IgG from mother to child across the placenta (2) protection of IgG from degradation thereby prolonging its serum half-life. It is thought that the receptor recycles IgG from endosomes. (See, Holliger et al, Nat Biotechnol 15(7):632-6 (1997).)
  • the invention also relates to ligands that comprise a toxin moiety or toxin.
  • Suitable toxin moieties comprise a toxin ⁇ e.g., surface active toxin, cytotoxin).
  • the toxin moiety or toxin can be linked or conjugated to the ligand using any suitable method.
  • the toxin moiety or toxin can be covalently bonded to the ligand directly or through a suitable linker.
  • Suitable linkers can include
  • noncleavable or cleavable linkers for example, pH cleavable linkers that comprise a cleavage site for a cellular enzyme ⁇ e.g., cellular esterases, cellular proteases such as cathepsin B).
  • cleavable linkers can be used to prepare a ligand that can release a toxin moiety or toxin after the ligand is internalized.
  • a variety of methods for linking or conjugating a toxin moiety or toxin to a ligand can be used. The particular method selected will depend on the toxin moiety or toxin and ligand to be linked or conjugated. If desired, linkers that contain terminal functional groups can be used to link the ligand and toxin moiety or toxin. Generally, conjugation is accomplished by reacting toxin moiety or toxin that contains a reactive functional group (or is modified to contain a reactive functional group) with a linker or directly with a ligand.
  • a suitable reactive chemical group can be added to ligand or to a linker using any suitable method. (See, e.g., Hermanson, G.
  • an amine group can react with an electrophilic group such as tosylate, mesylate, halo (chloro, bromo, fluoro, iodo), N-hydroxysuccinimidyl ester (NHS), and the like.
  • Thiols can react with maleimide, iodoacetyl, acrylolyl, pyridyl disulfides, 5-thiol-2-nitrobenzoic acid thiol (TNB-thiol), and the like.
  • An aldehyde functional group can be coupled to amine- or hydrazide-containing molecules, and an azide group can react with a trivalent phosphorous group to form
  • phosphoramidate or phosphorimide linkages Suitable methods to introduce activating groups into molecules are known in the art (see for example, Hermanson, G. T., Bioconjugate Techniques, Academic Press: San Diego, CA (1996)).
  • Suitable toxin moieties and toxins include, for example, a maytansinoid ⁇ e.g., maytansinol, e.g., DMl, DM4), a taxane, a calicheamicin, a duocarmycin, or derivatives thereof.
  • the maytansinoid can be, for example, maytansinol or a maytansinol analogue.
  • Examples of maytansinol analogues include those having a modified aromatic ring ⁇ e.g., C-19-decloro, C-20-demethoxy, C-20-acyloxy) and those having modifications at other positions ⁇ e.g., C-9-CH, C- 14-alkoxym ethyl, C- 14-hydroxymethyl or aceloxymethyl, C- 15-hydroxy/acyloxy, C- 15-methoxy, C-18- N-demethyl, 4,5-deoxy).
  • Maytansinol and maytansinol analogues are described, for example, in U.S. Patent Nos 5,208,020 and 6,333,410, the contents of which are incorporated herein by reference.
  • Maytansinol can be coupled to antibodies and antibody fragments using, e.g., an N-succinimidyl 3-(2-pyridyldithio) proprionate (also known as N-succinimidyl 4-(2-pyridyldithio)pentanoate or SPP), 4- succinimidyl-oxycarbonyl-a-(2-pyridyldithio)-toluene (SMPT), N-succinimidyl-3- (2-pyridyldithio)butyrate (SDPB), 2 iminothiolane, or S-acetylsuccinic anhydride.
  • N-succinimidyl 3-(2-pyridyldithio) proprionate also known as N-succinimidyl 4-(2-pyridyldithio)pentanoate or SPP
  • SPP N-succinimidyl 4-(2-pyridyl
  • the taxane can be, for example, a taxol, taxotere, or novel taxane (see, e.g., WO 01/38318).
  • the calicheamicin can be, for example, a bromo-complex calicheamicin (e.g., an alpha, beta or gamma bromo-complex), an iodo-complex calicheamicin (e.g., an alpha, beta or gamma iodo-complex), or analogs and mimics thereof.
  • Bromo-complex calicheamicins include Il -BR, I2-BR, I3-BR, I4-BR, Jl-BR, J2-BR and Kl-BR.
  • Iodo-complex calicheamicins include H-I, I2-I, I3-I, Jl-I, J2-I, Ll-I and Kl-BR.
  • Calicheamicin and mutants, analogs and mimics thereof are described, for example, in U.S. Patent Nos 4,970,198; 5,264,586; 5,550,246; 5,712,374, and 5,714,586, the contents of which are incorporated herein by reference.
  • Duocarmycin analogs e.g., KW-2189, DC88, DC89 CBI-TMI, and derivatives thereof are described, for example, in U.S. Patent No. 5,070,092, U.S. Patent No. 5,187,186, U.S. Patent No. 5,641,780, U.S. Patent No. 5,641,780, U.S. Patent No. 4,923,990, and U.S. Patent No. 5,101,038, the contents of which are incorporated herein by reference.
  • Examples of other toxins include, but are not limited to antimetabolites (e.g. , methotrexate, 6-mercaptopurine, 6-thioguanine, cytarabine, 5-fiuorouracil decarbazine), alkylating agents (e.g., mechlorethamine, thioepa chlorambucil, CC- 1065 (see US Patent Nos.
  • antimetabolites e.g. , methotrexate, 6-mercaptopurine, 6-thioguanine, cytarabine, 5-fiuorouracil decarbazine
  • alkylating agents e.g., mechlorethamine, thioepa chlorambucil, CC- 1065 (see US Patent Nos.
  • doxorubicin doxorubicin
  • antibiotics e.g., dactinomycin (formerly actinomycin), bleomycin, mithramycin, mitomycin, puromycin anthramycin (AMC)), duocarmycin and analogs or derivatives thereof
  • anti-mitotic agents e.g., vincristine, vinblastine, taxol, auristatins (e.g., auristatin E) and maytansinoids, and analogs or homologs thereof.
  • the toxin can also be a surface active toxin, such as a toxin that is a free radical generator (e.g. selenium containing toxin moieties), or radionuclide containing moiety.
  • Suitable radionuclide containing moieties include for example, moieties that contain radioactive iodine ( 131 I or 125 I), yttrium ( 90 Y), lutetium ( 177 Lu), actinium ( 225 Ac), praseodymium, astatine ( 211 At), rhenium ( 186 Re), bismuth ( 212 Bi or 213 Bi), indium ( 111 In), technetium (“mTc), phosphorus ( 32 P), rhodium ( 188 RIi), sulfur ( 35 S), carbon ( 14 C), tritium ( 3 H), chromium ( 51 Cr), chlorine ( 36 Cl), cobalt ( 57 Co or 58 Co), iron ( 59 Fe), selenium ( 75 Se), or gallium ( 67 Ga
  • the toxin can be a protein, polypeptide or peptide, from bacterial sources, e.g., diphtheria toxin, pseudomonas exotoxin (PE) and plant proteins, e.g., the A chain of ricin (RTA), the ribosome inactivating proteins (RIPs) gelonin, pokeweed antiviral protein, saporin, and dodecandron are contemplated for use as toxins.
  • bacterial sources e.g., diphtheria toxin, pseudomonas exotoxin (PE) and plant proteins, e.g., the A chain of ricin (RTA), the ribosome inactivating proteins (RIPs) gelonin, pokeweed antiviral protein, saporin, and dodecandron are contemplated for use as toxins.
  • PE pseudomonas exotoxin
  • RTA A chain of ricin
  • RIPs ribosome inactivating proteins
  • Antisense compounds of nucleic acids designed to bind, disable, promote degradation or prevent the production of the mRNA responsible for generating a particular target protein can also be used as a toxin.
  • Antisense compounds include antisense RNA or DNA, single or double stranded, oligonucleotides, or their analogs, which can hybridize specifically to individual mRNA species and prevent transcription and/or RNA processing of the mRNA species and/or translation of the encoded polypeptide and thereby effect a reduction in the amount of the respective encoded polypeptide. Ching, et al., Proc. Natl. Acad. Sd. U.S.A. 86: 10006-10010 (1989); Broder, et al, Ann. Int. Med. 113: 604-618 (1990); Loreau, et al, FEBS
  • Useful antisense therapeutics include for example: Veglin TM (VasGene) and OGX-011 (Oncogenix).
  • Toxins can also be photoactive agents.
  • Suitable photoactive agents include porphyrin-based materials such as porfimer sodium, the green porphyrins, chlorin E6, hematoporphyrin derivative itself, phthalocyanines, etiopurpurins, texaphrin, and the like.
  • the toxin can be an antibody or antibody fragment that binds an intracellular target ⁇ e.g., an intrabody), such as a dAb that binds an intracellular target.
  • an intrabody such as a dAb that binds an intracellular target.
  • dAbs can be directed to defined subcellular compartments or targets.
  • the antibodies or antibody fragments can bind an intracellular target selected from erbB2, EGFR, BCR-ABL, p21Ras, Caspase3, Caspase7, Bcl-2, p53, Cyclin E, ATF-1/CREB, HPV16 E7, HPl, Type IV collagenases, cathepsin L as well as others described in Kontermann, R.E., Methods, 34:163-170 (2004), incorporated herein by reference in its entirety.
  • polypeptide domains that Bind VEGF
  • polypeptide domains e.g., immunoglobulin single variable domains, dAb monomers
  • the polypeptide domain that has a binding site with binding specificity for VEGF competes for binding to VEGF with a dAb selected from the group consisting of TAR15-1 (SEQ ID NO:100), TAR15-3 (SEQ ID NO:101), TAR15-4 (SEQ ID NO:102), TAR15-9 (SEQ ID NO:103), TAR15-10 (SEQ ID NO:104), TAR15-11 (SEQ ID NO:105), TAR15-12 (SEQ ID NO:106), TAR15-13 (SEQ ID NO:107), TAR15-14 (SEQ ID NO:108), TAR15-15 (SEQ ID NO:109), TARI S-IO (SEQ ID NOI I IO) 5 TARI S-I V (SEQ ID NOI I I IX TARI S-I S (SEQ ID NO:112), TAR15-19 (SEQ ID NO:113), TAR15-20 (SEQ ID NO:114), TAR 15-22 (SEQ ID NO: 115), TARl 5-5 (SEQ ID NO
  • the polypeptide domain that has a binding site with binding specificity for VEGF competes for binding to VEGF with TARl 5-26-555 (SEQ ID NO:704).
  • the polypeptide domain that has a binding site with binding specificity for VEGF comprises an amino acid sequence that has at least about 80%, at least about 85%, at least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 94%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99% amino acid sequence identity with the amino acid sequence of a dAb selected from the group consisting of TAR15-1 (SEQ ID NO: 100), TAR15-3 (SEQ ID NO:101), TAR15-4 (SEQ ID NO:102), TAR15-9 (SEQ ID NO:103), TAR15-10 (SEQ ID NO: 104), TARl 5-11 (SEQ ID NO:105), TAR15-12 (SEQ ID NO:106), TAR15-13 (SEQ ID NO:107), TAR15-14 (SEQ ID NO:108), TAR15-15 (SEQ ID NO:109), TAR15-16 (SEQ ID NO:
  • TAR15-8-506 (SEQ ID NO:143), TAR15-8-507 (SEQ ID NO: 144), TAR15-8-508 (SEQ ID NO:145), TAR15-8-509 (SEQ ID NO:146), TAR15-8-510 (SEQ ID NO:147), TARl 5-8-511 (SEQ ID NO:148), TAR15-26-500 (SEQ ID NO:149), TAR15-26-501 (SEQ ID NO:150), TAR15-26-502 (SEQ ID NO:151), TAR15-26- 503 (SEQ ID NO: 152), TARl 5-26-504 (SEQ ID NO: 153), TARl 5-26-505 (SEQ ID NO: 154), TARl 5-26-506 (SEQ ID NO: 155), TARl 5-26-507 (SEQ ID NO: 156), TARl 5-26-508 (SEQ ID NO: 157), TARl 5-26-509 (SEQ ID NO: 158), TARl 5-26- 510 (S
  • the polypeptide domain that has a binding site with binding specificity for VEGF comprises an amino acid sequence that has at least about 80%, at least about 85%, at least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 94%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99% amino acid sequence identity with the amino acid sequence of TARl 5-26-555 (SEQ ID NO:704).
  • the polypeptide domain that has a binding site with binding specificity for VEGF comprises an amino acid sequence that has at least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 94%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99% amino acid sequence identity with the amino acid sequence of a dAb selected from the group consisting of TARl 5-6 (SEQ ID
  • the polypeptide domain that has a binding site with binding specificity for VEGF can comprise TARl 5-6 (SEQ ID NO: 117), TARl 5-8 (SEQ ID NO: 119), or TARl 5-26 (SEQ ID NO: 123).
  • the polypeptide domain that has a binding site with binding specificity for VEGF competes with any of the dAbs disclosed herein for binding to VEGF.
  • the polypeptide domain that has a binding site with binding specificity for VEGF is an immunoglobulin single variable domain.
  • the polypeptide domain that has a binding site with binding specificity for VEGF can comprise any suitable immunoglobulin variable domain, and preferably comprises a human variable domain or a variable domain that comprises human framework regions.
  • the polypeptide domain that has a binding site with binding specificity for VEGF comprises a universal framework, as described herein.
  • the universal framework can be a V L framework (V ⁇ or VK) , such as a framework that comprises the framework amino acid sequences encoded by the human germline DPKl, DPK2, DPK3, DPK4, DPK5, DPK6, DPK7, DPK8, DPK9, DPKlO, DPK12, DPK13, DPK15, DPKl 6, DPKl 8, DPK19, DPK20, DPK21, DPK22, DPK23, DPK24, DPK25, DPK26 or DPK 28 immunoglobulin gene segment.
  • the VL framework can further comprise the framework amino acid sequence encoded by the human germline J ⁇ l , J K 2, J K 3, J K 4, or J K 5
  • the universal framework can be a V H framework, such as a framework that comprises the framework amino acid sequences encoded by the human germline DP4, DP7, DP8, DP9, DPlO, DP31, DP33, DP38, DP45, DP46, DP47, DP49, DP50, DP51, DP53, DP54, DP65, DP66, DP67, DP68 or DP69 immunoglobulin gene segment.
  • the V H framework can further comprise the framework amino acid sequence encoded by the human germline J H I , J H 2, J H 3, J H 4, Jii4b, J H 5 and J H 6 immunoglobulin gene segment.
  • the polypeptide domain that has a binding site with binding specificity for VEGF comprises one or more framework regions comprising an amino acid sequence that is the same as the amino acid sequence of a
  • the amino acid sequences of FWl, FW2, FW3 and FW4 of the polypeptide domain that has a binding site with binding specificity for VEGF are the same as the amino acid sequences of corresponding framework regions encoded by a human germline antibody gene segment, or the amino acid sequences of FWl, FW2, FW3 and FW4 collectively contain up to 10 amino acid differences relative to the amino acid sequences of corresponding framework regions encoded by said human germline antibody gene segment.
  • the polypeptide domain that has a binding site with binding specificity for VEGF comprises FWl, FW2 and FW3 regions, and the amino acid sequence of said FWl, FW2 and FW3 regions are the same as the amino acid sequences of corresponding framework regions encoded by human germline antibody gene segments.
  • the polypeptide domain that has a binding site with binding specificity for VEGF comprises the DPK9 V L framework, or a V H framework selected from the group consisting of DP47, DP45 and DP38.
  • the polypeptide domain that has a binding site with binding specificity for VEGF can comprise a binding site for a generic ligand, such as protein A, protein L and protein G.
  • the polypeptide domain that has a binding site with binding specificity for VEGF is substantially resistant to aggregation.
  • Aggregation can be assessed using any suitable method, such as, by microscopy, assessing turbidity of a solution by visual inspection or spectroscopy or any other suitable method.
  • aggregation is assessed by dynamic light scattering.
  • Polypeptide domains that have a binding site with binding specificity for VEGF that are resistant to aggregation provide several advantages. For example, such polypeptide domains that have a binding site with binding specificity for VEGF can readily be produced in high yield as soluble proteins by expression using a suitable biological production system, such as E. coli, and can be formulated and/or stored at higher concentrations than conventional polypeptides, and with less aggregation and loss of activity.
  • the polypeptide domain that has a binding site with binding specificity for VEGF that are resistant to aggregation can be produced more economically than other antigen- or epitope-binding polypeptides (e.g. , conventional antibodies).
  • preparation of antigen- or epitope-binding polypeptides intended for in vivo applications includes processes (e.g., gel filtration) that remove aggregated polypeptides. Failure to remove such aggregates can result in a preparation that is not suitable for in vivo applications because, for example, aggregates of an antigen-binding polypeptide that is intended to act as an antagonist can function as an agonist by inducing cross-linking or clustering of the target antigen. Protein aggregates can also reduce the efficacy of therapeutic polypeptide by inducing an immune response in the subject to which they are administered.
  • the aggregation resistant polypeptide domain that has a binding site with binding specificity for VEGF of the invention can be prepared for in vivo applications without the need to include process steps that remove aggregates, and can be used in in vivo applications without the aforementioned disadvantages caused by polypeptide aggregates.
  • the polypeptide domain that has a binding site with binding specificity for VEGF unfolds reversibly when heated to a temperature (Ts) and cooled to a temperature (Tc), wherein Ts is greater than the melting temperature (Tm) of the polypeptide domain that has a binding site with binding specificity for VEGF, and Tc is lower than the melting temperature of the polypeptide domain that has a binding site with binding specificity for VEGF.
  • Ts is greater than the melting temperature (Tm) of the polypeptide domain that has a binding site with binding specificity for VEGF
  • Tc is lower than the melting temperature of the polypeptide domain that has a binding site with binding specificity for VEGF.
  • a polypeptide domain that has a binding site with binding specificity for VEGF can unfold reversibly when heated to 8O 0 C and cooled to about room temperature.
  • polypeptide that unfolds reversibly loses function when unfolded but regains function upon refolding are distinguished from polypeptides that aggregate when unfolded or that improperly refold (misfolded polypeptides), i.e., do not regain function.
  • Polypeptide unfolding and refolding can be assessed, for example, by directly or indirectly detecting polypeptide structure using any suitable method.
  • polypeptide structure can be detected by circular dichroism (CD) (e.g., far- UV CD, near-UV CD), fluorescence (e.g., fluorescence of tryptophan side chains), susceptibility to proteolysis, nuclear magnetic resonance (NMR), or by detecting or measuring a polypeptide function that is dependent upon proper folding (e.g., binding to target ligand, binding to generic ligand).
  • CD circular dichroism
  • fluorescence e.g., fluorescence of tryptophan side chains
  • susceptibility to proteolysis e.g., nuclear magnetic resonance (NMR)
  • NMR nuclear magnetic resonance
  • polypeptide unfolding is assessed using a functional assay in which loss of binding function (e.g., binding a generic and/or target ligand, binding a substrate) indicates that the polypeptide is unfolded.
  • the extent of unfolding and refolding of a polypeptide domain that has a binding site with binding specificity for VEGF can be determined using an unfolding or denaturation curve.
  • An unfolding curve can be produced by plotting temperature as the ordinate and the relative concentration of folded polypeptide as the abscissa.
  • the relative concentration of folded polypeptide domain that has a binding site with binding specificity for VEGF can be determined directly or indirectly using any suitable method (e.g., CD, fluorescence, binding assay).
  • a polypeptide domain that has a binding site with binding specificity for VEGF solution can be prepared and ellipticity of the solution determined by CD.
  • the ellipticity value obtained represents a relative concentration of folded ligand (e.g., dAb monomer) of 100%.
  • the polypeptide domain that has a binding site with binding specificity for VEGF in the solution is then unfolded by incrementally raising the temperature of the solution and ellipticity is determined at suitable increments (e.g., after each increase of one degree in temperature).
  • the polypeptide domain that has a binding site with binding specificity for VEGF in solution is then refolded by incrementally reducing the temperature of the solution and ellipticity is determined at suitable increments.
  • the data can be plotted to produce an unfolding curve and a refolding curve.
  • the unfolding and refolding curves have a
  • characteristic sigmoidal shape that includes a portion in which the polypeptide domain that has a binding site with binding specificity for VEGF molecules are folded, an unfolding/refolding transition in which polypeptide domain that has a binding site with binding specificity for VEGF molecules are unfolded to various degrees, and a portion in which polypeptide domain that has a binding site with binding specificity for VEGF are unfolded.
  • the y-axis intercept of the refolding curve is the relative amount of refolded polypeptide domain that has a binding site with binding specificity for VEGF recovered.
  • a recovery of at least about 50%, or at least about 60%, or at least about 70%, or at least about 75%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95% is indicative that the ligand or dAb monomer unfolds reversibly.
  • reversibility of unfolding of a polypeptide domain that has a binding site with binding specificity for VEGF is determined by preparing a polypeptide domain that has a binding site with binding specificity for VEGF solution and plotting heat unfolding and refolding curves.
  • the polypeptide domain that has a binding site with binding specificity for VEGF solution can be prepared in any suitable solvent, such as an aqueous buffer that has a pH suitable to allow polypeptide domain that has a binding site with binding specificity for VEGF to dissolve (e.g., pH that is about 3 units above or below the isoelectric point (pi)).
  • the polypeptide domain that has a binding site with binding specificity for VEGF solution is concentrated enough to allow unfolding/folding to be detected.
  • the ligand or dAb monomer solution can be about 0.1 ⁇ M to about 100 ⁇ M, or preferably about 1 ⁇ M to about 10 ⁇ M.
  • the solution can be heated to about ten degrees below the Tm (Tm-10) and folding assessed by ellipticity or
  • fluorescence e.g., far-UV CD scan from 200 ran to 250 ran, fixed wavelength CD at 235 ran or 225 nm; tryptophan fluorescent emission spectra at 300 to 450 nm with excitation at 298 nm
  • the solution is then heated to at least ten degrees above Tm (Tm+ 10) in predetermined increments (e.g., increases of about 0.1 to about 1 degree), and ellipticity or fluorescence is determined at each increment.
  • the polypeptide domain that has a binding site with binding specificity for VEGF is refolded by cooling to at least Tm-10 in predetermined increments and ellipticity or fluorescence determined at each increment.
  • the solution can be unfolded by incrementally heating from about 25 0 C to about 100 0 C and then refolded by incrementally cooling to at least about 25 0 C, and ellipticity or fluorescence at each heating and cooling increment is determined.
  • the data obtained can be plotted to produce an unfolding curve and a refolding curve, in which the y-axis intercept of the refolding curve is the relative amount of refolded protein recovered.
  • the polypeptide domain that has a binding site with binding specificity for VEGF does not comprise a Camelid
  • immunoglobulin variable domain or one or more framework amino acids that are unique to immunoglobulin variable domains encoded by Camelid germline antibody gene segments.
  • the polypeptide domain that has a binding site with binding specificity for VEGF is secreted in a quantity of at least about 0.5 mg/L when expressed in E. coli or in Pichia species (e.g., P. pastoris).
  • a polypeptide domain that has a binding site with binding specificity for VEGF is secreted in a quantity of at least about 0.75 mg/L, at least about 1 mg/L, at least about 4 mg/L, at least about 5 mg/L, at least about 10 mg/L, at least about 15 mg/L, at least about 20 mg/L, at least about 25 mg/L, at least about 30 mg/L, at least about 35 mg/L, at least about 40 mg/L, at least about 45 mg/L, or at least about 50 mg/L, or at least about 100 mg/L, or at least about 200 mg/L, or at least about 300 mg/L, or at least about 400 mg/L, or at least about 500 mg/L, or at least about 600 mg/L, or at least about 700 mg/L, or at least about 800 mg/L, at least about 900 mg/L, or at least about lg/L when expressed in E.
  • a polypeptide domain that has a binding site with binding specificity for VEGF is secreted in a quantity of at least about 1 mg/L to at least about lg/L, at least about 1 mg/L to at least about 750 mg/L, at least about 100 mg/L to at least about 1 g/L, at least about 200 mg/L to at least about 1 g/L, at least about 300 mg/L to at least about 1 g/L, at least about 400 mg/L to at least about 1 g/L, at least about 500 mg/L to at least about lg/L, at least about 600 mg/L to at least about 1 g/L, at least about 700 mg/L to at least about 1 g/L, at least about 800 mg/L to at least about lg/L, or at least about 900 mg/L to at least about lg/L when expressed in E.
  • a polypeptide domain that has a binding site with binding specificity for VEGF described herein can be secretable when expressed in E. coli or in Pichia species ⁇ e.g., P. pastoris), they can be produced using any suitable method, such as synthetic chemical methods or biological production methods that do not employ E. coli or Pichia species.
  • the invention provides polypeptide domains ⁇ e.g., dAb) that have a binding site with binding specificity for EGFR.
  • the polypeptide domain that has a binding site with binding specificity for EGFR competes for binding to EGFR with a dAb selected from the group consisting of DOM16-17 (SEQ ID NO:325), DOM16-18 (SEQ ID NO:326), DOM16-19 (SEQ ID NO:327), DOM16-20 (SEQ ID NO:328), D0M16- 21 (SEQ ID NO:329), DOM16-22 (SEQ ID NO:330), DOM16-23 (SEQ ID NO:
  • the polypeptide domain that has a binding site with binding specificity for EGFR competes for binding to EGFR with a dAb selected from the group consisting of DOM 16-39-210 (SEQ ID NO:541), DOM16-39-211 (SEQ ID NO:542), DOM16-39-212 (SEQ ID NO:543), DOM16-39-213 (SEQ ID NO:544), DOM16-39-214 (SEQ ID NO:545), DOM16-39-215 (SEQ ID NO:546), DOM16-39-216 (SEQ ID NO:547), DOM16-39-217 (SEQ ID NO:548), DOM16- 39-218 (SEQ ID NO:549), DOM16-39-219 (SEQ ID NO:550), DOM16-39-220 (SEQ ID NO:551), DOM16-39-221 (SEQ ID NO:552), DOM16-39-222 (SEQ ID NO:553), DOMl
  • the polypeptide domain that has a binding site with binding specificity for EGFR comprises an amino acid sequence that has at least about 80%, at least about 85%, at least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 94%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99% amino acid sequence identity with the amino acid sequence of a dAb selected from the group consisting of DOM16-17 (SEQ ID NO:325), DOM16-18 (SEQ ID NO:326), DOM16-19 (SEQ ID NO:327), DOM16-20 (SEQ ID NO:328), DOM16-21 (SEQ ID NO:329), DOM16- 22 (SEQ ID NO:330), DOM16-23 (SEQ ID NO:331), DOM16-24 (SEQ ID
  • DOM16-118 (SEQ ID NO:414), DOMl 6-119 (SEQ ID NO:415), DOMl 6-39-6 (SEQ ID NO:416), DOMl 6-39-8 (SEQ ID NO:417), DOMl 6-39-34 (SEQ ID NO:418), DOM16-39-48 (SEQ ID NO:419), DOM16-39-87 (SEQ ID NO:420), DOM16-39-90 (SEQ ID NO:421), DOM16-39-96 (SEQ ID NO:422), DOM16-39- 100 (SEQ ID NO:423), DOM16-39-101 (SEQ ID NO:424), DOM16-39-102 (SEQ ID NO:425), DOMl 6-39-103 (SEQ ID NO:426), DOMl 6-39-104 (SEQ ID NO:
  • the polypeptide domain that has a binding site with binding specificity for EGFR comprises an amino acid sequence that has at least about 80%, at least about 85%, at least about 90%, at least about 91 %, at least about 92%, at least about 93%, at least about 94%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99% amino acid sequence identity with the amino acid sequence of a dAb selected from the group consisting of DOM16-39-210 (SEQ ID NO:541), DOM16-39-211 (SEQ ID NO.542), D0M16- 39-212 (SEQ ID NO:543), DOM16-39-213 (SEQ ID NO:544), DOM16-39-214 (SEQ ID NO:545), DOM16-39-215 (SEQ ID NO:546), DOM16-39-216 (SEQ ID NO:547), DOM16-39-217 (SEQ ID NO:548),
  • the polypeptide domain that has a binding site with binding specificity for EGFR comprises an amino acid sequence that has at least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 94%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99% amino acid sequence identity with the amino acid sequence of DOM16-39 (SEQ ID NO:345).
  • the polypeptide domain that has a binding site with binding specificity for EGFR can comprise the amino acid sequence of DOMl 6-39-87 (SEQ ID NO:420), DOMl 6-39-100 (SEQ ID NO:423), DOM16-39-107 (SEQ ID NO:430), DOM16-39-109 (SEQ ID NO:432), DOM16-39-115 (SEQ ID NO:438), or DOM16-39-200 (SEQ ID NO:441).
  • the polypeptide domain that has a binding site with binding specificity for EGFR comprises an amino acid sequence that has at least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 94%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99% amino acid sequence identity with the amino acid sequence of DOM16-39-521 (SEQ ID NO:577), DOM16-39-541 (SEQ ID NO:585), DOM16-39-542 (SEQ ID NO:586), DOM16-39-551 (SEQ ID NO:591), D0M16- 39-601 (SEQ ID NO:608), DOM16-39-604 (SEQ ID NO:611), DOM16-39-618 (SEQ ID NO:621), and DOM 16-39-619 (SEQ ID NO:622).
  • the polypeptide domain that has a binding site with binding specificity for EGFR competes with any of the dAbs disclosed herein for binding to EGFR.
  • the polypeptide domain that has a binding site with binding specificity for EGFR is an immunoglobulin single variable domain.
  • polypeptide domain that has a binding site with binding specificity for EGFR can comprise any suitable immunoglobulin variable domain, and preferably comprises a human variable domain or a variable domain that comprises human framework regions.
  • the polypeptide domain that has a binding site with binding specificity for EGFR comprises a universal framework, as described herein.
  • the polypeptide domain that has a binding site with binding specificity for EGFR resists aggregation, unfolds reversibly comprises a framework region and/or is secreted as described above for the polypeptide domain that has a binding site with binding specificity for VEGF.
  • the ligands of the invention can further comprise a dAb monomer that binds serum albumin (SA) with a K ⁇ of InM to 500 ⁇ M (i.e., x 10 "9 to 5 x 10" 4 ), preferably 100 nM to 10 ⁇ M.
  • SA serum albumin
  • the binding (e.g. Kj and/or K 0 ff as measured by surface plasmon resonance, e.g., using BiaCore) of the ligand to its target(s) is from 1 to 100000 times (preferably 100 to 100000, more preferably 1000 to 100000, or 10000 to 100000 times) stronger than for SA.
  • the serum albumin is human serum albumin (HSA).
  • the first dAb (or a dAb monomer) binds SA (e.g., HSA) with a K ⁇ of approximately 50, preferably 70, and more preferably 100, 150 or 200 nM.
  • the dAb monomer that binds SA resists aggregation, unfolds reversibly and/or comprises a framework region as described above for dAb monomers that bind CD38.
  • the antigen-binding fragment of an antibody that binds serum albumin is a dAb that binds human serum albumin, hi certain embodiments, the dAb binds human serum albumin and competes for binding to albumin with a dAb selected from the group consisting of DOM7m-16 (SEQ ID NO: 473), DOM7m-12 (SEQ ID NO: 474), DOM7m-26 (SEQ ID NO: 475), DOM7r-l (SEQ ID NO: 476), DOM7r-3 (SEQ ID NO: 477), DOM7r-4 (SEQ ID NO: 478), DOM7r-5 (SEQ ID NO: 479), DOM7r-7 (SEQ ID NO: 480), DOM7r-8 (SEQ ID NO: 481),
  • the dAb binds human serum albumin and comprises an amino acid sequence that has at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 96%, or at least about 97%, or at least about 98%, or at least about 99% amino acid sequence identity with the amino acid sequence of a dAb selected from the group consisting of DOM7m-16 (SEQ ID NO: 473), DOM7m-12 (SEQ ID NO: 474), DOM7m-26 (SEQ ID NO: 475), DOM7r-l (SEQ ID NO: 476), DOM7r-3 (SEQ ID NO: 477), DOM7r-4 (SEQ ID NO: 478), DOM7r-5 (SEQ ID NO: 479), DOM7r-7 (SEQ ID NO: 480), DOM7r-8 (SEQ ID NO: 481), DOM7h-2 (SEQ ID NO: 482), DOM7h-3 (SEQ ID NO: 483),
  • the dAb that binds human serum albumin can comprise an amino acid sequence that has at least about 90%, or at least about 95%, or at least about 96%, or at least about 97%, or at least about 98%, or at least about 99% amino acid sequence identity with DOM7h-2 (SEQ ID NO:482), DOM7h-3 (SEQ ID NO:483), DOM7h-4 (SEQ ID NO:484), DOM7h-6 (SEQ ID NO:485), DOM7h-l (SEQ ID NO:486), DOM7h-7 (SEQ ID NO:487), DOM7h-8 (SEQ ID NO:496), DOM7r-13 (SEQ ID NO:497), DOM7r-14 (SEQ ID NO:498), DOM7h-22 (SEQ ID NO:489), DOM7h-23 (SEQ ID NO:490), DOM7h-24 (SEQ ID NO:491), DOM7h- 25 (SEQ ID NO:492), DOM7
  • Amino acid sequence identity is preferably determined using a suitable sequence alignment algorithm and default parameters, such as BLAST P (Karlin and Altschul, Proc. Natl. Acad. ScL USA S7(6):2264-2268 (1990)).
  • the dAb is a V ⁇ dAb that binds human serum albumin and has an amino acid sequence selected from the group consisting of DOM7h-2 (SEQ ID NO:482), DOM7h-3 (SEQ ID NO:483), DOM7h-4 (SEQ ID NO:484), DOM7h-6 (SEQ ID NO:485), DOM7h-l (SEQ ID NO:486), DOM7h-7 (SEQ ID NO:487), DOM7h-8 (SEQ ID NO:496), DOM7r-13 (SEQ ID NO:497), and DOM7r-14 (SEQ ID NO:498) , or a VH dAb that has an amino acid sequence selected from the group consisting of: DOM7h-22 (SEQ ID NO:489), DOM7h-23 (SEQ ID NO:490), DOM7h-24 (SEQ ID NO:491), DOM7h-25 (SEQ ID NO:492), DOM7h-26 (SEQ ID NO:
  • Suitable Camelid VjJiH tnat ⁇ n & seru rn albumin include those disclosed in
  • Sequence B SEQ ID NO:5119
  • Sequence C SEQ ID NO:520
  • Sequence D SEQ ID NO:521
  • Sequence E SEQ ID NO:522
  • Sequence F SEQ ID NO:
  • Sequence G SEQ ID NO:5234
  • Sequence H SEQ ID NO:525
  • Sequence I SEQ ID NO:526)
  • Sequence J SEQ ID NO:527
  • Sequence K SEQ ID NO:528
  • Sequence L SEQ ID NO:529
  • Sequence M SEQ ID NO:530
  • Sequence N SEQ ID NO:531
  • Sequence O SEQ ID NO:532
  • the Camelid VJJH binds human serum albumin and comprises an amino acid sequence that has at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 96%, or at least about 97%, or at least about 98%, or at least about 99% amino acid sequence identity with any one of SEQ ID NOS :518-534.
  • Amino acid sequence identity is preferably determined using a suitable sequence alignment algorithm and default parameters, such as BLAST P (Karlin and Altschul, Proc. Natl. Acad. ScL USA 57(6):2264-2268 (1990)).
  • the ligand comprises an anti-serum albumin dAb that competes with any anti-serum albumin dAb disclosed herein for binding to serum albumin ⁇ e.g., human serum albumin).
  • the invention also provides isolated and/or recombinant nucleic acid molecules encoding ligands ⁇ e.g., dual-specific ligands and multispecific ligands) as described herein.
  • Nucleic acids referred to herein as "isolated” are nucleic acids which have been separated away from the nucleic acids of the genomic DNA or cellular RNA of their source of origin ⁇ e.g., as it exists in cells or in a mixture of nucleic acids such as a library), and include nucleic acids obtained by methods described herein or other suitable methods, including essentially pure nucleic acids, nucleic acids produced by chemical synthesis, by combinations of biological and chemical methods, and recombinant nucleic acids which are isolated (see e.g., Daugherty, B.L. et al., Nucleic Acids Res., 19(9): 2471-2476 (1991); Lewis, A.P. and J.S. Crowe, Gene, 101: 297-302 (1991)).
  • Nucleic acids referred to herein as "recombinant” are nucleic acids which have been produced by recombinant DNA methodology, including those nucleic acids that are generated by procedures which rely upon a method of artificial recombination, such as the polymerase chain reaction (PCR) and/or cloning into a vector using restriction enzymes.
  • PCR polymerase chain reaction
  • the isolated and/or recombinant nucleic acid comprises a nucleotide sequence encoding a ligand, as described herein, wherein said ligand comprises an amino acid sequence that has at least about 80%, at least about 85%, at least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 94%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99% amino acid sequence identity with the amino acid sequence of a dAb that binds VEGF disclosed herein, or a dAb that binds EGFR disclosed herein.
  • the isolated and/or recombinant nucleic acid comprises a nucleotide sequence encoding a ligand that has binding specificity for VEGF, as described herein, wherein said ligand comprises an amino acid sequence that has at least about 80%, at least about 85%, at least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 94%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99% amino acid sequence identity with the amino acid sequence of a dAb selected .
  • TAR15-1 SEQ ID NOrIOO
  • TAR15-3 SEQ ID NO:101
  • TAR15-4 SEQ ID NO: 102
  • TAR15-9 SEQ ID NO:103
  • TAR15-10 SEQ ID NO:104
  • TARl 5-11 SEQ ID NO:105
  • TAR15-12 SEQ ID NO:106
  • TAR15-13 SEQ ID NO:107
  • TAR15-14 SEQ ID NO: 108
  • TAR15-15 SEQ ID NO:109
  • TAR15-16 SEQ ID NO:110
  • TAR15-17 SEQ ID NO:111
  • TAR15-18 SEQ ID NO:112
  • TAR15-19 SEQ ID NO:113
  • TAR15-20 SEQ ID NO:114
  • TAR 15-22 SEQ ID NO: 115
  • TARl 5-5 SEQ ID NO: 116
  • TARl 5-6 SEQ ID NO:117
  • TAR15-7 SEQ ID NO:118
  • TAR15-8 SEQ ID NO:119
  • the isolated and/or recombinant nucleic acid comprises a nucleotide sequence encoding a ligand that has binding specificity for VEGFA, as described herein, wherein said ligand comprises an amino acid sequence that has at least about 80%, at least about 85%, at least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 94%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99% amino acid sequence identity with the amino acid sequence of SEQ ID NO:705 (TARl 5-26-555).
  • the isolated and/or recombinant nucleic acid comprises a nucleotide sequence encoding a ligand that has binding specificity for EGFR, as described herein, wherein said ligand comprises an amino acid sequence that has at least about 80%, at least about 85%, at least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 94%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99% amino acid sequence identity with the amino acid sequence of a dAb selected from the group consisting of DOMl 6-17 (SEQ ID NO:325), DOMl 6-18 (SEQ ID NO:325), DOMl 6-18 (SEQ ID
  • DOM16-90 SEQ ID NO:387)
  • DOM16-91 SEQ ID NO:388
  • DOM16-92 SEQ ID NO:389
  • DOM16-94 SEQ ID NO:390
  • DOM16-95 SEQ ID NO:
  • DOM16-110 (SEQ ID NO:406), DOMl 6-111 (SEQ ID NO:407), DOMl 6-112 (SEQ ID NO:408), DOMl 6-113 (SEQ ID NO:409), DOMl 6-114 (SEQ ID NO:
  • the isolated and/or recombinant nucleic acid comprises a nucleotide sequence encoding a ligand that has binding specificity for EGFR, as described herein, wherein said ligand comprises an amino acid sequence that has at least about 80%, at least about 85%, at least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 94%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99% amino acid sequence identity with an amino acid sequence selected from the group consisting of SEQ ID NOS:623-703, 727 and 728.
  • the isolated and/or recombinant nucleic acid encoding a ligand that has binding specificity for VEGF comprises a nucleotide sequence that has at least about 80%, at least about 85%, at least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 94%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99% nucleotide sequence identity with a nucleotide sequence encoding an anti-VEGF dAb selected from the group consisting of TARl 5-1 (SEQ ID NO: 1), TAR15-3 (SEQ ID NO:2), TAR15-4 (SEQ ID NO:3), TAR15-9 (SEQ ID NO:4), TARl 5-10 (SEQ ID NO:5), TARl 5-11 (SEQ ID NO:6), TARl 5-12 (SEQ ID NO:7), TAR15-13 (SEQ ID NO:
  • TARl 5-26-509 SEQ ID NO:59
  • TARl 5-26-510 SEQ ID NO:60
  • TARl 5-26-511 SEQ ID NO:61
  • TAR15-26-512 SEQ ID NO:62
  • TAR15-26-513 SEQ ID NO:63
  • TARl 5-26-514 SEQ ID NO:64
  • TARl 5-26-515 SEQ ID NO:65
  • TARl 5-26-516 SEQ ID NO:66
  • TARl 5-26-517 SEQ ID NO:67
  • TARl 5-26-518 SEQ ID NO:68
  • TARl 5-26-519 SEQ ID NO:69
  • TARl 5-26-520 SEQ ID NO:70
  • TAR15-26-521 SEQ ID NO:71
  • TAR15-26-522 SEQ ID NO:72
  • TARl 5-26-523 SEQ ID NO:73
  • TARl 5-26-524 SEQ ID NO:74
  • TARl 5-26-525 SEQ ID NO:75
  • nucleotide sequence identity is determined over the whole length of the nucleotide sequence that encodes the selected anti-VEGF dAb.
  • the isolated and/or recombinant nucleic acid encoding a ligand that has binding specificity for VEGF comprises a nucleotide sequence that has at least about 80%, at least about 85%, at least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 94%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99% nucleotide sequence identity with a nucleotide sequence encoding TARl 5-26-555 (SEQ ID NO:705).
  • the isolated and/or recombinant nucleic acid encoding a ligand that has binding specificity for EGFR comprises a nucleotide sequence that has at least about 80%, at least about 85%, at least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 94%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99% nucleotide sequence identity with a nucleotide sequence encoding an anti-VEGF dAb selected from the group consisting of DOM16-17 (SEQ ID NO:199), DOM16-18 (SEQ ID NO:200), DOM16-19 (SEQ ID NO:201), D0M16- 20 (SEQ ID NO:202), DOMl 6-21 (SEQ ID NO:203), DOMl 6-22 (SEQ ID
  • DOM16-102 (SEQ ID NO:272)
  • DOM16-103 (SEQ ID NO:273)
  • DOM16-104 (SEQ ID NO:274)
  • DOMl 6-105 (SEQ ID NO:275)
  • DOMl 6-106 (SEQ ID NO:
  • the isolated and/or recombinant nucleic acid encoding a ligand that has binding specificity for EGFR comprises a nucleotide sequence that has at least about 80%, at least about 85%, at least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 94%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99% nucleotide sequence identity with a nucleotide sequence encoding an anti-EGFR dAb selected from the group consisting of SEQ ID NOS:623-703, 727 and 728.
  • the invention also provides a vector comprising a recombinant nucleic acid molecule of the invention.
  • the vector is an expression vector comprising one or more expression control elements or sequences that are operably linked to the recombinant nucleic acid of the invention.
  • the invention also provides a recombinant host cell comprising a recombinant nucleic acid molecule or vector of the invention.
  • Suitable vectors e.g., plasmids, phagmids
  • expression control elements, host cells and methods for producing recombinant host cells of the invention are well-known in the art, and examples are further described herein.
  • Suitable expression vectors can contain a number of components, for example, an origin of replication, a selectable marker gene, one or more expression control elements, such as a transcription control element (e.g., promoter, enhancer, terminator) and/or one or more translation signals, a signal sequence or leader sequence, and the like.
  • expression control elements and a signal sequence can be provided by the vector or other source.
  • the vector or other source for example, the
  • transcriptional and/or translational control sequences of a cloned nucleic acid encoding an antibody chain can be used to direct expression.
  • a promoter can be provided for expression in a desired host cell. Promoters can be constitutive or inducible. For example, a promoter can be operably linked to a nucleic acid encoding an antibody, antibody chain or portion thereof, such that it directs transcription of the nucleic acid.
  • suitable promoters for prokaryotic e.g., lac, tac, T3, T7 promoters for E. coli
  • expression vectors typically comprise a selectable marker for selection of host cells carrying the vector, and, in the case of a replicable expression vector, an origin or replication.
  • Genes encoding products which confer antibiotic or drug resistance are common selectable markers and may be used in procaryotic (e.g. , lactamase gene (ampicillin resistance), Tet gene for tetracycline resistance) and eucaryotic cells (e.g., neomycin (G418 or geneticin), gpt (mycophenolic acid), ampicillin, or hygromycin resistance genes).
  • Dihydrofolate reductase marker genes permit selection with methotrexate in a variety of hosts.
  • auxotrophic markers of the host e.g., LEU2, URA3, HIS3
  • vectors which are capable of integrating into the genome of the host cell such as retroviral vectors
  • Schnieder S2 cells, Sf9) and yeast are well-known in the art.
  • Suitable host cells can be prokaryotic, including bacterial cells such as E. coli, B. subtilis and/or other suitable bacteria; eukaryotic cells, such as fungal or yeast cells (e.g. , Pichia pastoris, Aspergillus sp. , Saccharomyces cerevisiae,
  • bacterial cells such as E. coli, B. subtilis and/or other suitable bacteria
  • eukaryotic cells such as fungal or yeast cells (e.g. , Pichia pastoris, Aspergillus sp. , Saccharomyces cerevisiae,
  • insects e.g., Drosophila Schnieder S2 cells, Sf9 insect cells (WO 94/26087 (O'Connor)
  • mammals e.g., COS cells, such as COS-I (ATCC Accession No. CRL-1650) and COS-7 (ATCC Accession
  • the host cell is an isolated host cell and is not part of a multicellular organism (e.g., plant or animal), hi preferred embodiments, the host cell is a non- human host cell.
  • the invention also provides a method for producing a ligand (e.g., dual- specific ligand, multispecific ligand) of the invention, comprising maintaining a recombinant host cell comprising a recombinant nucleic acid of the invention under conditions suitable for expression of the recombinant nucleic acid, whereby the recombinant nucleic acid is expressed and a ligand is produced.
  • a ligand e.g., dual- specific ligand, multispecific ligand
  • the method further comprises isolating the ligand.
  • Ligands e.g., dual specific ligands, multispecific ligands
  • Ligands can be prepared according to previously established techniques, used in the field of antibody engineering, for the preparation of scFv, "phage" antibodies and other engineered antibody molecules. Techniques for the preparation of antibodies are for example described in the following reviews and the references cited therein: Winter & Milstein, (1991) Nature 349:293-299; Pluckthun (1992) Immunological Reviews 13 0:151-188; Wright et al, (1992) CHt. Rev. Immunol 12:125-168; Holliger, P. & Winter, G. (1993) Curr. Opin. Biotechnol. 4, 446-449; Carter, et al (1995) J. Hematother. 4, 463-470; Chester, K.A. & Hawkins, R.E.
  • Suitable techniques employed for selection of antibody variable domains with a desired specificity employ libraries and selection procedures which are known in the art. Natural libraries (Marks et al (1991) J. MoI. Biol, 222: 581 ;
  • Vaughan et al. (1996) Nature Biotech., 14: 309) which use rearranged V genes harvested from human B cells are well known to those skilled in the art. Synthetic libraries (Hoogenboom & Winter (1992) J. MoI. Biol, 227: 381; Barbas et al. (1992) Proc. Natl. Acad. ScL USA, 89: 4457; Nissim et al. (1994) EMBO J, 13: 692;
  • Vjj and/or VL libraries may be selected against target antigens or epitopes separately, in which case single domain binding is directly selected for, or together.
  • Bacteriophage lambda expression systems may be screened directly as bacteriophage plaques or as colonies of lysogens, both as previously described (Huse et al. (1989) Science, 246: 1275; Caton and Koprowski (1990) Proc. Natl Acad. ScL U.S.A., 87; Mullinax et al. (1990) Proc. Natl. Acad. ScL U.S.A., 87: 8095; Persson et al. (1991) Proc. Natl. Acad. ScL U.S.A., 88: 2432) and are of use in the invention.
  • a selection display system is a system that permits the selection, by suitable display means, of the individual members of the library by binding the generic and/or target.
  • Selection protocols for isolating desired members of large libraries are known in the art, as typified by phage display techniques.
  • Such systems in which diverse peptide sequences are displayed on the surface of filamentous bacteriophage (Scott and Smith (1990) Science, 249: 386), have proven useful for creating libraries of antibody fragments (and the nucleotide sequences that encode them) for the in vitro selection and amplification of specific antibody fragments that bind a target antigen (McCafferty et al, WO 92/01047).
  • the nucleotide sequences encoding the variable regions are linked to gene fragments which encode leader signals that direct them to the periplasmic space of E.
  • phagebodies lambda phage capsids
  • An advantage of phage-based display systems is that, because they are biological systems, selected library members can be amplified simply by growing the phage containing the selected library member in bacterial cells. Furthermore, since the nucleotide sequence that encodes the polypeptide library member is contained on a phage or phagemid vector, sequencing, expression and subsequent genetic manipulation is relatively straightforward.
  • RNA molecules are selected by alternate rounds of selection against a target and PCR amplification (Tuerk and Gold (1990) Science, 249: 505; Ellington and Szostak (1990) Nature, 346: 818).
  • a similar technique may be used to identify DNA sequences which bind a predetermined human transcription factor (Thiesen and
  • a still further category of techniques involves the selection of repertoires in artificial compartments, which allow the linkage of a gene with its gene product.
  • a selection system in which nucleic acids encoding desirable gene products may be selected in microcapsules formed by water-in-oil emulsions is described in WO99/02671, WO00/40712 and Tawfik & Griffiths (1998) Nature Biotechnol 16(7), 652-6.
  • Genetic elements encoding a gene product having a desired activity are compartmentalised into microcapsules and then transcribed and/or translated to produce their respective gene products (RNA or protein) within the microcapsules.
  • Genetic elements which produce gene product having desired activity are subsequently sorted. This approach selects gene products of interest by detecting the desired activity by a variety of means.
  • Libraries intended for selection may be constructed using techniques known in the art, for example as set forth above, or may be purchased from commercial sources. Libraries which are useful in the present invention are described, for example, in WO99/20749.
  • PCR polymerase chain reaction
  • PCR is performed using template DNA (at least lfg; more usefully, 1-1000 ng) and at least 25 pmol of oligonucleotide primers; it maybe advantageous to use a larger amount of primer when the primer pool is heavily heterogeneous, as each sequence is represented by only a small fraction of the molecules of the pool, and amounts become limiting in the later amplification cycles.
  • a typical reaction mixture includes: 2 ⁇ l of DNA, 25 pmol of oligonucleotide primer, 2.5 ⁇ l of 1OX PCR buffer 1 (Perkin-Elmer, Foster City, CA), 0.4 ⁇ l of 1.25 ⁇ M dNTP, 0.15 ⁇ l (or 2.5 units) of Taq DNA polymerase (Perkin Elmer, Foster City, CA) and deionized water to a total volume of 25 ⁇ l.
  • Mineral oil is overlaid and the PCR is performed using a programmable thermal cycler. The length and temperature of each step of a PCR cycle, as well as the number of cycles, is adjusted in accordance to the stringency requirements in effect.
  • Annealing temperature and timing are determined both by the efficiency with which a primer is expected to anneal to a template and the degree of mismatch that is to be tolerated; obviously, when nucleic acid molecules are simultaneously amplified and mutagenised, mismatch is required, at least in the first round of synthesis.
  • the ability to optimise the stringency of primer annealing conditions is well within the knowledge of one of moderate skill in the art.
  • An annealing temperature of between 30 °C and 72 0 C is used.
  • Initial denaturation of the template molecules normally occurs at between 92 0 C and 99 0 C for 4 minutes, followed by 20-40 cycles consisting of denaturation (94-99 0 C for 15 seconds to 1 minute), annealing (temperature determined as discussed above; 1 -2 minutes), and extension (72 0 C for 1-5 minutes, depending on the length of the amplified product).
  • Final extension is generally for 4 minutes at 72 0 C, and may be followed by an indefinite (0-24 hour) step at 4 0 C.
  • Domains useful in the invention may be combined by a variety of methods known in the art, including covalent and non-covalent methods.
  • Preferred methods include the use of polypeptide linkers, as described, for example, in connection with scFv molecules (Bird et al , (1988) Science 2A2-A ⁇ h-A2 €).
  • Linkers are preferably flexible, allowing the two single domains to interact.
  • the linkers used in diabodies, which are less flexible, may also be employed (Holliger et al, (1993) Proc Nat Acad Sci USA 90:6444-6448).
  • the linker employed is not an immunoglobulin hinge region.
  • Variable domains may be combined using methods other than linkers. For example, the use of disulphide bridges, provided through naturally-occurring or engineered cysteine residues, may be exploited to stabilize V H ⁇ V H> V L 'V L or V H -V L dimers (Reiter et al., (1994) Protein Eng. 7:697-704) or by remodelling the interface between the variable domains to improve the "fit" and thus the stability of interaction (Ridgeway et al, (1996) Protein Eng. 7:617-621; Zhu et al, (1997) Protein Science 6:781-788). Other techniques for joining or stabilizing variable domains of immunoglobulins, and in particular antibody VH domains, may be employed as appropriate. Characterisation of Ligands
  • binding of a dual-specific ligand to the cell or the binding of each binding domain to each specific target can be tested by methods which will be familiar to those skilled in the art and include ELISA.
  • binding is tested using monoclonal phage ELISA.
  • Phage ELISA may be performed according to any suitable procedure: an exemplary protocol is set forth below.
  • phage produced at each round of selection can be screened for binding by ELISA to the selected antigen or epitope, to identify "polyclonal" phage antibodies. Phage from single infected bacterial colonies from these populations can then be screened by ELISA to identify "monoclonal” phage antibodies. It is also desirable to screen soluble antibody fragments for binding to antigen or epitope, and this can also be undertaken by ELISA using reagents, for example, against a C- or N-terminal tag (see for example Winter et al. (1994) Ann. Rev. Immunology 12, 433- 55 and references cited therein.
  • the diversity of the selected phage monoclonal antibodies may also be assessed by gel electrophoresis of PCR products (Marks et al. 1991, supra; Nissim et al. 1994 supra), probing (Tomlinson et al, 1992) J. MoI. Biol. 227, 776) or by sequencing of the vector DNA.
  • variable domains are selected from V-gene repertoires, selected for instance using phage display technology as herein described, then these variable domains comprise a universal framework region, such that they may be recognized by a specific generic dual-specific ligand as herein defined.
  • the use of universal frameworks, generic ligands and the like is described in WO99/20749.
  • variable domains are preferably located within the structural loops of the variable domains.
  • the polypeptide sequences of either variable domain may be altered by DNA shuffling or by mutation in order to enhance the interaction of each variable domain with its complementary pair.
  • DNA shuffling is known in the art and taught, for example, by Stemmer, 1994, Nature 370: 389-391 and U.S. Patent No. 6,297,053, both of which are incorporated herein by reference.
  • Other methods of mutagenesis are well known to those of skill in the art.
  • nucleic acid molecules and vector constructs required for selection, preparation and formatting dual-specific ligands may be constructed and manipulated as set forth in standard laboratory manuals, such as Sambrook et at. (1989) Molecular Cloning: A Laboratory Manual, Cold Spring Harbor,. USA.
  • vector refers to a discrete element that is used to introduce heterologous DNA into cells for the expression and/or replication thereof.
  • a vector of use according to the invention may be selected to
  • each vector contains various functional components, which generally include a cloning (or "polylinker") site, an origin of replication and at least one selectable marker gene. If the given vector is an expression vector, it additionally possesses one or more of the following: enhancer element, promoter, transcription termination and signal sequences, each positioned in the vicinity of the cloning site, such that they are operatively linked to the gene encoding a dual-specific ligand according to the invention.
  • Both cloning and expression vectors generally contain nucleic acid sequences that enable the vector to replicate in one or more selected host cells.
  • this sequence is one that enables the vector to replicate independently of the host chromosomal DNA and includes origins of replication or autonomously replicating sequences.
  • origins of replication or autonomously replicating sequences are well known for a variety of bacteria, yeast and viruses.
  • the origin of replication from the plasmid pBR322 is suitable for most Gram-negative bacteria, the 2 micron plasmid origin is suitable for yeast, and various viral origins (e.g. SV 40, adenovirus) are useful for cloning vectors in mammalian cells.
  • the origin of replication is not needed for mammalian expression vectors unless these are used in mammalian cells able to replicate high levels of DNA, such as COS cells.
  • a cloning or expression vector may contain a selection gene also referred to as a selectable marker.
  • This gene encodes a protein necessary for the survival or growth of transformed host cells grown in a selective culture medium. Host cells not transformed with the vector containing the selection gene will therefore not survive in the culture medium.
  • Typical selection genes encode proteins that confer resistance to antibiotics and other toxins, e.g. ampicillin, neomycin, methotrexate or tetracycline, complement auxotrophic deficiencies, or supply critical nutrients not available in the growth media.
  • an E. co/z-selectable marker for example, the ⁇ -lactamase gene that confers resistance to the antibiotic ampicillin.
  • E. coli plasmids such as pBR322 or a pUC plasmid such as pUCl 8 or pUCl 9.
  • Expression vectors usually contain a promoter that is recognised by the host organism and is operably linked to the coding sequence of interest. Such a promoter may be inducible or constitutive.
  • operably linked refers to a promoter that is recognised by the host organism and is operably linked to the coding sequence of interest.
  • a control sequence "operably linked" to a coding sequence is ligated in such a way that expression of the coding sequence is achieved under conditions compatible with the control sequences.
  • Promoters suitable for use with prokaryotic hosts include, for example, the ⁇ - lactamase and lactose promoter systems, alkaline phosphatase, the tryptophan (trp) promoter system and hybrid promoters such as the tac promoter. Promoters for use in bacterial systems will also generally contain a Shine-Delgarno sequence operably linked to the coding sequence.
  • the preferred vectors are expression vectors that enables the expression of a nucleotide sequence corresponding to a polypeptide library member.
  • selection with the first and/or second antigen or epitope can be performed by separate propagation and expression of a single clone expressing the polypeptide library member or by use of any selection display system.
  • the preferred selection display system is bacteriophage display.
  • phage or phagemid vectors may be used, (e.g., pITl or pIT2.
  • Leader sequences useful in the invention include pelB, stll, ompA, phoA, bla and pelA.
  • phagemid vectors which have an E. coli.
  • the vector contains a ⁇ -lactamase gene to confer selectivity on the phagemid and a lac promoter upstream of a expression cassette that consists (N to C terminal) of a pelB leader sequence (which directs the expressed polypeptide to the periplasmic space), a multiple cloning site (for cloning the nucleotide version of the library member), optionally, one or more peptide tag (for detection), optionally, one or more TAG stop codon and the phage protein pill.
  • a pelB leader sequence which directs the expressed polypeptide to the periplasmic space
  • a multiple cloning site for cloning the nucleotide version of the library member
  • one or more peptide tag for detection
  • TAG stop codon optionally, one or more TAG stop codon and the phage protein pill.
  • the vector is able to replicate as a plasmid with no expression, produce large quantities of the polypeptide library member only or produce phage, some of which contain at least one copy of the polypeptide-pIII fusion on their surface.
  • Construction of vectors encoding dual-specific ligands employs conventional ligation techniques. Isolated vectors or DNA fragments are cleaved, tailored, and religated in the form desired to generate the required vector. If desired, analysis to confirm that the correct sequences are present in the constructed vector can be performed in a known fashion. Suitable methods for constructing expression vectors, preparing in vitro transcripts, introducing DNA into host cells, and performing analyses for assessing expression and function are known to those skilled in the art.
  • Skeletons may be based on immunoglobulin molecules or may be non- immunoglobulin in origin as set forth above. Each domain of the dual-specific ligand may be a different skeleton.
  • Preferred immunoglobulin skeletons as herein defined includes any one or more of those selected from the following: an immunoglobulin molecule comprising at least (i) the CL (kappa or lambda subclass) domain of an antibody; or (ii) the CHl domain of an antibody heavy chain; an immunoglobulin molecule comprising the CHl and CH2 domains of an antibody heavy chain; an immunoglobulin molecule comprising the CHl, CH2 and CH3 domains of an antibody heavy chain; or any of the subset (ii) in conjunction with the CL (kappa or lambda subclass) domain of an antibody.
  • a hinge region domain may also be included.
  • Such combinations of domains may, for example, mimic natural antibodies, such as IgG or IgM, or fragments thereof, such as Fv, scFv, Fab or F(ab') 2 molecules. Those skilled in the art will be aware that this list is not intended to be exhaustive.
  • Each binding domain comprises a protein scaffold and one or more CDRs which are involved in the specific interaction of the domain with one or more epitopes.
  • an epitope binding domain according to the present invention comprises three CDRs.
  • Suitable protein scaffolds include any of those selected from the group consisting of the following: those based on immunoglobulin domains, those based on fibronectin, those based on affibodies, those based on CTLA4, those based on chaperones such as GroEL, those based on lipocallin and those based on the bacterial Fc receptors SpA and SpD. Those skilled in the art will appreciate that this list is not intended to be exhaustive.
  • the members of the immunoglobulin superfamily all share a similar fold for their polypeptide chain.
  • antibodies are highly diverse in terms of their primary sequence, comparison of sequences and crystallographic structures has revealed that, contrary to expectation, five of the six antigen binding loops of antibodies (Hl, H2, Ll, L2, L3) adopt a limited number of main-chain
  • H3 region is much more diverse in terms of sequence, length and structure (due to the use of D segments), it also forms a limited number of main-chain conformations for short loop lengths which depend on the length and the presence of particular residues, or types of residue, at key positions in the loop and the antibody framework (Martin et al. (1996) J. MoI Biol, 263: 800; Shirai et al. (1996) FEBS Letters, 399: 1).
  • Libraries of ligands and/or binding domains can be designed in which certain loop lengths and key residues have been chosen to ensure that the main-chain conformation of the members is known.
  • these are real
  • Germline V gene segments serve as one suitable basic framework for constructing antibody or T- cell receptor libraries; other sequences are also of use. Variations may occur at a low frequency, such that a small number of functional members may possess an altered main-chain conformation, which does not affect its function.
  • Canonical structure theory is also of use to assess the number of different main-chain conformations encoded by ligands, to predict the main-chain
  • Vx domain encodes a different range of canonical structures for the Ll, L2 and L3 loops and that V ⁇ and V ⁇ domains can pair with any V H domain which can encode several canonical structures for the Hl and H2 loops
  • the number of canonical structure combinations observed for these five loops is very large. This implies that the generation of diversity in the main-chain conformation may be essential for the production of a wide range of binding specificities.
  • by constructing an antibody library based on a single known main-chain conformation it has been found, contrary to expectation, that diversity in the main-chain conformation is not required to generate sufficient diversity to target substantially all antigens.
  • the single main-chain conformation need not be a consensus structure - a single naturally occurring conformation can be used as the basis for an entire library.
  • the ligands of the invention possess a single known main-chain conformation.
  • the single main-chain conformation that is chosen is preferably
  • the natural occurrence of the different main-chain conformations for each binding loop of an immunoglobulin domain are considered and then a naturally occurring variable domain is chosen which possesses the desired combination of main-chain conformations for the different loops. If none is available, the nearest equivalent may be chosen.
  • the desired combination of main- chain conformations for the different loops is created by selecting germline gene segments which encode the desired main-chain conformations. It is more preferable, that the selected germline gene segments are frequently expressed in nature, and most preferable that they are the most frequently expressed of all natural germline gene segments.
  • ligands e.g., ds-dAbs
  • the incidence of the different main-chain conformations for each of the six antigen binding loops may be considered separately.
  • Hl, H2, Ll, L2 and L3 a given conformation that is adopted by between 20% and 100% of the antigen binding loops of naturally occurring molecules is chosen.
  • its observed incidence is above 35% (i.e. between 35% and 100%) and, ideally, above 50% or even above 65%.
  • Hl - CS 1 (79% of the expressed repertoire), H2 - CS 3 (46%), Ll - CS 2 of V ⁇ (39%), L2 - CS 1 (100%), L3 - CS 1 of V ⁇ (36%) (calculation assumes a ⁇ : ⁇ ratio of 70:30, Hood et al. (1967) Cold Spring Harbor Symp. Quant. Biol, 48: 133).
  • H3 loops that have canonical structures a CDR3 length (Kabat et al. (1991) Sequences of proteins of immunological interest, U.S.
  • the natural occurrence of combinations of main-chain conformations is used as the basis for choosing the single main-chain conformation.
  • the natural occurrence of canonical structure combinations for any two, three, four, five or for all six of the antigen binding loops can be determined.
  • the chosen conformation is
  • dual-specific ligands e.g., ds-dAbs
  • libraries for use in the invention can be constructed by varying each binding site of the molecule in order to generate a repertoire with structural and/or functional diversity. This means that variants are generated such that they possess sufficient diversity in their structure and/or in their function so that they are capable of providing a range of activities.
  • the desired diversity is typically generated by varying the selected molecule at one or more positions.
  • the positions to be changed can be chosen at random or are preferably selected.
  • the variation can then be achieved either by randomisation, during which the resident amino acid is replaced by any amino acid or analogue thereof, natural or synthetic, producing a very large number of variants or by replacing the resident amino acid with one or more of a defined subset of amino acids, producing a more limited number of variants.
  • H3 region of a human tetanus toxoid-binding Fab has been randomised to create a range of new binding specificities (Barbas et al (1992) Proc. Natl. Acad. ScL USA, 89: 4457). Random or semi-random H3 and L3 regions have been appended to germline V gene segments to produce large libraries with unmutated framework regions (Hoogenboom & Winter (1992) J. MoI. Biol, 227: 381; Barbas et al (1992) Proc. Natl Acad. Sci.
  • loop randomization has the potential to create approximately more than 10 15 structures for H3 alone and a similarly large number of variants for the other five loops, it is not feasible using current transformation technology or even by using cell free systems to produce a library representing all possible combinations.
  • 6 x 10 1 different antibodies which is only a fraction of the potential diversity for a library of this design, were generated (Griffiths et al. (1994) supra).
  • each domain of the dual-specific ligand molecule Preferably, only the residues that are directly involved in creating or modifying the desired function of each domain of the dual-specific ligand molecule are diversified.
  • the function of each domain will be to bind a target and therefore diversity should be concentrated in the target binding site, while avoiding changing residues which are crucial to the overall packing of the molecule or to maintaining the chosen main-chain conformation.
  • the binding site for each target is most often the antigen binding site.
  • residues in the antigen binding site are varied.
  • These residues are extremely diverse in the human antibody repertoire and are known to make contacts in high-resolution antibody/antigen complexes.
  • positions 50 and 53 are diverse in naturally occurring antibodies and are observed to make contact with the antigen.
  • the conventional approach would have been to diversify all the residues in the corresponding Complementarity Determining Region (CDRl) as defined by Kabat et al. (1991, supra), some seven residues compared to the two diversified in the library for use according to the invention.
  • CDRl Complementarity Determining Region
  • antibody diversity is the result of two processes: somatic recombination of germline V, D and J gene segments to create a naive primary repertoire (so called germline and junctional diversity) and somatic hypermutation of the resulting rearranged V genes.
  • somatic recombination of germline V, D and J gene segments to create a naive primary repertoire (so called germline and junctional diversity)
  • somatic hypermutation of the resulting rearranged V genes The analysis of human antibody sequences has shown that diversity in the primary repertoire is focused at the centre of the antigen binding site whereas somatic hypermutation spreads diversity to regions at the periphery of the antigen binding site that are highly conserved in the primary repertoire (see Tomlinson et al. (1996) J. MoI. Biol, 256: 813).
  • an initial 'naive' repertoire can be created where some, but not all, of the residues in the antigen binding site are diversified.
  • the term "naive” refers to antibody molecules that have no pre-determined target. These molecules resemble those which are encoded by the immunoglobulin genes of an individual who has not undergone immune diversification, as is the case with fetal and newborn individuals, whose immune systems have not yet been challenged by a wide variety of antigenic stimuli.
  • This repertoire is then selected against a range of antigens or epitopes. If required, further diversity can then be introduced outside the region diversified in the initial repertoire. This matured repertoire can be selected for modified function, specificity or affinity.
  • Naive repertoires of binding domains for the construction of dual-specific ligands in which some or all of the residues in the antigen binding site are varied are known in the art. (See, WO 2004/058821, WO 2004/003019, and WO 03/002609).
  • the "primary" library mimics the natural primary repertoire, with diversity restricted to residues at the centre of the antigen binding site that are diverse in the germline V gene segments (germline diversity) or diversified during the recombination process (junctional diversity).
  • residues which are diversified include, but are not limited to, H50, H52, H52a, H53, H55, H56, H58, H95, H96, H97, H98, L50, L53, L91, L92, L93, L94 and L96.
  • diversity is restricted to residues that are diversified during the recombination process (junctional diversity) or are highly somatically mutated.
  • residues which are diversified include, but are not limited to: H31, H33, H35, H95, H96, H97, H98, L30, L31, L32, L34 and L96.
  • diversification of chosen positions is typically achieved at the nucleic acid level, by altering the coding sequence which specifies the sequence of the polypeptide such that a number of possible amino acids (all 20 or a subset thereof) can be incorporated at that position.
  • the most versatile codon is NNK, which encodes all amino acids as well as the TAG stop codon.
  • the NNK codon is preferably used in order to introduce the required diversity.
  • Other codons which achieve the same ends are also of use, including the NNN codon, which leads to the production of the additional stop codons TGA and TAA.
  • a feature of side-chain diversity in the antigen binding site of human antibodies is a pronounced bias which favors certain amino acid residues. If the amino acid composition of the ten most diverse positions in each of the V H , V K and V ⁇ regions are summed, more than 76% of the side-chain diversity comes from only seven different residues, these being, serine (24%), tyrosine (14%), asparagine (11%), glycine (9%), alanine (7%), aspartate (6%) and threonine (6%).
  • codons (AGT)(AGC)T, (AGT)(AGC)C and (AGT)(AGC)(CT) - that is, DVT, DVC and DVY, respectively using IUPAC nomenclature - are those closest to the desired amino acid profile: they encode 22% serine and 11% tyrosine, asparagine, glycine, alanine, aspartate, threonine and cysteine.
  • libraries are constructed using either the DVT, DVC or DVY codon at each of the diversified positions.
  • the invention provides compositions comprising the ligands of the invention and a pharmaceutically acceptable carrier, diluent or excipient, and therapeutic and diagnostic methods that employ the ligands or compositions of the invention.
  • a pharmaceutically acceptable carrier diluent or excipient
  • therapeutic and diagnostic methods that employ the ligands or compositions of the invention.
  • the ligands according to the method of the present invention may be employed in in vivo therapeutic and prophylactic applications, in vivo diagnostic applications and the like.
  • ligands of the invention involve the administration of ligands according to the invention to a recipient mammal, such as a human.
  • the ligands bind to targets with high affinity and/or avidity.
  • the ligands can allow recruitment of cytotoxic cells to mediate killing of camcer cells, for example by antibody dependent cellular cytoxicity.
  • Substantially pure ligands of at least 90 to 95% homogeneity are preferred for administration to a mammal, and 98 to 99% or more homogeneity is most preferred for pharmaceutical uses, especially when the mammal is a human.
  • the ligands may be used
  • the ligands of the present invention will typically find use in preventing, suppressing or treating disease states.
  • ligands can be administered to treat, suppress or prevent a chronic inflammatory disease, allergic hypersensitivity, cancer, bacterial or viral infection, autoimmune disorders (which include, but are not limited to, Type I diabetes, asthma, multiple sclerosis, rheumatoid arthritis, juvenile rheumatoid arthritis, psoriatic arthritis,
  • spondylarthropathy ⁇ e.g., ankylosing spondylitis
  • systemic lupus erythematosus inflammatory bowel disease ⁇ e.g., Crohn's disease, ulcerative colitis
  • myasthenia gravis and Behcet's syndrome myasthenia gravis and Behcet's syndrome
  • psoriasis endometriosis
  • abdominal adhesions ⁇ e.g., post abdominal surgery.
  • the ligands are useful for treating infectious diseases in which cells infected with an infectious agent contain higer levels of cell surface EGFR than uninfected cells or that contain one or more cell surface targets that are not present on ininfected cells, such as a protein that is encoded by the infectious agent ⁇ e.g., bacteria, virus).
  • Ligands according to the invention that are able to bind to EGFR can be internalized by cells that express EGFR ⁇ e.g., endocytosed), and can deliver therapeutic agents ⁇ e.g., a toxin) intracellular ⁇ ⁇ e.g., deliver a dAb that binds an intracellular target).
  • ligands provide a means by which a binding domain ⁇ e.g., a dAb monomer) that is specifically able to bind to an intracellular target can be delivered to an intracellular environment. This strategy requires, for example, a binding domain with physical properties that enable it to remain functional inside the cell. Alternatively, if the final destination intracellular compartment is oxidising, a well folding ligand may not need to be disulphide free.
  • prevention involves administration of the protective composition prior to the induction of the disease.
  • suppression refers to administration of the composition after an inductive event, but prior to the clinical appearance of the disease.
  • Treatment involves administration of the protective composition after disease symptoms become manifest. Treatment includes ameliorating symptoms associated with the disease, and also preventing or delaying the onset of the disease and also lessening the severity or frequency of symptoms of the disease.
  • cancer refers to the pathological condition in mammals that is typically characterized by dysregulated cellular proliferation or survival.
  • examples of cancer include, but are not limited to, carcinoma, lymphoma, blastoma, sarcoma, and leukemia and lymphoid malignancies. More particular examples of cancers include squamous cell cancer (e.g.
  • lung cancer e.g., small-cell lung carcinoma, non-small cell lung carcinoma, adenocarcinoma of the lung, squamous carcinoma of the lung
  • cancer of the peritoneum hepatocellular cancer
  • gastric or stomach cancer including gastrointestinal cancer, pancreatic cancer, glioblastoma, cervical cancer, ovarian cancer, liver cancer, bladder cancer, gall bladder cancer, hepatoma, breast cancer, colon cancer, rectal cancer, colorectal cancer, multiple myeloma, chronic myelogenous leukemia, acute myelogenous leukemia, endometrial or uterine carcinoma, salivary gland carcinoma, kidney or renal cancer, prostate cancer, vulval cancer, thyroid cancer, hepatic carcinoma, anal carcinoma, penile carcinoma, head and neck cancer, and the like.
  • Cancers characterized by expression of EGFR on the surface of cancerous cells include, for example, bladder cancer, ovarian cancer, colorectal cancer, breast cancer, lung cancer (e.g., non-small cell lung carcinoma), gastric cancer, pancreatic cancer, prostate cancer, head and neck cancer, renal cancer and gall bladder cancer.
  • Suitable models of cancer include, for example, xenograft and orthotopic models of human cancers in animal models, such as the SCID-hu myeloma model (Epstein J, and Yaccoby, S., Methods MoI Med. 773:183-90 (2005), Tassone P, et al, Clin Cancer Res.
  • mice models of human lung cancer e.g., Meu Giveaway R and Berns A, Genes Dev.l P(6):643-64 (2005)
  • mouse models of metastatic cancers e.g., Kubota T '., J Cell Biochem. 56(l):4-8 (1994)
  • the present ligands will be utilized in purified form together with pharmacologically appropriate carriers.
  • these carriers include aqueous or alcoholic/aqueous solutions, emulsions or suspensions, including saline and/or buffered media.
  • Parenteral vehicles include sodium chloride solution, Ringer's dextrose, dextrose and sodium chloride and lactated Ringer's. Suitable
  • physiologically-acceptable adjuvants if necessary to keep a polypeptide complex in suspension, may be chosen from thickeners such as carboxymethylcellulose, polyvinylpyrrolidone, gelatin and alginates.
  • Intravenous vehicles include fluid and nutrient replenishers and electrolyte replenishers, such as those based on Ringer's dextrose. Preservatives and other additives, such as antimicrobials, antioxidants, chelating agents and inert gases, may also be present (Mack (1982) Remington's Pharmaceutical Sciences, 16th Edition). A variety of suitable formulations can be used, including extended release formulations.
  • the ligands of the present invention may be used as separately administered compositions or in conjunction with other agents.
  • the ligands can be administered and or formulated together with one or more additional therapeutic or active agents.
  • additional therapeutic or active agents When a ligand is administered with an additional therapeutic agent, the ligand can be administered before, simultaneously with or subsequent to administration of the additional agent.
  • the ligand and additional agent are administered in a manner that provides an overlap of therapeutic effect.
  • Additional agents that can be administered or formulated with the ligand of the invention include, for example, various immunotherapeutic drugs, such as cylcosporine, methotrexate, adriamycin or cisplatinum, antibiotics, antimycotics, anti-viral agents and immunotoxins.
  • the antagonist when it is administered to prevent, suppress or treat lung inflammation or a respiratory disease, it can be administered in conjuction with phosphodiesterase inhibitors (e.g., inhibitors of phosphodiesterase 4),
  • bronchodilators e.g., beta2-agonists, anticholinergerics, theophylline
  • short-acting beta-agonists e.g., albuterol, salbutamol, bambuterol, fenoterol, isoetherine, isoproterenol, levalbuterol, metaproterenol, pirbuterol, terbutaline and tornlate
  • long-acting beta-agonists e.g., formoterol and salmeterol
  • short acting beta-agonists e.g., albuterol, salbutamol, bambuterol, fenoterol, isoetherine, isoproterenol, levalbuterol, metaproterenol, pirbuterol, terbutaline and tornlate
  • long-acting beta-agonists e.g., formoterol and salmeterol
  • anticholinergics e.g., ipratropium bromide and oxitropium bromide
  • long-acting anticholinergics e.g., tiotropium
  • theophylline e.g. short acting formulation, long acting formulation
  • inhaled steroids e.g., beclomethasone, beclometasone, budesonide, fiunisolide, fluticasone propionate and triamcinolone
  • oral steroids e.g., methylprednisolone, prednisolone, prednisolon and prednisone
  • combined short-acting beta-agonists with anticholinergics e.g.,
  • albuterol/salbutamol/ipratopium and fenoterol/ipratopium
  • combined long-acting beta-agonists with inhaled steroids e.g., salmeterol/fluticasone, and
  • mucolytic agents e.g., erdosteine, acetylcysteine, bromheksin, carbocysteine, guiafenesin and iodinated glycerol.
  • the ligands of the invention can be coadministered (e.g., to treat cancer, an inflammatory disease or other disease) with a variety of suitable co-therapeutic agents, including cytokines, analgesics/antipyretics, antiemetics, and
  • immunosuppressive agents selected from the group consisting of cyclosporine, azathioprine, mycophenolic acid, mycophenolate mofetil, corticosteroids, methotrexate, gold salts, sulfasalazine, antimalarials, brequinar, leflunomide, mizoribine, 15-deoxyspergualine, 6-mercaptopurine, cyclophosphamide, rapamycin, tacrolimus (FK-506), OKT3, and anti-thymocyte globulin, anti-inflammatory agents selected from the group consisting of aspirin, other salicylates, steroidal drugs, NSAIDs (nonsteroidal anti-inflammatory drugs), Cox-2 inhibitors, and DMARDs (disease modifying antirheumatic drugs); anti-psoriasis agents selected from the group consisting of coal tar, A vitamin, anthralin, calcipotrien, tarazotene, corticosteroids, met
  • Cytokines include, without limitation, a lymphokine, tumor necrosis factors, tumor necrosis factor-like cytokine, lymphotoxin, interferon, macrophage inflammatory protein, granulocyte monocyte colony stimulating factor, interleukin (including, without limitation, interleukin- 1, interleukin-2, interleukin-6, interleukin- 12, interleukin-15, interleukin- 18), growth factors, which include, without limitation, ⁇ e.g., growth hormone, insulin-like growth factor 1 and 2 (IGF-I and IGF-2), granulocyte colony stimulating factor (GCSF), platelet derived growth factor (PGDF), epidermal growth factor (EGF), and agents for erythropoiesis stimulation, e.g., recombinant human erythropoietin (Epoetin alfa), EPO, a hormonal agonist, hormonal antagonists ⁇ e.g., flutamide, tamoxifen, leuprolide
  • steroids ⁇ e.g., dexamethasone, retinoid, betamethasone, Cortisol, cortisone, prednisone, dehydrotestosterone, glucocorticoid, mineralocorticoid, estrogen, testosterone, progestin).
  • Analgesics/antipyretics can include, without limitation, e.g., aspirin, acetaminophen, ibuprofen, naproxen sodium, buprenorphine hydrochloride, propoxyphene hydrochloride, propoxyphene napsylate, meperidine hydrochloride, hydromorphone hydrochloride, morphine sulfate, oxycodone hydrochloride, codeine phosphate, dihydrocodeine bitartrate, pentazocine hydrochloride, hydrocodone bitartrate, levorphanol tartrate, diflunisal, trolamine salicylate, nalbuphine hydrochloride, mefenamic acid, butorphanol tartrate, choline salicylate, butalbital, phenyltoloxamine citrate, diphenhydramine citrate, methotrimeprazine,
  • Antiemetics can also be coadministered to prevent or treat nausea and vomiting e.g., suitable antiemetics include meclizine hydrochloride, nabilone, prochlorperazine, dimenhydrinate, promethazine hydrochloride, thiethylperazine, scopolamine, and the like.
  • Chemotherapeutic agents include, but are not limited to, for example antimicrotubule agents, e.g., taxol (paclitaxel), taxotere (docetaxel); alkylating agents, e.g., cyclophosphamide, carmustine, lomustine, and chlorambucil; cytotoxic antibiotics, e.g., dactinomycin, doxorubicin, mitomycin-C, and bleomycin; antimetabolites, e.g., cytarabine, gemcitatin, methotrexate, and 5- fluorouracil; antimiotics, e.g., vincristine vinca alkaloids, e.g., etoposide, vinblastine, and vincristine; and others such as cisplatin, dacarbazine, procarbazine, and hydroxyurea; and combinations thereof.
  • antimicrotubule agents e.g., taxol (
  • the ligands of the invention can be used to treat cancer in combination with another therapeutic agent.
  • a ligand of the invention can be
  • the invention provides a method of treating cancer comprising administering to a patient in need thereof a
  • chemotherapeutic agent administered at a low dose.
  • amount of chemotherapeutic agent that is coadministered with a ligand of the invention is about 80%, or about 70%, or about 60%, or about 50%, or about 40%, or about 30%, or about 20%, or about 10% or less, of the dose of chemotherapeutic agent alone that is normally administered to a patient.
  • cotherapy is particularly advantageous when the chemotherapeutic agent causes deleterious or undesirable side effects that may be reduced or eliminated at a lower doses.
  • compositions can include "cocktails" of various cytotoxic or other agents in conjunction with ligands of the present invention, or even combinations of ligands according to the present invention having different specificities, such as ligands selected using different target antigens or epitopes, whether or not they are pooled prior to administration.
  • the route of administration of pharmaceutical compositions according to the invention may be any suitable route, such as any of those commonly known to those of ordinary skill in the art.
  • For therapy including without limitation
  • the ligands of the invention can be administered to any patient in accordance with standard techniques.
  • the administration can be by any appropriate mode, including parenterally, intravenously, intramuscularly, intraperitoneally, transdermally, intrathecally, intraarticularly, via the pulmonary route, or also, appropriately, by direct infusion (e.g., with a catheter).
  • the dosage and frequency of administration will depend on the age, sex and condition of the patient, concurrent administration of other drugs, counterindications and other parameters to be taken into account by the clinician.
  • Administration can be local (e.g., local delivery to the lung by pulmonary administration,(e.g. , intranasal administration) or local injection directly into a tumor) or systemic as indicated.
  • the ligands of this invention can be lyophilised for storage and reconstituted in a suitable carrier prior to use. This technique has been shown to be effective with conventional immunoglobulins and art-known lyophilisation and reconstitution techniques can be employed. It will be appreciated by those skilled in the art that lyophilisation and reconstitution can lead to varying degrees of antibody activity loss (e.g. with conventional immunoglobulins, IgM antibodies tend to have greater activity loss than IgG antibodies) and that use levels may have to be adjusted upward to compensate.
  • compositions containing the ligands can be administered for any period of time.
  • the compositions containing the ligands can be administered for any period of time.
  • an adequate amount to accomplish at least partial inhibition, suppression, modulation, killing, or some other measurable parameter, of a population of selected cells is defined as a "therapeutically-effective dose”. Amounts needed to achieve this dosage will depend upon the severity of the disease and the general state of the patient's health, but generally range from 0.005 to 5.0 mg of ligand per kilogram of body weight, with doses of 0.05 to 2.0 mg/kg/dose being more commonly used.
  • compositions containing the present ligands or cocktails thereof may also be administered in similar or slightly lower dosages, to prevent, inhibit or delay onset of disease ⁇ e.g., to sustain remission or quiescence, or to prevent acute phase).
  • a ligand When a ligand is administered to treat, suppress or prevent a disease, it can be administered up to four times per day, twice weekly, once weekly, once every two weeks, once a month, or once every two months, at a dose of, for example, about 10 ⁇ g/kg to about 80 mg/kg, about 100 ⁇ g/kg to about 80 mg/kg, about 1 mg/kg to about 80 mg/kg, about 1 mg/kg to about 70 mg/kg, about 1 mg/kg to about 60 mg/kg, about 1 mg/kg to about 50 mg/kg, about 1 mg/kg to about 40 mg/kg, about 1 mg/kg to about 30 mg/kg, about 1 mg/kg to about 20 mg/kg , about 1 mg/kg to about 10 mg/kg, about 10 ⁇ g/kg to about 10 mg/kg, about 10 ⁇ g/kg to about 5 mg/kg, about 10 ⁇ g/kg to about 2.5 mg/kg, about 1 mg/kg
  • the dual-specific ligand is administered to treat, suppress or prevent a chronic inflammatory disease once every two weeks or once a month at a dose of about 10 ⁇ g/kg to about 10 mg/kg (e.g., about 10 ⁇ g/kg, about 100 ⁇ g/kg, about 1 mg/kg, about 2 mg/kg, about 3 mg/kg, about 4 mg/kg, about 5 mg/kg, about 6 mg/kg, about 7 mg/kg, about 8 mg/kg, about 9 mg/kg or about 10 mg/kg.)
  • the ligand of the invention is administered at a dose that provides saturation of EGFR or a desired serum concentration in vivo.
  • the skilled physician can determine appropriate dosing to achieve saturation, for example by titrating ligand and monitoring the amount of free binding sites on EGFR expressing cells or the serum concentration of ligand.
  • Therapeutic regiments that involve administering a therapeutic agent to achieve target saturation or a desired serum concentration of agent are common in the art, particularly in the field of oncology.
  • Treatment or therapy performed using the compositions described herein is considered “effective” if one or more symptoms are reduced ⁇ e.g., by at least 10% or at least one point on a clinical assessment scale), relative to such symptoms present before treatment, or relative to such symptoms in an individual (human or model animal) not treated with such composition or other suitable control. Symptoms will obviously vary depending upon the disease or disorder targeted, but can be measured by an ordinarily skilled clinician or technician.
  • Such symptoms can be measured, for example, by monitoring the level of one or more biochemical indicators of the disease or disorder ⁇ e.g., levels of an enzyme or metabolite correlated with the disease, affected cell numbers, etc.), by monitoring physical manifestations ⁇ e.g., inflammation, tumor size, etc.), or by an accepted clinical assessment scale, for example, the Expanded Disability Status Scale (for multiple sclerosis), the Irvine Inflammatory Bowel Disease Questionnaire (32 point assessment evaluates quality of life with respect to bowel function, systemic symptoms, social function and emotional status - score ranges from 32 to 224, with higher scores indicating a better quality of life), the Quality of Life Rheumatoid Arthritis Scale, or other accepted clinical assessment scale as known in the field.
  • biochemical indicators of the disease or disorder e.g., levels of an enzyme or metabolite correlated with the disease, affected cell numbers, etc.
  • physical manifestations e.g., inflammation, tumor size, etc.
  • an accepted clinical assessment scale for example, the
  • a sustained ⁇ e.g., one day or more, preferably longer) reduction in disease or disorder symptoms by at least 10% or by one or more points on a given clinical scale is indicative of "effective" treatment.
  • prophylaxis performed using a composition as described herein is
  • a composition containing ligands according to the present invention may be utilized in prophylactic and therapeutic settings to aid in the alteration, inactivation, killing or removal of a select target cell population in a mammal.
  • the ligands and selected repertoires of polypeptides described herein may be used extracorporeally or in vitro selectively to kill, deplete or otherwise effectively remove a target cell population from a heterogeneous collection of cells.
  • Blood from a mammal may be combined extracorporeally with the ligands, e.g. antibodies, cell- surface receptors or binding proteins thereof whereby the undesired cells are killed or otherwise removed from the blood for return to the mammal in accordance with standard techniques.
  • the invention relates to a method for delivering anti- angiogenic therapy (anti-VEGF therapy) to a site containing cells that express or overexpress EGFR, comprising administereing an effective amount of a ligand that has binding specificity for VEGF and for EGFR to a subject in need thereof.
  • anti-VEGF therapy anti-angiogenic therapy
  • the invention also relates to use of a ligand that has binding specificity for VEGF and for EGFR for delivering anti-angiogenic therapy (anti-VEGF therapy) to a site containing cells that express or overexpress EGFR.
  • the invention also relates to use of a ligand that has binding specificity for VEGF and for EGFR for the manufacture of a medicament for delivering anti-angiogenic therapy (anti-VEGF therapy) to a site containing cells that express or overexpress EGFR, or for inhibiting angiogenesis at a site containing cells that express of overexpress EGFR.
  • the invention relates to a method of treating cancer comprising administering to a subject in need thereof a therapeutically effective amount of a ligand, as described herein, that has binding specificity for VEGF and for EGFR.
  • the patient has an EGFR- expressing cancer, such as, bladder cancer, ovarian cancer, colorectal cancer, breast cancer, lung cancer ⁇ e.g., non-small cell lung carcinoma), gastric cancer, pancreatic cancer, prostate cancer, head and neck cancer, renal cancer and gall bladder cancer.
  • the invention relates to a method for treating cancer, comprising administering to a subject in need thereof a therapeutically effective amount of ligand, as described herein, ⁇ e.g., a ligand that has binding specificity for VEGF, a ligand that has binding specificity for EGFR, a ligand that has binding specificity for VEGF and EGFR) and an anti-neoplastic composition, wherein said anti-neoplastic composition comprises at least one chemotherapeutic agent selected from the group consisting of alkylating agents, antimetabolites, folic acid analogs, pyrimidine analogs, purine analogs and related inhibitors, vinca alkaloids, epipodopyyllotoxins, antibiotics, L-Asparaginase, topoisomerase inhibitor, interferons, platinum cooridnation complexes, anthracenedione substituted urea, methyl hydrazine derivatives, adrenocortical suppressant, a chemotherapeut
  • the chemotherapeutic agent is selected from the group consisting of cisplatin, dicarbazine, dactinomycin, mechlorethamine, streptozocin, cyclophosphamide, capecitabine, carmustine, lomustine, doxorubicin, daunorubicin, procarbazine, mitomycin, cytarabine, etoposide, methotrexate, 5-fluorouracil, vinblastine, vincristine, bleomycin, paclitaxel, docetaxel, doxetaxe, aldesleukin, asparaginase, busulfan, carboplatin, cladribine, dacarbazine, floxuridine, fludarabine, hydroxyurea, ifosfamide, interferon alpha, irinotecan, leuprolide, leucovorin, megestrol, melphalan, mercaptopurine, ox
  • the ligands of the invention can be assayed using any suitable in vitro or in vivo assay. For example, using the receptor binding assays or bioassays described herein.
  • This bioassay measures the ability of ligands ⁇ e.g., dAbs) to neutralise the
  • VEGF induced proliferation of HUVE human vascular endothelial cells.
  • HUVE cells plated in 96-well plates are incubated for 72 hours with pre-equilibrated VEGF and dAb protein. Cell number is then measured using a cell viability dye.
  • the assay is performed as follows. HUVE cells are trypsinized from a sub- confluent 175cm 2 flask. Medium is aspirated off, the cells are washed with 5ml trypsin and then incubated with 2ml trypsin at room temperature for 5min. The cells are gently dislodged from the base of the flask by knocking against your hand. 8ml of induction medium are then added to the flask, pipetting the cells to disperse any clumps. Viable cells are counted using trypan blue stain.
  • Cells are spun down and washed 2X in induction medium, spinning cells down and aspirating the medium after each wash. After the final aspiration the cells are diluted to 10 5 cells/ml (in induction medium) and plated at 100 ⁇ l per well into a 96 well plate (10,000 cells/well). The plate is incubated for >2h @ 37C to allow attachment of cells.
  • 60 ⁇ l dAb protein and 60 ⁇ l induction media containing 40ng/ml VEGF 165 (for a final concentration of 1 Ong/ml) is added to a v-bottom 96-well plate and sealed with film.
  • the dAb/VEGF mixture is then incubated at 37C for 0.5-1 hour.
  • the dAb/VEGF plate is removed from the incubator and lOO ⁇ l of solution added to each well of the HUVEC containing plate (final volume of 200 ⁇ l). This plate is then returned to the 37 0 C incubator for a period of at least 72 hours.
  • Control wells include the following: wells containing cells, but no VEGF; wells containing cells, a positive control neutralising anti-VEGF antibody and VEGF; and control wells containing cells and VEGF only.
  • Cell viability is assessed by adding 20 ⁇ l per well Celltiter96 reagent, and the plate is incubated at 37 0 C for 2-4h until a brown colour develops. The reaction is stopped by the addition of 20 ⁇ l per well of 10% (w/v) SDS. The absorbance is then read at 490nm using a Wallac microplate reader.
  • the absorbance of the no VEGF control wells is subtracted from all other values. Absorbance is proportional to cell number.
  • the control wells containing control anti-VEGF antibodies should also exhibit minimum cell proliferation.
  • the wells containing VEGF only should exhibit maximum cell proliferation.
  • VEGF is a specific mitogen for endothelial cells in vitro and a potent angiogenic factor in vivo, with high levels of the protein being expressed in various types of tumours. It is a 45kDa glycoprotein that is active as a homodimer.
  • isoforms have been described which occur through alternative mRNA splicing. Of these isoforms VEGF-121 and VEGF-165 appear to be the most abundant.
  • VEGF vascular endothelial cells
  • RTK receptor tyrosine kinases
  • FIt-I VEGF Rl
  • VEGF R2 receptor tyrosine kinases
  • This method describes a VEGF receptor binding assay for measuring the ability of ligands (e.g., dAbs) to prevent binding of VEGF- 165 to VEGF Receptor 2.
  • ligands e.g., dAbs
  • a recombinant human VEGF R2/Fc chimera was used in this assay, comprising the extracellular domain of human VEGF R2 fused to the Fc region of human IgG 1 . Briefly, the receptor was captured on an ELISA plate, then the plate was blocked to prevent nonspecific binding.
  • VEGF- 165 A mixture of VEGF- 165 and ligand was then added, the plate was washed and receptor bound VEGF-165 detected using a biotinylated anti-VEGF antibody and an Horse-radish Peroxidase (HRP) conjugated anti-biotin antibody. The plate was developed using a colorimetric substrate and the OD read at 450nm. If the dAb blocked VEGF binding to the receptor then no colour was detected.
  • HRP Horse-radish Peroxidase
  • the assay was performed as follows. A 96 well Nunc Maxisorp assay plate was coated overnight at 4°C with 100 ⁇ l per well of recombinant human VEGF R2/Fc (R&D Systems, Cat. No: 357-KD-050) at a concentration of 0.5 ⁇ g/ml in carbonate buffer. Wells were washed 3 times with 0.05%Tween/PBS and 3 times with PBS. 200 ⁇ l per well of 2% BSA in PBS was added to block the plate and the plate was incubated for a minimum of 1 hour at room temperature.
  • VEGF vascular endothelial growth factor
  • VEGF Ong/ml VEGF (diluent only); 3ng/ml VEGF (R&D Systems, Cat No: 293-VE-050); 3ng/ml VEGF with 0.1 ⁇ g/ml anti- VEGF neutralizing antibody (R&D Systems cat#MAB293).
  • the plate was washed (as above) and then lOO ⁇ l biotinylated anti-VEGF antibody (R&D Systems, Cat No: BAF293), 0.5 ⁇ g/ml in diluent, was added and incubated for 2 hours at room temperature.
  • biotinylated anti-VEGF antibody R&D Systems, Cat No: BAF293
  • the plate was washed (as above) ensuring any traces of Tween-20 were removed to limit background in the subsequent peroxidase assay and to help the prevention of bubbles in the assay plate wells that might give inaccurate OD readings.
  • hydrochloric acid (the blue colour turned yellow).
  • the OD, at 450nm, of the plate was read in a 96-well plate reader within 30 minutes of acid addition.
  • the OD450nm is proportional to the amount of bound streptavidin-HRP conjugate.
  • Protein L was added. Protein L cross links two dAb monomers.
  • 0ng/ml VEGF should give a low signal of ⁇ 0.15 OD
  • 3ng/ml VEGF should give a signal of >0.5 OD
  • 3ng/ml VEGF pre-incubated with 0.1 ⁇ g/ml neutralising antibody should give a signal ⁇ 0.2 OD.
  • VEGF Receptor 1 binding assay This assay measures the binding of VEGF-165 to VEGF Rl and the ability of ligands to block this interaction.
  • a recombinant human VEGF Rl/Fc chimera was used here, comprising the extracellular domain of human VEGF Rl fused to the Fc region of human IgG 1 .
  • the receptor was captured on an ELISA plate then the plate was blocked to prevent non-specific binding.
  • a mixture of VEGF-165 and ligand was then added, the plate was washed and receptor bound VEGF-165 detected using a biotinylated anti-VEGF antibody and an HRP conjugated anti- biotin antibody.
  • the plate was developed using a colorimetric substrate and the OD read at 450nm.
  • the assay was performed as follows. A 96-well Nunc Maxisorp assay plate was coated overnight at 4 0 C with lOO ⁇ l per well of recombinant human VEGF Rl/Fc (R&D Systems, Cat No: 321-FL-050) @ 0.1 ⁇ g/ml in carbonate buffer.
  • VEGF vascular endothelial growth factor
  • VEGF 500 ⁇ g/ml VEGF; and 500pg/ml VEGF with 1 ⁇ g/ml anti-VEGF antibody (R&D Systems cat#MAB293).
  • the plate was washed (as above) and then lOO ⁇ l biotinylated anti-VEGF antibody, 50ng/ml in diluent, was added and incubated for 1 hour at room
  • the plate was washed (as above), ensuring any traces of Tween-20 were removed to limit background in the subsequent peroxidase assay and to help the prevention of bubbles in the assay plate wells that might give inaccurate OD readings.
  • the OD, at 450nm, of the plate was read in a 96-well plate reader within 30 minutes of acid addition.
  • the OD450 nm is proportional to the amount of bound streptavidin-HRP conjugate.
  • 0 ng/ml VEGF should give a low signal of ⁇ 0.15 OD
  • 500pg/ml VEGF should give a signal of >0.8 OD
  • 500pg/ml VEGF pre-incubated with l ⁇ g/ml neutralising antibody should give a signal ⁇ 0.3 OD
  • the TARl 5-1 had a Kd of 50-80 nM when tested at various concentrations on a low density BIAcore chip.
  • Other VK dAbs were passed over the low density chip at one concentration (5OnM). Different dAbs showed different binding kinetics. Table 2
  • VH dAbs were passed over the low density VEGF chip on a BIAcore at one concentration (5OnM). Different dAbs showed different binding kinetics.
  • 25ul of ligand (e.g., dAb) were plated into a 96- well plate and then 25ul streptavidin-Alexa Fluor (lug/ml) (Molecular Probes) and 25 ul A431 cells (ATCC No. CRL-1555) (8xlO 5 /ml) were added. All reagents were prepared in PBS/1% BSA. The plate was incubated for 30 minutes at room temperature.
  • biotinylated EGF (Invitrogen) at 40ng/ml was added to each well, and the plate was incubated for three hours at room temperature. Fluoresecence was measured using the AB 8200 Cellular Detection System (Applied Biosystems).
  • Ligands e.g., dAbs
  • Ligands that inhibited the binding of biotinylated EGF to EGFR expressed on A431 cells resulted in lower fluorescence counts.
  • Wells without ligand provided a reference of the maximum fluorescence (i.e., biotinylated EGF binding) and wells without ligand or biotinylated EGF provided a reference of the
  • 5xlO 4 A431 cells (ATCC No. CRL-1555) were plated per well in RPMI- 1640 supplemented with 10% foetal calf serum. The plate was incubated overnight at 37°C/ 5% CO 2 to allow the cells to adhere, then the medium was replaced with RPMI-1640. The plate was incubated for 4 hours at 37°C / 5% CO 2 . The ligand (prepared in RPMI-1640) was added to the wells and the plate was incubated for 45 minutes at 37°C / 5% CO 2 . EGF (Invitrogen) was added to the wells to give a final concentration of 100ng/ml and the plate was incubated for 10 minutes at room temperature.
  • EGF Invitrogen
  • the wells were washed twice with ice cold PBS.
  • Cold lysis buffer (1 % NP-40, 2OmM Tris, 137mM NaCl, 10% glycerol, 2mM EDTA, ImM sodium ortho vanadate, lOug/ml aprotinin, lOug/ml leupeptin) was added and the plate was incubated on ice for 10 minutes.
  • the supernatants were transfered to an ELISA plate which had been coated overnight with anti-EGFR antibody (R&D Systems) at lug/ml in carbonate buffer.
  • the ELISA plate was incubated for 2 hours at room temperature.
  • the plate was washed three times with PBS/0.05% Tween 20.
  • Anti-phosphotyrosine antibody conjugated to horse-radish peroxidase (Upstate Biotechnology) at lug/ml was added and the plate was incubated for 1 hour at room temperature.
  • the plate was washed three times with PBS/Tween and three times with PBS.
  • the reaction was developed with SureBlue TMB 1 -component microwell peroxidase substrate (KPL) and the reaction was stopped with IM HCl after 25 minutes.
  • the absorbance was read using a Wallac plate reader.
  • Example 3 IgG-like fo ⁇ nats that have binding specificity for VEGF and EGFR.
  • the pBudCE4.1 backbone (Invitrogen) was used for cloning
  • immunoglobulin constant regions such as the IgGl heavy chain constant region and light chain kappa constant region (see FIG. 16 for overview).
  • An Ig Kappa chain leader was used to facilitate secretion of the expressed protein.
  • Ig constant regions human IgGl and CK were produced by GeneArt (Germany).
  • the heavy chain constant region and signal peptide were cloned into pBudCE4.1 as a Hind III/Bglll fragment into the Hindlll/BamH I restriction sites.
  • the light chain constant region and signal peptide were cloned into pBudCE4.1 as a Notl/MluI fragment. Cloning of dAb in IgG vectors and production of IgG-like format
  • VK dAb (specific to VEGF or EGFR) was cloned into IgG vector as a Sall/BsiWI fragment.
  • VH dAb (specific to VEGF or EGFR was cloned into IgG vector as a BamHI/XhoI fragment.
  • the plasmid was then transfected into HEK293T cells (ATCC CRL-11268) and IgG was expressed transiently for five days.
  • the IgG produced was purified using streamline Protein A.
  • Purified IgG was checked on a reducing and non-reducing SDS gel and bands of expected size were observed.
  • IgG-like formats that have binding specificity for VEGF and EGFR.
  • the IgG-like formats were prepared by producing constructs that encoded an IgG heavy chain wherein VH is a dAb, and a Kappa light chain wherein VK is a dAb.
  • the IgG-like formats that were prepared are shown in Table 4, and the results obtained for some of the IgG-like formats in assays are presented in Table 5. (Dummy VH and Dummy VK are germ line sequences that do not bind VEGF or EGFR).
  • fusion polypeptides that contained an anti- VEGFR dAb and an anti-EGFR dAb in a single polypeptide chain.
  • Some of the fusion polypeptides also included an antibody Fc region (-CH2-CH3 of human IgGl).
  • the positions of the foregoing fusions are listed as they appear in the fusion proteins from amino termin
  • DNA encoding dAbs was PCR amplified and cloned into expression vectors using standard methods.
  • Inline fusion polypeptides were produced by expressing the expression vectors in Pichia (fusion that did not contain an Fc region) or in HEK 293T cells (Fc region containing fusions). Inline fusions were batch bound and affinity purified on streamline protein A and streamline protein L resins for HEK 293T cells (Fc-tagged) and Pichia expressed constructs respectively.
  • the structure of the fusion proteins can be appreciated by reading the table from left to right.
  • the first fusion protein presented in Table 6 has the structure, from amino terminus to carboxy terminus, DOM15-10— Linker 1—
  • New linkers were designed that contain fewer Lys and Arg residues, which are cleavage points for trypsin and are abundant in the natural linker. Fusions that contained the engineered linkers (LVTVSSAST (SEQ ID NO:707)) or
  • Vk dAbs expressed on the light chain of IgG-like formats to reduce protease sensitivity.
  • the "natural linker” was GQGTKVEIKRTVAAPS (SEQ ID NO:709 which contains the carboxy- terminal amino acids of Vk and amino-terminal amino acids of Ck).
  • Variant linkers 1-3 were designed with amino acid replacements that replaced some or all of the positively charged residues in the natural linker with the most conservative substitutions that are not positively charged at physiological pH. It is likely that the arginine residue in the natural linker is less amenable to alteration due to ionic interactions it forms within the CL domain.
  • Variant linker 1 substitutes both lysines in the natural linker with asparagines.
  • Variant linker 1 and variant linker 2 (GQGTNVEINQTVAAPS (SEQ ID NO:711)), which additionally changes the arginine in the natural linker to glutamine, introduce an N-glycosylation site (NxT) into the linker.
  • NxT N-glycosylation site
  • GQGTLVTVSSTVAAPS (SEQ ID NO:713) replaces the six C-terminal amino acids of the Vk domain with the corresponding residues from a VH domain, and is devoid of positive charges.
  • IgG-like formats that contain variant linkers 1-4 revealed that IgG-like formats that contained engineered variant linkers were more protease resistant than an IgG-like format that contained the natural linker.
  • Example 6 DOM 16 dAb-anti-serum albumin d
  • DOMl 6 dAb- anti-serum albumin dAb fusions were designed and expressed as a fusion of an anti-EGFR dAb to an anti-serum albumin dAb (a D0M7 dAb).
  • the portions of the fusions are listed in Table 7 as they appear in the fusion proteins, from amino terminus to carboxy terminus. Accordingly, the structure of the fusion proteins can be appreciated by reading the table from left to right.
  • the first fusion protein presented in Table 7 has the structure, from amino terminus to carboxy terminus, DOM16-39-618(S12P)— Linker— DOM7h-14.
  • DOMl 6-39-618 contains a Serine to Proline mutation at position 12, which stops binding to protein L and prevents light chain aggregation.
  • the iD0M7 dAbs are mutated so as to abrogate binding to albumin, and thus are inactivated.
  • DOMl 6-39-200 was used as a reference in this study.
  • DOM16-39-200, and other dAbs designated DOM-16-39-X are affinity matured variants of DOM16-39. Accordingly, all dAbs in the D0M-16- 39 series will have substantially the same epitopic specificity, because affinity maturation produces dAbs that bind with higher affinity, but does not alter the specificity of the dAb.
  • DOM16-72, DOM16-79 and DOM16-112 competed with DOMl 6-39-200 for binding to EGFR, indicating that these dAbs bind overlapping epitopes.
  • DOMl 6-32, DOMl 6-52 and DOMl 6-80 were shown to bind to a different epitope.
  • ERBITUX cetuximab; Imclone Systems
  • cetuximab and EGF bind overlapping epitopes on EGFR.

Landscapes

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

Abstract

Disclosed are ligands that have binding specificity for vascular endothelial growth factor (VEGF), for epidermal growth factor receptor (EGFR), or for VEGF and EGFR. Also disclosed are methods of using these ligands. In particular, the use of these ligands for cancer therapy is described.

Description

LIGANDS THAT HAVE BINDING SPECIFICITY FOR EGFR AND/OR VEGF AND METHODS OF USE THEREFOR
RELATED APPLICATION
This application claims the benefit of U.S. Provisional Application No. 60/742,992, filed on December 6, 2005, and the benefit of U.S. Provisional
Application No. 60/758,355, filed on January 11, 2006. The entire teachings of the above applications are incorporated herein by reference.
BACKGROUND OF THE INVENTION
Cancer is a leading cause of mortality and morbidity. Approaches to treating cancer include surgical intervention to remove tumors and chemotherapy. These approaches can successfully cure some patients. However, even patients that appear to have been cured often suffer a recurrence of the cancer necessitating further therapy. Chemotherapeutic agents generally are nonselective agents that are toxic to cells, such as proliferating cells. Accordingly, such agents may effectively kill cancer cells but also kill healthy cells producing several deleterious side effects.
Certain cancer cells express or overexpress certain cellular components such as cell surface proteins, or express different cellular components when compared to normal cells. One approach to address the short comings of surgical and
chemotherapeutic approaches to cancer therapy and diagnosis involves targeting cancer cells, for example using antibodies or antibody fragments that bind to proteins that are expressed or overexpressed on cancerous cells. A number of such target proteins have been identified. Among such proteins is the epidermal growth factor receptor (EGFR).
EGFR is a member of the ErbBl family and transduces signals that lead to cellular proliferation and survival, and the elaboration of growth and angiogenic factors upon binding epidermal growth factor (EGF) and or transforming growth factor alpha (TGF alpha). Accordingly, EGFR has been demonstrated to be involved in tumor growth, metastasis and angiogenesis. Further, many cancers express EGFR, such as bladder cancer, ovarian cancer, colorectal cancer, breast cancer, lung cancer (e.g., non-small cell lung carcinoma), gastric cancer, pancreatic cancer, prostate cancer, head and neck cancer, renal cancer and gall bladder cancer. ERBITUX (cetuximab; Imclone Systems Inc) is a chimeric mouse/human antibody that binds human EGFR that has been approved for treating certain EGFR- expressing cancers in combination with irinotecan.
An important pathophysiological process that facilitates tumor formation, metastasis and recurrence is tumor angiogenesis. This process is mediated by the elaboration of angiogenic factors by the tumor, such as vascular endothelial growth factor (VEGF), which induce the formation of blood vessels that deliver nutrients to the tumor. Accordingly, another approach to treating certain cancers is to inhibit tumor angiogenesis mediated by VEGF, thereby starving the tumor. AVASTIN (bevacizumab; Genetech, Inc.) is a humanized antibody that binds human VEGF that has been approved for treating colorectal cancer. An antibody referred to as antibody 2C3 (ATCC Accession No. PTA 1595) is reported to bind VEGF and inhibit binding of VEGF to epidermal growth factor receptor 2.
Targeting EGFR or VEGF with currently available therapeutics is not effective in all patients, or for all cancers (e.g., EGFR-expressing cancers). Thus, a need exists for improved agents for treating cancer and other pathological conditions.
SUMMARY OF THE INVENTION
The invention relates to ligands that have binding specificity for VEGF (e.g., human VEGF), ligands that have binding specificity for EGFR (e.g., human EGFR), and to ligands that have binding specificity for VEGF and EGFR (e.g., human VEGF and human EGFR). For example, the ligand can comprise a polypeptide domain having a binding site with binding specificity for VEGF, a polypeptide domain having a binding site with binding specificity EGFR, or comprise a polypeptide domain having a binding site with binding specificity for VEGF and a polypeptide domain having a binding site with binding specificity EGFR.
In one aspect, the invention relates to a ligand that has binding specificity for
VEGF and for EGFR. Such ligands comprise at least one protein moiety that has a binding site with binding specificity for VEGF and at least one protein moiety that has a binding site with binding specificity for EGFR. The protein moiety that has a binding site with binding specificity for VEGF and the protein moiety that has a binding site with binding specificity for EGFR can each be any suitable binding moiety. The protein moieties can be a peptide moiety, polypeptide moiety or protein moiety. For example, the protein moieties can be provided by an antibody fragment that has a binding site with binding specificity for VEGF or EGFR, such as an immunoglobulin single variable domain that has binding specificity for VEGF or EGFR.
The ligand can comprise a protein moiety that has a binding site with binding specificity for VEGF that competes for binding to VEGF with AVASTIN
(bevacizumab; Genentech, Inc.) and/or antibody 2C3 (ATCC Accession No. PTA 1595). The ligand can comprise a protein moiety that has a binding site with binding specificity for EGFR that competes for binding to EGFR with ERBITUX (cetuximab; Imclone Systems, Inc.) and/or VECTIBIX (panitumumab; Amgen, Inc.). In some embodiments, the ligand comprises a protein moiety that has a binding site with binding specificity for VEGF that competes for binding to VEGF with bevacizumab and/or antibody 2C3 (ATCC Accession No. PTA 1595), and further comprises a protein moiety that has a binding site with binding specificity for EGFR that competes for binding to EGFR with cetuximab.
In some embodiments, the ligand comprises a protein moiety that has a binding site with binding specificity for VEGF {e.g., an immunoglobulin single variable domain) that competes for binding to VEGF with an anti-VEGF domain antibody (dAb) selected from the group consisting of TAR15-1 (SEQ ID NO:100), TARl 5-3 (SEQ ID NO: 101), TARl 5-4 (SEQ ID NO: 102), TARl 5-9 (SEQ ID NO:103), TAR15-10 (SEQ ID NO: 104), TAR15-11 (SEQ ID NO:105), TAR15-12 (SEQ ID NO:106), TAR15-13 (SEQ ID NO:107), TAR15-14 (SEQ ID NO:108), TAR15-15 (SEQ ID NO:109), TAR15-16 (SEQ ID NO:110), TAR15-17 (SEQ ID NO:111), TAR15-18 (SEQ ID NO:112), TAR15-19 (SEQ ID NO:113), TAR15-20 (SEQ ID NO: 114), TAR 15-22 (SEQ ID NO: 115), TARl 5-5 (SEQ ID NO: 116), TARl 5-6 (SEQ ID NO: 117), TARl 5-7 (SEQ ID NO: 118), TARl 5-8 (SEQ ID
NO:119), TARl 5-23 (SEQ ID NO: 120), TARl 5-24 (SEQ ID NO: 121), TARl 5-25 (SEQ ID NO: 122), TARl 5-26 (SEQ ID NO: 123), TARl 5-27 (SEQ ID NO: 124), - A -
TAR15-29 (SEQ ID NO.125), TAR15-30 (SEQ ID NO:126), TAR15-6-500 (SEQ ID NO:127), TAR15-6-501 (SEQ ID NO:128), TAR15-6-502 (SEQ ID NO:129), TAR15-6-503 (SEQ ID NO:130), TAR15-6-504 (SEQ ID NO:131), TAR15-6-505 (SEQ ID NO: 132), TARl 5-6-506 (SEQ ID NO: 133), TARl 5-6-507 (SEQ ID NO:134), TAR15-6-508 (SEQ ID NO:135), TAR15-6-509 (SEQ ID NO:136), TAR15-6-510 (SEQ ID NO:137), TAR15-8-500 (SEQ ID NO:138), TAR15-8-501 (SEQ ID NO:139), TAR15-8-502 (SEQ ID NO:140), TAR15-8-503 (SEQ ID NO:141), TAR15-8-505 (SEQ ID NO:142), TAR15-8-506 (SEQ ID NO:143), TAR15-8-507 (SEQ ID NO:144), TAR15-8-508 (SEQ ID NO:145), TAR15-8-509 (SEQ ID NO:146), TAR15-8-510 (SEQ ID NO:147), TARl 5-8-511 (SEQ ID NO:148), TAR15-26-500 (SEQ ID NO:149), TAR15-26-501 (SEQ ID NO:150), TAR15-26-502 (SEQ ID NO:151), TAR15-26-503 (SEQ ID NO: 152), TAR15-26- 504 (SEQ ID NO: 153), TARl 5-26-505 (SEQ ID NO: 154), TARl 5-26-506 (SEQ ID NO: 155), TARl 5-26-507 (SEQ ID NO: 156), TARl 5-26-508 (SEQ ID NO: 157), TARl 5-26-509 (SEQ ID NO: 158), TARl 5-26-510 (SEQ ID NO: 159), TARl 5-26- 511 (SEQ ID NO:160), TAR15-26-512 (SEQ ID NO:161), TAR15-26-513 (SEQ ID NO:162), TAR15-26-514 (SEQ ID NO:163), TAR15-26-515 (SEQ ID NO: 164), TARl 5-26-516 (SEQ ID NO: 165), TARl 5-26-517 (SEQ ID NO: 166), TARl 5-26- 518 (SEQ ID NO: 167), TARl 5-26-519 (SEQ ID NO: 168), TARl 5-26-520 (SEQ ID NO:169), TAR15-26-521 (SEQ ID NO: 170), TAR15-26-522 (SEQ ID NO:171), TAR15-26-523 (SEQ ID NO:172), TAR15-26-524 (SEQ ID NO:173), TAR15-26- 525 (SEQ ID NO: 174), TARl 5-26-526 (SEQ ID NO: 175), TARl 5-26-527 (SEQ ID NO: 176), TARl 5-26-528 (SEQ ID NO: 177), TARl 5-26-529 (SEQ ID NO: 178), TAR15-26-530 (SEQ ID NO: 179), TAR15-26-531 (SEQ ID NO:180), TAR15-26- 532 (SEQ ID NO: 181), TARl 5-26-533 (SEQ ID NO: 182), TARl 5-26-534 (SEQ ID NO: 183), TARl 5-26-535 (SEQ ID NO: 184), TARl 5-26-536 (SEQ ID NO: 185), TARl 5-26-537 (SEQ ID NO: 186), TARl 5-26-538 (SEQ ID NO: 187), TARl 5-26- 539 (SEQ ID NO: 188), TARl 5-26-540 (SEQ ID NO:189), TARl 5-26-541 (SEQ ID NO:190), TAR15-26-542 (SEQ ID NO:191), TAR15-26-543 (SEQ ID NO:192), TARl 5-26-544 (SEQ ID NO: 193), TARl 5-26-545 (SEQ ID NO: 194), TARl 5-26- 546 (SEQ ID NO: 195), TARl 5-26-547 (SEQ ID NO: 196), TARl 5-26-548 (SEQ ID NO:197), TAR15-26-549 (SEQ ID NO:198), TAR15-26-550 (SEQ ID NO:539), and TARl 5-26-551 (SEQ ID NO:540).
In some embodiments, the ligand comprises a protein moiety that has a binding site with binding specificity for VEGF (e.g., an immunoglobulin single variable domain) that competes for binding to VEGF with TARl 5-26-555 (SEQ ID NO:704).
Additionally, or in other embodiments, the ligand can comprise a protein moiety that has a binding site with binding specificity for EGFR (e.g., an immunoglobulin single variable domain) that competes for binding to EGFR with an anti-EGFR domain antibody (dAb) selected from the group consisting of DOM 16- 17 (SEQ ID NO:325), DOM16-18 (SEQ ID NO:326), DOM16-19 (SEQ ID
NO:327), DOM16-20 (SEQ ID NO:328), DOM16-21 (SEQ ID NO:329), D0M16- 22 (SEQ ID NO:330), DOMl 6-23 (SEQ ID NO:331), DOMl 6-24 (SEQ ID
NO:332), DOM16-25 (SEQ ID NO:333), DOM16-26 (SEQ ID NO:334), D0M16- 27 (SEQ ID NO:335), DOM16-28 (SEQ ID NO:336), DOM16-29 (SEQ ID
NO:337), DOM16-30 (SEQ ID NO:338), DOM16-31 (SEQ ID NO:339), D0M16- 32 (SEQ ID NO:340), DOM16-33 (SEQ ID NO:341), DOM16-35 (SEQ ID
NO:342), DOM16-37 (SEQ ID NO:343), DOM16-38 (SEQ ID NO:344), D0M16- 39 (SEQ ID NO:345), DOMl 6-40 (SEQ ID NO:346), DOMl 6-41 (SEQ ID
NO:347), DOMl 6-42 (SEQ ID NO:348), DOMl 6-43 (SEQ ID NO:349), DOMl 6- 44 (SEQ ID NO:350), DOM16-45 (SEQ ID NO:351), DOM16-46 (SEQ ID
NO:352), DOMl 6-47 (SEQ ID NO:353), DOMl 6-48 (SEQ ID NO:354), DOMl 6- 49 (SEQ ID NO:355), DOM16-50 (SEQ ID NO:356), DOM16-59 (SEQ ID
NO:357), DOM16-60 (SEQ ID NO:358), DOM16-61 (SEQ ID NO:359), D0M16- 62 (SEQ ID NO:360), DOM16-63 (SEQ ID NO:361), DOM16-64 (SEQ ID
NO:362), DOMl 6-65 (SEQ ID NO:363), DOMl 6-66 (SEQ ID NO:364), DOMl 6- 67 (SEQ ID NO:365), DOMl 6-68 (SEQ ID NO:366), DOMl 6-69 (SEQ ID
NO:367), DOM16-70 (SEQ ID NO:368), DOM16-71 (SEQ ID NO:369), D0M16- 72 (SEQ ID NO:370), DOM16-73 (SEQ ID NO:371), DOM16-74 (SEQ ID
NO:372), DOMl 6-75 (SEQ ID NO:373), DOMl 6-76 (SEQ ID NO:374), DOMl 6- 77 (SEQ ID NO:375), DOMl 6-78 (SEQ ID NO:376), DOMl 6-79 (SEQ ID
NO:377), DOM16-80 (SEQ ID NO:378), DOM16-81 (SEQ ID NO:379), D0M16- 82 (SEQ ID NO:380), DOM16-83 (SEQ ID NO:381), DOM16-84 (SEQ ID
NO:382), DOM16-85 (SEQ ID NO:383), DOM16-87 (SEQ ID NO:384), DOM16- 88 (SEQ ID NO:385), DOM16-89 (SEQ ID NO:386), DOM16-90 (SEQ ID
NO:387), DOM16-91 (SEQ ID NO:388), DOM16-92 (SEQ ID NO:389), DOM16- 94 (SEQ ID NO:390), DOMl 6-95 (SEQ ID NO:391), DOMl 6-96 (SEQ ID
NO:392), DOMl 6-97 (SEQ ID NO:393), DOMl 6-98 (SEQ ID NO:394), DOMl 6-
99 (SEQ ID NO:395), DOM16-100 (SEQ ID NO:396), DOM16-101 (SEQ ID NO:397), DOM16-102 (SEQ ID NO:398), DOM16-103 (SEQ ID NO:399), DOM16-104 (SEQ ID NO:400), DOM16-105 (SEQ ID NO:401), DOM16-106 (SEQ ID NO:402), DOMl 6-107 (SEQ ID NO:403), DOMl 6-108 (SEQ ID
NO:404), DOM16-109 (SEQ ID NO:405), DOM16-110 (SEQ ID NO:406), DOM16-111 (SEQ ID NO:407), DOM16-112 (SEQ ID NO:408), DOM16-113 (SEQ ID NO:409), DOM16-114 (SEQ ID NO:410), DOM16-115 (SEQ ID
NO:411), DOM16-116 (SEQ ID NO:412), DOM16-117 (SEQ ID NO:413), DOMl 6-118 (SEQ ID NO:414), DOMl 6-119 (SEQ ID NO:415), DOMl 6-39-6 (SEQ ID NO:416), DOM16-39-8 (SEQ ID NO:417), DOM16-39-34 (SEQ ID NO:418), DOM16-39-48 (SEQ ID NO:419), DOM16-39-87 (SEQ ID NO:420), DOM16-39-90 (SEQ ID NO:421), DOM16-39-96 (SEQ ID NO:422), DOM16-39-
100 (SEQ ID NO:423), DOM16-39-101 (SEQ ID NO:424), DOM16-39-102 (SEQ ID NO:425), DOM16-39-103 (SEQ ID NO:426), DOM16-39-104 (SEQ ID
NO:427), DOM16-39-105 (SEQ ID NO:428), DOM16-39-106 (SEQ ID NO:429), DOM16-39-107 (SEQ ID NO:430), DOM16-39-108 (SEQ ID NO:431), DOM16- 39-109 (SEQ ID NO:432), DOM16-39-110 (SEQ ID NO:433), DOM16-39-111 (SEQ ID NO:434), DOM16-39-112 (SEQ ID NO:435), DOM16-39-113 (SEQ ID NO:436), DOM16-39-114 (SEQ ID NO:437), DOM16-39-115 (SEQ ID NO:438), DOM16-39-116 (SEQ ID NO:439), DOM16-39-117 (SEQ ID NO:440), DOM16- 39-200 (SEQ ID NO:441), DOM16-39-201 (SEQ ID NO:442), DOM16-39-202 (SEQ ID NO:443), DOMl 6-39-203 (SEQ ID NO:444), DOMl 6-39-204 (SEQ ID NO:445), DOMl 6-39-205 (SEQ ID NO:446), DOMl 6-39-206 (SEQ ID NO:447), DOM16-39-207 (SEQ ID NO:448), DOM16-39-209 (SEQ ID NO:449), DOM16-52 (SEQ ID NO:450), NBl (SEQ ID NO:451), NB2 (SEQ ID NO:452), NB3 (SEQ ID NO:453), NB4 (SEQ ID NO:454), NB5 (SEQ ID NO:455), NB6 (SEQ ID NO:456), NB7 (SEQ ID NO:457), NB8 (SEQ ID NO:458), NB9 (SEQ ID NO:459), NBlO (SEQ ID NO:460), NBI l (SEQ ID NO:461), NB12 (SEQ ID NO:462), NB13 (SEQ ID NO:463), NB14 (SEQ ID NO:464), NB15 (SEQ ID NO:465), NB16 (SEQ ID NO:466), NB17 (SEQ ID NO:467), NB18 (SEQ ID NO:468), NB19 (SEQ ID NO:469), NB20 (SEQ ID NO:470), NB21 (SEQ ID NO:471), and NB22 (SEQ ID NO:472).
In particular embodiments, the ligand has binding specificity for VEGF and for EGFR and comprises a protein moiety that has a binding site with binding specificity for VEGF that competes for binding to VEGF with an anti-VEGF domain antibody (dAb) selected from the group consisting of TARl 5-6 (SEQ ID NO: 117), TAR15-8 (SEQ ID NO:119), and TAR15-26 (SEQ ID NO:123), and further comprises a protein moiety that has a binding site with binding specificity for EGFR that competes for binding to EGFR with an anti-EGFR domain antibody (dAb) selected from the group consisting of DOM16-39 (SEQ ID NO:345), DOM16-39-87 (SEQ ID NO:420), DOM16-39-100 (SEQ ID NO:423), DOM16-39-107 (SEQ ID NO:430), DOM16-39-109 (SEQ ID NO:432), DOM16-39-115 (SEQ ID NO:438), and DOMl 6-39-200 (SEQ ID NO:441).
In particular embodiments, the ligand has binding specificity for VEGF and for EGFR and comprises a protein moiety that has a binding site with binding specificity for VEGF that competes for binding to VEGF with an anti-VEGF domain antibody (dAb) selected from the group consisting of TARl 5-6 (SEQ ID NO:117), TAR15-8 (SEQ ID NO:119), and TAR15-26 (SEQ ID NO:123), and further comprises a protein moiety that has a binding site with binding specificity for EGFR that competes for binding to EGFR with an anti-EGFR domain antibody (dAb) selected from the group consisting of DOMl 6-39-521 (SEQ ID NO:577), D0M16- 39-541 (SEQ ID NO:585), DOM16-39-542 (SEQ ID NO:586), DOM16-39-551 (SEQ ID NO:591), DOM16-39-601 (SEQ ID NO:608), DOM16-39-604 (SEQ ID NO:611), DOMl 6-39-618 (SEQ ID NO:621), and DOMl 6-39-619 (SEQ ID NO:622).
In more particular embodiments, the ligand has binding specificity for VEGF and for EGFR and comprises at least one immunoglobulin single variable domain with binding specificity for VEGF and at least one immunoglobulin single variable domain with binding specificity for EGFR, wherein an immunoglobulin single variable domain with binding specificity for VEGF competes for binding to VEGF with an anti-VEGF domain antibody (dAb) selected from the group consisting of TAR15-1 (SEQ ID NO:100), TAR15-3 (SEQ ID NOiIOl), TAR15-4 (SEQ ID NO: 102), TARl 5-9 (SEQ ID NO: 103), TARl 5-10 (SEQ ID NO: 104), TARl 5-11 (SEQ ID NO: 105), TARl 5-12 (SEQ ID NO: 106), TARl 5-13 (SEQ ID NO: 107), TAR15-14 (SEQ ID NO:108), TAR15-15 (SEQ ID NO: 109), TAR15-16 (SEQ ID NO-.110), TAR15-17 (SEQ ID NO:111), TAR15-18 (SEQ ID NO:112), TAR15-19 (SEQ ID NO:113), TAR15-20 (SEQ ID NO:114), TAR 15-22 (SEQ ID NO:115), TARl 5-5 (SEQ ID NO: 116), TARl 5-6 (SEQ ID NO: 117), TARl 5-7 (SEQ ID
NO:118), TAR15-8 (SEQ ID NO:119), TAR15-23 (SEQ ID NO: 120), TAR15-24 (SEQ ID NO:121), TAR15-25 (SEQ ID NO:122), TAR15-26 (SEQ ID NO:123), TAR15-27 (SEQ ID NO: 124), TAR15-29 (SEQ ID NO:125), TAR15-30 (SEQ ID NO:126), TAR15-6-500 (SEQ ID NO: 127), TAR15-6-501 (SEQ ID NO:128), TAR15-6-502 (SEQ ID NO:129), TAR15-6-503 (SEQ ID NO:130), TAR15-6-504 (SEQ ID NO:131), TAR15-6-505 (SEQ ID NO:132), TAR15-6-506 (SEQ ID NO: 133), TARl 5-6-507 (SEQ ID NO: 134), TARl 5-6-508 (SEQ ID NO: 135), TAR15-6-509 (SEQ ID NO:136), TAR15-6-510 (SEQ ID NO:137), TAR15-8-500 (SEQ ID NO:138), TAR15-8-501 (SEQ ID NO:139), TAR15-8-502 (SEQ ID NO:140), TAR15-8-503 (SEQ ID NO:141), TAR15-8-505 (SEQ ID NO: 142),
TARl 5-8-506 (SEQ ID NO: 143), TARl 5-8-507 (SEQ ID NO: 144), TARl 5-8-508 (SEQ ID NO:145), TAR15-8-509 (SEQ ID NO:146), TAR15-8-510 (SEQ ID NO:147), TARl 5-8-511 (SEQ ID NO:148), TAR15-26-500 (SEQ ID NO: 149), TAR15-26-501 (SEQ ID NO:150), TAR15-26-502 (SEQ ID NO:151), TAR15-26- 503 (SEQ ID NO: 152), TARl 5-26-504 (SEQ ID NO: 153), TARl 5-26-505 (SEQ ID NO: 154), TARl 5-26-506 (SEQ ID NO: 155), TARl 5-26-507 (SEQ ID NO: 156), TARl 5-26-508 (SEQ ID NO: 157), TARl 5-26-509 (SEQ ID NO: 158), TARl 5-26- 510 (SEQ ID NO: 159), TARl 5-26-511 (SEQ ID NO: 160), TARl 5-26-512 (SEQ ID NO:161), TAR15-26-513 (SEQ ID NO: 162), TAR15-26-514 (SEQ ID NO:163), TARl 5-26-515 (SEQ ID NO: 164), TARl 5-26-516 (SEQ ID NO: 165), TARl 5-26- 517 (SEQ ID NO: 166), TARl 5-26-518 (SEQ ID NO: 167), TARl 5-26-519 (SEQ ID NO:168), TAR15-26-520 (SEQ ID NO:169), TAR15-26-521 (SEQ ID NO:170), TAR15-26-522 (SEQ ID NO:171), TAR15-26-523 (SEQ ID NO.172), TAR15-26- 524 (SEQ ID NO: 173), TARl 5-26-525 (SEQ ID NO: 174), TARl 5-26-526 (SEQ ID NO:175), TAR15-26-527 (SEQ ID NO:176), TAR15-26-528 (SEQ ID NO:177), TARl 5-26-529 (SEQ ID NO: 178), TARl 5-26-530 (SEQ ID NO: 179), TARl 5-26- 531 (SEQ ID NO: 180), TARl 5-26-532 (SEQ ID NO: 181), TARl 5-26-533 (SEQ ID NO: 182), TARl 5-26-534 (SEQ ID NO: 183), TARl 5-26-535 (SEQ ID NO: 184), TARl 5-26-536 (SEQ ID NO: 185), TARl 5-26-537 (SEQ ID NO: 186), TARl 5-26- 538 (SEQ ID NO:187), TAR15-26-539 (SEQ ID NO:188), TAR15-26-540 (SEQ ID NO:189), TAR15-26-541 (SEQ ID NO:190), TAR15-26-542 (SEQ ID NO:191), TARl 5-26-543 (SEQ ID NO: 192), TARl 5-26-544 (SEQ ID NO: 193), TARl 5-26- 545 (SEQ ID NO: 194), TARl 5-26-546 (SEQ ID NO: 195), TARl 5-26-547 (SEQ ID NO: 196), TARl 5-26-548 (SEQ ID NO: 197), and TARl 5-26-549 (SEQ ID NO: 198), TARl 5-26-550 (SEQ ID NO:539), and TARl 5-26-551 (SEQ ID NO:540).
For example, the immunoglobulin single variable domain with binding specificity for VEGF can comprise an amino acid sequence that has at least about 85% amino acid sequence identity with the amino acid sequence of a dAb selected from the group consisting of TARl 5-1 (SEQ ID NO: 100), TARl 5-3 (SEQ ID NO:101), TAR15-4 (SEQ ID NO:102), TAR15-9 (SEQ ID NO:103), TAR15-10 (SEQ ID NO:104), TAR15-11 (SEQ ID NO:105), TAR15-12 (SEQ ID NO:106), TAR15-13 (SEQ ID NO:107), TAR15-14 (SEQ ID NO:108), TAR15-15 (SEQ ID NO:109), TAR15-16 (SEQ ID NO:110), TAR15-17 (SEQ ID NO:111), TAR15-18 (SEQ ID NO:112), TAR15-19 (SEQ ID NO:113), TAR15-20 (SEQ ID NO:114), TAR 15-22 (SEQ ID NO: 115), TARl 5-5 (SEQ ID NO: 116), TARl 5-6 (SEQ ID NO: 117), TARl 5-7 (SEQ ID NO: 118), TARl 5-8 (SEQ ID NO: 119), TARl 5-23 (SEQ ID NO:120), TAR15-24 (SEQ ID NO:121), TAR15-25 (SEQ ID NO:122), TAR15-26 (SEQ ID NO:123), TAR15-27 (SEQ ID NO:124), TAR15-29 (SEQ ID NO:125), TAR15-30 (SEQ ID NO:126), TAR15-6-500 (SEQ ID NO:127), TAR15- 6-501 (SEQ ID NO:128), TAR15-6-502 (SEQ ID NO:129), TAR15-6-503 (SEQ ID NO:130), TAR15-6-504 (SEQ ID NO:131), TAR15-6-505 (SEQ ID NO:132), TARl 5-6-506 (SEQ ID NO: 133), TARl 5-6-507 (SEQ ID NO: 134), TARl 5-6-508 (SEQ ID NO:135), TAR15-6-509 (SEQ ID NO:136), TAR15-6-510 (SEQ ID NO:137), TAR15-8-500 (SEQ ID NO:138), TAR15-8-501 (SEQ ID NO:139), TAR15-8-502 (SEQ ID NO: 140), TAR15-8-503 (SEQ ID NO:141), TAR15-8-505 (SEQ ID NO: 142), TARl 5-8-506 (SEQ ID NO: 143), TARl 5-8-507 (SEQ ID NO: 144), TAR15-8-508 (SEQ ID NO:145), TAR15-8-509 (SEQ ID NO:146), TARl 5-8-510 (SEQ ID NO: 147), TARl 5-8-511 (SEQ ID NO: 148), TARl 5-26-500 (SEQ ID NO: 149), TARl 5-26-501 (SEQ ID NO: 150), TARl 5-26-502 (SEQ ID NO: 151), TARl 5-26-503 (SEQ ID NO: 152), TARl 5-26-504 (SEQ ID NO: 153), TARl 5-26-505 (SEQ ID NO: 154), TARl 5-26-506 (SEQ ID NO: 155), TARl 5-26- 507 (SEQ ID NO: 156), TARl 5-26-508 (SEQ ID NO: 157), TARl 5-26-509 (SEQ ID NO:158), TAR15-26-510 (SEQ ID NO:159), TAR15-26-511 (SEQ ID NO: 160), TAR15-26-512 (SEQ ID NO:161), TAR15-26-513 (SEQ ID NO:162), TAR15-26- 514 (SEQ ID NO:163), TAR15-26-515 (SEQ ID NO:164), TAR15-26-516 (SEQ ID NO: 165), TARl 5-26-517 (SEQ ID NO: 166), TARl 5-26-518 (SEQ ID NO: 167), TARl 5-26-519 (SEQ ID NO: 168), TARl 5-26-520 (SEQ ID NO: 169), TARl 5-26- 521 (SEQ ID NO:170), TAR15-26-522 (SEQ ID NO:171), TAR15-26-523 (SEQ ID NO:172), TAR15-26-524 (SEQ ID NO:173), TAR15-26-525 (SEQ ID NO: 174), TARl 5-26-526 (SEQ ID NO: 175), TARl 5-26-527 (SEQ ID NO: 176), TARl 5-26- 528 (SEQ ID NO: 177), TARl 5-26-529 (SEQ ID NO: 178), TARl 5-26-530 (SEQ ID NO: 179), TARl 5-26-531 (SEQ ID NO: 180), TARl 5-26-532 (SEQ ID NO: 181), TARl 5-26-533 (SEQ ID NO: 182), TARl 5-26-534 (SEQ ID NO: 183), TARl 5-26- 535 (SEQ ID NO: 184), TAR15-26-536 (SEQ ID NO:185), TAR15-26-537 (SEQ ID NO: 186), TARl 5-26-538 (SEQ ID NO: 187), TARl 5-26-539 (SEQ ID NO: 188), TARl 5-26-540 (SEQ ID NO: 189), TARl 5-26-541 (SEQ ID NO: 190), TARl 5-26- 542 (SEQ ID NO:191), TAR15-26-543 (SEQ ID NO:192), TAR15-26-544 (SEQ ID NO: 193), TARl 5-26-545 (SEQ ID NO: 194), TARl 5-26-546 (SEQ ID NO: 195), TARl 5-26-547 (SEQ ID NO: 196), TARl 5-26-548 (SEQ ID NO: 197), and TARl 5- 26-549 (SEQ ID NO:198), TAR15-26-550 (SEQ ID NO:539), and TAR15-26-551 (SEQ ID NO:540).
In other particular embodiments, the ligand has binding specificity for VEGF and for EGFR and comprises at least one immunoglobulin single variable domain with binding specificity for VEGF and at least one immunoglobulin single variable domain with binding specificity for EGFR, wherein an immunoglobulin single variable domain with binding specificity for EGFR competes for binding to EGFR with an anti-EGFR domain antibody (dAb) selected from the group consisting of DOM16-17 (SEQ ID NO:325), DOM16-18 (SEQ ID NO:326), DOM16-19 (SEQ ID NO:327), DOM16-20 (SEQ ID NO:328), DOM16-21 (SEQ ID NO:329), DOM16- 22 (SEQ ID NO:330), DOM16-23 (SEQ ID NO:331), DOM16-24 (SEQ ID
NO:332), DOMl 6-25 (SEQ ID NO:333), DOMl 6-26 (SEQ ID NO:334), DOMl 6- 27 (SEQ ID NO:335), DOMl 6-28 (SEQ ID NO:336), DOMl 6-29 (SEQ ID
NO:337), DOM16-30 (SEQ ID NO:338), DOM16-31 (SEQ ID NO:339), D0M16- 32 (SEQ ID NO:340), DOM16-33 (SEQ ID NO:341), DOM16-35 (SEQ ID
NO:342), DOM16-37 (SEQ ID NO:343), DOM16-38 (SEQ ID NO:344), D0M16- 39 (SEQ ID NO:345), DOMl 6-40 (SEQ ID NO:346), DOMl 6-41 (SEQ ID
NO:347), DOMl 6-42 (SEQ ID NO:348), DOMl 6-43 (SEQ ID NO:349), DOMl 6- 44 (SEQ ID NO:350), DOM16-45 (SEQ ID NO:351), DOM16-46 (SEQ ID
NO:352), DOM16-47 (SEQ ID NO:353), DOM16-48 (SEQ ID NO:354), D0M16- 49 (SEQ ID NO:355), DOM16-50 (SEQ ID NO:356), DOM16-59 (SEQ ID
NO:357), DOM16-60 (SEQ ID NO:358), DOM16-61 (SEQ ID NO:359), D0M16- 62 (SEQ ID NO:360), DOM16-63 (SEQ ID NO:361), DOM16-64 (SEQ ID
NO:362), DOMl 6-65 (SEQ ID NO:363), DOMl 6-66 (SEQ ID NO:364), DOMl 6- 67 (SEQ ID NO:365), DOMl 6-68 (SEQ ID NO:366), DOMl 6-69 (SEQ ID NO:367), DOM16-70 (SEQ ID NO:368), DOM16-71 (SEQ ID NO:369), DOM16- 72 (SEQ ID NO:370), DOM16-73 (SEQ ID NO:371), DOM16-74 (SEQ ID
NO:372), DOMl 6-75 (SEQ ID NO:373), DOMl 6-76 (SEQ ID NO:374), DOMl 6- 77 (SEQ ID NO:375), DOMl 6-78 (SEQ ID NO:376), DOMl 6-79 (SEQ ID NO:377), DOM16-80 (SEQ ID NO:378), DOM16-81 (SEQ ID NO:379), D0M16- 82 (SEQ ID NO:380), DOM16-83 (SEQ ID NO:381), DOM16-84 (SEQ ID NO:382), DOMl 6-85 (SEQ ID NO:383), DOMl 6-87 (SEQ ID NO:384), DOMl 6- 88 (SEQ ID NO:385), DOM16-89 (SEQ ID NO:386), DOM16-90 (SEQ ID NO:387), DOM16-91 (SEQ ID NO:388), DOM16-92 (SEQ ID NO:389), D0M16- 94 (SEQ ID NO:390), DOM16-95 (SEQ ID NO:391), DOM16-96 (SEQ ID NO:392), DOM16-97 (SEQ ID NO:393), DOM16-98 (SEQ ID NO:394), D0M16- 99 (SEQ ID NO:395), DOM16-100 (SEQ ID NO:396), DOM16-101 (SEQ ID NO:397), DOM16-102 (SEQ ID NO:398), DOM16-103 (SEQ ID NO:399), DOM16-104 (SEQ ID NO:400), DOM16-105 (SEQ ID NO:401), DOM16-106 (SEQ ID NO:402), DOM16-107 (SEQ ID NO:403), DOM16-108 (SEQ ID
NO:404), DOM16-109 (SEQ ID NO:405), DOM16-110 (SEQ ID NO:406), DOMl 6-111 (SEQ ID NO:407), DOMl 6-112 (SEQ ID NO:408), DOMl 6-113 (SEQ ID NO:409), DOM16-114 (SEQ ID NO:410), DOM16-115 (SEQ ID
NO:411), DOM16-116 (SEQ ID NO:412), DOM16-117 (SEQ ID NO:413), DOMl 6-118 (SEQ ID NO:414), DOM16-119 (SEQ ID NO:415), DOM16-39-6 (SEQ ID NO:416), DOMl 6-39-8 (SEQ ID NO:417), DOMl 6-39-34 (SEQ ID NO:418), DOM16-39-48 (SEQ ID NO:419), DOM16-39-87 (SEQ ID NO:420), DOM16-39-90 (SEQ ID NO:421), DOM16-39-96 (SEQ ID NO:422), DOM16-39- 100 (SEQ ID NO:423), DOM16-39-101 (SEQ ID NO:424), DOM16-39-102 (SEQ ID NO:425), DOMl 6-39-103 (SEQ ID NO:426), DOMl 6-39-104 (SEQ ID
NO:427), DOM16-39-105 (SEQ ID NO:428), DOM16-39-106 (SEQ ID NO:429), DOM16-39-107 (SEQ ID NO:430), DOM16-39-108 (SEQ ID NO:431), DOM16- 39-109 (SEQ ID NO:432), DOM16-39-110 (SEQ D_λNO:433), DOMl 6-39-111 (SEQ ID NO:434), DOM16-39-112 (SEQ ID NO:435), DOM16-39-113 (SEQ ID NO:436), DOM16-39-114 (SEQ ID NO:437), DOM16-39-115 (SEQ ID NO:438), DOMl 6-39-116 (SEQ ID NO:439), DOMl 6-39-117 (SEQ ID NO:440), DOM16- 39-200 (SEQ ID NO:441), DOMl 6-39-201 (SEQ ID NO:442), DOMl 6-39-202 (SEQ ID NO:443), DOMl 6-39-203 (SEQ ID NO:444), DOMl 6-39-204 (SEQ ID NO:445), DOMl 6-39-205 (SEQ ID NO:446), DOMl 6-39-206 (SEQ ID NO:447), DOMl 6-39-207 (SEQ ID NO:448), DOMl 6-39-209 (SEQ ID NO:449), DOMl 6-52 (SEQ ID NO:450), NBl (SEQ ID NO:451), NB2 (SEQ ID NO:452), NB3 (SEQ ID NO:453), NB4 (SEQ ID NO:454), NB5 (SEQ ID NO:455), NB6 (SEQ ID NO:456), NB7 (SEQ ID NO:457), NB8 (SEQ ID NO:458), NB9 (SEQ ID NO:459), NBlO (SEQ ID NO:460), NBIl (SEQ ID NO:461), NB12 (SEQ ID NO:462), NB13 (SEQ ID NO:463), NB14 (SEQ ID NO:464), NB15 (SEQ ID NO:465), NB16 (SEQ ID NO:466), NB17 (SEQ ID NO:467), NBl 8 (SEQ ID NO:468), NB19 (SEQ ID NO:469), NB20 (SEQ ID NO:470), NB21 (SEQ ID NO:471), and NB22 (SEQ ID NO:472).
In other particular embodiments, the ligand has binding specificity for VEGF and for EGFR and comprises at least one immunoglobulin single variable domain with binding specificity for VEGF and at least one immunoglobulin single variable domain with binding specificity for EGFR, wherein an immunoglobulin single variable domain with binding specificity for EGFR competes for binding to EGFR with an anti-EGFR domain antibody (dAb) selected from the group consisting of DOM16-39-210 (SEQ ID NO:541), DOM16-39-211 (SEQ ID NO:542), D0M16- 39-212 (SEQ ID NO:543), DOM16-39-213 (SEQ ID NO:544), DOM16-39-214 (SEQ ID NO:545), DOM16-39-215 (SEQ ID NO:546), DOM16-39-216 (SEQ ID NO:547), DOM16-39-217 (SEQ ID NO:548), DOM16-39-218 (SEQ ID NO:549), DOM16-39-219 (SEQ ID NO:550), DOM16-39-220 (SEQ ID NO:551), D0M16- 39-221 (SEQ ID NO:552), DOM16-39-222 (SEQ ID NO:553), DOM16-39-223 (SEQ ID NO:554), DOM16-39-224 (SEQ ID NO:555), DOM16-39-225 (SEQ ID NO:556), DOMl 6-39-226 (SEQ ID NO:557), DOMl 6-39-227 (SEQ ID NO:558), DOM16-39-228 (SEQ ID NO:559), DOM16-39-229 (SEQ ID NO:560), DOM16- 39-230 (SEQ ID NO:561), DOM16-39-231 (SEQ ID NO:562), DOM16-39-232 (SEQ ID NO:563), DOM16-39-233 (SEQ ID NO:564), DOM16-39-234 (SEQ ID NO:565), DOM16-39-235 (SEQ ID NO:566), DOM16-39-500 (SEQ ID NO:725), DOM16-39-502(SEQ ID NO:726), DOM16-39-503 (SEQ ID NO:567), DOM16-39- 504 (SEQ ID NO:568), DOMl 6-39-505 (SEQ ID NO:569), DOMl 6-39-506 (SEQ ID NO:570), DOM16-39-507 (SEQ ID NO:571), DOM16-39-508 (SEQ ID
NO:572), DOM16-39-509 (SEQ ID NO:573), DOM16-39-510 (SEQ ID NO:574), DOM16-39-511 (SEQ ID NO:575), DOM16-39-512 (SEQ ID NO:576), DOM16- 39-521 (SEQ ID NO:577), DOM16-39-522 (SEQ ID NO:578), DOM16-39-523 (SEQ ID NO:579), DOMl 6-39-524 (SEQ ID NO:580), DOMl 6-39-527 (SEQ ID NO:581), DOM16-39-525 (SEQ ID NO:582), DOM16-39-526 (SEQ ID NO:583), DOM16-39-540 (SEQ ID NO:584), DOM16-39-541 (SEQ ID NO:585), DOM16- 39-542 (SEQ ID NO:586), DOM16-39-543 (SEQ ID NO:587), DOM16-39-544 (SEQ ID NO:588), DOM16-39-545 (SEQ ID NO:589), DOM16-39-550 (SEQ ID NO:590), DOM16-39-551 (SEQ ID NO:591), DOM16-39-552 (SEQ ID NO:592), DOM16-39-553 (SEQ ID NO:593), DOM16-39-554 (SEQ ID NO:594), DOM16- 39-555 (SEQ ID NO:595), DOMl 6-39-561 (SEQ ID NO:596), DOMl 6-39-562 (SEQ ID NO:597), DOMl 6-39-563 (SEQ ID NO:598), DOMl 6-39-564 (SEQ ID NO:599), DOM16-39-571 (SEQ ID NO:600), DOM16-39-572 (SEQ ID NO:601), DOM16-39-573 (SEQ ID NO:602), DOM16-39-574 (SEQ ID NO:603), D0M16- 39-580 (SEQ ID NO:604), DOM16-39-591 (SEQ ID NO:605), DOM16-39-592 (SEQ ID NO:606), DOMl 6-39-593 (SEQ ID NO:607), DOMl 6-39-601 (SEQ ID NO:608), DOM16-39-602 (SEQ ID NO:609), DOM16-39-603 (SEQ ID NO:610), DOM16-39-604 (SEQ ID NO:611), DOM16-39-605 (SEQ ID NO:612), DOM16- 39-607 (SEQ ID NO:613), DOM16-39-611 (SEQ ID NO:614), DOM16-39-612 (SEQ ID NO:615), DOM16-39-613 (SEQ ID NO:616), DOM16-39-614 (SEQ ID NO:617), DOM16-39-615 (SEQ ID NO:618), DOM16-39-616 (SEQ ID NO:619), DOM16-39-617 (SEQ ID NO:620), DOM16-39-618 (SEQ ID NO:621), and DOMl 6-39-619 (SEQ ID NO:622).
For example, the immunoglobulin single variable domain with binding specificity for EGFR can comprise an amino acid sequence that has at least about 85% amino acid sequence identity with the amino acid sequence of a dAb selected from the group consisting of DOM16-17 (SEQ ID NO:325), DOM16-18 (SEQ ID NO:326), DOM16-19 (SEQ ID NO:327), DOM16-20 (SEQ ID NO:328), DOM16- 21 (SEQ ID NO:329), DOM16-22 (SEQ ID NO:330), DOM16-23 (SEQ ID
NO:331), DOMl 6-24 (SEQ ID NO:332), DOMl 6-25 (SEQ ID NO:333), DOMl 6- 26 (SEQ ID NO:334), DOMl 6-27 (SEQ ID NO:335), DOMl 6-28 (SEQ ID NO:336), DOM16-29 (SEQ ID NO:337), DOM16-30 (SEQ ID NO:338), DOM16- 31 (SEQ ID NO:339), DOM16-32 (SEQ ID NO:340), DOM16-33 (SEQ ID NO:341), DOM16-35 (SEQ ID NO:342), DOM16-37 (SEQ ID NO:343), DOM16- 38 (SEQ ID NO:344), DOMl 6-39 (SEQ ID NO:345), DOMl 6-40 (SEQ ID NO:346), DOMl 6-41 (SEQ ID NO:347), DOMl 6-42 (SEQ ID NO:348), DOMl 6- 43 (SEQ ID NO:349), DOM16-44 (SEQ ID NO:350), DOM16-45 (SEQ ID NO:351), DOM16-46 (SEQ ID NO:352), DOM16-47 (SEQ ID NO:353), DOM16- 48 (SEQ ID NO:354), DOM16-49 (SEQ ID NO:355), DOM16-50 (SEQ ID
NO:356), DOMl 6-59 (SEQ ID NO:357), DOMl 6-60 (SEQ ID NO:358), DOMl 6- 61 (SEQ ID NO:359), DOM16-62 (SEQ ID NO:360), DOM16-63 (SEQ ID NO:361), DOM16-64 (SEQ ID NO:362), DOM16-65 (SEQ ID NO:363), D0M16- 66 (SEQ ID NO:364), DOMl 6-67 (SEQ ID NO:365), DOMl 6-68 (SEQ ID NO:366), DOM16-69 (SEQ ID NO:367), DOM16-70 (SEQ ID NO:368), D0M16- 71 (SEQ ID NO:369), DOM16-72 (SEQ ID NO:370), DOM16-73 (SEQ ID NO:371), DOMl 6-74 (SEQ ID NO:372), DOMl 6-75 (SEQ ID NO:373), DOMl 6- 76 (SEQ ID NO:374), DOMl 6-77 (SEQ ID NO:375), DOMl 6-78 (SEQ ID
NO:376), DOMl 6-79 (SEQ ID NO:377), DOMl 6-80 (SEQ ID NO:378), DOMl 6- 81 (SEQ ID NO:379), DOMl 6-82 (SEQ ID NO:380), DOM16-83 (SEQ ID
NO:381), DOM16-84 (SEQ ID NO:382), DOM16-85 (SEQ ID NO:383), DOM16- 87 (SEQ ID NO:384), DOMl 6-88 (SEQ ID NO:385), DOMl 6-89 (SEQ ID
NO:386), DOMl 6-90 (SEQ ID NO:387), DOMl 6-91 (SEQ ID NO:388), DOMl 6- 92 (SEQ ID NO:389), DOM16-94 (SEQ ID NO:390), DOM16-95 (SEQ ID
NO:391), DOMl 6-96 (SEQ ID NO:392), DOMl 6-97 (SEQ ID NO:393), DOMl 6- 98 (SEQ ID NO:394), DOM16-99 (SEQ ID NO:395), DOM16-100 (SEQ ID NO:396), DOM16-101 (SEQ ID NO:397), DOM16-102 (SEQ ID NO:398), DOM16-103 (SEQ ID NO:399), DOM16-104 (SEQ ID NO:400), DOM16.-105 (SEQ ID NO:401), DOM16-106 (SEQ ID NO:402), DOM16-107 (SEQ ID
NO:403), DOM16-108 (SEQ ID NO:404), DOM16-109 (SEQ ID NO:405), D0M16-110 (SEQ ID NO:406), DOM16-111 (SEQ ID NO:407), DOM16-112 (SEQ ID NO:408), DOMl 6-113 (SEQ ID NO:409), DOMl 6-114 (SEQ ID
NO:410), DOM16-115 (SEQ ID N0:411), D0M16-116 (SEQ ID NO:412), DOM16-117 (SEQ ID NO:413), DOM16-118 (SEQ ID NO:414), DOM16-119 (SEQ ID NO:415), DOMl 6-39-6 (SEQ ID NO:416), DOMl 6-39-8 (SEQ ID NO:417), DOM16-39-34 (SEQ ID NO:418), DOM16-39-48 (SEQ ID NO:419), DOMl 6-39-87 (SEQ ID NO:420), DOMl 6-39-90 (SEQ ID NO:421), DOMl 6-39- 96 (SEQ ID NO:422), DOM16-39-100 (SEQ ID NO:423), DOM16-39-101 (SEQ ID NO:424), DOM16-39-102 (SEQ ID NO:425), DOM16-39-103 (SEQ ID NO:426), DOM16-39-104 (SEQ ID NO:427), DOM16-39-105 (SEQ ID NO:428), DOM16- 39-106 (SEQ ID NO:429), DOM16-39-107 (SEQ ID NO:430), DOM16-39-108 (SEQ ID NO:431), DOMl 6-39-109 (SEQ ID NO:432), DOMl 6-39-110 (SEQ ID NO:433), DOM16-39-111 (SEQ ID NO:434), DOM16-39-112 (SEQ ID NO:435), DOM16-39-113 (SEQ ID NO:436), DOM16-39-114 (SEQ ID NO:437), DOM16- 39-115 (SEQ ID NO:438), DOMl 6-39-116 (SEQ ID NO:439), DOMl 6-39-117 (SEQ ID NO:440), DOM16-39-200 (SEQ ID NO:441), DOM16-39-201 (SEQ ID NO:442), DOMl 6-39-202 (SEQ ID NO:443), DOMl 6-39-203 (SEQ ID NO:444), DOM16-39-204 (SEQ ID NO:445), DOM16-39-205 (SEQ ID NO:446), DOM16- 39-206 (SEQ ID NO:447), DOM16-39-207 (SEQ ID NO:448), DOM16-39-209 (SEQ ID NO:449), DOM16-52 (SEQ ID NO:450), NBl (SEQ ID NO:451), NB2 (SEQ ID NO:452), NB3 (SEQ ID NO:453), NB4 (SEQ ID NO:454), NB5 (SEQ ID NO:455), NB6 (SEQ ID NO:456), NB7 (SEQ ID NO:457), NB8 (SEQ ID NO:458), NB9 (SEQ ID NO:459), NBlO (SEQ ID NO:460), NBI l (SEQ ID NO:461), NB12 (SEQ ID NO:462), NB13 (SEQ ID NO:463), NB14 (SEQ ID NO:464), NB15 (SEQ ID NO:465), NB 16 (SEQ ID NO:466), NB 17 (SEQ ID NO:467), NB 18 (SEQ ID NO:468), NB19 (SEQ ID NO:469), NB20 (SEQ ID NO:470), NB21 (SEQ ID NO:471), and NB22 (SEQ ID NO:472).
For example, the immunoglobulin single variable domain with binding specificity for EGFR can comprise an amino acid sequence that has at least about 85% amino acid sequence identity with the amino acid sequence of a dAb selected from the group consisting of DOM16-39-210 (SEQ ID NO:541), DOM16-39-21I (SEQ ID NO:542), DOM16-39-212 (SEQ ID NO:543), DOM16-39-213 (SEQ ID NO:544), DOM16-39-214 (SEQ ID NO:545), DOM16-39-215 (SEQ ID NO:546), DOM16-39-216 (SEQ ID NO:547), DOM16-39-217 (SEQ ID NO:548), DOM16- 39-218 (SEQ ID NO:549), DOM16-39-219 (SEQ ID NO:550), DOM16-39-220 (SEQ ID NO:551), DOM16-39-221 (SEQ ID NO:552), DOM16-39-222 (SEQ ID NO:553), DOM16-39-223 (SEQ ID NO:554), DOM16-39-224 (SEQ ID NO:555), DOMl 6-39-225 (SEQ ID NO:556), DOMl 6-39-226 (SEQ ID NO:557), DOMl 6- 39-227 (SEQ ID NO:558), DOMl 6-39-228 (SEQ ID NO:559), DOMl 6-39-229 (SEQ ID NO:560), DOMl 6-39-230 (SEQ ID NO:561), DOMl 6-39-231 (SEQ ID NO:562), DOMl 6-39-232 (SEQ ID NO:563), DOMl 6-39-233 (SEQ ID NO:564), DOMl 6-39-234 (SEQ ID NO:565), DOMl 6-39-235 (SEQ ID NO:566), DOMl 6- 39-500 (SEQ ID NO:725), DOMl 6-39-502(SEQ ID NO:726), DOMl 6-39-503 (SEQ ID NO:567), DOMl 6-39-504 (SEQ ID NO:568), DOMl 6-39-505 (SEQ ID NO:569), DOMl 6-39-506 (SEQ ID NO:570), DOMl 6-39-507 (SEQ ID NO:571), DOMl 6-39-508 (SEQ ID NO:572), DOMl 6-39-509 (SEQ ID NO:573), DOMl 6- 39-510 (SEQ ID NO:574), DOM16-39-511 (SEQ ID NO:575), DOM16-39-512 (SEQ ID NO:576), DOM16-39-521 (SEQ ID NO:577), DOM16-39-522 (SEQ ID NO:578), DOMl 6-39-523 (SEQ ID NO:579), DOMl 6-39-524 (SEQ ID NO:580), DOM16-39-527 (SEQ ID NO:581), DOM16-39-525 (SEQ ID NO:582), D0M16- 39-526 (SEQ ID NO:583), DOMl 6-39-540 (SEQ ID NO:584), DOMl 6-39-541 (SEQ ID NO:585), DOM 16-39-542 (SEQ ID NO:586), DOM16-39-543 (SEQ ID NO:587), DOM16-39-544 (SEQ ID NO:588), DOM16-39-545 (SEQ ID NO:589), DOM16-39-550 (SEQ ID NO:590), DOM16-39-551 (SEQ ID NO:591), DOM16- 39-552 (SEQ ID NO:592), DOM16-39-553 (SEQ ID NO:593), DOM16-39-554 (SEQ ID NO:594), DOM16-39-555 (SEQ ID NO:595), DOM16-39-561 (SEQ ID NO:596), DOM16-39-562 (SEQ ID NO:597), DOMl 6-39-563 (SEQ ID NO:598), DOM16-39-564 (SEQ ID NO:599), DOM16-39-571 (SEQ ID NO:600), DOM16- 39-572 (SEQ ID NO:601), DOMl 6-39-573 (SEQ ID NO:602), DOMl 6-39-574 (SEQ ID NO:603), DOM16-39-580 (SEQ ID NO:604), DOM16-39-591 (SEQ ID NO:605), DOM16-39-592 (SEQ ID NO:606), DOM16-39-593 (SEQ ID NO:607), DOM16-39-601 (SEQ ID NO:608), DOM16-39-602 (SEQ ID NO:609), DOM16- 39-603 (SEQ ID NO:610), DOM16-39-604 (SEQ ID NO:611), DOM16-39-605 (SEQ ID NO:612), DOM16-39-607 (SEQ ID NO:613), DOM16-39-611 (SEQ ID , NO:614), DOM16-39-612 (SEQ ID NO:615), DOM16-39-613 (SEQ ID NO:616), DOM16-39-614 (SEQ ID NO:617), DOM16-39-615 (SEQ ID NO:618), DOM16- 39-616 (SEQ ID NO:619), DOM16-39-617 (SEQ ID NO:620), DOM16-39-618 (SEQ ID NO:621), and DOM16-39-619 (SEQ ID NO:622).
In some embodiments, the ligand has binding specificity for VEGF and for EGFR and comprises at least one immunoglobulin single variable domain with binding specificity for VEGF and at least one immunoglobulin single variable domain with binding specificity for EGFR, wherein an immunoglobulin single variable domain with binding specificity for VEGF competes for binding to VEGF with an anti-VEGF domain antibody (dAb) selected from the group consisting of TARl 5-1 (SEQ ID NO: 100), TARl 5-3 (SEQ ID NO: 101), TARl 5-4 (SEQ ID NO:102), TAR15-9 (SEQ ID NO:103), TAR15-10 (SEQ ID NO: 104), TARl 5-11 (SEQ ID NO:105), TAR15-12 (SEQ ID NO:106), TAR15-13 (SEQ ID NO:107), TARl 5-14 (SEQ ID NO: 108), TARl 5-15 (SEQ ID NO: 109), TARl 5-16 (SEQ ID NO:110), TAR15-17 (SEQ ID NO:111), TAR15-18 (SEQ ID NO:112), TAR15-19 (SEQ ID NO: 113), TARl 5-20 (SEQ ID NO: 114), TAR 15-22 (SEQ ID NO: 115), TARl 5-5 (SEQ ID NO:116), TARl 5-6 (SEQ ID NO: 117), TARl 5-7 (SEQ ID NO: 118), TARl 5-8 (SEQ ID NO: 119), TARl 5-23 (SEQ ID NO: 120), TARl 5-24 (SEQ ID NO:121), TAR15-25 (SEQ ID NO: 122), TAR15-26 (SEQ ID NO:123), TAR15-27 (SEQ ID NO:124), TAR15-29 (SEQ ID NO:125), TAR15-30 (SEQ ID NO:126), TAR15-6-500 (SEQ ID NO:127), TAR15-6-501 (SEQ ID NO:128), TAR15-6-502 (SEQ ID NO:129), TAR15-6-503 (SEQ ID NO:130), TAR15-6-504 (SEQ ID NO:131), TAR15-6-505 (SEQ ID NO:132), TAR15-6-506 (SEQ ID NO:133), TAR15-6-507 (SEQ ID NO:134), TAR15-6-508 (SEQ ID NO:135), TARl 5-6-509 (SEQ ID NO: 136), TARl 5-6-510 (SEQ ID NO: 137), TARl 5-8-500 (SEQ ID NO:138), TAR15-8-501 (SEQ ID NO:139), TAR15-8-502 (SEQ ID NO: 140), TARl 5-8-503 (SEQ ID NO: 141), TARl 5-8-505 (SEQ ID NO: 142), TAR15-8-506 (SEQ ID NO:143), TAR15-8-507 (SEQ ID NO:144), TAR15-8-508 (SEQ ID NO: 145), TARl 5-8-509 (SEQ ID NO: 146), TARl 5-8-510 (SEQ ID NO:147), TAR15-8-511 (SEQ ID NO:148), TAR15-26-500 (SEQ ID NO:149), TAR15-26-501 (SEQ ID NO:150), TAR15-26-502 (SEQ ID NO:151), TAR15-26- 503 (SEQ ID NO: 152), TARl 5-26-504 (SEQ ID NO: 153), TARl 5-26-505 (SEQ ID NO: 154), TARl 5-26-506 (SEQ ID NO: 155), TARl 5-26-507 (SEQ ID NO: 156), TAR15-26-508 (SEQ ID NO:157), TAR15-26-509 (SEQ ID NO:158), TAR15-26- 510 (SEQ ID NO: 159), TARl 5-26-511 (SEQ ID NO: 160), TARl 5-26-512 (SEQ ID NO:161), TAR15-26-513 (SEQ ID NO:162), TAR15-26-514 (SEQ ID NO:163), TARl 5-26-515 (SEQ ID NO: 164), TARl 5-26-516 (SEQ ID NO: 165), TARl 5-26- 517 (SEQ ID NO: 166), TARl 5-26-518 (SEQ ID NO: 167), TARl 5-26-519 (SEQ ID NO:168), TAR15-26-520 (SEQ ID NO:169), TAR15-26-521 (SEQ ID NO:170), TAR15-26-522 (SEQ ID NO:171), TAR15-26-523 (SEQ ID NO:172), TAR15-26- 524 (SEQ ID NO: 173), TARl 5-26-525 (SEQ ID NO: 174), TARl 5-26-526 (SEQ ID NO: 175), TARl 5-26-527 (SEQ ID NO: 176), TARl 5-26-528 (SEQ ID NO: 177), TAR15-26-529 (SEQ ID NO:178), TAR15-26-530 (SEQ ID NO:179), TAR15-26- 531 (SEQ ID NO: 180), TARl 5-26-532 (SEQ ID NO: 181), TARl 5-26-533 (SEQ ID NO: 182), TARl 5-26-534 (SEQ ID NO: 183), TARl 5-26-535 (SEQ ID NO: 184), TARl 5-26-536 (SEQ ID NO: 185), TARl 5-26-537 (SEQ ID NO: 186), TARl 5-26- 538 (SEQ ID NO:187), TAR15-26-539 (SEQ ID NO:188), TAR15-26-540 (SEQ ID NO:189), TAR15-26-541 (SEQ ID NO:190), TAR15-26-542 (SEQ ID NO:191), TAR15-26-543 (SEQ ID NO:192), TAR15-26-544 (SEQ ID NO:193), TAR15-26- 545 (SEQ ID NO: 194), TARl 5-26-546 (SEQ ID NO: 195), TARl 5-26-547 (SEQ ID NO: 196), TARl 5-26-548 (SEQ ID NO: 197), and TARl 5-26-549 (SEQ ID NO: 198), TARl 5-26-550 (SEQ ID NO:539), and TARl 5-26-551 (SEQ ID NO:540); and wherein an immunoglobulin single variable domain with binding specificity for EGFR competes for binding to EGFR with an anti-EGFR domain antibody (dAb) selected from the group consisting of DOM16-17 (SEQ ID NO:325), DOM16-18 (SEQ ID NO:326), DOMl 6-19 (SEQ ID NO:327), DOMl 6-20 (SEQ ID NO:328), DOM16-21 (SEQ ID NO:329), DOM16-22 (SEQ ID NO:330), DOM16-23 (SEQ ID NO:331), DOM16-24 (SEQ ID NO:332), DOM16-25 (SEQ ID NO:333), DOM16- 26 (SEQ ID NO:334), DOMl 6-27 (SEQ ID NO:335), DOMl 6-28 (SEQ ID
NO:336), DOM16-29 (SEQ ID NO:337), DOM16-30 (SEQ ID NO:338), DOM16- 31 (SEQ ID NO:339), DOMl 6-32 (SEQ ID NO:340), DOMl 6-33 (SEQ ID
NO:341), DOM16-35 (SEQ ID NO:342), DOM16-37 (SEQ ID NO:343), D0M16- 38 (SEQ ID NO:344), DOMl 6-39 (SEQ ID NO:345), DOMl 6-40 (SEQ ID
NO:346), DOMl 6-41 (SEQ ID NO:347), DOMl 6-42 (SEQ ID NO:348), DOMl 6- 43 (SEQ ID NO:349), DOMl 6-44 (SEQ ID NO:350), DOMl 6-45 (SEQ ID
NO:351), DOM16-46 (SEQ ID NO:352), DOM16-47 (SEQ ID NO:353), DOM16- 48 (SEQ ID NO:354), DOMl 6-49 (SEQ ID NO:355), DOMl 6-50 (SEQ ID
NO:356), DOM16-59 (SEQ ID NO:357), DOM16-60 (SEQ ID NO:358), DOM16- 61 (SEQ ID NO:359), DOM16-62 (SEQ ID NO:360), DOM16-63 (SEQ ID
NO:361), DOMl 6-64 (SEQ ID NO:362), DOMl 6-65 (SEQ ID NO:363), DOMl 6- 66 (SEQ ID NO:364), DOM16-67 (SEQ ID NO:365), DOM16-68 (SEQ ID
NO:366), DOMl 6-69 (SEQ ID NO:367), DOMl 6-70 (SEQ ID NO:368), DOMl 6- 71 (SEQ ID NO:369), DOMl 6-72 (SEQ ID NO:370), DOMl 6-73 (SEQ ID
NO:371), DOM16-74 (SEQ ID NO:372), DOM16-75 (SEQ ID NO:373), D0M16- 76 (SEQ ID NO:374), DOMl 6-77 (SEQ ID NO:375), DOMl 6-78 (SEQ ID
NO:376), DOM16-79 (SEQ ID NO:377), DOM16-80 (SEQ ID NO:378), DOM16- 81 (SEQ ID NO:379), DOM16-82 (SEQ ID NO:380), DOM16-83 (SEQ ID
NO:381), DOM16-84 (SEQ ID NO:382), DOM16-85 (SEQ ID NO:383), D0M16- 87 (SEQ ID NO:384), DOM16-88 (SEQ ID NO:385), DOM16-89 (SEQ ID
NO:386), DOM16-90 (SEQ ID NO:387), DOM16-91 (SEQ ID NO:388), DOM16- 92 (SEQ ID NO:389), DOM16-94 (SEQ ID NO:390), DOM16-95 (SEQ ID
NO:391), DOM16-96 (SEQ ID NO:392), DOM16-97 (SEQ ID NO:393), D0M16- 98 (SEQ ID NO.-394), DOM16-99 (SEQ ID NO:395), DOM16-100 (SEQ ID NO:396), DOM16-101 (SEQ ID NO:397), DOM16-102 (SEQ ID NO:398), DOMl 6-103 (SEQ ID NO:399), DOMl 6-104 (SEQ ID NO:400), DOMl 6-105 (SEQ ID NO:401), DOM16-106 (SEQ ID NO:402), DOM16-107 (SEQ ID
NO:403), DOM16-108 (SEQ ID NO:404), DOM16-109 (SEQ ID NO:405), DOM16-110 (SEQ ID NO:406), DOM16-111 (SEQ ID NO:407), DOM16-112 (SEQ ID NO:408), DOMl 6-113 (SEQ ID NO:409), DOMl 6-114 (SEQ ID
NO:410), DOM16-115 (SEQ ID NO:411), DOM16-116 (SEQ ID NO:412), DOM16-117 (SEQ ID NO:413), DOM16-118 (SEQ ID NO:414), DOM16-119 (SEQ ID NO:415), DOM16-39-6 (SEQ ID NO:416), DOM16-39-8 (SEQ ID NO:417), DOM16-39-34 (SEQ ID NO:418), DOM16-39-48 (SEQ ID NO:419), DOM16-39-87 (SEQ ID NO:420), DOM16-39-90 (SEQ ID NO:421), DOM16-39- 96 (SEQ ID NO:422), DOM16-39-100 (SEQ ID NO:423), DOM16-39-101 (SEQ ID NO:424), DOM16-39-102 (SEQ ID NO:425), DOM16-39-103 (SEQ ID NO:426), DOM16-39-104 (SEQ ID NO:427), DOM16-39-105 (SEQ ID NO:428), DOM16- 39-106 (SEQ ID NO:429), DOM16-39-107 (SEQ ID NO:430), DOM16-39-108 (SEQ ID NO:431), DOMl 6-39-109 (SEQ ID NO:432), DOM16-39-110 (SEQ ID NO:433), DOM16-39-111 (SEQ ID NO:434), DOM16-39-112 (SEQ ID NO:435), DOM16-39-113 (SEQ ID NO:436), DOM16-39-114 (SEQ ID NO:437), DOM16- 39-115 (SEQ ID NO:438), DOMl 6-39-116 (SEQ ID NO:439), DOMl 6-39-117 (SEQ ID NO:440), DOM16-39-200 (SEQ ID NO:441), DOM16-39-201 (SEQ ID NO:442), DOMl 6-39-202 (SEQ ID NO:443), DOMl 6-39-203 (SEQ ID NO:444), DOM16-39-204 (SEQ ID NO:445), DOM16-39-205 (SEQ ID NO:446), DOM16- 39-206 (SEQ ID NO:447), DOM16-39-207 (SEQ ID NO:448), DOM16-39-209 (SEQ ID NO:449), DOM16-52 (SEQ ID NO:450), NBl (SEQ ID NO:451), NB2 (SEQ ID NO:452), NB3 (SEQ ID NO:453), NB4 (SEQ ID NO:454), NB5 (SEQ ID NO:455), NB6 (SEQ ID NO:456), NB7 (SEQ ID NO:457), NB8 (SEQ ID NO:458), NB9 (SEQ ID NO:459), NBlO (SEQ ID NO:460), NBI l (SEQ ID NO:461), NB12 (SEQ ID NO:462), NB13 (SEQ ID NO:463), NB14 (SEQ ID NO:464), NB15 (SEQ ID NO:465), NB 16 (SEQ ID NO:466), NB 17 (SEQ ID NO:467), NB 18 (SEQ ID NO:468), NB19 (SEQ ID NO:469), NB20 (SEQ ID NO:470), NB21 (SEQ ID NO:471), and NB22 (SEQ ID NO:472). In additional embodiments, the ligand has binding specificity for VEGF and for EGFR and comprises at least one immunoglobulin single variable domain with binding specificity for VEGF and at least one immunoglobulin single variable domain with binding specificity for EGFR, wherein an immunoglobulin single variable domain with binding specificity for VEGF competes for binding to VEGF with an anti-VEGF domain antibody (dAb) selected from the group consisting of TAR15-1 (SEQ ID NO:100), TAR15-3 (SEQ ID NOrIOl), TAR15-4 (SEQ ID NO:102), TAR15-9 (SEQ ID NOrI 03), TAR15-10 (SEQ ID NOrI 04), TAR15-11 (SEQ ID NO:105), TAR15-12 (SEQ ID NO:106), TAR15-13 (SEQ ID NO: 107), TAR15-14 (SEQ ID NO:108), TAR15-15 (SEQ ID NO:109), TAR15-16 (SEQ ID NOrI lO), TAR15-17 (SEQ ID NO: 111), TARl 5-18 (SEQ ID NOrI 12), TAR15-19 (SEQ ID NOrI 13), TARl 5-20 (SEQ ID NOrI 14), TAR 15-22 (SEQ ID NOrI 15), TARl 5-5 (SEQ ID NO: 116), TARl 5-6 (SEQ ID NO: 117), TARl 5-7 (SEQ ID NO: 118), TARl 5-8 (SEQ ID NO: 119), TARl 5-23 (SEQ ID NO: 120), TARl 5-24 (SEQ ID NO:121), TAR15-25 (SEQ ID NO:122), TAR15-26 (SEQ ID NO:123), TAR15-27 (SEQ ID NO: 124), TAR15-29 (SEQ ID NO:125), TAR15-30 (SEQ ID NO: 126), TARl 5-6-500 (SEQ ID NO: 127), TARl 5-6-501 (SEQ ID NO: 128), TAR15-6-502 (SEQ ID NO:129), TAR15-6-503 (SEQ ID NO: 130), TAR15-6-504 (SEQ ID NO: 131), TARl 5-6-505 (SEQ ID NO: 132), TARl 5-6-506 (SEQ ID NO:133), TAR15-6-507 (SEQ ID NO:134), TAR15-6-508 (SEQ ID NO:135),
TARl 5-6-509 (SEQ ID NO: 136), TARl 5-6-510 (SEQ ID NO: 137), TARl 5-8-500 (SEQ ID NO: 138), TARl 5-8-501 (SEQ ID NO: 139), TARl 5-8-502 (SEQ ID NO:140), TAR15-8-503 (SEQ ID NO:141), TAR15-8-505 (SEQ ID NO: 142), TARl 5-8-506 (SEQ ID NO: 143), TARl 5-8-507 (SEQ ID NO: 144), TARl 5-8-508 (SEQ ID NO: 145), TARl 5-8-509 (SEQ ID NO: 146), TARl 5-8-510 (SEQ ID NO: 147), TARl 5-8-511 (SEQ ID NO: 148), TARl 5-26-500 (SEQ ID NO: 149), TARl 5-26-501 (SEQ ID NO: 150), TARl 5-26-502 (SEQ ID NO:151), TARl 5-26- 503 (SEQ ID NO: 152), TARl 5-26-504 (SEQ ID NO: 153), TARl 5-26-505 (SEQ ID NO: 154), TARl 5-26-506 (SEQ ID NO: 155), TARl 5-26-507 (SEQ ID NO: 156), TARl 5-26-508 (SEQ ID NO: 157), TARl 5-26-509 (SEQ ID NO: 158), TARl 5-26- 510 (SEQ ID NO:159), TAR15-26-511 (SEQ ID NO: 160), TAR15-26-512 (SEQ ID NO:161), TAR15-26-513 (SEQ ID NO:162), TAR15-26-514 (SEQ ID NO:163), TARl 5-26-515 (SEQ ID NO: 164), TARl 5-26-516 (SEQ ID NO: 165), TARl 5-26- 517 (SEQ ID NO:166), TAR15-26-518 (SEQ ID NO:167), TAR15-26-519 (SEQ ID NO:168), TAR15-26-520 (SEQ ID NO:169), TAR15-26-521 (SEQ ID NO:170), TAR15-26-522 (SEQ ID NO:171), TAR15-26-523 (SEQ ID NO: 172), TAR15-26- 524 (SEQ ID NO: 173), TARl 5-26-525 (SEQ ID NO: 174), TARl 5-26-526 (SEQ ID NO: 175), TARl 5-26-527 (SEQ ID NO: 176), TARl 5-26-528 (SEQ ID NO: 177), TARl 5-26-529 (SEQ ID NO: 178), TARl 5-26-530 (SEQ ID NO: 179), TARl 5-26- 531 (SEQ ID NO:180), TAR15-26-532 (SEQ ID NO:181), TAR15-26-533 (SEQ ID NO: 182), TARl 5-26-534 (SEQ ID NO: 183), TARl 5-26-535 (SEQ ID NO: 184), TARl 5-26-536 (SEQ ID NO: 185), TARl 5-26-537 (SEQ ID NO: 186), TARl 5-26- 538 (SEQ ID NO: 187), TARl 5-26-539 (SEQ ID NO: 188), TARl 5-26-540 (SEQ ID NO:189), TAR15-26-541 (SEQ E) NO: 190), TAR15-26-542 (SEQ E) NO:191), TARl 5-26-543 (SEQ ID NO: 192), TARl 5-26-544 (SEQ ID NO: 193), TARl 5-26- 545 (SEQ ID NO: 194), TARl 5-26-546 (SEQ ID NO: 195), TARl 5-26-547 (SEQ ID NO: 196), TARl 5-26-548 (SEQ ID NO: 197), TARl 5-26-549 (SEQ ID NO: 198), TARl 5-26-550 (SEQ ID NO:539), and TARl 5-26-551 (SEQ ID NO:540); and wherein an immunoglobulin single variable domain with binding specificity for EGFR competes for binding to EGFR with an anti-EGFR domain antibody (dAb) selected from the group consisting of DOM 16-39-210 (SEQ ID NO: 541), DOMl 6- 39-211 (SEQ ID NO:542), DOM16-39-212 (SEQ ID NO:543), DOM16-39-213 (SEQ ID NO:544), DOMl 6-39-214 (SEQ ID NO:545), DOMl 6-39-215 (SEQ ID NO:546), DOM16-39-216 (SEQ ID NO:547), DOM16-39-217 (SEQ ID NO:548), DOM16-39-218 (SEQ ID NO:549), DOM16-39-219 (SEQ ID NO:550), DOM16- 39-220 (SEQ ID NO:551), DOM16-39-221 (SEQ ED NO:552), DOM16-39-222 (SEQ ID NO:553), DOMl 6-39-223 (SEQ ID NO:554), DOMl 6-39-224 (SEQ ID NO:555), DOMl 6-39-225 (SEQ ID NO:556), DOMl 6-39-226 (SEQ ID NO:557), DOMl 6-39-227 (SEQ ID NO:558), DOMl 6-39-228 (SEQ ID NO:559), DOMl 6- 39-229 (SEQ ID NO:560), DOM16-39-230 (SEQ ID NO:561), DOM16-39-231 (SEQ ID NO:562), DOM16-39-232 (SEQ ID NO:563), DOM16-39-233 (SEQ ID NO:564), DOMl 6-39-234 (SEQ ID NO:565), DOMl 6-39-235 (SEQ ID NO:566), DOMl 6-39-500 (SEQ ID NO:725), DOMl 6-39-502(SEQ ID NO:726), DOMl 6-39- 503 (SEQ ID NO:567), DOMl 6-39-504 (SEQ ID NO:568), DOMl 6-39-505 (SEQ ID NO:569), DOMl 6-39-506 (SEQ ID NO:570), DOMl 6-39-507 (SEQ ID NO:571), DOM16-39-508 (SEQ ID NO:572), DOM16-39-509 (SEQ ID NO:573), DOM16-39-510 (SEQ ID NO:574), DOM16-39-511 (SEQ ID NO:575), DOM16- 39-512 (SEQ ID NO:576), DOM16-39-521 (SEQ ID NO:577), DOM16-39-522 (SEQ ID NO:578), DOM16-39-523 (SEQ ID NO:579), DOM16-39-524 (SEQ ID NO:580), DOM16-39-527 (SEQ ID NO:581), DOM16-39-525 (SEQ ID NO:582), DOM16-39-526 (SEQ ID NO:583), DOM16-39-540 (SEQ ID NO:584), DOM16- 39-541 (SEQ ID NO:585), DOM16-39-542 (SEQ ID NO:586), DOM16-39-543 (SEQ ID NO:587), DOMl 6-39-544 (SEQ ID NO:588), DOMl 6-39-545 (SEQ ID NO:589), DOM16-39-550 (SEQ ID NO:590), DOM16-39-551 (SEQ ID NO:591), DOMl 6-39-552 (SEQ ID NO:592), DOMl 6-39-553 (SEQ ID NO:593), DOMl 6- 39-554 (SEQ ID NO:594), DOM16-39-555 (SEQ ID NO:595), DOM16-39-561 (SEQ ID NO:596), DOMl 6-39-562 (SEQ ID NO:597), DOMl 6-39-563 (SEQ ID NO:598), DOM16-39-564 (SEQ ID NO:599), DOM16-39-571 (SEQ ID NO:600), DOM16-39-572 (SEQ ID NO:601), DOM16-39-573 (SEQ ID NO:602), DOM16- 39-574 (SEQ ID NO:603), DOM16-39-580 (SEQ ID NO:604), DOM16-39-591 (SEQ ID NO:605), DOMl 6-39-592 (SEQ ID NO:606), DOMl 6-39-593 (SEQ ID NO:607), DOM16-39-601 (SEQ ID NO:608), DOM16-39-602 (SEQ ID NO:609), DOM16-39-603 (SEQ ID NO:610), DOM16-39-604 (SEQ ID NO:611), DOM16- 39-605 (SEQ ID NO:612), DOMl 6-39-607 (SEQ ID NO:613), DOMl 6-39-611 (SEQ ID NO:614), DOM16-39-612 (SEQ ID NO:615), DOM16-39-613 (SEQ ID NO:616), DOM16-39-614 (SEQ ID NO:617), DOM16-39-615 (SEQ ID NO:618), DOM16-39-616 (SEQ ID NO:619), DOM16-39-617 (SEQ ID NO:620), DOM16- 39-618 (SEQ ID NO:621), and DOM16-39-619 (SEQ ID NO:622).
For example, the ligand can comprise an immunoglobulin single variable domain with binding specificity for VEGF that comprises an amino acid sequence that has at least about 85% amino acid sequence identity with the amino acid sequence of a dAb selected from the group consisting of TARl 5-1 (SEQ ID NOrIOO), TAR15-3 (SEQ ID NO:101), TAR15-4 (SEQ ID NO:102), TAR15-9 (SEQ ID NO:103), TAR15-10 (SEQ ID NO:104), TAR15-11 (SEQ ID NO: 105), TAR15-12 (SEQ ID NO:106), TAR15-13 (SEQ ID NO:107), TAR15-14 (SEQ ID NO: 108), TAR15-15 (SEQ ID NO: 109), TAR15-16 (SEQ ID NO:110), TAR15-17 (SEQ ID NO:111), TAR15-18 (SEQ ID NO:112), TAR15-19 (SEQ ID NO:113), TARl 5-20 (SEQ ID NO: 114), TAR 15-22 (SEQ ID NO: 115), TARl 5-5 (SEQ ID NO: 116), TARl 5-6 (SEQ ID NO: 117), TARl 5-7 (SEQ ID NO: 118), TARl 5-8 (SEQ ID NO:119), TAR15-23 (SEQ ID NO:120), TAR15-24 (SEQ ID NO:121), TARl 5-25 (SEQ ID NO: 122), TARl 5-26 (SEQ ID NO: 123), TARl 5-27 (SEQ ID NO:124), TAR15-29 (SEQ ID NO:125), TAR15-30 (SEQ ID NO:126), TAR15-6- 500 (SEQ ID NO:127), TAR15-6-501 (SEQ ID NO:128), TAR15-6-502 (SEQ ID NO:129), TAR15-6-503 (SEQ ID NO:130), TAR15-6-504 (SEQ ID NO:131), TARl 5-6-505 (SEQ ID NO: 132), TARl 5-6-506 (SEQ ID NO: 133), TARl 5-6-507 (SEQ ID NO:134), TAR15-6-508 (SEQ ID NO:135), TAR15-6-509 (SEQ ID NO:136), TAR15-6-510 (SEQ ID NO:137), TAR15-8-500 (SEQ ID NO:138), TAR15-8-501 (SEQ ID NO:139), TAR15-8-502 (SEQ ID NO:140), TAR15-8-503 (SEQ ID NO:141), TAR15-8-505 (SEQ ID NO:142), TAR15-8-506 (SEQ ID NO:143), TAR15-8-507 (SEQ ID NO:144), TAR15-8-508 (SEQ ID NO:145), TARl 5-8-509 (SEQ ID NO: 146), TARl 5-8-510 (SEQ ID NO: 147), TARl 5-8-511 (SEQ ID NO:148), TAR15-26-500 (SEQ ID NO:149), TAR15-26-501 (SEQ ID NO:150), TAR15-26-502 (SEQ ID NO:151), TAR15-26-503 (SEQ ID NO:152), TARl 5-26-504 (SEQ ID NO: 153), TARl 5-26-505 (SEQ ID NO: 154), TARl 5-26- 506 (SEQ ID NO: 155), TARl 5-26-507 (SEQ ID NO: 156), TARl 5-26-508 (SEQ ID NO: 157), TARl 5-26-509 (SEQ ID NO: 158), TARl 5-26-510 (SEQ ID NO: 159), TAR15-26-511 (SEQ ID NO: 160), TARl 5-26-512 (SEQ ID NO:161), TAR15-26- 513 (SEQ ID NO:162), TAR15-26-514 (SEQ ID NO:163), TAR15-26-515 (SEQ ID NO: 164), TARl 5-26-516 (SEQ ID NO: 165), TARl 5-26-517 (SEQ ID NO: 166), TARl 5-26-518 (SEQ ID NO: 167), TARl 5-26-519 (SEQ ID NO: 168), TARl 5-26- 520 (SEQ ID NO:169), TAR15-26-521 (SEQ ID NO:170), TAR15-26-522 (SEQ ID NO:171), TAR15-26-523 (SEQ ID NO:172), TAR15-26-524 (SEQ ID NO:173), TARl 5-26-525 (SEQ ID NO: 174), TARl 5-26-526 (SEQ ID NO: 175), TARl 5-26- 527 (SEQ ID NO: 176), TARl 5-26-528 (SEQ ID NO: 177), TARl 5-26-529 (SEQ ID NO: 178), TARl 5-26-530 (SEQ ID NO: 179), TARl 5-26-531 (SEQ ID NO: 180), TARl 5-26-532 (SEQ ID NO: 181), TARl 5-26-533 (SEQ ID NO: 182), TARl 5-26- 534 (SEQ ID NO: 183), TARl 5-26-535 (SEQ ID NO: 184), TARl 5-26-536 (SEQ ID NO: 185), TARl 5-26-537 (SEQ ID NO: 186), TARl 5-26-538 (SEQ ID NO: 187), TARl 5-26-539 (SEQ ID NO: 188), TARl 5-26-540 (SEQ ID NO: 189), TARl 5-26- 541 (SEQ ID NO:190), TAR15-26-542 (SEQ ID NO:191), TAR15-26-543 (SEQ ID NO: 192), TARl 5-26-544 (SEQ ID NO: 193), TARl 5-26-545 (SEQ ID NO: 194), TARl 5-26-546 (SEQ ID NO: 195), TARl 5-26-547 (SEQ ID NO: 196), TARl 5-26- 548 (SEQ ID NO: 197), and TARl 5-26-549 (SEQ ID NO: 198), TARl 5-26-550 (SEQ ID NO:539), and TARl 5-26-551 (SEQ ID NO:540); and further comprise an immunoglobulin single variable domain with binding specificity for EGFR that comprise an amino acid sequence that has at least about 85% amino acid sequence identity with the amino acid sequence of a dAb selected from the group consisting of DOM16-17 (SEQ ID NO:325), DOM16-18 (SEQ ID NO:326), DOM16-19 (SEQ ID NO:327), DOM16-20 (SEQ ID NO:328), DOM16-21 (SEQ ID NO:329), DOM16- 22 (SEQ ID NO:330), DOMl 6-23 (SEQ ID NO:331), DOMl 6-24 (SEQ ID
NO:332), DOM16-25 (SEQ ID NO:333), DOM16-26 (SEQ ID NO:334), DOM16- 27 (SEQ ID NO:335), DOM16-28 (SEQ ID NO:336), DOM16-29 (SEQ ID
NO:337), DOM16-30 (SEQ ID NO:338), DOM16-31 (SEQ ID NO:339), DOM16- 32 (SEQ ID NO:340), DOM16-33 (SEQ ID NO:341), DOM16-35 (SEQ ID
NO:342), DOM16-37 (SEQ ID NO:343), DOM16-38 (SEQ ID NO:344), DOM16- 39 (SEQ ID NO:345), DOM16-40 (SEQ ID NO:346), DOM16-41 (SEQ ID
NO:347), DOMl 6-42 (SEQ ID NO:348), DOMl 6-43 (SEQ ID NO:349), DOMl 6- 44 (SEQ ID NO:350), DOM16-45 (SEQ ID NO:351), DOM16-46 (SEQ ID
NO:352), DOM16-47 (SEQ ID NO:353), DOM16-48 (SEQ ID NO:354), DOM16- 49 (SEQ ID NO:355), DOM16-50 (SEQ ID NO:356), DOM16-59 (SEQ ID
NO:357), DOM16-60 (SEQ ID NO:358), DOM16-61 (SEQ ID NO:359), DOM16- 62 (SEQ ID NO:360), DOMl 6-63 (SEQ ID NO:361), DOMl 6-64 (SEQ ID
NO:362), DOM16-65 (SEQ ID NO:363), DOM16-66 (SEQ ID NO:364), DOM16- 67 (SEQ ID NO:365), DOMl 6-68 (SEQ ID NO:366), DOMl 6-69 (SEQ ID
NO:367), DOM16-70 (SEQ ID NO:368), DOM16-71 (SEQ ID NO:369), D0M16- 72 (SEQ ID NO:370), DOM16-73 (SEQ ID NO:371), DOM16-74 (SEQ ID
NO:372), DOMl 6-75 (SEQ ID NO:373), DOMl 6-76 (SEQ ID NO:374), DOMl 6- 77 (SEQ ID NO:375), DOMl 6-78 (SEQ ID NO:376), DOMl 6-79 (SEQ ID
NO:377), DOM16-80 (SEQ ID NO:378), DOM16-81 (SEQ ID NO:379), D0M16- 82 (SEQ ID NO:380), DOM16-83 (SEQ ID NO:381), DOM16-84 (SEQ ID NO:382), DOM16-85 (SEQ ID NO:383), DOM16-87 (SEQ ID NO:384), DOM16- 88 (SEQ ID NO:385), DOM16-89 (SEQ ID NO:386), DOM16-90 (SEQ ID
NO:387), DOM16-91 (SEQ ID NO:388), DOM16-92 (SEQ ID NO:389), DOM16- 94 (SEQ ID NO:390), DOM16-95 (SEQ ID NO:391), DOM16-96 (SEQ ID
NO:392), DOM16-97 (SEQ ID NO:393), DOM16-98 (SEQ ID NO:394), DOM16-
99 (SEQ ID NO:395), DOM16-100 (SEQ ID NO:396), DOMl 6-101 (SEQ ID NO:397), DOM16-102 (SEQ ID NO:398), DOM16-103 (SEQ ID NO:399), DOM16-104 (SEQ ID NO:400), DOM16-105 (SEQ ID NO:401), DOM16-106 (SEQ ID NO:402), DOM16-107 (SEQ ID NO:403), DOM16-108 (SEQ ID
NO:404), DOM16-109 (SEQ ID NO:405), DOM16-110 (SEQ ID NO:406), DOMl 6-111 (SEQ ID NO:407), DOM16-112 (SEQ ID NO:408), DOM16-113 (SEQ ID NO:409), DOM16-114 (SEQ ID NO:410), DOM16-115 (SEQ ID
NO:411), DOM16-116 (SEQ ID NO:412), DOM16-117 (SEQ ID NO:413), DOMl 6-118 (SEQ ID NO:414), DOM16-119 (SEQ ID NO:415), DOM16-39-6 (SEQ ID NO:416), DOMl 6-39-8 (SEQ ID NO:417), DOMl 6-39-34 (SEQ ID NO:418), DOMl 6-39-48 (SEQ ID NO:419), DOMl 6-39-87 (SEQ ID NO:420), DOM16-39-90 (SEQ ID NO:421), DOM16-39-96 (SEQ ID NO:422), DOM16-39-
100 (SEQ ID NO:423), DOM16-39-101 (SEQ ID NO:424), DOM16-39-102 (SEQ ID NO:425), DOMl 6-39-103 (SEQ ID NO:426), DOMl 6-39-104 (SEQ ID
NO:427), DOM16-39-105 (SEQ ID NO:428), DOM16-39-106 (SEQ ID NO:429), DOM16-39-107 (SEQ ID NO:430), DOM16-39-108 (SEQ ID NO:431), DOM16- 39-109 (SEQ ID NO:432), DOM16-39-110 (SEQ ID NO:433), DOM16-39-111 (SEQ ID NO:434), DOM16-39-112 (SEQ ID NO:435), DOM16-39-113 (SEQ ID NO:436), DOM16-39-114 (SEQ ID NO:437), DOM16-39-115 (SEQ ID NO:438), DOM16-39-116 (SEQ ID NO:439), DOM16-39-117 (SEQ ID NO:440), DOM16- 39-200 (SEQ ID NO:441), DOM16-39-201 (SEQ ID NO:442), DOMl 6-39-202 (SEQ ID NO:443), DOMl 6-39-203 (SEQ ID NO:444), DOMl 6-39-204 (SEQ ID NO:445), DOMl 6-39-205 (SEQ ID NO:446), DOMl 6-39-206 (SEQ ID NO:447), DOMl 6-39-207 (SEQ ID NO:448), DOMl 6-39-209 (SEQ ID NO:449), DOMl 6-52 (SEQ ID NO:450), NBl (SEQ ID NO:451), NB2 (SEQ ID NO:452), NB3 (SEQ ID NO:453), NB4 (SEQ ID NO:454), NB5 (SEQ ID NO:455), NB6 (SEQ ID NO:456), NB7 (SEQ ID NO:457), NB8 (SEQ ID NO:458), NB9 (SEQ ID NO:459), NBlO (SEQ ID NO:460), NBl 1 (SEQ ID NO:461), NB12 (SEQ ID NO:462), NB13 (SEQ ID NO:463), NB14 (SEQ ID NO:464), NB15 (SEQ ID NO:465), NB16 (SEQ ID NO:466), NB17 (SEQ ID NO:467), NBl 8 (SEQ ID NO:468), NB19 (SEQ ID
NO:469), NB20 (SEQ ID NO:470), NB21 (SEQ ID NO:471), and NB22 (SEQ ID NO:472).
For example, the ligand can comprise an immunoglobulin single variable domain with binding specificity for VEGF that comprises an amino acid sequence that has at least about 85% amino acid sequence identity with the amino acid sequence of a dAb selected from the group consisting of TARl 5-1 (SEQ ID
NOII OO), TAR15-3 (SEQ ID NOrIOl), TAR15-4 (SEQ ID NO:102), TAR15-9 (SEQ ID NO: 103), TARl 5-10 (SEQ ID NO: 104), TARl 5-11 (SEQ ID NO: 105), TAR15-12 (SEQ ID NO:106), TAR15-13 (SEQ ID NO:107), TAR15-14 (SEQ ID NO:108), TAR15-15 (SEQ ID NO: 109), TAR15-16 (SEQ ID NOrI lO), TAR15-17 . (SEQ ID NO:111), TAR15-18 (SEQ ID NO:112), TAR15-19 (SEQ ID NO:113), TAR15-20 (SEQ ID NOrI 14), TAR 15-22 (SEQ ID NOrI 15), TAR15-5 (SEQ ID NOrI 16), TARl 5-6 (SEQ ID NOrI 17), TARl 5-7 (SEQ ID NOrI 18), TARl 5-8 (SEQ ID NOrI 19), TAR15-23 (SEQ ID NO: 120), TAR15-24 (SEQ ID NO:121), TARl 5-25 (SEQ ID NO: 122), TARl 5-26 (SEQ ID NO: 123), TARl 5-27 (SEQ ID NO:124), TAR15-29 (SEQ ID NO:125), TAR15-30 (SEQ ID NO: 126), TAR15-6- 500 (SEQ ID NO:127), TAR15-6-501 (SEQ ID NO:128), TAR15-6-502 (SEQ ID NO:129), TAR15-6-503 (SEQ ID NO:130), TAR15-6-504 (SEQ ID NO:131), TARl 5-6-505 (SEQ ID NO: 132), TARl 5-6-506 (SEQ ID NO: 133), TARl 5-6-507 (SEQ ID NO:134), TAR15-6-508 (SEQ ID NO:135), TAR15-6-509 (SEQ ID
NO:136), TAR15-6-510 (SEQ ID NO:137), TAR15-8-500 (SEQ ID NO: 138), TAR15-8-501 (SEQ ID NO:139), TAR15-8-502 (SEQ ID NO: 140), TAR15-8-503 (SEQ ID NO:141), TAR15-8-505 (SEQ ID NO: 142), TAR15-8-506 (SEQ ID
NO:143), TAR15-8-507 (SEQ ID NO: 144), TAR15-8-508 (SEQ ID NO: 145), TARl 5-8-509 (SEQ ID NO: 146), TARl 5-8-510 (SEQ ID NO: 147), TARl 5-8-511 (SEQ ID NO:148), TAR15-26-500 (SEQ ID NO: 149), TAR15-26-501 (SEQ ID NO: 150), TARl 5-26-502 (SEQ ID NO:151), TAR15-26-503 (SEQ ID NO: 152), TARl 5-26-504 (SEQ ID NO: 153), TARl 5-26-505 (SEQ ID NO: 154), TARl 5-26- 506 (SEQ ID NO: 155), TARl 5-26-507 (SEQ ID NO: 156), TARl 5-26-508 (SEQ ID NO:157), TAR15-26-509 (SEQ ID NO:158), TAR15-26-510 (SEQ ID NO:159), TAR15-26-511 (SEQ ID NO: 160), TAR15-26-512 (SEQ ID NO: 161), TARl 5-26- 513 (SEQ ID NO:162), TAR15-26-514 (SEQ ID NO:163), TAR15-26-515 (SEQ ID NO: 164), TARl 5-26-516 (SEQ ID NO: 165), TARl 5-26-517 (SEQ ID NO: 166), TAR15-26-518 (SEQ ID NO: 167), TAR15-26-519 (SEQ ID NO:168), TAR15-26- 520 (SEQ ID NO: 169), TARl 5-26-521 (SEQ ID NO: 170), TARl 5-26-522 (SEQ ID NO:171), TAR15-26-523 (SEQ ID NO: 172), TAR15-26-524 (SEQ ID NO: 173), TARl 5-26-525 (SEQ ID NO: 174), TARl 5-26-526 (SEQ ID NO: 175), TARl 5-26- 527 (SEQ ID NO: 176), TARl 5-26-528 (SEQ ID NO: 177), TARl 5-26-529 (SEQ ID NO: 178), TARl 5-26-530 (SEQ ID NO: 179), TARl 5-26-531 (SEQ ID NO: 180), TARl 5-26-532 (SEQ ID NO: 181), TARl 5-26-533 (SEQ ID NO: 182), TARl 5-26- 534 (SEQ ID NO: 183), TARl 5-26-535 (SEQ ID NO: 184), TARl 5-26-536 (SEQ ID NO: 185), TARl 5-26-537 (SEQ ID NO: 186), TARl 5-26-538 (SEQ ID NO: 187), TARl 5-26-539 (SEQ ID NO: 188), TARl 5-26-540 (SEQ ID NO: 189), TARl 5-26- 541 (SEQ ID NO: 190), TARl 5-26-542 (SEQ ID NO: 191), TARl 5-26-543 (SEQ ID NO: 192), TARl 5-26-544 (SEQ ID NO: 193), TARl 5-26-545 (SEQ ID NO: 194), TARl 5-26-546 (SEQ ID NO: 195), TARl 5-26-547 (SEQ ID NO: 196), TARl 5-26- 548 (SEQ ID NO: 197), TARl 5-26-549 (SEQ ID NO: 198), TARl 5-26-550 (SEQ ID NO:539), and TARl 5-26-551 (SEQ ID NO:540); and further comprise an immunoglobulin single variable domain with binding specificity for EGFR that comprise an amino acid sequence that has at least about 85% amino acid sequence identity with the amino acid sequence of a dAb selected from the group consisting of DOM16-39-210 (SEQ ID NO:541), DOM16-39-211 (SEQ ID NO:542), DOM16- 39-212 (SEQ ID NO:543), DOM16-39-213 (SEQ ID NO:544), DOM16-39-214 (SEQ ID NO:545), DOMl 6-39-215 (SEQ ID NO:546), DOMl 6-39-216 (SEQ ID NO:547), DOMl 6-39-217 (SEQ ID NO:548), DOMl 6-39-218 (SEQ ID NO:549), DOMl 6-39-219 (SEQ ID NO:550), DOMl 6-39-220 (SEQ ID NO:551), DOMl 6- 39-221 (SEQ ID NO:552), DOM16-39-222 (SEQ ID NO:553), DOM16-39-223 (SEQ ID NO:554), DOM16-39-224 (SEQ ID NO:555), DOM16-39-225 (SEQ ID NO:556), DOMl 6-39-226 (SEQ ID NO:557), DOMl 6-39-227 (SEQ ID NO:558), DOMl 6-39-228 (SEQ ID NO:559), DOMl 6-39-229 (SEQ ID NO:560), DOMl 6- 39-230 (SEQ ID NO:561), DOMl 6-39-231 (SEQ ID NO:562), DOMl 6-39-232 (SEQ ID NO:563), DOMl 6-39-233 (SEQ ID NO:564), DOMl 6-39-234 (SEQ ID NO:565), DOM16-39-235 (SEQ ID NO:566), DOM16-39-500 (SEQ ID NO:725), DOMl 6-39-502(SEQ ID NO:726), DOMl 6-39-503 (SEQ ID NO:567), DOMl 6-39- 504 (SEQ ID NO:568), DOMl 6-39-505 (SEQ ID NO:569), DOMl 6-39-506 (SEQ ID NO:570), DOM16-39-507 (SEQ ID NO:571), DOM16-39-508 (SEQ ID
NO:572), DOMl 6-39-509 (SEQ ID NO:573), DOMl 6-39-510 (SEQ ID NO:574), DOM16-39-511 (SEQ ID NO:575), DOM16-39-512 (SEQ ID NO:576), DOM16- 39-521 (SEQ ID NO:577), DOM16-39-522 (SEQ ID NO:578), DOM16-39-523 (SEQ ID NO:579), DOMl 6-39-524 (SEQ ID NO:580), DOMl 6-39-527 (SEQ ID NO:581), DOM16-39-525 (SEQ ID NO:582), DOM16-39-526 (SEQ ID NO:583), DOM16-39-540 (SEQ ID NO:584), DOM16-39-541 (SEQ ID NO:585), DOM16- 39-542 (SEQ ID NO:586), DOM16-39-543 (SEQ ID NO:587), DOM16-39-544 (SEQ ID NO:588), DOM16-39-545 (SEQ ID NO:589), DOM16-39-550 (SEQ ID NO:590), DOM16-39-551 (SEQ ID NO:591), DOM16-39-552 (SEQ ID NO:592), DOM16-39-553 (SEQ ID NO:593), DOM16-39-554 (SEQ ID NO:594), DOM16- 39-555 (SEQ ID NO:595), DOM16-39-561 (SEQ ID NO:596), DOM16-39-562 (SEQ ID NO:597), DOM16-39-563 (SEQ ID NO:598), DOM16-39-564 (SEQ ID NO:599), DOM16-39-571 (SEQ ID NO:600), DOM16-39-572 (SEQ ID NO:601), DOM16-39-573 (SEQ ID NO:602), DOM16-39-574 (SEQ ID NO:603), DOM16- 39-580 (SEQ ID NO:604), DOM16-39-591 (SEQ ID NO:605), DOM16-39-592 (SEQ ID NO:606), DOM16-39-593 (SEQ ID NO:607), DOM16-39-601 (SEQ ID NO:608), DOM16-39-602 (SEQ ID NO:609), DOM16-39-603 (SEQ ID NO:610), DOMl 6-39-604 (SEQ ID NO:611), DOMl 6-39-605 (SEQ ID NO:612), DOMl 6- 39-607 (SEQ ID NO:613), DOM16-39-611 (SEQ ID NO:614), DOM16-39-612 (SEQ ID NO:615), DOM16-39-613 (SEQ ID NO:616), DOM16-39-614 (SEQ ID NO:617), DOM16-39-615 (SEQ ID NO:618), DOM16-39-616 (SEQ ID NO:619), DOM16-39-617 (SEQ ID NO:620), DOM16-39-618 (SEQ ID NO:621), and DOMl 6-39-619 (SEQ ID NO:622).
In some embodiments, the ligand has binding specificity for VEGF and for EGFR and comprises at least one immunoglobulin single variable domain with binding specificity for VEGF and at least one immunoglobulin single variable domain with binding specificity for EGFR, wherein an immunoglobulin single variable domain with binding specificity for VEGF competes for binding to VEGF with an anti-VEGF domain antibody (dAb) selected from the group consisting of TAR15-1 (SEQ ID NOrIOO), TAR15-3 (SEQ ID NOrIOl), TAR15-4 (SEQ ID NO:102), TAR15-9 (SEQ ID NO:103), TAR15-10 (SEQ ID NO:104), TAR15-11 (SEQ ID NO:105), TAR15-12 (SEQ ID NO:106), TAR15-13 (SEQ ID NO: 107), TAR15-14 (SEQ ID NO:108), TAR15-15 (SEQ ID NO:109), TAR15-16 (SEQ ID NO:110), TAR15-17 (SEQ ID NO:111), TAR15-18 (SEQ ID NO:112), TAR15-19 (SEQ ID NO: 113), TARl 5-20 (SEQ ID NO:114), TAR 15-22 (SEQ ID NO:115), TARl 5-5 (SEQ ID NO: 116), TARl 5-6 (SEQ ID NO: 117), TARl 5-7 (SEQ ID NO:118), TAR15-8 (SEQ E) NOrIl 9), TAR15-23 (SEQ ID NO:120), TAR15-24 (SEQ ID NO: 121), TARl 5-25 (SEQ ID NO: 122), TARl 5-26 (SEQ ID NO: 123), TAR15-27 (SEQ ID NOr 124), TAR15-29 (SEQ ID NO:125), TAR15-30 (SEQ ID NO:126), TAR15-6-500 (SEQ ID NOrl27), TAR15-6-501 (SEQ ID NO:128), TAR15-6-502 (SEQ ID NO:129), TAR15-6-503 (SEQ ID NO:130), TAR15-6-504 (SEQ ID NO:131), TAR15-6-505 (SEQ ID NOr 132), TAR15-6-506 (SEQ ID NO:133), TAR15-6-507 (SEQ E) NO: 134), TAR15-6-508 (SEQ ID NO:135), TARl 5-6-509 (SEQ ID NO: 136), TARl 5-6-510 (SEQ ID NO: 137), TARl 5-8-500 (SEQ ID NO:138), TAR15-8-501 (SEQ ID NO:139), TAR15-8-502 (SEQ ID NO:140), TAR15-8-503 (SEQ ID NOrHl), TAR15-8-505 (SEQ ID NO: 142), TAR15-8-506 (SEQ ID NO:143), TAR15-8-507 (SEQ ID NO: 144), TAR15-8-508 (SEQ ID NO:145), TAR15-8-509 (SEQ ID NO:146), TAR15-8-510 (SEQ ID NO:147), TARl 5-8-511 (SEQ E) NO: 148), TAR15-26-500 (SEQ E) NO: 149), TARl 5-26-501 (SEQ ID NO: 150), TARl 5-26-502 (SEQ ID NOrI 51), TARl 5-26- 503 (SEQ ID NO: 152), TARl 5-26-504 (SEQ ID NO: 153), TARl 5-26-505 (SEQ ID NO: 154), TARl 5-26-506 (SEQ ID NO: 155), TARl 5-26-507 (SEQ ID NO: 156), TARl 5-26-508 (SEQ ID NO: 157), TARl 5-26-509 (SEQ ID NO: 158), TARl 5-26- 510 (SEQ ID NO: 159), TARl 5-26-511 (SEQ ID NO: 160), TARl 5-26-512 (SEQ ID NO:161), TAR15-26-513 (SEQ ID NO:162), TAR15-26-514 (SEQ ID NO: 163), TARl 5-26-515 (SEQ ID NO: 164), TARl 5-26-516 (SEQ ID NO: 165), TARl 5-26- 517 (SEQ ID NO:166), TAR15-26-518 (SEQ ID NO: 167), TARl 5-26-519 (SEQ ID NO:168), TAR15-26-520 (SEQ E) NO: 169), TAR15-26-521 (SEQ ID NO: 170), TARl 5-26-522 (SEQ ID NOrI 71), TARl 5-26-523 (SEQ ID NO: 172), TARl 5-26- 524 (SEQ ID NO: 173), TARl 5-26-525 (SEQ ID NO: 174), TARl 5-26-526 (SEQ ID NO: 175), TARl 5-26-527 (SEQ ID NO: 176), TARl 5-26-528 (SEQ ID NO: 177), TARl 5-26-529 (SEQ ID NO: 178), TARl 5-26-530 (SEQ ID NO: 179), TARl 5-26- 531 (SEQ ID NO:180), TAR15-26-532 (SEQ ID NO:181), TAR15-26-533 (SEQ ID NO:182), TAR15-26-534 (SEQ ID NO:183), TAR15-26-535 (SEQ ID NO:184), TARl 5-26-536 (SEQ ID NO: 185), TARl 5-26-537 (SEQ ID NO: 186), TARl 5-26- 538 (SEQ ID NO:187), TAR15-26-539 (SEQ ID NO:188), TAR15-26-540 (SEQ ID NO:189), TAR15-26-541 (SEQ ID NO:190), TAR15-26-542 (SEQ ID NO:191), TAR15-26-543 (SEQ ID NO:192), TAR15-26-544 (SEQ ID NO:193), TAR15-26- 545 (SEQ ID NO: 194), TARl 5-26-546 (SEQ ID NO: 195), TARl 5-26-547 (SEQ ID NO:196), TAR15-26-548 (SEQ ID NO:197), and TAR15-26-549 (SEQ ID NO:198), TARl 5-26-550 (SEQ ID NO:539), and TARl 5-26-551 (SEQ ID NO:540); and an immunoglobulin single variable domain with binding specificity for EGFR competes for binding to EGFR with cetuximab.
For example, the immunoglobulin single variable domain with binding specificity for VEGF can comprise an amino acid sequence that has at least about 85% amino acid sequence identity with the amino acid sequence of a dAb selected from the group consisting of TAR15-1 (SEQ ID NO:100), TAR15-3 (SEQ ID NO:101), TAR15-4 (SEQ ID NO:102), TAR15-9 (SEQ ID NO:103), TAR15-10 (SEQ ID NO:104), TAR15-11 (SEQ ID NO:105), TAR15-12 (SEQ ID NO:106), TAR15-13 (SEQ ID NO:107), TAR15-14 (SEQ ID NO:108), TAR15-15 (SEQ ID NO:109), TAR15-16 (SEQ ID NO:110), TAR15-17 (SEQ ID NO:111), TAR15-18 (SEQ ID NO:112), TAR15-19 (SEQ ID NO:113), TAR15-20 (SEQ ID NO:114), , TAR 15-22 (SEQ ID NO: 115), TARl 5-5 (SEQ ID NO: 116), TARl 5-6 (SEQ ID NO: 117), TARl 5-7 (SEQ ID NO: 118), TARl 5-8 (SEQ ID NO: 119), TARl 5-23 (SEQ ID NO:120), TAR15-24 (SEQ ID NO:121), TAR15-25 (SEQ ID NO:122), TAR15-26 (SEQ ID NO:123), TAR15-27 (SEQ ID NO:124), TAR15-29 (SEQ ID NO:125), TAR15-30 (SEQ ID NO:126), TAR15-6-500 (SEQ ID NO:127), TAR15- 6-501 (SEQ ID NO:128), TAR15-6-502 (SEQ ID NO:129), TAR15-6-503 (SEQ ID NO:130), TAR15-6-504 (SEQ ID NO:131), TAR15-6-505 (SEQ ID NO:132),
TARl 5-6-506 (SEQ ID NO: 133), TARl 5-6-507 (SEQ ID NO: 134), TARl 5-6-508 (SEQ ID NO: 135), TARl 5-6-509 (SEQ ID NO: 136), TARl 5-6-510 (SEQ ID NO:137), TAR15-8-500 (SEQ ID NO:138), TAR15-8-501 (SEQ ID NO:139), TAR15-8-502 (SEQ ID NO: 140), TAR15-8-503 (SEQ ID NO:141), TAR15-8-505 (SEQ ID NO:142), TAR15-8-506 (SEQ ID NO:143), TAR15-8-507 (SEQ ID NO:144), TAR15-8-508 (SEQ ID NO: 145), TAR15-8-509 (SEQ ID NO: 146), TAR15-8-510 (SEQ ID NO:147), TAR15-8-511 (SEQ ID NO:148), TAR15-26-500 (SEQ ID NO:149), TAR15-26-501 (SEQ ID NO:150), TAR15-26-502 (SEQ ID NO:151), TAR15-26-503 (SEQ ID NO:152), TAR15-26-504 (SEQ ID NO:153), TARl 5-26-505 (SEQ ID NO: 154), TARl 5-26-506 (SEQ ID NO: 155), TARl 5-26- 507 (SEQ ID NO: 156), TARl 5-26-508 (SEQ ID NO: 157), TARl 5-26-509 (SEQ ID NO: 158), TARl 5-26-510 (SEQ ID NO: 159), TARl 5-26-511 (SEQ ID NO: 160), TAR15-26-512 (SEQ ID NO:161), TAR15-26-513 (SEQ ID NO: 162), TAR15-26- 514 (SEQ ID NO:163), TAR15-26-515 (SEQ ID NO: 164), TAR15-26-516 (SEQ ID NO: 165), TARl 5-26-517 (SEQ ID NO: 166), TARl 5-26-518 (SEQ ID NO: 167), TARl 5-26-519 (SEQ ID NO: 168), TARl 5-26-520 (SEQ ID NO: 169), TARl 5-26- 521 (SEQ ID NO:170), TAR15-26-522 (SEQ ID NO:171), TAR15-26-523 (SEQ ID NO: 172), TARl 5-26-524 (SEQ ID NO: 173), TARl 5-26-525 (SEQ ID NO: 174), TARl 5-26-526 (SEQ ID NO: 175), TARl 5-26-527 (SEQ ID NO: 176), TARl 5-26- 528 (SEQ ID NO: 177), TARl 5-26-529 (SEQ ID NO: 178), TARl 5-26-530 (SEQ ID NO:179), TAR15-26-531 (SEQ ID NO:180), TAR15-26-532 (SEQ ID NO:181), TARl 5-26-533 (SEQ ID NO: 182), TARl 5-26-534 (SEQ ID NO: 183), TARl 5-26- 535 (SEQ ID NO:184), TAR15-26-536 (SEQ ID NO:185), TAR15-26-537 (SEQ ID NO: 186), TARl 5-26-538 (SEQ ID NO: 187), TARl 5-26-539 (SEQ ID NO:188), TAR15-26-540 (SEQ ID NO:189), TAR15-26-541 (SEQ ID NO:190), TAR15-26- 542 (SEQ ID NO:191), TAR15-26-543 (SEQ ID NO: 192), TAR15-26-544 (SEQ ID NO:193), TAR15-26-545 (SEQ ID NO: 194), TAR15-26-546 (SEQ ID NO:195), TARl 5-26-547 (SEQ ID NO: 196), TARl 5-26-548 (SEQ ID NO: 197), and TARl 5- 26-549 (SEQ ID NO: 198), TARl 5-26-550 (SEQ ID NO:539), and TARl 5-26-551 (SEQ ID NO:540).
In other embodiments, the ligand has binding specificity for VEGF and for EGFR and comprises at least one immunoglobulin single variable domain with binding specificity for VEGF and at least one immunoglobulin single variable domain with binding specificity for EGFR, wherein an immunoglobulin single variable domain with binding specificity for VEGF competes for binding to VEGF with bevacizumab and/or antibody 2C3 (ATCC Accession No. PTA 1595); and an immunoglobulin single variable domain with binding specificity for EGFR competes for binding to EGFR with an anti-EGFR domain antibody (dAb) selected from the group consisting of DOMl 6-17 (SEQ ID NO:325), DOMl 6-18 (SEQ ID NO:326), DOMl 6-19 (SEQ ID NO:327), DOMl 6-20 (SEQ ID NO:328), DOMl 6- 21 (SEQ ID NO:329), DOM16-22 (SEQ ID NO:330), DOM16-23 (SEQ ID NO:331), DOM16-24 (SEQ ID NO:332), DOM16-25 (SEQ ID NO:333), D0M16- 26 (SEQ ID NO:334), DOMl 6-27 (SEQ ID NO:335), DOMl 6-28 (SEQ ID NO:336), DOM16-29 (SEQ ID NO:337), DOM16-30 (SEQ ID NO:338), D0M16- 31 (SEQ ID NO:339), DOM16-32 (SEQ ID NO:340), DOM16-33 (SEQ ID NO:341), DOM16-35 (SEQ ID NO:342), DOM16-37 (SEQ ID NO:343), DOM16- 38 (SEQ ID NO:344), DOMl 6-39 (SEQ ID NO:345), DOMl 6-40 (SEQ ID NO:346), DOMl 6-41 (SEQ ID NO:347), DOMl 6-42 (SEQ ID NO:348), DOMl 6- 43 (SEQ ID NO:349), DOM16-44 (SEQ ID NO:350), DOM16-45 (SEQ ID
NO:351), DOM16-46 (SEQ ID NO:352), DOM16-47 (SEQ ID NO:353), D0M16- 48 (SEQ ID NO:354), DOM16-49 (SEQ ID NO:355), DOM16-50 (SEQ ID NO:356), DOM16-59 (SEQ ID NO:357), DOM16-60 (SEQ ID NO:358), D0M16- 61 (SEQ ID NO:359), DOM16-62 (SEQ ID NO:360), DOM16-63 (SEQ ID NO:361), DOM16-64 (SEQ ID NO:362), DOM16-65 (SEQ ID NO:363), DOM16- 66 (SEQ ID NO:364), DOM16-67 (SEQ ID NO:365), DOM16-68 (SEQ ID NO:366), DOMl 6-69 (SEQ ID NO:367), DOMl 6-70 (SEQ ID NO:368), DOMl 6- 71 (SEQ ID NO:369), DOM16-72 (SEQ ID NO:370), DOM16-73 (SEQ ID NO:371), DOM16-74 (SEQ ID NO:372), DOM16-75 (SEQ ID NO:373), D0M16- 76 (SEQ ID NO:374), DOMl 6-77 (SEQ ID NO:375), DOMl 6-78 (SEQ ID
NO:376), DOMl 6-79 (SEQ ID NO:377), DOMl 6-80 (SEQ ID NO:378), DOMl 6- 81 (SEQ ID NO:379), DOM16-82 (SEQ ID NO:380), DOM16-83 (SEQ ID NO:381), DOM16-84 (SEQ ID NO:382), DOM16-85 (SEQ ID NO:383), DOM16- 87 (SEQ ID NO:384), DOM16-88 (SEQ ID NO:385), DOM16-89 (SEQ ID NO:386), DOM16-90 (SEQ ID NO:387), DOM16-91 (SEQ ID NO:388), D0M16- 92 (SEQ ID NO:389), DOMl 6-94 (SEQ ID NO:390), DOMl 6-95 (SEQ ID NO:391), DOM16-96 (SEQ ID NO:392), DOM16-97 (SEQ ID NO:393), D0M16- 98 (SEQ ID NO:394), DOMl 6-99 (SEQ ID NO:395), DOMl 6-100 (SEQ ID NO:396), DOMl 6-101 (SEQ ID NO:397), DOM16-102 (SEQ ID NO:398), DOMl 6-103 (SEQ ID NO:399), DOMl 6-104 (SEQ ID NO:400), DOMl 6-105 (SEQ ID NO:401), DOM16-106 (SEQ ID NO:402), DOM16-107 (SEQ ID
NO:403), DOM16-108 (SEQ ID NO:404), DOM16-109 (SEQ ID NO:405), DOMl 6-110 (SEQ ID NO:406), DOM16-111 (SEQ ID NO:407), DOM16-112 (SEQ ID NO:408), DOM16-113 (SEQ ID NO:409), DOM16-114 (SEQ ID
NO:410), DOM16-115 (SEQ ID NO:411), DOM16-116 (SEQ ID NO:412), DOM16-117 (SEQ ID NO:413), DOM16-118 (SEQ ID NO:414), DOM16-119 (SEQ ID NO:415), DOM16-39-6 (SEQ ID NO:416), DOM16-39-8 (SEQ ID NO:417), DOM16-39-34 (SEQ ID NO:418), DOM16-39-48 (SEQ ID NO:419), DOM16-39-87 (SEQ ID NO:420), DOM16-39-90 (SEQ ID NO:421), DOM16-39- 96 (SEQ ID NO:422), DOM16-39-100 (SEQ ID NO:423), DOM16-39-101 (SEQ ID NO:424), DOMl 6-39-102 (SEQ ID NO:425), DOMl 6-39-103 (SEQ ID NO:426), DOM16-39-104 (SEQ ID NO:427), DOM16-39-105 (SEQ ID NO:428), DOM16- 39-106 (SEQ ID NO:429), DOM16-39-107 (SEQ ID NO:430), DOM16-39-108 (SEQ ID NO:431), DOM16-39-109 (SEQ ID NO:432), DOM16-39-110 (SEQ ID NO:433), DOMl 6-39-111 (SEQ ID NO:434), DOM16-39-112 (SEQ ID NO:435), DOM16-39-113 (SEQ ID NO:436), DOM16-39-114 (SEQ ID NO:437), DOM16- 39-115 (SEQ ID NO:438), DOM16-39-116 (SEQ ID NO:439), DOM16-39-117 (SEQ ID NO:440), DOM16-39-200 (SEQ ID NO:441), DOM16-39-201 (SEQ ID NO:442), DOM16-39-202 (SEQ ID NO:443), DOM16-39-203 (SEQ ID NO:444), DOM16-39-204 (SEQ ID NO:445), DOM16-39-205 (SEQ ID NO:446), DOM16- 39-206 (SEQ ID NO:447), DOM16-39-207 (SEQ ID NO:448), DOM16-39-209 (SEQ ID NO:449), DOM16-52 (SEQ ID NO:450), NBl (SEQ ID NO:451), NB2 (SEQ ID NO:452), NB3 (SEQ ID NO:453), NB4 (SEQ ID NO:454), NB5 (SEQ ID NO:455), NB6 (SEQ ID NO:456), NB7 (SEQ ID NO:457), NB8 (SEQ ID NO:458), NB9 (SEQ ID NO:459), NBlO (SEQ ID NO:460), NBI l (SEQ ID NO:461), NB12 (SEQ ID NO:462), NB 13 (SEQ ID NO:463), NB14 (SEQ ID NO:464), NB 15 (SEQ ID NO:465), NB16 (SEQ ID NO:466), NB17 (SEQ ID NO:467), NB18 (SEQ ID NO:468), NB19 (SEQ ID NO:469), NB20 (SEQ ID NO:470), NB21 (SEQ ID NO:471), and NB22 (SEQ ID NO:472). In other embodiments, the ligand has binding specificity for VEGF and for EGFR and comprises at least one immunoglobulin single variable domain with binding specificity for VEGF and at least one immunoglobulin single variable domain with binding specificity for EGFR, wherein an immunoglobulin single variable domain with binding specificity for VEGF competes for binding to VEGF with bevacizumab and/or antibody 2C3 (ATCC Accession No. PTA 1595); and an immunoglobulin single variable domain with binding specificity for EGFR competes for binding to EGFR with an anti-EGFR domain antibody (dAb) selected from the group consisting of DOM16-39-210 (SEQ ID NO:541), DOM16-39-211 (SEQ ID NO:542), DOM16-39-212 (SEQ ID NO:543), DOM16-39-213 (SEQ ID NO:544), DOM16-39-214 (SEQ ID NO:545), DOM16-39-215 (SEQ ID NO:546), DOM16-39-216 (SEQ ID NO:547), DOM16-39-217 (SEQ ID NO:548), DOM16- 39-218 (SEQ ID NO:549), DOM16-39-219 (SEQ ID NO:550), DOM16-39-220 (SEQ ID NO:551), DOM16-39-221 (SEQ ID NO:552), DOM16-39-222 (SEQ ID NO:553), DOMl 6-39-223 (SEQ ID NO:554), DOMl 6-39-224 (SEQ ID NO:555), DOM16-39-225 (SEQ ID NO:556), DOM16-39-226 (SEQ ID NO:557), D0M16- 39-227 (SEQ ID NO:558), DOMl 6-39-228 (SEQ ID NO:559), DOMl 6-39-229 (SEQ ID NO:560), DOM16-39-230 (SEQ ID NO:561), DOM16-39-231 (SEQ ID NO:562), DOMl 6-39-232 (SEQ ID NO:563), DOMl 6-39-233 (SEQ ID NO:564), DOMl 6-39-234 (SEQ ID NO:565), DOMl 6-39-235 (SEQ ID NO:566), DOMl 6- 39-500 (SEQ ID NO:725), DOM16-39-502(SEQ ID NO:726), DOM16-39-503 (SEQ ID NO:567), DOM16-39-504 (SEQ ID NO:568), DOM16-39-505 (SEQ ID NO:569), DOM16-39-506 (SEQ ID NO:570), DOM16-39-507 (SEQ ID NO:571), DOMl 6-39-508 (SEQ ID NO:572), DOMl 6-39-509 (SEQ ID NO:573), DOMl 6- 39-510 (SEQ ID NO:574), DOM16-39-511 (SEQ ID NO:575), DOM16-39-512 (SEQ ID NO:576), DOM16-39-521 (SEQ ID NO:577), DOM16-39-522 (SEQ ID NO:578), DOM16-39-523 (SEQ ID NO:579), DOM16-39-524 (SEQ ID NO:580), DOM16-39-527 (SEQ ID NO:581), DOM16-39-525 (SEQ ID NO:582), D0M16- 39-526 (SEQ ID NO:583), DOM16-39-540 (SEQ ID NO:584), DOM16-39-541 (SEQ ID NO:585), DOM16-39-542 (SEQ ID NO:586), DOM16-39-543 (SEQ ID NO:587), DOM16-39-544 (SEQ ID NO:588), DOM16-39-545 (SEQ ID NO:589), DOM16-39-550 (SEQ ID NO:590), DOM16-39-551 (SEQ ID NO:591), D0M16- 39-552 (SEQ ID NO:592), DOM16-39-553 (SEQ ID NO:593), DOM16-39-554 (SEQ ID NO:594), DOM16-39-555 (SEQ ID NO:595), DOM16-39-561 (SEQ ID NO:596), DOM16-39-562 (SEQ ID NO:597), DOM16-39-563 (SEQ ID NO:598), DOM16-39-564 (SEQ ID NO:599), DOM16-39-571 (SEQ ID NO:600), DOM16- 39-572 (SEQ ID NO:601), DOM16-39-573 (SEQ ID NO:602), DOM16-39-574 (SEQ ID NO:603), DOM16-39-580 (SEQ ID NO:604), DOM16-39-591 (SEQ ID NO:605), DOM16-39-592 (SEQ ID NO:606), DOM16-39-593 (SEQ ID NO:607), DOM16-39-601 (SEQ ID NO:608), DOM16-39-602 (SEQ ID NO:609), DOM16- 39-603 (SEQ ID NO:610), DOM16-39-604 (SEQ ID NO:611), DOM16-39-605 (SEQ ID NO:612), DOM16-39-607 (SEQ ID NO:613), DOM16-39-611 (SEQ ID NO:614), DOM16-39-612 (SEQ ID NO:615), DOM16-39-613 (SEQ ID NO:616), DOM16-39-614 (SEQ ID NO:617), DOMl 6-39-615 (SEQ ID NO:618), DOM16- 39-616 (SEQ ID NO:619), DOM16-39-617 (SEQ ID NO:620), DOM16-39-618 (SEQ ID NO:621), and DOM16-39-619 (SEQ ID NO:622).
For example, the immunoglobulin single variable domain with binding specificity for EGFR can comprise an amino acid sequence that has at least about 85% amino acid sequence identity with the amino acid sequence of a dAb selected from the group consisting of DOM16-17 (SEQ ID NO:325), DOM16-18 (SEQ ID NO:326), DOM16-19 (SEQ ID NO:327), DOM16-20 (SEQ ID NO:328), DOM16- 21 (SEQ ID NO:329), DOM16-22 (SEQ ID NO:330), DOM16-23 (SEQ ID
NO:331), DOM16-24 (SEQ ID NO:332), DOM16-25 (SEQ ID NO:333), DOM16- 26 (SEQ ID NO:334), DOM16-27 (SEQ ID NO:335), DOM16-28 (SEQ ID NO:336), DOM16-29 (SEQ ID NO:337), DOM16-30 (SEQ ID NO:338), DOM16- 31 (SEQ ID NO:339), DOMl 6-32 (SEQ ID NO:340), DOMl 6-33 (SEQ ID NO:341), DOM16-35 (SEQ ID NO:342), DOM16-37 (SEQ ID NO:343), D0M16- 38 (SEQ ID NO:344), DOMl 6-39 (SEQ ID NO:345), DOMl 6-40 (SEQ ID NO:346), DOM16-41 (SEQ ID NO:347), DOM16-42 (SEQ ID NO:348), D0M16- 43 (SEQ ID NO:349), DOM16-44 (SEQ ID NO:350), DOM16-45 (SEQ ID NO:351), DOM16-46 (SEQ ID NO:352), DOM16-47 (SEQ ID NO:353), D0M16- 48 (SEQ ID NO:354), DOMl 6-49 (SEQ ID NO:355), DOMl 6-50 (SEQ ID
NO:356), DOM16-59 (SEQ ID NO:357), DOM16-60 (SEQ ID NO:358), D0M16- 61 (SEQ ID NO:359), DOM16-62 (SEQ ID NO:360), DOM16-63 (SEQ ID NO:361), DOM16-64 (SEQ ID NO:362), DOM16-65 (SEQ ID NO:363), DOM16- 66 (SEQ ID NO:364), DOMl 6-67 (SEQ ID NO:365), DOMl 6-68 (SEQ ID
NO:366), DOM16-69 (SEQ ID NO:367), DOM16-70 (SEQ ID NO:368), DOM16- 71 (SEQ ID NO:369), DOMl 6-72 (SEQ ID NO:370), DOMl 6-73 (SEQ ID
NO:371), DOMl 6-74 (SEQ ID NO:372), DOMl 6-75 (SEQ ID NO:373), DOMl 6- 76 (SEQ ID NO:374), DOMl 6-77 (SEQ ID NO:375), DOMl 6-78 (SEQ ID
NO:376), DOMl 6-79 (SEQ ID NO:377), DOMl 6-80 (SEQ ID NO:378), DOMl 6- 81 (SEQ ID NO:379), DOMl 6-82 (SEQ ID NO:380), DOMl 6-83 (SEQ ID
NO:381), DOM16-84 (SEQ ID NO:382), DOM16-85 (SEQ ID NO:383), D0M16- 87 (SEQ ID NO:384), DOMl 6-88 (SEQ ID NO:385), DOMl 6-89 (SEQ ID
NO:386), DOM16-90 (SEQ ID NO:387), DOM16-91 (SEQ ID NO:388), D0M16- 92 (SEQ ID NO:389), DOM16-94 (SEQ ID NO:390), DOM16-95 (SEQ ID
NO:391), DOM16-96 (SEQ ID NO:392), DOM16-97 (SEQ ID NO:393), D0M16- 98 (SEQ ID NO:394), DOM16-99 (SEQ ID NO:395), DOM16-100 (SEQ ID NO:396), DOM16-101 (SEQ ID NO:397), DOM16-102 (SEQ ID NO:398), DOMl 6-103 (SEQ ID NO:399), DOMl 6-104 (SEQ ID NO:400), DOMl 6-105 (SEQ ID NO:401), DOM16-106 (SEQ ID NO:402), DOM16-107 (SEQ ID
NO:403), DOMl 6-108 (SEQ ID NO:404), DOMl 6-109 (SEQ ID NO:405), DOMl 6-110 (SEQ ID NO:406), DOMl 6-111 (SEQ ID NO:407), DOMl 6-112 (SEQ ID NO:408), DOM16-113 (SEQ ID NO:409), DOM16-114 (SEQ ID
NO:410), DOM16-115 (SEQ ID NO:411), DOM16-116 (SEQ ID NO:412), DOM16-117 (SEQ ID NO:413), DOM16-118 (SEQ ID NO:414), DOMl 6-119 (SEQ ID NO:415), DOM16-39-6 (SEQ ID NO:416), DOM16-39-8 (SEQ ID NO:417), DOM16-39-34 (SEQ ID NO:418), DOM16-39-48 (SEQ ID NO:419), DOM16-39-87 (SEQ ID NO:420), DOM16-39-90 (SEQ ID NO:421), DOM16-39- 96 (SEQ ID NO:422), DOM16-39-100 (SEQ ID NO:423), DOM16-39-101 (SEQ ID NO:424), DOM16-39-102 (SEQ ID NO:425), DOM16-39-103 (SEQ ID NO:426), DOM16-39-104 (SEQ ID NO:427), DOM16-39-105 (SEQ ID NO:428), DOM16- 39-106 (SEQ ID NO:429), DOM16-39-107 (SEQ ID NO:430), DOM16-39-108 (SEQ ID NO:431), DOM16-39-109 (SEQ ID NO:432), DOM16-39-110 (SEQ ID NO:433), DOM16-39-111 (SEQ ID NO:434), DOM16-39-112 (SEQ ID NO:435), DOM16-39-113 (SEQ ID NO:436), DOM16-39-114 (SEQ ID NO:437), DOM16- 39-115 (SEQ ID NO:438), DOM16-39-116 (SEQ ID NO:439), DOMl 6-39-117 (SEQ ID NO:440), DOM16-39-200 (SEQ ID NO:441), DOM16-39-201 (SEQ ID NO:442), DOMl 6-39-202 (SEQ ID NO:443), DOMl 6-39-203 (SEQ ID NO:444), DOMl 6-39-204 (SEQ ID NO:445), DOMl 6-39-205 (SEQ ID NO:446), DOMl 6- 39-206 (SEQ ID NO:447), DOMl 6-39-207 (SEQ ID NO:448), DOMl 6-39-209 (SEQ ID NO:449), DOM16-52 (SEQ ID NO:450), NBl (SEQ ID NO:451), NB2 (SEQ ID NO:452), NB3 (SEQ ID NO:453), NB4 (SEQ ID NO:454), NB5 (SEQ ID NO:455), NB6 (SEQ ID NO:456), NB7 (SEQ ID NO:457), NB8 (SEQ ID NO:458), NB9 (SEQ ID NO:459), NBlO (SEQ ID NO:460), NBl 1 (SEQ ID NO:461), NB12 (SEQ ID NO:462), NB13 (SEQ ID NO:463), NB14 (SEQ ID NO:464), NB15 (SEQ ID NO:465), NB16 (SEQ ID NO:466), NB17 (SEQ ID NO:467), NB18 (SEQ ID NO:468), NB19 (SEQ ID NO:469), NB20 (SEQ ID NO:470), NB21 (SEQ ID NO:471), and NB22 (SEQ ID NO:472).
For example, the immunoglobulin single variable domain with binding specificity for EGFR can comprise an amino acid sequence that has at least about 85% amino acid sequence identity with the amino acid sequence of a dAb selected from the group consisting of DOM16-39-210 (SEQ ID NO:541), DOM16-39-211 (SEQ ID NO:542), DOM16-39-212 (SEQ ID NO:543), DOM16-39-213 (SEQ ID NO:544), DOM16-39-214 (SEQ ID NO:545), DOM16-39-215 (SEQ ID NO:546), DOM16-39-216 (SEQ ID NO:547), DOM16-39-217 (SEQ ID NO:548), DOM16- 39-218 (SEQ ID NO:549), DOM16-39-219 (SEQ ID NO:550), DOM16-39-220 (SEQ ID NO:551), DOM16-39-221 (SEQ ID NO:552), DOM16-39-222 (SEQ ID NO:553), DOM16-39-223 (SEQ ID NO:554), DOM16-39-224 (SEQ ID NO:555), DOM16-39-225 (SEQ ID NO:556), DOM16-39-226 (SEQ ID NO:557), D0M16- 39-227 (SEQ ID NO:558), DOM16-39-228 (SEQ ID NO:559), DOM16-39-229 (SEQ ID NO:560), DOM16-39-230 (SEQ ID NO:561), DOM16-39-231 (SEQ ID NO:562), DOM16-39-232 (SEQ ID NO:563), DOM16-39-233 (SEQ ID NO:564), DOMl 6-39-234 (SEQ ID NO:565), DOMl 6-39-235 (SEQ ID NO:566), DOMl 6- 39-500 (SEQ ID NO:725), DOMl 6-39-502(SEQ ID NO:726), DOMl 6-39-503 (SEQ ID NO:567), DOMl 6-39-504 (SEQ ID NO:568), DOMl 6-39-505 (SEQ ID NO:569), DOM16-39-506 (SEQ ID NO:570), DOM16-39-507 (SEQ ID NO:571), DOM16-39-508 (SEQ ID NO:572), DOM16-39-509 (SEQ ID NO:573), DOM16- 39-510 (SEQ ID NO:574), DOM16-39-511 (SEQ ID NO:575), DOM16-39-512 (SEQ ID NO:576), DOM16-39-521 (SEQ ID NO:577), DOM16-39-522 (SEQ ID NO:578), DOMl 6-39-523 (SEQ ID NO:579), DOMl 6-39-524 (SEQ ID NO:580), DOM16-39-527 (SEQ ID NO:581), DOM16-39-525 (SEQ ID NO:582), DOM16- 39-526 (SEQ ID NO:583), DOM16-39-540 (SEQ ID NO:584), DOM16-39-541 (SEQ ID NO:585), DOMl 6-39-542 (SEQ ID NO:586), DOMl 6-39-543 (SEQ ID NO:587), DOM16-39-544 (SEQ ID NO:588), DOM16-39-545 (SEQ ID NO:589), DOM16-39-550 (SEQ ID NO:590), DOM16-39-551 (SEQ ID NO:591), DOM16- 39-552 (SEQ ID NO:592), DOMl 6-39-553 (SEQ ID NO:593), DOMl 6-39-554 (SEQ ID NO:594), DOM16-39-555 (SEQ ID NO:595), DOM16-39-561 (SEQ ID NO:596), DOMl 6-39-562 (SEQ ID NO:597), DOMl 6-39-563 (SEQ ID NO:598), DOMl 6-39-564 (SEQ ID NO:599), DOMl 6-39-571 (SEQ ID NO:600), DOMl 6- 39-572 (SEQ ID NO:601), DOMl 6-39-573 (SEQ ID NO:602), DOMl 6-39-574 (SEQ ID NO:603), DOM16-39-580 (SEQ ID NO:604), DOM16-39-591 (SEQ ID NO:605), DOMl 6-39-592 (SEQ ID NO:606), DOMl 6-39-593 (SEQ ID NO:607), DOMl 6-39-601 (SEQ ID NO:608), DOMl 6-39-602 (SEQ ID NO:609), DOMl 6- 39-603 (SEQ ID NO:610), DOM16-39-604 (SEQ ID NO:611), DOM16-39-605 (SEQ ID NO:612), DOM16-39-607 (SEQ ID NO:613), DOM16-39-611 (SEQ ID NO:614), DOM16-39-612 (SEQ ID NO:615), DOM16-39-613 (SEQ ID NO:616), DOM16-39-614 (SEQ ID NO:617), DOM16-39-615 (SEQ ID NO:618), DOM16- 39-616 (SEQ ID NO:619), DOM16-39-617 (SEQ ID NO:620), DOM16-39-618 (SEQ ID NO:621), and DOM16-39-619 (SEQ ID NO:622).
In other embodiments, the ligand that has binding specificity for VEGF and for EGFR comprises a first immunoglobulin single variable domain with binding specificity for VEGF and a second immunoglobulin single variable domain with binding specificity for EGFR, wherein said first immunoglobulin single variable domain competes for binding to VEGF with bevacizumab and/or antibody 2C3 (ATCC Accession No. PTA 1595); and said second immunoglobulin single variable domain competes for binding to EGFR with cetuximab.
In particular embodiments, the ligand has binding specificity for VEGF and for EGFR and comprises at least one immunoglobulin single variable domain with binding specificity for VEGF and at least one immunoglobulin single variable domain with binding specificity for EGFR, wherein the ligand comprises an immunoglobulin single variable domain with binding specificity for VEGF that comprises an amino acid sequence that has at least 90% amino acid sequence identity with the amino acid sequence of an anti-VEGF dAb selected from the group consisting of TARl 5-6 (SEQ ID NO: 117), TARl 5-8 (SEQ ID NO: 119), and TAR15-26 (SEQ ID NO:123), and further comprises an immunoglobulin single variable domain with binding specificity for EGFR that comprises an amino acid sequence that has at least 90% amino acid sequence identity with an amino acid sequence selected from the group consisting of DOM16-39 (SEQ ID NO:345), DOM16-39-87 (SEQ ID NO:420), DOM16-39-100 (SEQ ID NO:423), DOM16-39- 107 (SEQ ID NO:430), DOM16-39-109 (SEQ ID NO:432), DOM16-39-115 (SEQ ID NO:438), or DOM16-39-200 (SEQ ID NO:441).
In particular embodiments, the ligand has binding specificity for VEGF and for EGFR and comprises at least one immunoglobulin single variable domain with binding specificity for VEGF and at least one immunoglobulin single variable domain with binding specificity for EGFR, wherein the ligand comprises an immunoglobulin single variable domain with binding specificity for VEGF that comprises an amino acid sequence that has at least 90% amino acid sequence identity with the amino acid sequence of an anti-VEGF dAb selected from the group consisting of TAR15-6 (SEQ ID NO:117), TAR15-8 (SEQ ID NO:119), and TAR15-26 (SEQ ID NO: 123), and further comprises an immunoglobulin single variable domain with binding specificity for EGFR that comprises an amino acid sequence that has at least 90% amino acid sequence identity with an amino acid sequence selected from the group consisting of DOM16-39-521 (SEQ ID NO:577), DOM16-39-541 (SEQ ID NO:585), DOM16-39-542 (SEQ ID NO:586), DOM16- 39-551 (SEQ ID NO:591), DOMl 6-39-601 (SEQ ID NO:608), DOMl 6-39-604 (SEQ ID NO:611), DOMl 6-39-618 (SEQ ID NO:621), and DOM 16-39-619 (SEQ ID NO:622).
The ligand that has binding specificity for VEGF and for EGFR can inhibit binding of epidermal growth factor (EGF) and/or transforming growth factor alpha (TGFalpha) to EGFR, inhibit the activity of EGFR, and/or inhibit the activity of EGFR without substantially inhibiting binding of epidermal growth factor (EGF) and/or transforming growth factor alpha (TGFalpha) to EGFR. In addition, or alternatively, the ligand that has binding specificity for VEGF and for EGFR can inhibit binding of VEGF to vascular endothelial growth factor receptor 1 (VEGFRl) and/or vascular endothelial growth factor receptor 2 (VEGFR2), inhibit the activity of VEGF and/or inhibit the activity of VEGF without substantially inhibiting binding of VEGF to VEGFRl and/or VEGFR2.
The ligand that has binding specificity for VEGF and for EGFR can contain a protein binding moiety (e.g., immunoglobulin single variable domain) with binding specificity for VEGF that binds VEGF with an affinity (KD) that is between about 100 nM and about 1 pM, as determined by surface plasmon resonance.
The ligand that has binding specificity for VEGF and for EGFR can contain a protein binding moiety (e.g., immunoglobulin single variable domain) with binding specificity for EGFR that binds EGFR with an affinity (KD) that is between about 100 nM and about 1 pM or about 10 nM to about 100 pM, as determined by surface plasmon resonance.
The ligand that has binding specificity for VEGF and for EGFR can bind VEGF with an affinity (KD) that is between about 100 nM and about 1 pM, as determined by surface plasmon resonance.
The ligand that has binding specificity for VEGF and for EGFR can bind EGFR with an affinity (KD) that is between about 100 nM and about 1 pM or about 10 nM to about 100 pM, as determined by surface plasmon resonance.
The ligand that has binding specificity for VEGF and for EGFR can comprise an immunoglobulin single variable domain with binding specificity for VEGF that is a VHH and/or an immunoglobulin single variable domain with bining specificity for EGFR that is a VHH.
The ligand that has binding specificity for VEGF and for EGFR can comprise an immunoglobulin single variable domain with binding specificity for VEGF and an immunoglobulin single variable domain with binding specificity for EGFR, wherein the immunoglobuoin single domains are selected from the group consisting of a human VH and a human VL.
In some embodiments, the ligand that has binding specificity for VEGF and for EGFR can be an IgG-like format comprising two immunoglobulin single variable domains with binding specificity for VEGF, and two immunoglobulin single variable domains with binding specificity for EGFR.
In some embodiments, the ligand that has binding specificity for VEGF and for EGFR can comprise an antibody Fc region.
The invention also relates to a ligand that has binding specificity for VEGF, comprising at least one immunoglobulin single variable domain with binding specificity for VEGF, wherein an immunoglobulin single variable domain with binding specificity for VEGF competes for binding to VEGF with an anti-VEGF domain antibody (dAb) selected from the group consisting of TARl 5-1 (SEQ ID NO:100), TAR15-3 (SEQ ID NO:101), TAR15-4 (SEQ ID NO:102), TAR15-9 (SEQ ID NO: 103), TARl 5-10 (SEQ ID NO: 104), TARl 5-11 (SEQ ID NO: 105), TAR15-12 (SEQ ID NO:106), TAR15-13 (SEQ ID NO:107), TAR15-14 (SEQ ID NO:108), TAR15-15 (SEQ ID NO:109), TAR15-16 (SEQ ID NO:110), TAR15-17 (SEQ ID NO:111), TAR15-18 (SEQ ID NO:112), TAR15-19 (SEQ ID NO:113), TARl 5-20 (SEQ ID NO: 114), TAR 15-22 (SEQ ID NO: 115), TARl 5-5 (SEQ ID NO: 116), TARl 5-6 (SEQ ID NO: 117), TARl 5-7 (SEQ ID NO: 118), TARl 5-8 (SEQ ID NO:119), TAR15-23 (SEQ ID NO:120), TAR15-24 (SEQ ID NO:121), TAR15-25 (SEQ ID NO:122), TAR15-26 (SEQ ID NO:123), TAR15-27 (SEQ ID NO:124), TAR15-29 (SEQ ID NO:125), TAR15-30 (SEQ ID NO:126), TAR15-6- 500 (SEQ ID NO:127), TAR15-6-501 (SEQ ID NO:128), TAR15-6-502 (SEQ ID NO:129), TAR15-6-503 (SEQ ID NO:130), TAR15-6-504 (SEQ ID NO:131), TARl 5-6-505 (SEQ ID NO: 132), TARl 5-6-506 (SEQ ID NO: 133), TARl 5-6-507 (SEQ ID NO: 134), TARl 5-6-508 (SEQ ID NO: 135), TARl 5-6-509 (SEQ ID NO:136), TAR15-6-510 (SEQ ID NO:137), TAR15-8-500 (SEQ ID NO:138), TARl 5-8-501 (SEQ ID NO: 139), TARl 5-8-502 (SEQ ID NO: 140), TARl 5-8-503 (SEQ ID NO:141), TAR15-8-505 (SEQ ID NO:142), TAR15-8-506 (SEQ ID NO:143), TAR15-8-507 (SEQ ID NO:144), TAR15-8-508 (SEQ ID NO:145), TAR15-8-509 (SEQ ID NO:146), TAR15-8-510 (SEQ ID NO:147), TAR15-8-511 (SEQ ID NO:148), TAR15-26-500 (SEQ ID NO:149), TAR15-26-501 (SEQ ID NO: 150), TARl 5-26-502 (SEQ ID NO:151), TARl 5-26-503 (SEQ ID NO: 152), TARl 5-26-504 (SEQ ID NO: 153), TARl 5-26-505 (SEQ ID NO: 154), TARl 5-26- 506 (SEQ ID NO: 155), TARl 5-26-507 (SEQ ID NO: 156), TARl 5-26-508 (SEQ ID NO: 157), TARl 5-26-509 (SEQ ID NO: 158), TARl 5-26-510 (SEQ ID NO: 159), TAR15-26-511 (SEQ ID NO:160), TAR15-26-512 (SEQ ID NO:161), TAR15-26- 513 (SEQ ID NO:162), TAR15-26-514 (SEQ ID NO:163), TAR15-26-515 (SEQ ID NO: 164), TARl 5-26-516 (SEQ ID NO: 165), TARl 5-26-517 (SEQ ID NO: 166), TARl 5-26-518 (SEQ ID NO: 167), TARl 5-26-519 (SEQ ID NO: 168), TARl 5-26- 520 (SEQ ID NO:169), TAR15-26-521 (SEQ ID NO: 170), TAR15-26-522 (SEQ ID NO:171), TAR15-26-523 (SEQ ID NO:172), TAR15-26-524 (SEQ ID NO:173), TARl 5-26-525 (SEQ ID NO: 174), TARl 5-26-526 (SEQ ID NO: 175), TARl 5-26- 527 (SEQ ID NO: 176), TARl 5-26-528 (SEQ ID NO: 177), TARl 5-26-529 (SEQ ID NO: 178), TARl 5-26-530 (SEQ ID NO: 179), TARl 5-26-531 (SEQ ID NO: 180), TAR15-26-532 (SEQ ID NO:181), TAR15-26-533 (SEQ ID NO: 182), TAR15-26- 534 (SEQ ID NO:183), TAR15-26-535 (SEQ ID NO:184), TAR15-26-536 (SEQ ID NO: 185), TARl 5-26-537 (SEQ ID NO: 186), TARl 5-26-538 (SEQ ID NO: 187), TARl 5-26-539 (SEQ ID NO: 188), TARl 5-26-540 (SEQ ID NO: 189), TARl 5-26- 541 (SEQ ID NO:190), TAR15-26-542 (SEQ ID NO:191), TAR15-26-543 (SEQ ID NO: 192), TARl 5-26-544 (SEQ ID NO: 193), TARl 5-26-545 (SEQ ID NO: 194), TARl 5-26-546 (SEQ ID NO: 195), TARl 5-26-547 (SEQ ID NO: 196), TARl 5-26- 548 (SEQ ID NO: 197), and TARl 5-26-549 (SEQ ID NO: 198), TARl 5-26-550 (SEQ ID NO:539), and TARl 5-26-551 (SEQ ID NO:540).
For example, an immunoglobulin single variable domain with binding specificity for VEGF can comprise an amino acid sequence that has at least about 85% amino acid sequence identity with the amino acid sequence of a dAb selected from the group consisting of TAR15-1 (SEQ ID NO:100), TAR15-3 (SEQ ID NO:101), TAR15-4 (SEQ ID NO: 102), TAR15-9 (SEQ ID NO:103), TAR15-10 (SEQ ID NO: 104), TARl 5-11 (SEQ ID NO:105), TAR15-12 (SEQ ID NO:106), TAR15-13 (SEQ ID NO:107), TAR15-14 (SEQ ID NO: 108), TAR15-15 (SEQ ID NO:109), TAR15-16 (SEQ ID NO:110), TAR15-17 (SEQ ID NO:111), TAR15-18 (SEQ ID NO:112), TARl 5-19 (SEQ ID NO:113), TAR15-20 (SEQ ID NO:114), TAR 15-22 (SEQ ID NO: 115), TARl 5-5 (SEQ ID NO: 116), TARl 5-6 (SEQ ID NO: 117), TARl 5-7 (SEQ ID NO: 118), TARl 5-8 (SEQ ID NO: 119), TARl 5-23 (SEQ ID NO:120), TAR15-24 (SEQ ID NO:121), TAR15-25 (SEQ ID NO:122), TARl 5-26 (SEQ ID NO: 123), TARl 5-27 (SEQ ID NO: 124), TARl 5-29 (SEQ ID NO:125), TAR15-30 (SEQ ID NO:126), TAR15-6-500 (SEQ ID NO:127), TAR15- 6-501 (SEQ ID NO:128), TAR15-6-502 (SEQ ID NO: 129), TAR15-6-503 (SEQ ID NO:130), TAR15-6-504 (SEQ ID NO:131), TAR15-6-505 (SEQ ID NO:132), TARl 5-6-506 (SEQ ID NO: 133), TARl 5-6-507 (SEQ ID NO: 134), TARl 5-6-508 (SEQ ID NO:135), TAR15-6-509 (SEQ ID NO:136), TAR15-6-510 (SEQ ID NO:137), TAR15-8-500 (SEQ ID NO:138), TAR15-8-501 (SEQ ID NO:139), TAR15-8-502 (SEQ ID NO:140), TAR15-8-503 (SEQ ID NO:141), TAR15-8-505 (SEQ ID NO:142), TAR15-8-506 (SEQ ID NO:143), TAR15-8-507 (SEQ ID NO:144), TAR15-8-508 (SEQ ID NO:145), TAR15-8-509 (SEQ ID NO:146), TAR15-8-510 (SEQ ID NO:147), TARl 5-8-511 (SEQ ID NO:148), TAR15-26-500 (SEQ ID NO:149), TAR15-26-501 (SEQ ID NO:150), TAR15-26-502 (SEQ ID NO:151), TAR15-26-503 (SEQ ID NO:152), TAR15-26-504 (SEQ ID NO:153), TARl 5-26-505 (SEQ ID NO: 154), TARl 5-26-506 (SEQ ID NO: 155), TARl 5-26- 507 (SEQ ID NO:156), TAR15-26-508 (SEQ ID NO:157), TAR15-26-509 (SEQ ID NO: 158), TARl 5-26-510 (SEQ ID NO: 159), TARl 5-26-511 (SEQ ID NO: 160), TAR15-26-512 (SEQ ID NO:161), TAR15-26-513 (SEQ ID NO:162), TAR15-26- 514 (SEQ ID NO:163), TAR15-26-515 (SEQ ID NO:164), TAR15-26-516 (SEQ ID NO: 165), TARl 5-26-517 (SEQ ID NO: 166), TARl 5-26-518 (SEQ ID NO: 167), TARl 5-26-519 (SEQ ID NO: 168), TARl 5-26-520 (SEQ ID NO: 169), TARl 5-26- 521 (SEQ ID NO: 170), TARl 5-26-522 (SEQ ID NO: 171), TARl 5-26-523 (SEQ ID NO:172), TAR15-26-524 (SEQ ID NO:173), TAR15-26-525 (SEQ ID NO: 174), TARl 5-26-526 (SEQ ID NO: 175), TARl 5-26-527 (SEQ ID NO: 176), TARl 5-26- 528 (SEQ ID NO: 177), TARl 5-26-529 (SEQ ID NO: 178), TARl 5-26-530 (SEQ ID NO: 179), TARl 5-26-531 (SEQ ID NO: 180), TARl 5-26-532 (SEQ ID NO: 181), TARl 5-26-533 (SEQ ID NO: 182), TARl 5-26-534 (SEQ ID NO: 183), TARl 5-26- 535 (SEQ ID NO: 184), TARl 5-26-536 (SEQ ID NO: 185), TARl 5-26-537 (SEQ ID NO: 186), TARl 5-26-538 (SEQ ID NO: 187), TARl 5-26-539 (SEQ ID NO: 188), TAR15-26-540 (SEQ ID NO:189), TAR15-26-541 (SEQ ID NO:190), TAR15-26- 542 (SEQ ID NO:191), TAR15-26-543 (SEQ ID NO:192), TAR15-26-544 (SEQ ID NO: 193), TARl 5-26-545 (SEQ ID NO: 194), TARl 5-26-546 (SEQ ID NO: 195), TARl 5-26-547 (SEQ ID NO: 196), TARl 5-26-548 (SEQ ID NO: 197), and TARl 5- 26-549 (SEQ ID NO:198), TAR15-26-550 (SEQ ID NO:539), and TAR15-26-551 (SEQ ID NO:540).
The ligand that has binding specificity for VEGF can inhibit binding of VEGF to vascular endothelial growth factor receptor 1 (VEGFRl) and/or vascular endothelial growth factor receptor 2 (VEGFR2), inhibit the activity of VEGF and/or inhibit the activity of VEGF without substantially inhibiting binding of VEGF to VEGFRl and/or VEGFR2.
The ligand that has binding specificity for VEGF can contain an
immunoglobulin single variable domain with binding specificity for VEGF that binds VEGF with an affinity (KD) that is between about 100 nM and about 1 pM, as determined by surface plasmon resonance.
The ligand that has binding specificity for VEGF can bind VEGF with an affinity (KD) that is between about 100 nM and about 1 pM, as determined by surface plasmon resonance.
The ligand that has binding specificity for VEGF can comprise an immunoglobulin single variable domain with binding specificity for VEGF that is a
VHH.
The ligand that has binding specificity for VEGF can comprise an immunoglobulin single variable domain with binding specificity for VEGF that is selected from the group consisting of a human VH and a human VL-
In some embodiments, the ligand that has binding specificity for VEGF is an IgG-like format comprising at least two immunoglobulin single variable domains with binding specificity for VEGF.
In some embodiments, the ligand that has binding specificity for VEGF comprises an antibody Fc region.
The invention also relates to a ligand that has binding specificity for EGFR comprising at least one immunoglobulin single variable domain with binding specificity for EGFR, wherein an immunoglobulin single variable domain with binding specificity for EGFR competes for binding to EGFR with an anti-EGFR domain antibody (dAb) selected from the group consisting of DOM16-17 (SEQ ID NO:325), DOM16-18 (SEQ ID NO:326), DOM16-19 (SEQ ID NO:327), DOM16- 20 (SEQ ID NO:328), DOM16-21 (SEQ ID NO:329), DOM16-22 (SEQ ID NO:330), DOMl 6-23 (SEQ ID NO:331), DOMl 6-24 (SEQ ID NO:332), DOMl 6- 25 (SEQ ID NO:333), DOMl 6-26 (SEQ ID NO:334), DOMl 6-27 (SEQ ID NO:335), DOM16-28 (SEQ ID NO:336), DOM16-29 (SEQ ID NO:337), DOM16- 30 (SEQ ID NO:338), DOMl 6-31 (SEQ ID NO:339), DOMl 6-32 (SEQ ID NO:340), DOM16-33 (SEQ ID NO:341), DOM16-35 (SEQ ID NO:342), DOM16- 37 (SEQ ID NO:343), DOM16-38 (SEQ ID NO:344), DOM16-39 (SEQ ID NO:345), DOMl 6-40 (SEQ ID NO:346), DOMl 6-41 (SEQ ID NO:347), DOMl 6- 42 (SEQ ID NO:348), DOMl 6-43 (SEQ ID NO:349), DOMl 6-44 (SEQ ID NO:350), DOM16-45 (SEQ ID NO:351), DOM16-46 (SEQ ID NO:352), DOM16- 47 (SEQ ID NO:353), DOM16-48 (SEQ ID NO:354), DOM16-49 (SEQ ID
NO:355), DOM16-50 (SEQ ID NO:356), DOM16-59 (SEQ ID NO:357), DOM16- 60 (SEQ ID NO:358), DOM16-61 (SEQ ID NO:359), DOM16-62 (SEQ ID NO:360), DOM16-63 (SEQ ID NO:361), DOM16-64 (SEQ ID NO:362), DOM16- 65 (SEQ ID NO:363), DOMl 6-66 (SEQ ID NO:364), DOMl 6-67 (SEQ ID NO:365), DOMl 6-68 (SEQ ID NO:366), DOMl 6-69 (SEQ ID NO:367), DOMl 6- 70 (SEQ ID NO:368), DOM16-71 (SEQ ID NO:369), DOM16-72 (SEQ ID NO:370), DOM16-73 (SEQ ID NO:371), DOM16-74 (SEQ ID NO:372), DOM16- 75 (SEQ ID NO:373), DOMl 6-76 (SEQ ID NO:374), DOMl 6-77 (SEQ ID NO:375), DOM16-78 (SEQ ID NO:376), DOM16-79 (SEQ ID NO:377), DOM16- 80 (SEQ ID NO:378), DOM16-81 (SEQ ID NO:379), DOM16-82 (SEQ ID
NO:380), DOM16-83 (SEQ ID NO:381), DOM16-84 (SEQ ID NO:382), DOM16- 85 (SEQ ID NO:383), DOM16-87 (SEQ ID NO:384), DOM16-88 (SEQ ID NO:385), DOM16-89 (SEQ ID NO:386), DOM16-90 (SEQ ID NO:387), DOM16- 91 (SEQ ID NO:388), DOM16-92 (SEQ ID NO:389), DOM16-94 (SEQ ID NO:390), DOMl 6-95 (SEQ ID NO:391), DOMl 6-96 (SEQ ID NO:392), DOMl 6- 97 (SEQ ID NO:393), DOMl 6-98 (SEQ ID NO:394), DOMl 6-99 (SEQ ID NO:395), DOM16-100 (SEQ ID NO:396), DOM16-101 (SEQ ID NO:397), DOM16-102 (SEQ ID NO:398), DOM16-103 (SEQ ID NO:399), DOM16-104 (SEQ ID NO:400), DOM16-105 (SEQ ID NO:401), DOM16-106 (SEQ ID
NO:402), DOM16-107 (SEQ ID NO:403), DOM16-108 (SEQ ID NO:404), DOM16-109 (SEQ ID NO:405), DOM16-110 (SEQ ID NO:406), DOM16-111 (SEQ ID NO:407), DOM16-112 (SEQ ID NO:408), DOM16-113 (SEQ ID NO:409), DOM16-114 (SEQ ID NO:410), DOM16-115 (SEQ ID NO:411), DOM16-116 (SEQ ID NO:412), DOM16-117 (SEQ ID NO:413), DOM16-118 (SEQ ID NO:414), DOM16-119 (SEQ ID NO:415), DOM16-39-6 (SEQ ID
NO:416), DOMl 6-39-8 (SEQ ID NO:417), DOMl 6-39-34 (SEQ ID NO:418), DOMl 6-39-48 (SEQ ID NO:419), DOMl 6-39-87 (SEQ ID NO:420), DOMl 6-39- 90 (SEQ ID NO:421), DOM16-39-96 (SEQ ID NO:422), DOM16-39-100 (SEQ ID NO:423), DOM16-39-101 (SEQ ID NO:424), DOM16-39-102 (SEQ ID NO:425), DOM16-39-103 (SEQ ID NO:426), DOM16-39-104 (SEQ ID NO:427), DOM16- 39-105 (SEQ ID NO:428), DOM16-39-106 (SEQ ID NO:429), DOM16-39-107 (SEQ ID NO:430), DOM16-39-108 (SEQ ID NO:431), DOM16-39-109 (SEQ ID NO:432), DOM16-39-110 (SEQ ID NO:433), DOM16-39-111 (SEQ ID NO:434), DOM16-39-112 (SEQ ID NO:435), DOM16-39-113 (SEQ ID NO:436), DOM16- 39-114 (SEQ ID NO:437), DOM16-39-115 (SEQ ID NO:438), DOM16-39-116 (SEQ ID NO:439), DOM16-39-117 (SEQ ID NO:440), DOM16-39-200 (SEQ ID NO:441), DOM16-39-201 (SEQ ID NO:442), DOM16-39-202 (SEQ ID NO:443), DOMl 6-39-203 (SEQ ID NO:444), DOMl 6-39-204 (SEQ ID NO:445), DOMl 6- 39-205 (SEQ ID NO:446), DOM16-39-206 (SEQ ID NO:447), DOM16-39-207 (SEQ ID NO:448), DOMl 6-39-209 (SEQ ID NO:449), DOMl 6-52 (SEQ ID NO:450), NBl (SEQ ID NO:451), NB2 (SEQ ID NO:452), NB3 (SEQ ID NO:453), NB4 (SEQ ID NO:454), NB5 (SEQ ID NO:455), NB6 (SEQ ID NO:456), NB7
(SEQ ID NO:457), NB8 (SEQ ID NO:458), NB9 (SEQ ID NO:459), NBlO (SEQ ID NO:460), NBl 1 (SEQ ID NO:461), NB12 (SEQ ID NO:462), NB13 (SEQ ID NO:463), NB14 (SEQ ID NO:464), NB15 (SEQ ID NO:465), NB16 (SEQ ID NO:466), NB17 (SEQ ID NO:467), NB18 (SEQ ID NO:468), NB19 (SEQ ID NO:469), NB20 (SEQ ID NO:470), NB21 (SEQ ID NO:471), and NB22 (SEQ ID NO:472).
The invention also relates to a ligand that has binding specificity for EGFR comprising at least one immunoglobulin single variable domain with binding specificity for EGFR, wherein an immunoglobulin single variable domain with binding specificity for EGFR competes for binding to EGFR with an anti-EGFR domain antibody (dAb) selected from the group consisting of DOMl 6-39-210 (SEQ ID NO:541), DOM16-39-211 (SEQ ID NO:542), DOM16-39-212 (SEQ ID NO:543), DOM 16-39-213 (SEQ ID NO:544), DOM16-39-214 (SEQ ID NO:545), DOM16-39-215 (SEQ ID NO:546), DOM16-39-216 (SEQ ID NO:547), DOM16- 39-217 (SEQ ID NO:548), DOM16-39-218 (SEQ ID NO:549), DOM16-39-219 (SEQ ID NO:550), DOM16-39-220 (SEQ ID NO:551), DOM16-39-221 (SEQ ID NO:552), DOMl 6-39-222 (SEQ ID NO:553), DOMl 6-39-223 (SEQ ID NO:554), DOMl 6-39-224 (SEQ ID NO:555), DOMl 6-39-225 (SEQ ID NO:556), DOMl 6- 39-226 (SEQ ID NO:557), DOM16-39-227 (SEQ ID NO:558), DOM16-39-228 (SEQ ID NO:559), DOMl 6-39-229 (SEQ ID NO:560), DOMl 6-39-230 (SEQ ID NO:561), DOM16-39-231 (SEQ ID NO:562), DOM16-39-232 (SEQ ID NO:563), DOM16-39-233 (SEQ ID NO:564), DOM16-39-234 (SEQ ID NO:565), DOM16- 39-235 (SEQ ID NO:566), DOM16-39-500 (SEQ ID NO:725), DOM16-39- 502(SEQ ID NO:726), DOMl 6-39-503 (SEQ ID NO:567), DOMl 6-39-504 (SEQ ID NO:568), DOMl 6-39-505 (SEQ ID NO:569), DOMl 6-39-506 (SEQ ID NO:570), DOM16-39-507 (SEQ ID NO:571), DOM16-39-508 (SEQ ID NO:572), DOM16-39-509 (SEQ ID NO:573), DOM16-39-510 (SEQ ID NO:574), DOM16- 39-511 (SEQ ID NO:575), DOM16-39-512 (SEQ ID NO:576), DOM16-39-521 (SEQ ID NO:577), DOM16-39-522 (SEQ ID NO:578), DOM16-39-523 (SEQ ID NO:579), DOM16-39-524 (SEQ ID NO:580), DOM16-39-527 (SEQ ID NO:581), DOM16-39-525 (SEQ ID NO:582), DOM16-39-526 (SEQ ID NO:583), DOM16- 39-540 (SEQ ID NO:584), DOM16-39-541 (SEQ ID NO:585), DOM16-39-542 (SEQ ID NO:586), DOMl 6-39-543 (SEQ ID NO:587), DOMl 6-39-544 (SEQ ID NO:588), DOMl 6-39-545 (SEQ ID NO:589), DOMl 6-39-550 (SEQ ID NO:590), DOM16-39-551 (SEQ ID NO:591), DOM16-39-552 (SEQ ID NO:592), DOM16- 39-553 (SEQ ID NO:593), DOM16-39-554 (SEQ ID NO:594), DOM16-39-555 (SEQ ID NO:595), DOMl 6-39-561 (SEQ ID NO:596), DOMl 6-39-562 (SEQ ID NO:597), DOM16-39-563 (SEQ ID NO:598), DOM16-39-564 (SEQ ID NO:599), DOM16-39-571 (SEQ ID NO:600), DOM16-39-572 (SEQ ID NO:601), DOM16- 39-573 (SEQ ID NO:602), DOMl 6-39-574 (SEQ ID NO:603), DOMl 6-39-580 (SEQ ID NO:604), DOM16-39-591 (SEQ ID NO:605), DOM16-39-592 (SEQ ID NO:606), DOM16-39-593 (SEQ ID NO:607), DOM16-39-601 (SEQ ID NO:608), DOMl 6-39-602 (SEQ ID NO:609), DOMl 6-39-603 (SEQ ID NO:610), DOMl 6- 39-604 (SEQ ID NO:611), DOM16-39-605 (SEQ ID NO:612), DOM16-39-607 (SEQ ID NO:613), DOM16-39-611 (SEQ ID NO:614), DOM16-39-612 (SEQ ID NO:615), DOM16-39-613 (SEQ ID NO:616), DOM16-39-614 (SEQ ID NO:617), DOM16-39-615 (SEQ ID NO:618), DOM16-39-616 (SEQ ID NO:619), DOM16- 39-617 (SEQ ID NO:620), DOM16-39-618 (SEQ ID NO:621), and DOM16-39-619 (SEQ ID NO:622).
For example, the immunoglobulin single variable domain with binding specificity for EGFR can comprise an amino acid sequence that has at least about 85% amino acid sequence identity with the amino acid sequence of a dAb selected from the group consisting of DOM16-17 (SEQ ID NO.325), DOM16-18 (SEQ ID NO:326), DOMl 6-19 (SEQ ID NO:327), DOMl 6-20 (SEQ ID NO:328), DOMl 6- 21 (SEQ ID NO:329), DOMl 6-22 (SEQ ID NO:330), DOMl 6-23 (SEQ ID
NO:331), DOMl 6-24 (SEQ ID NO:332), DOMl 6-25 (SEQ ID NO:333), DOMl 6- 26 (SEQ ID NO:334), DOM16-27 (SEQ ID NO:335), DOMl 6-28 (SEQ ID
NO:336), DOM16-29 (SEQ ID NO:337), DOM16-30 (SEQ ID NO:338), DOM16- 31 (SEQ ID NO:339), DOMl 6-32 (SEQ ID NO:340), DOMl 6-33 (SEQ ID
NO:341), DOM16-35 (SEQ ID NO:342), DOM16-37 (SEQ ID NO:343), DOM16- 38 (SEQ ID NO:344), DOMl 6-39 (SEQ ID NO:345), DOMl 6-40 (SEQ ID
NO:346), DOMl 6-41 (SEQ ID NO:347), DOMl 6-42 (SEQ ID NO:348), DOMl 6- 43 (SEQ ID NO:349), DOM16-44 (SEQ ID NO:350), DOM16-45 (SEQ ID
NO:351), DOMl 6-46 (SEQ ID NO;352), DOMl 6-47 (SEQ ID NO:353), DOMl 6- 48 (SEQ ID NO:354), DOM16-49 (SEQ ID NO:355), DOM16-50 (SEQ ID
NO:356), DOM16-59 (SEQ ID NO:357), DOM16-60 (SEQ ID NO:358), DOM16- 61 (SEQ ID NO:359), DOMl 6-62 (SEQ ID NO:360), DOMl 6-63 (SEQ ID
NO:361), DOM16-64 (SEQ ID NO:362), DOM16-65 (SEQ ID NO:363), D0M16- 66 (SEQ ID NO:364), DOMl 6-67 (SEQ ID NO:365), DOMl 6-68 (SEQ ID
NO:366), DOM16-69 (SEQ ID NO:367), DOMl 6-70 (SEQ ID NO:368), D0M16- 71 (SEQ ID NO:369), DOMl 6-72 (SEQ ID NO:370), DOMl 6-73 (SEQ ID
NO:371), DOM16-74 (SEQ ID NO:372), DOM16-75 (SEQ ID NO:373), DOM16- 76 (SEQ ID NO:374), DOMl 6-77 (SEQ ID NO:375), DOMl 6-78 (SEQ ID
NO:376), DOM16-79 (SEQ ID NO:377), DOM16-80 (SEQ ID NO:378), D0M16- 81 (SEQ ID NO:379), DOM16-82 (SEQ ID NO:380), DOM16-83 (SEQ ID
NO:381), DOM16-84 (SEQ ID NO:382), DOM16-85 (SEQ ID NO:383), D0M16- 87 (SEQ ID NO:384), DOM16-88 (SEQ ID NO:385), DOM16-89 (SEQ ID
NO:386), DOM16-90 (SEQ ID NO:387), DOM16-91 (SEQ ID NO:388), DOM16- 92 (SEQ ID NO:389), DOM16-94 (SEQ ID NO:390), DOM16-95 (SEQ ID
NO:391), DOM16-96 (SEQ ID NO:392), DOM16-97 (SEQ ID NO:393), DOM16- 98 (SEQ ID NO:394), DOM16-99 (SEQ ID NO:395), DOM16-100 (SEQ ID NO:396), DOM16-101 (SEQ ID NO:397), DOM16-102 (SEQ ID NO:398), DOM16-103 (SEQ ID NO:399), DOM16-104 (SEQ ID NO:400), DOM16-105 (SEQ ID NO:401), DOM16-106 (SEQ ID NO:402), DOM16-107 (SEQ ID
NO:403), DOM16-108 (SEQ ID NO:404), DOM16-109 (SEQ ID NO:405), DOM16-110 (SEQ ID NO:406), DOM16-111 (SEQ ID NO:407), DOM16-112 (SEQ ID NO:408), DOMl 6-113 (SEQ ID NO:409), DOMl 6-114 (SEQ ID
NO:410), DOM16-115 (SEQ ID NO:411), DOM16-116 (SEQ ID NO:412), DOMl 6-117 (SEQ ID NO:413), DOM16-118 (SEQ ID NO:414), DOM16-119 (SEQ ID NO:415), DOMl 6-39-6 (SEQ ID NO:416), DOMl 6-39-8 (SEQ ID NO:417), DOM16-39-34 (SEQ ID NO:418), DOM16-39-48 (SEQ ID NO:419), DOM16-39-87 (SEQ ID NO:420), DOM16-39-90 (SEQ ID NO:421), DOM16-39- 96 (SEQ ID NO:422), DOM16-39-100 (SEQ ID NO:423), DOM16-39-101 (SEQ ID NO:424), DOM16-39-102 (SEQ ID NO:425), DOM16-39-103 (SEQ ID NO:426), DOM16-39-104 (SEQ ID NO:427), DOM16-39-105 (SEQ ID NO:428), DOM16- 39-106 (SEQ ID NO:429), DOM16-39-107 (SEQ ID NO:430), DOM16-39-108 (SEQ ID NO:431), DOM16-39-109 (SEQ ID NO:432), DOM16-39-110 (SEQ ID NO:433), DOM16-39-111 (SEQ ID NO:434), DOM16-39-112 (SEQ ID NO:435), DOM16-39-113 (SEQ ID NO:436), DOM16-39-114 (SEQ ID NO:437), DOM16- 39-115 (SEQ ID NO:438), DOM16-39-116 (SEQ ID NO:439), DOM16-39-117 (SEQ ID NO:440), DOM16-39-200 (SEQ ID NO:441), DOM16-39-201 (SEQ ID NO:442), DOMl 6-39-202 (SEQ ID NO:443), DOMl 6-39-203 (SEQ ID NO:444), DOM16-39-204 (SEQ ID NO:445), DOM16-39-205 (SEQ ID NO:446), DOM16- 39-206 (SEQ ID NO:447), DOMl 6-39-207 (SEQ ID NO:448), DOMl 6-39-209 (SEQ ID NO:449), DOMl 6-52 (SEQ ID NO:450), NBl (SEQ ID NO:451), NB2 (SEQ ID NO:452), NB3 (SEQ ID NO:453), NB4 (SEQ ID NO:454), NB5 (SEQ ID NO:455), NB6 (SEQ ID NO:456), NB7 (SEQ ID NO:457), NB8 (SEQ ID NO:458), NB9 (SEQ ID NO:459), NBlO (SEQ ID NO:460), NBI l (SEQ ID NO:461), NB12 (SEQ ID NO:462), NB 13 (SEQ ID NO:463), NB 14 (SEQ ID NO:464), NB 15 (SEQ ID NO:465), NB16 (SEQ ID NO:466), NB17 (SEQ ID NO:467), NB18 (SEQ ID NO:468), NB19 (SEQ ID NO:469), NB20 (SEQ ID NO:470), NB21 (SEQ ID NO:471), and NB22 (SEQ ID NO:472).
For example, the immunoglobulin single variable domain with binding specificity for EGFR can comprise an amino acid sequence that has at least about 85% amino acid sequence identity with the amino acid sequence of a dAb selected from the group consisting of DOM 16-39-210 (SEQ ID NO:541), DOM16-39-211 (SEQ ID NO:542), DOM16-39-212 (SEQ ID NO:543), DOM16-39-213 (SEQ ID NO:544), DOM16-39-214 (SEQ ID NO:545), DOM16-39-215 (SEQ ID NO:546), DOM16-39-216 (SEQ ID NO:547), DOM16-39-217 (SEQ ID NO:548), DOM16- 39-218 (SEQ ID NO:549), DOM16-39-219 (SEQ ID NO:550), DOM16-39-220 (SEQ ID NO:551), DOM16-39-221 (SEQ ID NO:552), DOM16-39-222 (SEQ ID NO:553), DOMl 6-39-223 (SEQ ID NO:554), DOMl 6-39-224 (SEQ ID NO:555), DOM16-39-225 (SEQ ID NO:556), DOM16-39-226 (SEQ ID NO:557), D0M16- 39-227 (SEQ ID NO:558), DOMl 6-39-228 (SEQ ID NO:559), DOMl 6-39-229 (SEQ ID NO:560), DOM16-39-230 (SEQ ID NO:561), DOM16-39-231 (SEQ ID NO:562), DOM16-39-232 (SEQ ID NO:563), DOM16-39-233 (SEQ ID NO:564), DOM16-39-234 (SEQ ID NO:565), DOM16-39-235 (SEQ ID NO:566), D0M16- 39-500 (SEQ ID NO:725), DOMl 6-39-502(SEQ ID NO:726), DOM16-39-503 (SEQ ID NO:567), DOM16-39-504 (SEQ ID NO:568), DOM16-39-505 (SEQ ID NO:569), DOM16-39-506 (SEQ ID NO:570), DOM16-39-507 (SEQ ID NO:571), DOMl 6-39-508 (SEQ ID NO:572), DOMl 6-39-509 (SEQ ID NO:573), DOMl 6- 39-510 (SEQ ID NO:574), DOM16-39-511 (SEQ ID NO:575), DOM16-39-512 (SEQ ID NO:576), DOM16-39-521 (SEQ ID NO:577), DOM16-39-522 (SEQ ID NO:578), DOM16-39-523 (SEQ ID NO:579), DOM16-39-524 (SEQ ID NO:580), DOM16-39-527 (SEQ ID NO:581), DOM16-39-525 (SEQ ID NO:582), DOM16- 39-526 (SEQ ID NO:583), DOMl 6-39-540 (SEQ ID NO:584), DOMl 6-39-541 (SEQ ID NO:585), DOM16-39-542 (SEQ ID NO:586), DOM16-39-543 (SEQ ID NO:587), DOM16-39-544 (SEQ ID NO:588), DOM16-39-545 (SEQ ID NO:589), DOM16-39-550 (SEQ ID NO:590), DOM16-39-551 (SEQ ID NO:591), DOM16- 39-552 (SEQ ID NO:592), DOMl 6-39-553 (SEQ ID NO:593), DOMl 6-39-554 (SEQ ID NO:594), DOM16-39-555 (SEQ ID NO:595), DOM16-39-561 (SEQ ID NO:596), DOMl 6-39-562 (SEQ ID NO:597), DOMl 6-39-563 (SEQ ID NO:598), DOM16-39-564 (SEQ ID NO:599), DOM16-39-571 (SEQ ID NO:600), DOM16- 39-572 (SEQ ID NO:601), DOMl 6-39-573 (SEQ ID NO:602), DOMl 6-39-574 (SEQ ID NO:603), DOM16-39-580 (SEQ ID NO:604), DOM16-39-591 (SEQ ID NO:605), DOMl 6-39-592 (SEQ ID NO:606), DOMl 6-39-593 (SEQ ID NO:607), DOM16-39-601 (SEQ ID NO:608), DOM16-39-602 (SEQ ID NO:609), D0M16- 39-603 (SEQ ID NO:610), DOM16-39-604 (SEQ ID NO:611), DOM16-39-605 (SEQ ID NO:612), DOM16-39-607 (SEQ ID NO:613), DOM16-39-611 (SEQ ID NO:614), DOM16-39-612 (SEQ ID NO:615), DOM16-39-613 (SEQ ID NO:616), DOM16-39-614 (SEQ ID NO:617), DOM16-39-615 (SEQ ID NO:618), DOM16- 39-616 (SEQ ID NO:619), DOM16-39-617 (SEQ ID NO:620), DOM16-39-618 (SEQ ID NO:621), and DOM16-39-619 (SEQ ID NO:622).
The ligand that has binding specificity for EGFR can inhibit binding of epidermal growth factor (EGF) and/or transforming growth factor alpha (TGF alpha) to EGFR, inhibit the activity of EGFR, and/or inhibit the activity of EGFR without substantially inhibiting binding of epidermal growth factor (EGF) and/or transforming growth factor alpha (TGF alpha) to EGFR.
The ligand that has binding specificity for EGFR can contain an
immunoglobulin single variable domain with binding specificity for EGFR that binds EGFR with an affinity (KD) that is between about 100 nM and about 1 pM or about 10 nM to about 100 pM, as determined by surface plasmon resonance.
The ligand that has binding specificity for VEGF and for EGFR can bind EGFR with an affinity (KD) that is between about 100 nM and about 1 pM or about 10 nM to about 100 pM, as determined by surface plasmon resonance.
The ligand that has binding specificity for EGFR can comprise an immunoglobulin single variable domain with binding specificity for EGFR that is a
VHH.
The ligand that has binding specificity for EGFR can comprise an immunoglobulin single variable domain with binding specificity for EGFR that is selected from the group consisting of a human VH and a human VL. In some embodiments, the ligand that has binding specificity for EGFR is an IgG-like format comprising at least two immunoglobulin single variable domains with binding specificity for EGFR.
In some embodiments, the ligand that has binding specificity for EGFR comprises an antibody Fc region.
In some embodiments, the ligand comprises a single immunoglobulin variable domain polypeptide that antagonizes (inhibits) human EGFR binding to a receptor, wherein said single immunoglobulin variable domain polypeptide comprises a CDR3 sequence that is the same sequence of CDR3 of an anti-EGFR dAb disclosed herein.
In other embodiments, the ligand comprises a single immunoglobulin variable domain polypeptide that binds EGFR, wherein the polypeptide has an amino acid sequence that is identical to the amino acid sequence of an anti-EGFR dAb disclosed herein, or differs from the amino acid sequence of an anti-EGFR dAb disclosed herein at no more than 25 amino acid positions and has a CDRl sequence that has at least 50% identity to the CDRl sequence of the anti-EGFR dAb.
In other embodiments, the ligand comprises single immunoglobulin variable domain polypeptide that binds to EGFR, wherein the polypeptide has an amino acid sequence that is identical to the amino acid sequence of anti-EGFR dAb disclosed herein, or differs from the amino acid sequence of anti-EGFR dAb disclosed herein at no more than 25 amino acid positions and has a CDR2 sequence that has at least 50% identity to the CDR2 sequence of the anti-EGFR dAb.
In other embodiments, the ligand comprises an immunoglobulin single variable domain polypeptide that binds EGFR, wherein the polypeptide has an amino acid sequence that is identical to the amino acid sequence of anti-EGFR dAb disclosed herein, or differs from the amino acid sequence of anti-EGFR dAb disclosed herein at no more than 25 amino acid positions and has a CDR3 sequence that has at least 50% identity to the CDR3 sequence of the anti-EGFR dAb.
In other embodiments, the ligand comprises an immunoglobulin single variable domain polypeptide that binds EGFR, wherein the polypeptide has an amino acid sequence that is identical to the amino acid sequence of an anti-EGFR dAb disclosed herein, or differs from the amino acid sequence of an anti-EGFR dAb disclosed herein at no more than 25 amino acid positions and has a CDRl sequence that has at least 50% identity to the CDRl sequence of the anti-EGFR dAb and has a CDR2 sequence has at least 50% identity to the CDR2 sequence of the anti-EGFR dAb.
In other embodiments, the ligand comprises an immunoglobulin single variable domain polypeptide that binds EGFR, wherein the polypeptide has an amino acid sequence that is identical to the amino acid sequence of an anti-EGFR dAb disclosed herein, or differs from the amino acid sequence of an anti-EGFR dAb disclosed herein at no more than 25 amino acid positions and has a CDR2 sequence that has at least 50% identity to the CDR2 sequence of the anti-EGFR dAb and has a CDR3 sequence that has at least 50% identity to the CDR3 sequence of the anti- EGFR dAb.
In other embodiments, the ligand comprises an immunoglobulin single variable domain polypeptide that binds EGFR, wherein the polypeptide has an amino acid sequence that is identical to the amino acid sequence of an anti-EGFR dAb disclosed herein, or differs from the amino acid sequence of an anti-EGFR dAb disclosed herein at no more than 25 amino acid positions and has a CDRl sequence that has at least 50% identity to the CDRl sequence of the anti-EGFR dAb and has a CDR3 sequence that has at least 50% identity to the CDR3 sequence of the anti- EGFR dAb.
In other embodiments, the ligand comprises an immunoglobulin single variable domain polypeptide that binds EGFR, wherein the polypeptide has an amino acid sequence that is identical to the amino acid sequence of an anti-EGFR dAb disclosed herein, or differs from the amino acid sequence of an anti-EGFR dAb disclosed herein at no more than 25 amino acid positions and has a CDRl sequence that has at least 50% identity to the CDRl sequence of the anti-EGFR dAb and has a CDR2 sequence that has at least 50% identity to the CDR2 sequence of the anti- EGFR dAb and has a CDR3 sequence that has at least 50% identity to the CDR3 sequence of the anti-EGFR dAb.
In another embodiment, the invention is an EGFR antagonist having a CDRl sequence that has at least 50% identity to the CDRl sequence of an anti-EGFR dAb disclosed herein. In another embodiment, the invention is an EGFR antagonist having a CDR2 sequence that has at least 50% identity to the CDR2 sequence of an anti-EGFR dAb disclosed herein.
In another embodiment, the invention is an EGFR antagonist having a CDR3 sequence that has at least 50% identity to the CDR3 sequence of an anti-EGFR dAb disclosed herein.
In another embodiment, the invention is an EGFR antagonist having a CDRl sequence that has at least 50% identity to the CDRl sequence of an anti-EGFR dAb disclosed herein and a CDR2 sequence that has at least 50% identity to the CDR2 sequence of the anti-EGFR dAb.
In another embodiment, the invention is an EGFR antagonist having a CDR2 sequence that has at least 50% identity to the CDR2 sequence of an anti-EGFR dAb disclosed herein and a CDR3 sequence that has at least 50% identity to the CDR3 sequence of the anti-EGFR dAb .
In another embodiment, the invention is an EGFR antagonist having a CDRl sequence that has at least 50% identity to the CDRl sequence of an anti-EGFR dAb disclosed herein and a CDR3 sequence that has at least 50% identity to the CDR3 sequence of the anti-EGFR dAb.
In another embodiment, the invention is an EGFR antagonist having a CDRl sequence that has at least 50% identity to the CDRl sequence of an anti-EGFR dAb disclosed herein and a CDR2 sequence that has at least 50% identity to the CDR2 sequence of the anti-EGFR dAb and a CDR3 sequence that has at least 50% identity to the CDR3 sequence of the anti-EGFR dAb.
In some embodiments, the ligand comprises a single immunoglobulin variable domain polypeptide that antagonizes (inhibits) human VEGF binding to a receptor, wherein said single immunoglobulin variable domain polypeptide comprises a CDR3 sequence that is the same sequence of CDR3 of an anti-VEGF dAb disclosed herein.
In other embodiments, the ligand comprises a single immunoglobulin variable domain polypeptide that binds VEGF, wherein the polypeptide has an amino acid sequence that is identical to the amino acid sequence of an anti-VEGF dAb disclosed herein, or differs from the amino acid sequence of an anti-VEGF dAb disclosed herein at no more than 25 amino acid positions and has a CDRl sequence that has at least 50% identity to the CDRl sequence of the anti-VEGF dAb.
In other embodiments, the ligand comprises single immunoglobulin variable domain polypeptide that binds to VEGF, wherein the polypeptide has an amino acid sequence that is identical to the amino acid sequence of anti-VEGF dAb disclosed herein, or differs from the amino acid sequence of anti-VEGF dAb disclosed herein at no more than 25 amino acid positions and has a CDR2 sequence that has at least 50% identity to the CDR2 sequence of the anti-VEGF dAb.
In other embodiments, the ligand comprises an immunoglobulin single variable domain polypeptide that binds VEGF, wherein the polypeptide has an amino acid sequence that is identical to the amino acid sequence of anti-VEGF dAb disclosed herein, or differs from the amino acid sequence of anti-VEGF dAb disclosed herein at no more than 25 amino acid positions and has a CDR3 sequence that has at least 50% identity to the CDR3 sequence of the anti-VEGF dAb.
In other embodiments, the ligand comprises an immunoglobulin single variable domain polypeptide that binds VEGF, wherein the polypeptide has an amino acid sequence that is identical to the amino acid sequence of an anti-VEGF dAb disclosed herein, or differs from the amino acid sequence of an anti-VEGF dAb disclosed herein at no more than 25 amino acid positions and has a CDRl sequence that has at least 50% identity to the CDRl sequence of the anti-VEGF dAb and has a CDR2 sequence has at least 50% identity to the CDR2 sequence of the anti-VEGF dAb.
In other embodiments, the ligand comprises an immunoglobulin single variable domain polypeptide that binds VEGF, wherein the polypeptide has an amino acid sequence that is identical to the amino acid sequence of an anti-VEGF dAb disclosed herein, or differs from the amino acid sequence of an anti-VEGF dAb disclosed herein at no more than 25 amino acid positions and has a CDR2 sequence that has at least 50% identity to the CDR2 sequence of the anti-VEGF dAb and has a CDR3 sequence that has at least 50% identity to the CDR3 sequence of the anti- VEGF dAb.
In other embodiments, the ligand comprises an immunoglobulin single variable domain polypeptide that binds VEGF, wherein the polypeptide has an amino acid sequence that is identical to the amino acid sequence of an anti-VEGF dAb disclosed herein, or differs from the amino acid sequence of an anti-VEGF dAb disclosed herein at no more than 25 amino acid positions and has a CDRl sequence that has at least 50% identity to the CDRl sequence of the anti-VEGF dAb and has a CDR3 sequence that has at least 50% identity to the CDR3 sequence of the anti- VEGF dAb.
In other embodiments, the ligand comprises an immunoglobulin single variable domain polypeptide that binds VEGF, wherein the polypeptide has an amino acid sequence that is identical to the amino acid sequence of an anti-VEGF dAb disclosed herein, or differs from the amino acid sequence of an anti-VEGF dAb disclosed herein at no more than 25 amino acid positions and has a CDRl sequence that has at least 50% identity to the CDRl sequence of the anti-VEGF dAb and has a CDR2 sequence that has at least 50% identity to the CDR2 sequence of the anti- VEGF dAb and has a CDR3 sequence that has at least 50% identity to the CDR3 sequence of the anti-VEGF dAb .
In another embodiment, the invention is an VEGF antagonist having a CDRl sequence that has at least 50% identity to the CDRl sequence of an anti-VEGF dAb disclosed herein.
In another embodiment, the invention is an VEGF antagonist having a CDR2 sequence that has at least 50% identity to the CDR2 sequence of an anti-VEGF dAb disclosed herein.
In another embodiment, the invention is an VEGF antagonist having a CDR3 sequence that has at least 50% identity to the CDR3 sequence of an anti-VEGF dAb disclosed herein.
In another embodiment, the invention is an VEGF antagonist having a CDRl sequence that has at least 50% identity to the CDRl sequence of an anti-VEGF dAb disclosed herein and a CDR2 sequence that has at least 50% identity to the CDR2 sequence of the anti-VEGF dAb.
In another embodiment, the invention is an VEGF antagonist having a CDR2 sequence that has at least 50% identity to the CDR2 sequence of an anti-VEGF dAb disclosed herein and a CDR3 sequence that has at least 50% identity to the CDR3 sequence of the anti-VEGF dAb . In another embodiment, the invention is an VEGF antagonist having a CDRl sequence that has at least 50% identity to the CDRl sequence of an anti-VEGF dAb disclosed herein and a CDR3 sequence that has at least 50% identity to the CDR3 sequence of the anti-VEGF dAb.
In another embodiment, the invention is an VEGF antagonist having a CDRl sequence that has at least 50% identity to the CDRl sequence of an anti-VEGF dAb disclosed herein and a CDR2 sequence that has at least 50% identity to the CDR2 sequence of the anti-VEGF dAb and a CDR3 sequence that has at least 50% identity to the CDR3 sequence of the anti-VEGF dAb.
In additional embodiments, any of the ligands described herein further comprise a toxin, such as a cytotoxin, free radical generator, antimetabolite, protein, polypeptide, peptide, photoactive agent, antisense compound, chemotherapeutic, radionuclide or intrabody. In particular embodiments, the toxin is a surface active toxin (e.g., a free radical generator, a radionuclide).
In other embodiments, the ligand further comprises a half-life extending moiety, such as a polyalkylene glycol moiety, serum albumin or a fragment thereof, transferrin receptor or a transferrin-binding portion thereof, or a moiety comprising a binding site for a polypeptide that enhances half-life in vivo. In some embodiments, the half-life extending moiety is a moiety comprising a binding site for a polypeptide that enhances half-life in vivo selected from the group consisting of an affibody, an SpA domain, an LDL receptor class A domain, an EGF domain, and an avimer.
In other embodiments, the half-life extending moiety is an antibody or antibody fragment (e.g., an immunoglobulin single variable domain) comprising a binding site for serum albumin or neonatal Fc receptor.
In particular embodiments, the half-life extending moiety is an
immunoglobulin single variable domain comprising a binding site for serum albumin that competes for binding to human serum albumin with a dAb selected from the group consisting of DOM7m-16 (SEQ ID NO: 473), DOM7m-12 (SEQ ID NO: 474), DOM7m-26 (SEQ ID NO: 475), DOM7r-l (SEQ ID NO: 476), DOM7r-3 (SEQ ID NO: 477), DOM7r-4 (SEQ ID NO: 478), DOM7r-5 (SEQ ID NO: 479), DOM7r-7 (SEQ ID NO: 480), DOM7r-8 (SEQ ID NO: 481), DOM7h-2 (SEQ ID NO: 482), DOM7h-3 (SEQ ID NO: 483), DOM7h-4 (SEQ ID NO: 484), DOM7h-6 (SEQ ID NO: 485), DOM7h-l (SEQ ID NO: 486), DOM7h-7 (SEQ ID NO: 487), DOM7h-22 (SEQ ID NO: 489), DOM7h-23 (SEQ ID NO: 490), DOM7h-24 (SEQ ID NO: 491), DOM7h-25 (SEQ ID NO: 492), DOM7h-26 (SEQ ID NO: 493), DOM7h-21 (SEQ ID NO: 494), DOM7h-27 (SEQ ID NO: 495), DOM7h-8 (SEQ ID NO: 496), DOM7r-13 (SEQ ID NO: 497), DOM7r-14 (SEQ ID NO: 498), DOM7r- 15 (SEQ ID NO: 499), DOM7r-16 (SEQ ID NO: 500), DOM7r-17 (SEQ ID NO: 501), DOM7r-18 (SEQ ID NO: 502), DOM7r-19 (SEQ ID NO: 503), DOM7r-20 (SEQ ID NO: 504), DOM7r-21 (SEQ ID NO: 505), DOM7r-22 (SEQ ID NO: 506), DOM7r-23 (SEQ ID NO: 507), DOM7r-24 (SEQ ID NO: 508), DOM7r-25 (SEQ ID NO: 509), DOM7r-26 (SEQ ID NO: 510), DOM7r-27 (SEQ ID NO: 511), DOM7r- 28 (SEQ ID NO: 512), DOM7r-29 (SEQ ID NO: 513), DOM7r-30 (SEQ ID NO: 514), DOM7r-31 (SEQ ID NO: 515), DOM7r-32 (SEQ ID NO: 516), and DOM7r- 33 (SEQ ID NO: 517).
For example, the immunoglobulin single variable domain comprising a binding site for serum albumin can comprise an amino acid sequence that has at least 85% amino acid sequence identity with the amino acid sequence of a dAb selected from the group consisting of DOM7m-16 (SEQ ID NO: 473), DOM7m-12 (SEQ ID NO: 474), DOM7m-26 (SEQ ID NO: 475), DOM7r-l (SEQ ID NO: 476), DOM7r-3 (SEQ ID NO: 477), DOM7r-4 (SEQ ID NO: 478), DOM7r-5 (SEQ ID NO: 479), DOM7r-7 (SEQ ID NO: 480), DOM7r-8 (SEQ ID NO: 481), DOM7h-2 (SEQ ID NO: 482), DOM7h-3 (SEQ ID NO: 483), DOM7h-4 (SEQ ID NO: 484), DOM7h-6 (SEQ ID NO: 485), DOM7h-l (SEQ ID NO: 486), DOM7h-7 (SEQ ID NO: 487), DOM7h-22 (SEQ ID NO: 489), DOM7h-23 (SEQ ID NO: 490), DOM7h- 24 (SEQ ID NO: 491), DOM7h-25 (SEQ ID NO: 492), DOM7h-26 (SEQ ID NO: 493), DOM7h-21 (SEQ ID NO: 494), DOM7h-27 (SEQ ID NO: 495), DOM7h-8 (SEQ ID NO: 496), DOM7r-13 (SEQ ID NO: 497), DOM7r-14 (SEQ ID NO: 498), DOM7r-15 (SEQ ID NO: 499), DOM7r-16 (SEQ ID NO: 500), DOM7r-17 (SEQ ID NO: 501), DOM7r-18 (SEQ ID NO: 502), DOM7r-19 (SEQ ID NO: 503), DOM7r- 20 (SEQ ID NO: 504), DOM7r-21 (SEQ ID NO: 505), DOM7r-22 (SEQ ID NO: 506), DOM7r-23 (SEQ ID NO: 507), DOM7r-24 (SEQ ID NO: 508), DOM7r-25 (SEQ ID NO: 509), DOM7r-26 (SEQ ID NO: 510), DOM7r-27 (SEQ ID NO: 511), DOM7r-28 (SEQ ID NO: 512), DOM7r-29 (SEQ ID NO: 513), DOM7r-30 (SEQ ID NO: 514), DOM7r-31 (SEQ ID NO: 515), DOM7r-32 (SEQ ID NO: 516), and
DOM7r-33 (SEQ ID NO: 517).
The invention also relates to an isolated or recombinant nucleic acid encoding a ligand described herein, and to a vector (e.g., recombinant vector) comprising the recombinant nucleic acid. The invention also relates to a host cell
(e.g., recombinant host cell, isolated host cell) comprising a recombinant nucleic acid or vector of the invention. The invention also relates to a method for producing a ligand, comprising maintaining a host cell of the invention under conditions suitable for expression of said nucleic acid or vector, whereby a ligand is produced. In some embodiments, the method further comprises isolating the ligand.
The invention also relates to a ligand of the invention for use in therapy or diagnosis, and to the use of a ligand of the invention for the manufacture of a medicament for treatment, prevention or suppression of a disease described herein
(e.g., cancer).
The invention also relates to a pharmaceutical compositon for the treatment, prevention or suppression of a disease described herein (e.g., cancer) comprising as an active ingredient a ligand of the invention.
In some embodiments, the invention relates to a ligand for use in treating cancer, or cancer cells that overexpress EGFR and/or VEGF.
In other embodiments, the invention relates to use of a ligand for the manufacture of a medicament for killing cells (e.g., selectively killing cancer cells over normal cells).
In other embodiments, the invention relates to use of a ligand for the manufacture of a medicament for treating cancer cells that overexpress EGFR and/or VEGF.
The invention also relates to therapeutic methods that comprise
administering a therapeutically effective amount of a ligand of the invention to a subject in need thereof. In one embodiment, the invention relates to a method for treating cancer comprising administering to a subject in need thereof a
therapeutically effective amount of ligand of the invention. In some embodiments, the method further comprises administering to the subject a chemotherapeutic agent
(e.g., at a low dose). In other embodiments, the method for treating cancer comprises
administering to a subject in need thereof a therapeutically effective amount of ligand of the invention and an anti-neoplastic composition, wherein said antineoplastic composition comprises at least one chemotherapeutic agent. The chemotherapeutic agent can be selected from the group consisting of alkylating agents, antimetabolites, folic acid analogs, pyrimidine analogs, purine analogs and related inhibitors, vinca alkaloids, epipodopyyllotoxins, antibiotics, L-Asparaginase, topoisomerase inhibitor, interferons, platinum cooridnation complexes,
anthracenedione substituted urea, methyl hydrazine derivatives, adrenocortical suppressant, adrenocorticosteroides, progestins, estrogens, antiestrogen, androgens, antiandrogen, and gonadotropin-releasing hormone analog. In some embodiments, the chemotherapeutic agent is selected from the group consisting of cisplatin, dicarbazine, dactinomycin, mechlorethamine, streptozocin, cyclophosphamide, capecitabine, carmustine, lomustine, doxorubicin, daunorubicin, procarbazine, mitomycin, cytarabine, etoposide, methotrexate, 5-fluorouracil, vinblastine, vincristine, bleomycin, paclitaxel, docetaxel, doxetaxe, aldesleukin, asparaginase, busulfan, carboplatin, cladribine, dacarbazine, fioxuridine, fludarabine, hydroxyurea, ifosfamide, interferon alpha, irinotecan, leuprolide, leucovorin, megestrol, melphalan, mercaptopurine, oxaliplatin, plicamycin, mitotane, pegaspargase, pentostatin, pipobroman, plicamycin, streptozocin, tamoxifen, teniposide, testolactone, thioguanine, thiotepa, uracil mustard, vinorelbine, chlorambucil, taxol, an additional growth factor receptor antagonist, and a combination of any of the foregoing.
In some embodiments, the method is a method of treating a cancer selected from the group consisting of bladder cancer, ovarian cancer, colorectal cancer
(colorectal carcinoma), breast cancer, lung cancer (non-small cell lung carcinoma), gastric cancer, pancreatic cancer, prostate cancer, head and neck cancer, renal cancer and gall bladder cancer.
The invention also relates to a method of administering to a subject anti- VEGF treatment and anti-EGFR treatment, the method comprising simultaneous administration of an anti-VEGF treatment and an anti-EGFR treatment by administering to said subject a therapeutically effective amount of a ligand that has binding specificity for VEGF and EGFR.
The invention also relates to a composition {e.g., pharmaceutical
composition) comprising a ligand of the invention and a physiologically or pharmaceutically acceptable carrier. In some embodiments, the composition comprises a vehicle for intravenous, intramuscular, intraperitoneal, intraarterial, intrathecal, intraarticular subcutaneous administration, pulmonary, intranasal, vaginal, or rectal administration.
The invention also relates to a drug delivery device comprising the composition {e.g., pharmaceutical composition) of the invention or a ligand of the invention. In one embodiment, the drug delivery device is for simultaneously administering to a subject anti-VEGF treatment and anti-EGFR treatment, and the device comprising a ligand that has binding specificity for VEGF and EGFR. In some embodiments, the drug device comprises a plurality of therapeutically effective doses of ligand.
In other embodiments, the drug delivery device is selected from the group consisting of a parenteral delivery device, intravenous delivery device, intramuscular delivery device, intraperitoneal delivery device, transdermal delivery device, pulmonary delivery device, intraarterial delivery device, intrathecal delivery device, intraarticular delivery device, subcutaneous delivery device, intranasal delivery device, vaginal delivery device, rectal delivery device, a syringe, a transdermal delivery device, a capsule, a tablet, a nebulizer, an inhaler, an atomizer, an aerosolizer, a mister, a dry powder inhaler, a metered dose inhaler, a metered dose sprayer, a metered dose mister, a metered dose atomizer, a catheter.
The invention also relates to a ligand that has binding specificity for vascular endothelial growth factor (VEGF) and epidermal growth factor receptor (EGFR), comprising at least one protein moiety that has a binding site with binding specificity for VEGF, at least one protein moiety that has a binding site with binding specificity for EGFR, and an Fc region of an antibody. Such ligands can consist of a single polypeptide. In other embodiments, two ligands that contain Fc regions are bonded together, for example through a disulfide bond {e.g., in the hinge region), to form a dimer. In some embodiments, the ligand that has binding specificity for VEGF can comprise a protein moiety that has a binding site with binding specificity for VEGF and an Fc region of an antibody. In some embodiments, the protein moiety having binding specificity for VEGF is fused to an Fc region of an antibody.
In other embodiments, the ligand that has binding specificity for EGFR can comprise a protein moiety that has a binding site with binding specificity for EGFR and an Fc region of an antibody. In some embodiments, the protein moiety having binding specificity for EGFR is fused to an Fc region of an antibody. For example, the ligand can comprise two protein moieties that have binding sites with binding specificity for EGFR and an Fc region of an antibody.
Additionally, or in other embodiments, the ligand that has binding specificity for VEGF and EGFR, comprises a single variable domain with binding specificity for VEGF, a single variable domain with binding specificity for EGFR, and optionally a linker. In such embodiments, the single variable domain with binding specificity for EGFR can be bonded via the linker to the immunoglobulin single variable domain with binding specificity for VEGF. Suitable linkers include SEQ ID NO:706, SEQ ID NO:707, SEQ ID NO:708, SEQ ID NO:709, SEQ ID NO:710, SEQ ID NO:711, SEQ ID NO:712, SEQ ID NO:713, SEQ ID NO:714, SEQ ID NO:723 and SEQ ID NO:724. The ligand can also comprise an Fc region of an antibody if desired. When the ligand further comprises an Fc region of an antibody, a linker can bind an immunoglobulin variable domain to the Fc region. In other embodiments, two ligands that contain Fc regions are bonded together, for example through a disulfide bond (e.g., in the hinge region), to form a dimer.
Additionally, or in other embodiments, the ligand that has binding specificity for VEGF and EGFR, comprises a single variable domain with binding specificity for VEGF directly fused to a single variable domain with binding specificity for EGFR.
In embodiments where the ligand comprises a single variable domain with binding specificity for VEGF, and a single variable domain with binding specificity for EGFR, and optionally one or more linkers, the single variable domains can independently be a light chain variable domain or a heavy chain variable domain. For example, the ligand can comprise a) a single variable domain with binding specificity for VEGF that is a heavy chain variable region, and the immunoglobulin single variable domain with binding specificity for EGFR that is a light chain variable region; b) a single variable domain with binding specificity for VEGF that is a light chain variable domain, and a single variable domain with binding specificity for EGFR that is a heavy chain variable domain; c) a single variable domain with binding specificity for VEGF that is a heavy chain variable domain, and a single variable domain with binding specificity for EGFR that is a heavy chain variable domain; or d) a single variable domain with binding specificity for VEGF that is a light chain variable domain, and a single variable domain with binding specificity for EGFR that is a light chain variable domain. In particular
embodiments, the heavy chain variable region is a VH or VHH- In further
embodiments, the VH is a human VH. In other embodiments, the light chain variable region is a VK-
In another aspect of the invention, a ligand that has binding specificity for VEGF and EGFR comprises at least one immunoglobulin single variable domain with binding specificity for VEGF and at least one immunoglobulin single variable domain with binding specificity for EGFR, wherein the immunoglobulin single variable domain with binding specificity for EGFR is bonded via a disulfide bond to the immunoglobulin single variable domain with binding specificity for VEGF. Alternatively, a ligand that has binding specificity for VEGF and EGFR can comprises at least one immunoglobulin single variable domain with binding specificity for VEGF and at least one immunoglobulin single variable domain with binding specificity for EGFR, wherein the immunoglobulin single variable domain with binding specificity for EGFR is directly fused to the immunoglobulin single variable domain with binding specificity for VEGF (z. e. , a single polypeptide comprising two dAbs).
In other particular embodiments, the ligand is a fusion of a dAb to an antiserum albumin dAb (a DOM7 dAb). For example, the ligand can have the structure, from amino-terminal to carboxy-terminal, DOMl 5-10— DOMl 6-39— anti-serum albumin dAb, DOM16-39— DOM15-10— anti-serum albumin dAb, DOM15-26- 501— DOM16-39— anti-serum albumin dAb, or DOM16-39— DOM15-26-501— anti-serum albumin dAb. In additional embodiments, the ligand that has a binding site with binding specificity for EGFR can compete for binding to EGFR with cetuximab and/or panitumumab and is fused to an anti-serum albumin dAb.
Additionally, or in other embodiments, the ligand can comprise two or more dAbs (e.g. anti-EGFR dAbs) fused to an anti-serum albumin dAb.
BRIEF DESCRIPTION OF THE DRAWINGS
FIG. IA- IE illustrates twenty-seven nucleotide sequences that encode human (Homo sapiens) domain antibodies (dAbs) that specifically bind human VEGF. The nucleotide sequences presented are SEQ ID NOS:l-27, 535 and 536.
FIG. 2A-2C is a alignment of twelve nucleotide sequences that encode human dAbs that bind human VEGF. The nucleotide sequences presented are SEQ ID NO: 18 and SEQ ID NOS:28-38.
FIG. 3A-3D is a alignment of twelve nucleotide sequences that encode human dAbs that bind human VEGF. The nucleotide sequences presented are SEQ ID NO:20 and SEQ ID NOS:39-49.
FIG. 4A-4J is a alignment of fifty-three nucleotide sequences that encode human dAbs that bind human VEGF. The nucleotide sequences presented are SEQ ID NO:24, 50-99, 537 and 538.
FIG. 5A-5C illustrates the amino acid sequences of dAbs encoded by several of the nucleic acid sequences shown in FIG. 1A-1E. The amino acid sequences presented are SEQ ID NOS: 100-126.
FIG. 6 is an alignment of the amino acid sequences of the dAbs encoded by the nucleic acid sequences shown in FIG. 2A-2C. The amino acid sequences presented are SEQ ID NO:117 AND SEQ ID NOS:127-137.
FIG. 7A-7B is an alignment of the amino acid sequences of the dAbs encoded by the nucleic acid sequences shown in FIG. 3A-2D. The symbol ~ has been inserted into the sequence of TARl 5-8-500 to facilitate alignment. The amino acid sequences presented are SEQ ID NO:119 and SEQ ID NOS:138-148.
FIG. 8A-8D is an alignment of the amino acid sequences of the dAbs encoded by the nucleic acid sequences shown in FIG. 4A-4J. The amino acid sequences presented are SEQ ID NO:123, 149-198, 539 and 540. FIG. 9A-9O illustrates several nucleotide sequences that encode human (Homo sapiens) domain antibodies (dAbs) that specifically bind human EGFR. The nucleotide sequences presented are SEQ ID NOS: 199-324.
FIG. 10A- 101 illustrates the amino acid sequences of the dAbs encoded by the nucleic acid sequences shown in FIG. 9A-9O. The amino acid sequences presented are SEQ ID NOS:325-450.
FIG. 1 IA-I IB illustrates the amino acid sequences of several Camelid VHHS that bind EGFR that are disclosed in WO 2005/044858. NBl (SEQ ID NO:451), NB2 (SEQ ID NO:452), NB3 (SEQ ID NO:453), NB4 (SEQ ID NO:454), NB5 (SEQ ID NO:455), NB6 (SEQ ID NO:456), NB7 (SEQ ID NO:457), NB8 (SEQ ID NO:458), NB9 (SEQ ID NO:459), NBlO (SEQ ID NO:460), NBI l (SEQ ID NO:461), NB12 (SEQ ID NO:462), NB13 (SEQ ID NO:463), NB14 (SEQ ID NO:464), NB15 (SEQ ID NO:465), NB16 (SEQ ID NO:466), NB17 (SEQ ID NO:467), NB 18 (SEQ ID NO:468), NB 19 (SEQ ID NO:469), NB20 (SEQ ID NO:470), NB21 (SEQ ID NO:471), NB22 (SEQ ID NO:472).
FIG. 12A is an alignment of the amino acid sequences of three Vies that bind mouse serum albumin (MSA). The aligned amino acid sequences are from VKS designated MSA16, which is also referred to as DOM7m-16 (SEQ ID NO: 473), MSA 12, which is also referred to as DOM7m-12 (SEQ ID NO: 474), and MSA 26, which is also referred to as DOM7m-26 (SEQ ID NO: 475).
FIG. 12B is an alignment of the amino acid sequences of six VKS that bind rat serum albumin (RSA). The aligned amino acid sequences are from VKS designated DOM7r-l (SEQ ID NO: 476), DOM7r-3 (SEQ ID NO: 477), DOM7r-4 (SEQ ID NO: 478), DOM7r-5 (SEQ ID NO: 479), DOM7r-7 (SEQ ID NO: 480), and DOM7r-8 (SEQ ID NO: 481).
FIG. 12C is an alignment of the amino acid sequences of six VKS that bind human serum albumin (HSA). The aligned amino acid sequences are from VKS designated DOM7h-2 (SEQ ID NO: 482), DOM7h-3 (SEQ ID NO: 483), DOM7h-4 (SEQ ID NO: 484), DOM7h-6 (SEQ ID NO: 485), DOM7h-l (SEQ ID NO: 486), and DOM7h-7 (SEQ ID NO: 487).
FIG. 12D is an alignment of the amino acid sequences of seven VHS that bind human serum albumin and a consensus sequence (SEQ ID NO: 488). The aligned sequences are from VHs designated DOM7h-22 (SEQ ID NO: 489), DOM7h-23 (SEQ ID NO: 490), DOM7h-24 (SEQ ID NO: 491), DOM7h-25 (SEQ ID NO: 492), DOM7h-26 (SEQ ID NO: 493), DOM7h-21 (SEQ ID NO: 494), and DOM7h-27 (SEQ ID NO: 495).
FIG. 12E is an alignment of the amino acid sequences of three VKS that bind human serum albumin and rat serum albumin. The aligned amino acid sequences are from VKS designated DOM7h-8 (SEQ ID NO: 496), DOM7r-13 (SEQ ID NO: 497), and DOM7r-14 (SEQ ID NO: 498).
FIG. 13 is an illustration of the amino acid sequences of Vies that bind rat serum albumin (RSA). The illustrated sequences are from VKS designated DOM7r- 15 (SEQ ID NO: 499), DOM7r-16 (SEQ ID NO: 500), DOM7r-17 (SEQ ID NO: 501), DOM7r-18 (SEQ ID NO: 502), DOM7r-19 (SEQ ID NO: 503).
FIG. 14A-14B is an illustration of the amino acid sequences of VHS that bind rat serum albumin (RSA). The illustrated sequences are from VHS designated DOM7r-20 (SEQ ID NO: 504), DOM7r-21 (SEQ ID NO: 505), DOM7r-22 (SEQ ID NO: 506), DOM7r-23 (SEQ ID NO: 507), DOM7r-24 (SEQ ID NO: 508), DOM7r- 25 (SEQ ID NO: 509), DOM7r-26 (SEQ ID NO: 510), DOM7r-27 (SEQ ID NO: 511), DOM7r-28 (SEQ ID NO: 512), DOM7r-29 (SEQ ID NO: 513), DOM7r-30 (SEQ ID NO: 514), DOM7r-31 (SEQ ID NO: 515), DOM7r-32 (SEQ ID NO: 516), and DOM7r-33 (SEQ ID NO: 517).
FIG. 15 illustrates the amino acid sequences of several Camelid VHHS that bind mouse serum albumin that are disclosed in WO 2004/041862. Sequence A (SEQ ID NO: 518), Sequence B (SEQ ID NO: 519), Sequence C (SEQ ID NO: 520), Sequence D (SEQ ID NO: 521), Sequence E (SEQ ID NO: 522), Sequence F (SEQ ID NO: 523), Sequence G (SEQ ID NO: 524), Sequence H (SEQ ID NO: 525),
Sequence I (SEQ ID NO:526), Sequence J (SEQ ID NO:527), Sequence K (SEQ ID NO: 528), Sequence L (SEQ ID NO:529), Sequence M (SEQ ID NO:530), Sequence N (SEQ ID NO:531), Sequence O (SEQ ID NO: 532), Sequence P (SEQ ID
NO:533), Sequence Q (SEQ ID NO:534).
FIG. 16 is a map of a vector used to prepare IgG-like formats.
FIG. 17A-17F illustrates the amino acid sequences of human dAbs that bind human EGFR. The amino acid sequences presented are SEQ ID NOS:541-622, 725 and 726. The sequences are continuous with no gaps, the symbols ~, ~~ and ~~~ have been inserted to show the locations of the CDRs. CDRl is flanked by ~, CDR2 is flanked by ~~, and CDR3 is flanked by ~~~.
FIG. 18A-18L illustrates nucleotide sequences that encode the dAbs shown in FIG. 17A-17F. The nucleotide sequences presented are SEQ ID NOS:623-703, 727 and 728.
FIG. 19 illustrates the amino acid sequence (SEQ ID NO:704) of a human dAb that binds VEGF, and a nucleotide sequence (SEQ ID NO:705) that encodes the dAb. The sequences are continuous with no gaps, the symbols ~,— and ~~~ have been inserted to show the locations of the CDRs. CDRl is flanked by ~, CDR2 is flanked by— , and CDR3 is flanked by ~~~.
DETAILED DESCRIPTION OF THE INVENTION
Within this specification embodiments have been described in a way which enables a clear and concise specification to be written, but it is intended and will be appreciated that embodiments may be variously combined or separated without parting from the invention.
As used herein, the term "ligand" refers to a compound that comprises at least one peptide, polypeptide or protein moiety that has a binding site with binding specificity for a desired endogenous target compound. The ligands according to the invention preferably comprise immunoglobulin variable domains which have different binding specificities, and do not contain variable domain pairs which have the same specificity. Preferably each domain which has a binding site that has binding specificity for a cell surface target is an immunoglobulin single variable domain {e.g., immunoglobulin single heavy chain variable domain {e.g., VH, VHH) immunoglobulin single light chain variable domain {e.g., VL)) that has binding specificity for a desired cell surface target {e.g., a membrane protein, such as a receptor protein). Each polypeptide domain which has a binding site that has binding specificity for a cell surface target can also comprise one or more complementarity determining regions (CDRs) of an antibody or antibody fragment {e.g., an immunoglobulin single variable domain) that has binding specificity for a desired cell surface target in a suitable format, such that the binding domain has binding specificity for the cell surface target. For example, the CDRs can be grafted onto a suitable protein scaffold or skeleton, such as an affibody, an SpA scaffold, an LDL receptor class A domain, or an EGF domain. Further, the ligand can be bivalent (heterobivalent) or multivalent (heteromultivalent) as described herein. The first and second domains lack domains that share the same specificity. Thus, "ligands" include polypeptides that comprise two dAbs wherein each dAb binds to a different cell surface target. Ligands also include polypeptides that comprise at least two dAbs that bind different cell surface targets (or the CDRs of a dAbs) in a suitable format, such as an antibody format (e.g., IgG-like format, scFv, Fab, Fab', F(ab')2) or a suitable protein scaffold or skeleton, such as an affibody, an SpA scaffold, an LDL receptor class A domain, an EGF domain, avimer and
multispecific ligands as described herein. The polypeptide domain which has a binding site that has binding specificity for a cell surface target (i.e., first or second cell surface target) can also be a protein domain comprising a binding site for a desired target, e.g., a protein domain is selected from an affibody, an SpA domain, an LDL receptor class A domain, an avimer (see, e.g., U.S. Patent Application Publication Nos. 2005/0053973, 2005/0089932, 2005/0164301). If desired, a "ligand" can further comprise one or more additional moieties, that can each independently be a peptide, polypeptide or protein moiety or a non-peptidic moiety (e.g., a polyalkylene glycol, a lipid, a carbohydrate). For example, the ligand can further comprise a half-life extending moiety as described herein (e.g., a
polyalkylene glycol moiety, a moiety comprising albumin, an albumin fragment or albumin variant, a moiety comprising transferrin, a transferrin fragment or transferrin variant, a moiety that binds albumin, a moiety that binds neonatal Fc receptor).
As used herein, the phrase "target" refers to a biological molecule (e.g., peptide, polypeptide, protein, lipid, carbohydrate) to which a polypeptide domain which has a binding site can bind. The target can be, for example, an intracellular target (e.g., an intracellular protein target) or a cell surface target (e.g., a membrane protein, a receptor protein). Preferably, the target is VEGF or EGFR.
The phrase "immunoglobulin single variable domain" refers to an antibody variable region (VH, VHH, VL) that specifically binds a target, antigen or epitope independently of other V domains; however, as the term is used herein, an immuno globulin single variable domain can be present in a format (e.g., hetero- multimer) with other variable regions or variable domains where the other regions or domains are not required for antigen binding by the single immunoglobulin variable domain (i.e., where the immunoglobulin single variable domain binds antigen independently of the additional variable domains). Each "Immunoglobulin single variable domain" encompasses not only an isolated antibody single variable domain polypeptide, but also larger polypeptides that comprise one or more monomers of an antibody single variable domain polypeptide sequence. A "domain antibody" or "dAb" is the same as an "immunoglobulin single variable domain" polypeptide as the term is used herein. An immunoglobulin single variable domain polypeptide, as used herein refers to a mammalian immunoglobulin single variable domain polypeptide, preferably human, but also includes rodent (for example, as disclosed in WO 00/29004, the contents of which are incorporated herein by reference in their entirety) or camelid VHH dAbs. As used herein, camelid dAbs are immunoglobulin single variable domain polypeptides which are derived from species including camel, llama, alpaca, dromedary, and guanaco, and comprise heavy chain antibodies naturally devoid of light chain (VHH)- Similar dAbs, can be obtained for single chain antibodies from other species, such as nurse shark. Preferred ligands comprises at least two different immunoglobulin single variable domain polypeptides or at least two different dAbs.
A "human" immunoglobulin single variable domain (e.g., dAb, VH, VL, VK, Vλ) can be derived from an antibody of human origin or from a library prepared using human antibody variable region genes. For example, as described herein, human immunoglobulin single variable domains have one or more framework regions that are encoded by a human germline antibody gene segment, or that have up to 5 amino acid differences relative to the amino acid sequence encoded by a human germline antibody gene segment. Preferably, the amino acid sequences of FWl, FW2, FW3 and FW4 are each encoded by a human germline antibody gene segment, or collectively contain up to 10 amino acid differences relative to the amino acid sequences of the corresponding framework regions encoded by the human germline antibody gene segment. As used herein "vascular endothelial growth factor" (VEGF) refers to naturally occurring or endogenous mammalian VEGF-A proteins and to proteins having an amino acid sequence which is the same as that of a naturally occurring or endogenous corresponding mammalian VEGF-A protein (e.g., recombinant proteins, synthetic proteins (i.e., produced using the methods of synthetic organic chemistry)). Accordingly, as defined herein, the term includes mature VEGF-A protein, polymorphic or allelic variants, and other isoforms of a VEGF-A (e.g., produced by alternative splicing or other cellular processes), and modified or unmodified forms of the foregoing (e.g., lipidated, glycosylated). Alternative splicing of RNA encoding human (Homo sapiens) VEGF-A yield several isoforms of human VEGF- A that differ in the number of amino acids in the protein sequence. For example, isoforms referred to as VEGF-121, VEGF-165, VEGF-189 and VEGF-206 are produced in humans. (See, e.g., Ferrara, N., Endocrine Reviews, 25(4):581-611 (2004).) These isoforms and other naturally occurring isoforms are expressly encompassed by the term "VEGF". Naturally occurring or endogenous VEGF-A include wild type proteins such as mature VEGF-A, polymorphic or allelic variants and other isoforms which occur naturally in mammals (e.g., humans, non-human primates). Such proteins can be recovered or isolated from a source which naturally produces VEGF-A, for example. These proteins and proteins having the same amino acid sequence as a naturally occurring or endogenous corresponding VEGF, are referred to by the name of the corresponding mammal. For example, where the corresponding mammal is a human, the protein is designated as a human VEGF.
A ligand (e.g., immunoglobulin single variable domain) that inhibits binding of VEGF to VEGFRl or VEGFR2 inhibits binding in the VEGFRl binding assay or VEGFR2 assay described herein by at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 85%, at least about 90%, or at least about 95% when the ligand is assayed at a concentration of about 1 nM, about 10 nM, about 50 nM, about 100 nM, about 1 μM, about 10 μM or about 100 μM. A ligand that inhibits binding of VEGF to VEGFRl or VEGFR2, can also or alternatively, inhibit binding in the VEGFRl binding assay or VEGFR2 assay with an IC50 of about 1 μM or less, about 500 nM or less, about 100 nM or less, about 75 nM or less, about 50 nM or less, about 10 nM or less or about 1 nM or less. A ligand (e.g., immunoglobulin single variable domain) that inhibits activity of VEGF inhibits viability in the VEGF bioassay described herein by at least about 20%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 85%, at least about 90%, or at least about 95%.
A ligand (e.g., immunoglobulin single variable domain) that does not substantially inhibit binding of VEGF to VEGFRl or VEGFR2 does not
significantly inhibit binding in the VEGFRl binding assay or VEGFR2 assay described herein. For example, such a ligand might inhibit binding of VEGF in the VEGFRl binding assay or VEGFR2 assay described herein with an IC50 of about 1 mM or higher, or inhibit binding by no more than about 20%, no more than about 15%, no more than about 10% or no more than about 5%.
As used herein "epidermal growth factor receptor" (EGFR, ErbBl, HERl) refers to naturally occurring or endogenous mammalian EGFR proteins and to proteins having an amino acid sequence which is the same as that of a naturally occurring or endogenous corresponding mammalian EGFR protein (e.g.,
recombinant proteins, synthetic proteins (i.e., produced using the methods of synthetic organic chemistry)). Accordingly, as defined herein, the term includes mature EGFR protein, polymorphic or allelic variants, and other isoforms of a EGFR (e.g., produced by alternative splicing or other cellular processes), and modified or unmodified forms of the foregoing (e.g., lipidated, glycosylated).
Naturally occurring or endogenous EGFR include wild type proteins such as mature EGFR, polymorphic or allelic variants and other isoforms which occur naturally in mammals (e.g., humans, non-human primates). Such proteins can be recovered or isolated from a source which naturally produces EGFR, for example. These proteins and proteins having the same amino acid sequence as a naturally occurring or endogenous corresponding EGFR, are referred to by the name of the corresponding mammal. For example, where the corresponding mammal is a human, the protein is designated as a human EGFR.
A ligand (e.g., immunoglobulin single variable domain) that inhibits binding of EGF and/or TGF alpha to EGFR inhibits binding in the EGFR binding assay or EGFR kinase assay described herein with an IC50 of about 1 μM or less, about 500 nM or less, about 100 nM or less, about 75 nM or less, about 50 nM or less, about 10 nM or less or about 1 nM or less.
A ligand {e.g., immunoglobulin single variable domain) that inhibits activity of EGFR inhibits kinase activity of EGFR in the EGFR kinase assay described herein with an IC50 of about 1 μM or less, about 500 nM or less, about 100 nM or less, about 75 nM or less, about 50 nM or less, about 10 nM or less or about 1 nM or less.
A ligand {e.g., immunoglobulin single variable domain) that does not substantially inhibit binding of EGF or TGF alpha to EGFR does not significantly inhibit binding of EGF and/or TGF alpha to EGFR in the receptor binding assay or kinase assay described herein. For example, such a ligand might inhibit binding of EGF or TGF alpha to EGFR in the receptor binding assay or kinase assay described herein with an IC50 of about 1 mM or higher.
"Affinity" and "avidity" are terms of art that describe the strength of a binding interaction. With respect to the ligands of the invention, avidity refers to the overall strength of binding between the targets {e.g., first cell surface target and second cell surface target) on the cell and the ligand. Avidity is more than the sum of the individual affinities for the individual targets.
As used herein, "toxin moiety" refers to a moiety that comprises a toxin. A toxin is an agent that has deleterious effects on or alters cellular physiology {e.g., causes cellular necrosis, apoptosis or inhibits cellular division).
As used herein, the term "dose" refers to the quantity of ligand administered to a subject all at one time (unit dose), or in two or more administrations over a defined time interval. For example, dose can refer to the quantity of ligand {e.g., ligand comprising an immunoglobulin single variable domain that binds VEGF and an immunoglobulin single variable domain that binds EGFR) administered to a subject over the course of one day (24 hours) (daily dose), two days, one week, two weeks, three weeks or one or more months {e.g., by a single administration, or by two or more administrations). The interval between doses can be any desired amount of time.
As used herein "complementary" refers to when two immunoglobulin domains belong to families of structures which form cognate pairs or groups or are derived from such families and retain this feature. For example, a Vpj domain and a VL domain of an antibody are complementary; two Vg domains are not
complementary, and two VL domains are not complementary. Complementary domains may be found in other members of the immunoglobulin superfamily, such as the Vα and Vβ (or γ and δ) domains of the T-cell receptor. Domains which are artificial, such as domains based on protein scaffolds which do not bind epitopes unless engineered to do so, are non- complementary. Likewise, two domains based on (for example) an immunoglobulin domain and a fibronectin domain are not complementary.
As used herein, "immunoglobulin" refers to a family of polypeptides which retain the immunoglobulin fold characteristic of antibody molecules, which contains two β sheets and, usually, a conserved disulphide bond. Members of the
immunoglobulin superfamily are involved in many aspects of cellular and non- cellular interactions in vivo, including widespread roles in the immune system (for example, antibodies, T-cell receptor molecules and the like), involvement in cell adhesion (for example the ICAM molecules) and intracellular signaling (for example, receptor molecules, such as the PDGF receptor). The present invention is applicable to all immunoglobulin superfamily molecules which possess binding domains. Preferably, the present invention relates to antibodies.
As used herein "domain" refers to a folded protein structure which retains its tertiary structure independently of the rest of the protein. Generally, domains are responsible for discrete functional properties of proteins, and in many cases maybe added, removed or transferred to other proteins without loss of function of the remainder of the protein and/or of the domain. By single antibody variable domain is meant a folded polypeptide domain comprising sequences characteristic of antibody variable domains. It therefore includes complete antibody variable domains and modified variable domains, for example in which one or more loops have been replaced by sequences which are not characteristic of antibody variable domains, or antibody variable domains which have been truncated or comprise N- or C-terminal extensions, as well as folded fragments of variable domains which retain at least in part the binding activity and specificity of the full-length domain. Thus, each ligand comprises at least two different domains. "Repertoire" A collection of diverse variants, for example polypeptide variants which differ in their primary sequence. A library used in the present invention will encompass a repertoire of polypeptides comprising at least 1000 members.
"Library" The term library refers to a mixture of heterogeneous polypeptides or nucleic acids. The library is composed of members, each of which have a single polypeptide or nucleic acid sequence. To this extent, library is synonymous with repertoire. Sequence differences between library members are responsible for the diversity present in the library. The library may take the form of a simple mixture of polypeptides or nucleic acids, or may be in the form of organisms or cells, for example bacteria, viruses, animal or plant cells and the like, transformed with a library of nucleic acids. Preferably, each individual organism or cell contains only one or a limited number of library members. Advantageously, the nucleic acids are incorporated into expression vectors, in order to allow expression of the polypeptides encoded by the nucleic acids. In a preferred aspect, therefore, a library may take the form of a population of host organisms, each organism containing one or more copies of an expression vector containing a single member of the library in nucleic acid form which can be expressed to produce its
corresponding polypeptide member. Thus, the population of host organisms has the potential to encode a large repertoire of genetically diverse polypeptide variants.
As used herein an antibody refers to IgG, IgM, IgA, IgD or IgE or a fragment (such as a Fab , F(ab')2, Fv, disulphide linked Fv, scFv, closed conformation multispecific antibody, disulphide-linked scFv, diabody) whether derived from any species naturally producing an antibody, or created by recombinant DNA
technology; whether isolated from serum, B-cells, hybridomas, transfectomas, yeast or bacteria.
As described herein an "antigen' is a molecule that is bound by a binding domain according to the present invention. Typically, antigens are bound by antibody ligands and are capable of raising an antibody response in vivo. It may be a polypeptide, protein, nucleic acid or other molecule. Generally, the dual-specific ligands according to the invention are selected for target specificity against two particular targets {e.g., antigens). In the case of conventional antibodies and fragments thereof, the antibody binding site defined by the variable loops (Ll , L2, L3 and Hl, H2, H3) is capable of binding to the antigen.
An "epitope" is a unit of structure conventionally bound by an
immunoglobulin VJJ/VL pair- Epitopes define the minimum binding site for an antibody, and thus represent the target of specificity of an antibody. In the case of a single domain antibody, an epitope represents the unit of structure bound by a variable domain in isolation.
"Universal framework" refers to a single antibody framework sequence corresponding to the regions of an antibody conserved in sequence as defined by Kabat ("Sequences of Proteins of Immunological Interest", US Department of Health and Human Services) or corresponding to the human germline
immunoglobulin repertoire or structure as defined by Chothia and Lesk, (1987) J. MoI. Biol. 196:910-917. The invention provides for the use of a single framework, or a set of such frameworks, which has been found to permit the derivation of virtually any binding specificity though variation in the hypervariable regions alone.
The phrase, "half-life," refers to the time taken for the serum concentration of the ligand to reduce by 50%, in vivo, for example due to degradation of the ligand and/or clearance or sequestration of the dual-specific ligand by natural mechanisms. The ligands of the invention are stabilized in vivo and their half-life increased by binding to molecules which resist degradation and/or clearance or sequestration.
Typically, such molecules are naturally occurring proteins which themselves have a long half-life in vivo. The half-life of a ligand is increased if its functional activity persists, in vivo, for a longer period than a similar ligand which is not specific for the half-life increasing molecule. Thus a ligand specific for HSA and two target molecules is compared with the same ligand wherein the specificity to HSA is not present, that is does not bind HSA but binds another molecule. For example, it may bind a third target on the cell. Typically, the half-life is increased by 10%, 20%, 30%, 40%, 50% or more. Increases in the range of 2x, 3x, 4x, 5x, 10x, 2Ox, 30x, 40x, 5Ox or more of the half-life are possible. Alternatively, or in addition, increases in the range of up to 3Ox, 4Ox, 5Ox, 6Ox, 7Ox, 80x, 9Ox, 10Ox, 15Ox of the half-life are possible. As referred to herein, the term "competes" means that the binding of a first target to its cognate target binding domain is inhibited when a second target is bound to its cognate target binding domain. For example, binding may be inhibited sterically, for example by physical blocking of a binding domain or by alteration of the structure or environment of a binding domain such that its affinity or avidity for a target is reduced. A protein moiety competes for binding to a target (e.g., EGFR, VEGF, serum albumin) with another agent, when the protein moiety inhibits binding of the other agent to the target in a competitive binding assay (e.g., a competitive ELISA or other suitable binding assay). For example, the protein moiety can inhibit binding of another agent that binds a target (e.g., EGFR, VEGF, serum albumin) in a competitive binding assay by at least about 25%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, or at least about 95%.
As used herein, the terms "low stringency," "medium stringency," "high stringency," or "very high stringency conditions" describe conditions for nucleic acid hybridization and washing. Guidance for performing hybridization reactions can be found in Current Protocols in Molecular Biology, John Wiley & Sons, N.Y. (1989), 6.3.1-6.3.6, which is incorporated herein by reference in its entirety.
Aqueous and nonaqueous methods are described in that reference and either can be used. Specific hybridization conditions referred to herein are as follows: (1) low stringency hybridization conditions in 6X sodium chloride/sodium citrate (SSC) at about 45C, followed by two washes in 0.2X SSC, 0.1% SDS at least at 5OC (the temperature of the washes can be increased to 55C for low stringency conditions); (2) medium stringency hybridization conditions in 6X SSC at about 45C, followed by one or more washes in 0.2X SSC, 0.1 % SDS at 6OC; (3) high stringency hybridization conditions in 6X SSC at about 45C, followed by one or more washes in 0.2X SSC, 0.1% SDS at 65C; and preferably (4) very high stringency
hybridization conditions are 0.5M sodium phosphate, 7% SDS at 65C, followed by one or more washes at 0.2X SSC, 1% SDS at 65C. Very high stringency conditions (4) are the preferred conditions and the ones that should be used unless otherwise specified. Sequences similar or homologous (e.g., at least about 70% sequence identity) to the sequences disclosed herein are also part of the invention. In some
embodiments, the sequence identity at the amino acid level can be about 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or higher. At the nucleic acid level, the sequence identity can be about 70%, 75%, 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or higher. Alternatively, substantial identity exists when the nucleic acid segments will hybridize under selective hybridization conditions (e.g., very high stringency hybridization conditions), to the complement of the strand. The nucleic acids may be present in whole cells, in a cell lysate, or in a partially purified or substantially pure form.
Calculations of "homology" or "sequence identity" or "similarity" between two sequences (the terms are used interchangeably herein) are performed as follows. The sequences are aligned for optimal comparison purposes (e.g., gaps can be introduced in one or both of a first and a second amino acid or nucleic acid sequence for optimal alignment and non-homologous sequences can be disregarded for comparison purposes). In a preferred embodiment, the length of a reference sequence aligned for comparison purposes is at least 30%, preferably at least 40%, more preferably at least 50%, even more preferably at least 60%, and even more preferably at least 70%, 80%, 90%, or 100% of the length of the reference sequence. The amino acid residues or nucleotides at corresponding amino acid positions or nucleotide positions are then compared. When a position in the first sequence is occupied by the same amino acid residue or nucleotide as the corresponding position in the second sequence, then the molecules are identical at that position (as used herein amino acid or nucleic acid " homology" is equivalent to amino acid or nucleic acid "identity"). The percent identity between the two sequences is a function of the number of identical positions shared by the sequences, taking into account the number of gaps, and the length of each gap, which need to be introduced for optimal alignment of the two sequences.
Amino acid and nucleotide sequence alignments and homology, similarity or identity, as defined herein are preferably prepared and determined using the algorithm BLAST 2 Sequences, using default parameters (Tatusova, T. A. et al., FEMS Microbiol Lett, 174:187-188 (1999)). Alternatively, the BLAST algorithm (version 2.0) is employed for sequence alignment, with parameters set to default values. BLAST (Basic Local Alignment Search Tool) is the heuristic search algorithm employed by the programs blastp, blastn, blastx, tblastn, and tblastx; these programs ascribe significance to their findings using the statistical methods of Karlin and Altschul, 1990, Proc. Natl. Acad. Sd. USA 87(6):2264-8.
Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art {e.g., in cell culture, molecular genetics, nucleic acid chemistry, hybridization techniques and biochemistry). Standard techniques are used for molecular, genetic and biochemical methods (see generally, Sambrook et al, Molecular Cloning: A
Laboratory Manual, 2d ed. (1989) Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N. Y. and Ausubel et al, Short Protocols in Molecular Biology (1999) 4th Ed, John Wiley & Sons, Inc. which are incorporated herein by reference) and chemical methods.
The invention relates to ligands that have binding specificity for VEGF {e.g. , human VEGF), ligands that have binding specificity for EGFR {e.g., human EGFR), and to ligands that have binding specificity for VEGF and EGFR {e.g., human VEGF and human EGFR). For example, the ligand can comprise a polypeptide domain having a binding site with binding specificity for VEGF, a polypeptide domain having a binding site with binding specificity for EGFR, or comprise a polypeptide domain having a binding site with binding specificity for VEGF and a polypeptide domain having a binding site with binding specificity for EGFR.
The ligands of the invention provide several advantages. For example, as described herein, the ligand can be tailored to have a desired in vivo serum half-life. Thus, the ligands can be used to control, reduce, or eliminate general toxicity of therapeutic agents, such as cytotoxin used to treat cancer. Further, dAbs are much smaller than conventional antibodies, and can be administered to achieve better tissue penetration than conventional antibodies. Thus, dAbs and ligands that comprise a dAb provide advantages over conventional antibodies when administered to treat cancer, for example by targeting solid tumors. Moreover, many cancers overexpress EGFR, and ligands that have binding specificity for EGFR and VEGF can be administered to target VEGF-inhibitory activity to tumors or the environment of cancer cells. This approach provides two beneficial activities directly at the site of a tumor or cancer, i.e., direct anti-cancer activity by binding to EGFR and inhibiting binding of ligands {e.g., EGF, TGF alpha) to the receptor, and inhibition of angiogenesis that supports tumor formation and development. Accordingly, ligands that have binding specificity for VEGF and EGFR can be administered to a patient with cancer {e.g., EGFR-expressing cancer) to provide superior therapy using a single therapeutic agent.
Further, signals transduced through EGFR can lead to the production of angiogenic factors, such as VEGF. Cancer cells {e.g., in a tumor) that express or overexpress EGFR can produce a high level of VEGF that acts locally to induce formation of tumor vasculature. Accordingly, the ligands of the invention that have binding specificity for VEGF and EGFR can be administered to a subject to target delivery of the VEGF inhibitory activity of the ligand to cells that overexpress EGFR. Accordingly, anti-angiogenic therapy can be delivered specifically to sites where VEGF is being produced {e.g., to cells that overexpress EGFR).
In some embodiments, the ligand has binding specificity for VEGF and comprises an (at least one) immunoglobulin single variable domain with binding specificity for VEGF. In other embodiments, the ligand has binding specificity for EGFR and comprises an (at least one) immunoglobulin single variable domain with binding specificity for EGFR. In certain embodiments, the ligand has binding specificity for VEGF and EGFR, and comprises an (at least one) immunoglobulin single variable domain with binding specificity for VEGF and an (at least one) immunoglobulin single variable domain with binding specificity for EGFR.
The ligand of the invention can be formatted as described herein For example, the ligand of the invention can be formatted to tailor in vivo serum half- life. If desired, the ligand can further comprise a toxin or a toxin moiety as described herein. In some embodiments, the ligand comprises a surface active toxin, such as a free radical generator (e.g., selenium containing toxin) or a radionuclide. In other embodiments, the toxin or toxin moiety is a polypeptide domain (e.g., a dAb) having a binding site with binding specificity for an intracellular target. In particular embodiments, the ligand is an IgG-like format that has binding specificity for VEGF and EGFR (e.g., human VEGF and human EGFR). Ligand Formats
The ligand of the invention can be formatted as a monospecific, dual specific or multispecific ligand as described herein. See, also WO 03/002609, the entire teachings of which are incorporated herein by reference, regarding ligand formatting. Such dual specific ligands comprise immunoglobulin single variable domains that have different binding specificities. Such dual specific ligands can comprise combinations of heavy and light chain domains. For example, the dual specific ligand may comprise a VH domain and a VL domain, which may be linked together in the form of an scFv (e.g., using a suitable linker such as GIy4S er), or formatted into a bispecifϊc antibody or antigen-binding fragment thereof (e.g. F(ab')2 fragment). The dual specific ligands do not comprise complementary VJ/VL pairs which form a conventional two chain antibody antigen-binding site that binds antigen or epitope co-operatively. Instead, the dual format ligands comprise a VH/VL complementary pair, wherein the V domains have different binding specificities.
In addition, the dual specific ligands may comprise one or more CH or CL domains if desired. A hinge region may also be included if desired. Such combinations of domains may, for example, mimic natural antibodies, such as IgG or IgM, or fragments thereof, such as Fv, scFv, Fab or F(ab')2 molecules. Other structures, such as a single arm of an IgG molecule comprising VH, VL, CHI and CL domains, are envisaged. Preferably, the dual specific ligand of the invention comprises only two variable domains although several such ligands may be incorporated together into the same protein, for example two such ligands can be incorporated into an IgG or a multimeric immunoglobulin, such as IgM.
Alternatively, in another embodiment a plurality of dual specific ligands are combined to form a multimer. For example, two different dual specific ligands are combined to create a tetra-specific molecule. It will be appreciated by one skilled in the art that the light and heavy variable regions of a dual-specific ligand produced according to the method of the present invention may be on the same polypeptide chain, or alternatively, on different polypeptide chains. Pn the case that the variable regions are on different polypeptide chains, then they may be linked via a linker, generally a flexible linker (such as a polypeptide chain), a chemical linking group, or any other method known in the art.
In some embodiments, the linker may be a "natural linker" that contains carboxy-terminal amino acids of an antibody variable domain and amino-terminal amino acids of an antibody constant domain. For example, a natural linker can contain the carboxy-terminal amino acids of Vk and amino-terminal amino acids of Ck (e.g. KVEIKRTVAAPS (SEQ ID NO:706)). In other embodiments, the linker can contain fewer Lys and Arg residues than the natural linker (e.g., LVTVSSAST (SEQ ID NO:707) or (LVTVS S GGGGS GGGS (SEQ ID NO:708)). If desired, the linker can be mutated to substitute some or all of the positively charged residues
(e.g., in a natural linker), such as Lys and/or Arg with residues that are not positively charged at physiological pH. For example, Lys and/or Arg residues can be replaced with Asn, Leu, GIn or Ser. This type of linker provides the advantage of reducing protease sensitivity (e.g., serine protease, cysteine protease, matrix metalloprotease, pepsin, trypsin, elastase, chymotrypsin, carboxypeptidase, cathepsin (e.g., cathepsin G), proteinase 3). Examples of such linkers include, GQGTNVEINRTVAAPS (SEQ ID NO:710), GQGTNVEINQTVAAPS (SEQ ID NO:711),
GQGTNVEIQRTVAAPS (SEQ ID NO:712), or GQGTLVTVSSTVAAPS (SEQ ID NO:713).
Proteases (e.g., a serine protease, cysteine protease, matrix metalloprotease, pepsin, trypsin, elastase, chymotrypsin, carboxypeptidase, cathepsin (e.g., cathepsin G), proteinase 3) function in the normal turn over and metabolism of proteins.
However, in certain physiological states, such as inflammatory states (e.g., COPD) and cancer, the amount of proteases present in a tissue, organ or animal (e.g., in the lung, in or adjacent to a tumor) can increase. This increase in proteases can result in accelerated degradation and inactivation of endogenous proteins and of therapeutic or diagnostic peptides, polypeptides and proteins that are administered. In fact, some agents that have potential for in vivo use (e.g., use in treating, diagnosing or preventing disease) have only limited efficacy because they are rapidly degraded and inactivated by proteases .
The invention relates to ligand comprising a linker that is resistant to protease degradation. The protease resistant ligands of the invention provide several advantages. For example, a protease resistant ligand can be administered to a subject and remain active in vivo longer than protease sensitive agents.
Accordingly, protease resistant ligand will remain functional for a period of time that is sufficient to produce biological effects.
A ligand or linker that is resistant to protease degradation is not substantially degraded by a protease when incubated with the protease under conditions suitable for protease activity for at least about 2 hours, at least about 3 hours, at least about 4 hours, at least about 5 hours, at least about 6 hours, at least about 7 hours, at least about 8 hours, at least about 9 hours, at least about 10 hours, at least about 11 hours, at least about 12 hours, at least about 24 hours, at least about 36 hours, or at least about 48 hours. A ligand or linker is not substantially degraded when no more than about 25%, no more than about 20%, no more than about 15%, no more than about 14%, no more than about 13%, no more than about 12%, no more than about 11%, no more than about 10%, no more than about 9%, no more than about 8%, no more than about 7% no more than about 6%, no more than about 5%, no more than about 4%, no more than about 3%, no more than about 2%, no more than about 1%, or substantially none of the ligand or linker is degraded by protease after incubation with the protease for at least about 2 hours. Protein degradation can be assessed using any suitable method, for example, by SDS-PAGE.
Protease resistance can be assessed using any suitable method. For example, a protease can be added to a solution of ligand or linker in a suitable buffer {e.g., PBS) to produce a ligand or linker/protease solution, such as a solution of at least about 0.01% (w/w) protease, about 0.01% to about 5% (w/w) protease, about 0.05% to about 5% (w/w) protease, about 0.1% to about 5% (w/w) protease, about 0.5% to about 5% (w/w) protease, about 1% to about 5% (w/w) protease, at least about 0.01% (w/w) protease, at least about 0.02% (w/w) protease, at least about 0.03% (w/w) protease, at least about 0.04% (w/w) protease, at least about 0.05% (w/w) protease, at least about 0.06% (w/w) protease, at least about 0.07% (w/w) protease, at least about 0.08% (w/w) protease, at least about 0.09% (w/w) protease, at least about 0.1% (w/w) protease, at least about 0.2% (w/w) protease, at least about 0.3% (w/w) protease, at least about 0.4% (w/w) protease, at least about 0.5% (w/w) protease, at least about 0.6% (w/w) protease, at least about 0.7% (w/w) protease, at least about 0.8% (w/w) protease, at least about 0.9% (w/w) protease, at least about 1% (w/w) protease, at least about 2% (w/w) protease, at least about 3% (w/w) protease, at least about 4% (w/w) protease, or about 5% (w/w) protease. The ligand or linker/protease mixture can be incubated at a suitable temperature for protease activity (e.g., at 370C) and samples can be taken at time intervals (e.g., at 1 hour, 2 hours, 3 hours, etc.) and the protease reaction stopped. The samples can then be analyzed for protein degradation using any suitable method, such as SDS-PAGE analysis. The results can be used to establish a time course of degradation.
Ligands can be formatted as bi- or multispecific antibodies or antibody fragments or into bi- or multispecific non-antibody structures. Suitable formats include, any suitable polypeptide structure in which an antibody variable domain or one or more of the CDRs thereof can be incorporated so as to confer binding specificity for antigen on the structure. A variety of suitable antibody formats are known in the art, such as, bispecific IgG-like formats (e.g., chimeric antibodies, humanized antibodies, human antibodies, single chain antibodies, heterodimers of antibody heavy chains and/or light chains, antigen-binding fragments of any of the foregoing (e.g., a Fv fragment (e.g., single chain Fv (scFv), a disulfide bonded Fv), a Fab fragment, a Fab' fragment, a F(ab')2 fragment), a single variable domain (e.g., VH, VL, VHH), a dAb), and modified versions of any of the foregoing (e.g., modified by the covalent attachment of polyalkylene glycol (e.g., polyethylene glycol, polypropylene glycol, polybutylene glycol) or other suitable polymer). See, PCT/GB03/002804, filed June 30, 2003, which designated the United States, (WO 2004/081026) regarding PEGylated single variable domains and dAbs, suitable methods for preparing same, increased in vivo half-life of the PEGylated single variable domains and dAb monomers and multimers, suitable PEGs, preferred hydrodynamic sizes of PEGs, and preferred hydrodynamic sizes of PEGylated single variable domains and dAb monomers and multimers. The entire teaching of PCT/GB03/002804 (WO 2004/081026), including the portions referred to above, are incorporated herein by reference.
The ligand can be formatted using a suitable linker such as (Gly4Ser)n, where n = from 1 to 8, e.g., 2, 3, 4, 5, 6 or 7. If desired, ligands, including dAb monomers, dimers and trimers, can be linked to an antibody Fc region, comprising one or both of CH2 and CH3 domains, and optionally a hinge region. For example, vectors encoding ligands linked as a single nucleotide sequence to an Fc region may be used to prepare such polypeptides. In some embodiments, the ligand comprises one, two or more dAbs of the same or different binding specificities, and CH2, CH3, CH2-CH3, hinge-CjH[2, hinge-CH3, hinge-Cπ2-CH3, a portion of hinge-CH2, a portion of hinge- CH3, or a portion of hinge-CH2-CH3. In such embodiments, CH2, CH3, CH2-CH3, hinge-Cκ2, hinge-Cπ3, hinge-CH2-Cπ3, the portion of hinge-Cπ2, the portion of hinge-CH3, and the portion of hinge-CH2-CH3 can be from any desired antibody, such as a human IgG, such as a human IgGl or a human IgG4.
In some embodiments, the ligand of the invention comprises an anti-EGFR dAb or an anti-VEGF dAb that is fused (e.g., directly or through a linker) to an Fc region of an antibody. In some embodiments, the ligand is an Fc fusion that comprises an anti-VEGF dAb that is disulfide bonded to an anti-EGFR dAb. In particular examples, the ligand comprises two or more dAbs (e.g., two dAbs that bind EGFR, two dAbs that bind VEGF, a dAb that binds EGFR and a dAb that binds VEGF) and an Fc region, and the ligand has the structure, from amino terminus to carboxy terminus, VH-VH-FC, VL-VL-FC, VH-VL-FC, VL-VH-FC. For example, the ligand can have the structure VH-VK-hinge-CH2-CH3, VK-VH-hinge-CH2-CH3, Vκ- VK-hinge-CH2-CH3, or VH-VH-hinge-CH2-CH3. If desired, the VH shown in any of the foregoing formulae can be a VHH- TWO ligands that contain Fc regions can be bonded together to form a dimer, for example, through a disulfide bond (e.g., in the hinge region).
Generally the orientation of the polypeptide domains that have a binding site with binding specificity for a target (e.g., dAbs), and whether the ligand comprises a linker, is a matter of design choice. However, some orientations, with or without linkers, may provide preferred binding characteristics in comparison to other orientations. AU orientations (e.g., dAbl -dAb2-Fc; dAb2 -dAb 1-Fc) are encompassed by the invention, and ligands that contain an orientation that provides desired binding characteristics can be easily identified by routine screening.
Ligands and dAb monomers can also be combined and/or formatted into non-antibody multi-ligand structures to form multivalent complexes, which bind target molecules with the same antigen, thereby providing superior avidity. For example natural bacterial receptors such as SpA can be used as scaffolds for the grafting of CDRs to generate ligands which bind specifically to one or more epitopes. Details of this procedure are described in US 5,831,012. Other suitable scaffolds include those based on fibronectin and affibodies. Details of suitable procedures are described in WO 98/58965. Other suitable scaffolds include lipocallin and CTLA4, as described in van den Beuken et at., J. MoI. Biol. 310:591- 601 (2001), and scaffolds such as those described in WO 00/69907 (Medical Research Council), which are based for example on the ring structure of bacterial GroEL or other chaperone polypeptides. Protein scaffolds may be combined; for example, CDRs may be grafted on to a CTLA4 scaffold and used together with immunoglobulin VH or VL domains to form a ligand. Likewise, fibronectin, lipocallin and other scaffolds may be combined
A variety of suitable methods for preparing any desired format are known in the art. For example, antibody chains and formats {e.g., bispecific IgG-like formats, chimeric antibodies, humanized antibodies, human antibodies, single chain antibodies, homodimers and heterodimers of antibody heavy chains and/or light chains) can be prepared by expression of suitable expression constructs and/or culture of suitable cells {e.g., hybridomas, heterohybridomas, recombinant host cells containing recombinant constructs encoding the format). Further, formats such as antigen-binding fragments of antibodies or antibody chains {e.g., bispecific binding fragments, such as a Fv fragment {e.g., single chain Fv (scFv), a disulfide bonded Fv), a Fab fragment, a Fab' fragment, a F(ab')2 fragment), can be prepared by expression of suitable expression constructs or by enzymatic digestion of antibodies, for example using papain or pepsin.
The ligand can be formatted as a multispecific ligand, for example as described in WO 03/002609, the entire teachings of which are incorporated herein by reference. Such multispecific ligands possess more than one epitope binding specificity. Generally, the multi-specific ligand comprises two or more epitope binding domains, such as dAbs or non-antibody protein domain comprising a binding site for an epitope, e.g., an affibody, an SpA domain, an LDL receptor class A domain, an EGF domain, an avimer. Multispecific ligands can be formatted further as described herein. In some embodiments, the ligand is an IgG-like format. Such formats have the conventional four chain structure of an IgG molecule (2 heavy chains and two light chains), in which one or more of the variable regions (VH and or VL) have been replaced with a dAb or single variable domain of a desired specificity. Preferably, each of the variable regions (2 VH regions and 2 VL regions) is replaced with a dAb or single variable domain. The dAb(s) or single variable domain(s) that are included in an IgG-like format can have the same specificity or different specificities. In some embodiments, the IgG-like format is tetravalent and can have one, two, three or four specificities. For example, the IgG-like format can be monospecific and comprises 4 dAbs that have the same specificity; bispecific and comprises 3 dAbs that have the same specificity and another dAb that has a different specificity;
bispecific and comprise two dAbs that have the same specificity and two dAbs that have a common but different specificity; trispecific and comprises first and second dAbs that have the same specificity, a third dAb with a different specificity and a fourth dAb with a different specificity from the first, second and third dAbs; or tetraspecific and comprise four dAbs that each have a different specificity. Antigen- binding fragments of IgG-like formats (e.g., Fab, F(ab')2, Fab', Fv, scFy) can be prepared. In addition, a particular constant region of Fc portion (e.g., of an IgG, such as IgGl), variant or portion thereof can be selected in order to tailor effector function. For example, if complement activation and/or antibody dependent cellular cytotoxicity (ADCC) function is desired, the ligand can be an IgGl -like format. If desired, the IgG-like format can comprise a mutated constant region (variant IgG heavy chain constant region) to minimize binding to Fc receptors and/or ability to fix complement, (see e.g. Winter et al, GB 2,209,757 B; Morrison et al, WO 89/07142;
Morgan et al, WO 94/29351 , December 22, 1994).
In some embodiments, the IgG-like formats can comprise an anti-EGFR dAb (e.g., DOM16-39-542, DOM16-39-618 or DOM16-39-619), an anti-VEGF dAb (e.g., DOMl 5-26-501), or an anti-EGFR dAb and an anti-VEGF dAb.
The ligands of the invention can be formatted as a fusion protein that contains a first immunoglobulin single variable domain that is fused directly to a second immunoglobulin single variable domain. If desired such a format can further comprise a half-life extending moiety. For example, the ligand can comprise a first immunoglobulin single variable domain that is fused directly to a second
immunoglobulin single variable domain that is fused directly to an immunoglobulin single variable domain that binds serum albumin. For example, the ligand can be an in line fusion of two or more protein moieties that have a binding site with binding specificity for EGFR that competes for binding to EGFR with an anti-EGFR domain antibody (e.g., any of the DOMl 6 dAbs disclosed herein) and fused to an anti-serum albumin dAb (e.g., any of the D0M7 dAbs disclosed herein). In some
embodiments, the protein moieties that have a binding site with binding specificity for EGFR (e.g., anti-EGFR dAbs) have different epitopic specificities. In other examples, the ligand is an in line fusion protein comprising a protein moiety that has a binding site with binding specificity for EGFR (e.g., anti-EGFR dAb), a protein moiety that has a binding site with binding specificity for VEGF, and an anti-serum albumin dAb.
In particular embodiments, such an in line fusion comprises DOMl 6-39-618 dAb and/or DOM16-39-619 and an anti-serum albumin dAb (e.g., DOM16-39- 618— DOM7h-14, DOM7h-14— DOMl 6-39-618, DOM16-39-619— DOM7h-14, DOM7h-14— DOM16-39-619). In other embodiments, the in line fusion comprises a protein moiety (e.g., dAb) that has at least about 80%, 85%, 87%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% amino acid sequence identiy with the amino acid sequence of DOMl 6-39-618, a protein moiety that has at least about 80%, 85%, 87%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% amino acid sequence identity with the amino acid sequence of DOM 16-39-619, and/or a protein moiety (e.g., dAb) that has at least about 80%, 85%, 87%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% amino acid sequence identiy with the amino acid sequence of an anti-serum albumin dAb disclosed herein, such as DOM7h-14.
In other particular embodiments, the ligand comprises an anti-VEGF dAb, an anti-EGFR dAb, and an anti-serum albumin dAb (e.g., DOMl 5-10— DOMl 6-39— anti-serum albumin dAb, DOMl 6-39— DOMl 5-10— anti-serum albumin dAb, DOM15-26-501— DOM16-39— anti-serum albumin dAb, DOM16-39— D0M15- 26-501— anti-serum albumin dAb). In other embodiments, the in line fusion comprises a protein moiety (e.g., dAb) that has at least about 80%, 85%, 87%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% amino acid sequence identiy with the amino acid sequence of an anti-VEGF dAb disclosed herein, such as DOMl 5-10 or DOMl 5-25-501, and/or a protein moiety that has at least about 80%, 85%, 87%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% amino acid sequence identity with the amino acid sequence of an anti-EGFR dAb disclosed herein, such as DOM16-39, and/or a protein moiety (e.g., dAb) that has at least about 80%, 85%, 87%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% amino acid sequence identiy with the amino acid sequence of an anti-serum albumin dAb disclosed herein, such as DOM7h-14.
Generally the orientation of the polypeptide domains that have a binding site with binding specificity for a target, and whether the ligand comprises a linker, is a matter of design choice. However, some orientations, with or without linkers, may provide better binding characteristics than other orientations. AU orientations (e.g., dAbl-linker-dAb2; dAb2-linker-dAbl) are encompassed by the invention are ligands that contain an orientation that provides desired binding characteristics can be easily identified by screening.
Half-life Extended Formats
The ligands and dAb monomers disclosed herein can be formatted to extend their in vivo serum half-life. Increased in vivo half-life is useful in in vivo applications of immunoglobulins, especially antibodies and most especially antibody fragments of small size such as dAbs. Such fragments (Fvs, disulphide bonded Fvs, Fabs, scFvs, dAbs) are rapidly cleared from the body, which can limit clinical applications.
A ligand can be formatted as a larger antigen-binding fragment of an antibody or as an antibody (e.g., formatted as a Fab, Fab', F(ab)2, F(ab')2, IgG, scFv) that has larger hydrodynamic size. Ligands can also be formatted to have a larger hydrodynamic size, for example, by attachment of a polyalkyleneglycol group (e.g. polyethyleneglycol (PEG) group, polypropylene glycol, polybutylene glycol), serum albumin, transferrin, transferrin receptor or at least the transferrin-binding portion thereof, an antibody Fc region, or by conjugation to an antibody domain. In some embodiments, the ligand (e.g., dAb monomer) is PEGylated. Preferably the PEGylated ligand (e.g., dAb monomer) binds VEGF and/or EGFR with substantially the same affinity or avidity as the same ligand that is not PEGylated. For example, the ligand can be a PEGylated ligand comprising a dAb that binds VEGF or EGFR with an affinity or avidity that differs from the avidity of ligand in unPEGylated form by no more than a factor of about 1000, preferably no more than a factor of about 100, more preferably no more than a factor of about 10, or with affinity or avidity substantially unchanged relative to the unPEGylated form. See,
PCT/GB03/002804, filed June 30, 2003, which designated the United States, (WO 2004/081026) regarding PEGylated single variable domains and dAbs, suitable methods for preparing same, increased in vivo half-life of the PEGylated single variable domains and dAb monomers and multimers, suitable PEGs, preferred hydrodynamic sizes of PEGs, and preferred hydrodynamic sizes of PEGylated single variable domains and dAb monomers and multimers. The entire teaching of PCT/GB03/002804 (WO 2004/081026), including the portions referred to above, are incorporated herein by reference.
Hydrodynamic size of the ligands (e.g., dAb monomers and multimers) of the invention may be determined using methods which are well known in the art. For example, gel filtration chromatography may be used to determine the hydrodynamic size of a ligand. Suitable gel filtration matrices for determining the hydrodynamic sizes of ligands, such as cross-linked agarose matrices, are well known and readily available.
The size of a ligand format (e.g., the size of a PEG moiety attached to a dAb monomer), can be varied depending on the desired application. For example, where ligand is intended to leave the circulation and enter into peripheral tissues, it is desirable to keep the hydrodynamic size of the ligand low to facilitate extravazation from the blood stream. Alternatively, where it is desired to have the ligand remain in the systemic circulation for a longer period of time the size of the ligand can be increased, for example by formatting as an Ig like protein or by addition of a 30 to 60 kDa PEG moiety (e.g., linear or branched PEG 30 to 40 IcDa PEG, such as addition of two 20IcDa PEG moieties.) The size of the ligand format can be tailored to achieve a desired in vivo serum half-life, for example to control exposure to a toxin and/or to reduce side effects of toxic agents. The hydrodynamic size of ligand (e.g., dAb monomer) and its serum half-life can also be increased by conjugating or linking the ligand to a binding domain (e.g., antibody or antibody fragment) that binds an antigen or epitope that increases half- life in vivo, as described herein. For example, the ligand (e.g., dAb monomer) can be conjugated or linked to an anti-serum albumin or anti-neonatal Fc receptor antibody or antibody fragment, e.g., an anti-SA or anti-neonatal Fc receptor dAb, Fab, Fab' or scFv, or to an anti-SA affibody or anti-neonatal Fc receptor affibody.
Examples of suitable albumin, albumin fragments or albumin variants for use in a ligand according to the invention are described in WO 2005/077042A2, which is incorporated herein by reference in its entirety. In particular, the following albumin, albumin fragments or albumin variants can be used in the present invention:
• SEQ ID NO: 1 as disclosed in WO 2005/077042A2, (this sequence being explicitly incorporated into the present disclosure by reference);
• Albumin fragment or variant comprising or consisting of amino acids 1-387 of SEQ ID NO:1 in WO 2005/077042A2;
• Albumin, or fragment or variant thereof, comprising an amino acid sequence selected from the group consisting of: (a) amino acids 54 to 61 of SEQ ID NO:1 in WO 2005/077042 A2; (b) amino acids 76 to 89 of SEQ ID NO:1 in WO 2005/077042A2; (c) amino acids 92 to 100 of SEQ ID NO: 1 in WO
2005/077042A2; (d) amino acids 170 to 176 of SEQ ID NO:1 in WO 2005/077042A2; (e) amino acids 247 to 252 of SEQ ID NO:1 in WO 2005/077042A2; (f) amino acids 266 to 277 of SEQ ID NO:1 in WO
2005/077042A2; (g) amino acids 280 to 288 of SEQ ID NO:1 in WO 2005/077042A2; (h) amino acids 362 to 368 of SEQ ID NO:1 in WO
2005/077042A2; (i) amino acids 439 to 447 of SEQ ID NO:1 in WO
2005/077042A2 (j) amino acids 462 to 475 of SEQ ID NO:1 in WO
2005/077042A2; (k) amino acids 478 to 486 of SEQ ID NO:1 in WO 2005/077042A2; and (1) amino acids 560 to 566 of SEQ ID NO:1 in WO 2005/077042A2.
Further examples of suitable albumin, fragments and analogs for use in a ligand according to the invention are described in WO 03/076567A2, which is incorporated herein by reference in its entirety. In particular, the following albumin, fragments or variants can be used in the present invention:
• Human serum albumin as described in WO 03/076567 A2, e.g., in figure
3 (this sequence information being explicitly incorporated into the present disclosure by reference);
• Human serum albumin (HA) consisting of a single non-glycosylated
polypeptide chain of 585 amino acids with a formula molecular weight of 66,500 (See, Meloun, et al, FEBS Letters 55:136 (1975); Behrens, et al, Fed. Proc. 34:591 (1975); Lawn, et al., Nucleic Acids Research P:6102-6114 (1981); Minghetti, et al., J. Biol. Chem. 261:6747 (1986));
• A polymorphic variant or analog or fragment of albumin as described in Weitkamp, et al, Ann. Hum. Genet. 37:219 (1973);
• An albumin fragment or variant as described in EP 322094, e.g., HA(l-373., HA(l-388), HA(l-389), HA(l-369), and HA(1-419) and fragments between 1-369 and 1-419;
• An albumin fragment or variant as described in EP 399666, e.g., HA(1-177) and HA(I -200) and fragments between HA(I-X), where X is any number from 178 to 199.
Where a (one or more) half-life extending moiety {e.g., albumin, transferrin and fragments and analogues thereof) is used in the ligands of the invention, it can be conjugated to the ligand using any suitable method, such as, by direct fusion to the target-binding moiety (e.g., dAb or antibody fragment), for example by using a single nucleotide construct that encodes a fusion protein, wherein the fusion protein is encoded as a single polypeptide chain with the half-life extending moiety located N- or C-terminally to the cell surface target binding moieties. Alternatively, conjugation can be achieved by using a peptide linker between moieties, e.g., a peptide linker as described in WO 03/076567A2 or WO 2004/003019 (these linker disclosures being incorporated by reference in the present disclosure to provide examples for use in the present invention). Typically, a polypeptide that enhances serum half-life in vivo is a polypeptide which occurs naturally in vivo and which resists degradation or removal by endogenous mechanisms which remove unwanted material from the organism {e.g., human). For example, a polypeptide that enhances seram half-life in vivo can be selected from proteins from the extracellular matrix, proteins found in blood, proteins found at the blood brain barrier or in neural tissue, proteins localized to the kidney, liver, lung, heart, skin or bone, stress proteins, disease-specific proteins, or proteins involved in Fc transport.
Suitable polypeptides that enhance serum half-life in vivo include, for example, transferrin receptor specific ligand-neuropharmaceutical agent fusion proteins (see U.S. Patent No. 5,977,307, the teachings of which are incorporated herein by reference), brain capillary endothelial cell receptor, transferrin, transferrin receptor {e.g., soluble transferrin receptor), insulin, insulin-like growth factor 1 (IGF 1) receptor, insulin-like growth factor 2 (IGF 2) receptor, insulin receptor, blood coagulation factor X, αl-antitrypsin and HNF lα. Suitable polypeptides that enhance serum half-life also include alpha- 1 glycoprotein (orosomucoid; AAG), alpha-1 antichymotrypsin (ACT), alpha-1 microglobulin (protein HC; AIM), antithrombin III (AT III), apolipoprotein A-I (Apo A-I), apolipoprotein B (Apo B), ceruloplasmin (Cp), complement component C3 (C3), complement component C4 (C4), Cl esterase inhibitor (Cl INH), C-reactive protein (CRP), ferritin (FER), hemopexin (HPX), lipoprotein(a) (Lp(a)), mannose-binding protein (MBP), myoglobin (Myo), prealbumin (transthyretin; PAL), retinol-binding protein (RBP), and rheumatoid factor (RF).
Suitable proteins from the extracellular matrix include, for example, collagens, laminins, integrins and fibronectin. Collagens are the major proteins of the extracellular matrix. About 15 types of collagen molecules are currently known, found in different parts of the body, e.g. type I collagen (accounting for 90% of body collagen) found in bone, skin, tendon, ligaments, cornea, internal organs or type II collagen found in cartilage, vertebral disc, notochord, and vitreous humor of the eye.
Suitable proteins from the blood include, for example, plasma proteins {e.g., fibrin, α-2 macroglobulin, serum albumin, fibrinogen {e.g., fibrinogen A, fibrinogen B), serum amyloid protein A, haptoglobin, profilin, ubiquitin, uteroglobulin and β-2- microglobulin), enzymes and enzyme inhibitors {e.g., plasminogen, lysozyme, cystatin C, alpha-1 -antitrypsin and pancreatic trypsin inhibitor), proteins of the immune system, such as immunoglobulin proteins {e.g., IgA, IgD, IgE, IgG, IgM, immunoglobulin light chains (kappa/lambda)), transport proteins {e.g., retinol binding protein, α-1 microglobulin), defensins {e.g., beta-defensin 1, neutrophil defensin 1 , neutrophil defensin 2 and neutrophil defensin 3) and the like.
Suitable proteins found at the blood brain barrier or in neural tissue include, for example, melanocortin receptor, myelin, ascorbate transporter and the like.
Suitable polypeptides that enhance serum half-life in vivo also include proteins localized to the kidney {e.g., polycystin, type IV collagen, organic anion transporter Kl, Heymann's antigen), proteins localized to the liver {e.g., alcohol dehydrogenase, G250), proteins localized to the lung {e.g., secretory component, which binds IgA), proteins localized to the heart {e.g., HSP 27, which is associated with dilated cardiomyopathy), proteins localized to the skin {e.g., keratin), bone specific proteins such as morphogenic proteins (BMPs), which are a subset of the transforming growth factor β superfamily of proteins that demonstrate osteogenic activity {e.g., BMP-2, BMP-4, BMP-5, BMP-6, BMP-7, BMP-8), tumor specific proteins {e.g., trophoblast antigen, herceptin receptor, oestrogen receptor, cathepsins {e.g., cathepsin B, which can be found in liver and spleen)).
Suitable disease-specific proteins include, for example, antigens expressed only on activated T-cells, including LAG-3 (lymphocyte activation gene), osteoprotegerin ligand (OPGL; see Nature 402, 304-309 (1999)), OX40 (a member of the TNF receptor family, expressed on activated T cells and specifically up- regulated in human T cell leukemia virus type-I (HTLV-I)-producing cells; see Immunol. 165 (l):263-70 (2000)). Suitable disease-specific proteins also include, for example, metalloproteases (associated with arthritis/cancers) including CG6512 Drosophila, human paraplegin, human FtsH, human AFG3L2, murine ftsH; and angiogenic growth factors, including acidic fibroblast growth factor (FGF-I), basic fibroblast growth factor (FGF-2), vascular endothelial growth factor/vascular permeability factor (VEGF/VPF), transforming growth factor-α (TGF α), tumor necrosis factor-alpha (TNF-α), angiogenin, interleukin-3 (IL-3), interleukin-8 (IL- 8), platelet-derived endothelial growth factor (PD-ECGF), placental growth factor (PlGF), midkine platelet-derived growth factor-BB (PDGF), and fractalkine.
Suitable polypeptides that enhance serum half-life in vivo also include stress proteins such as heat shock proteins (HSPs). HSPs are normally found
intracellularly. When they are found extracellularly, it is an indicator that a cell has died and spilled out its contents. This unprogrammed cell death (necrosis) occurs when as a result of trauma, disease or injury, extracellular HSPs trigger a response from the immune system. Binding to extracellular HSP can result in localizing the compositions of the invention to a disease site.
Suitable proteins involved in Fc transport include, for example, Brambell receptor (also known as FcRB). This Fc receptor has two functions, both of which are potentially useful for delivery. The functions are (1) transport of IgG from mother to child across the placenta (2) protection of IgG from degradation thereby prolonging its serum half-life. It is thought that the receptor recycles IgG from endosomes. (See, Holliger et al, Nat Biotechnol 15(7):632-6 (1997).)
Methods for pharmacokinetic analysis and determination of ligand half-life will be familiar to those skilled in the art. Details may be found in Kenneth, A et al: Chemical Stability of Pharmaceuticals: A Handbook for Pharmacists and in Peters et al, Pharmacokinetc analysis: A Practical Approach (1996). Reference is also made to "Pharmacokinetics", M Gibaldi & D Perron, published by Marcel Dekker, 2nd
Rev. ex edition (1982), which describes pharmacokinetic parameters such as t alpha and t beta half lives and area under the curve (AUC).
Ligands that Contain a Toxin Moiety or Toxin
The invention also relates to ligands that comprise a toxin moiety or toxin.
Suitable toxin moieties comprise a toxin {e.g., surface active toxin, cytotoxin). The toxin moiety or toxin can be linked or conjugated to the ligand using any suitable method. For example, the toxin moiety or toxin can be covalently bonded to the ligand directly or through a suitable linker. Suitable linkers can include
noncleavable or cleavable linkers, for example, pH cleavable linkers that comprise a cleavage site for a cellular enzyme {e.g., cellular esterases, cellular proteases such as cathepsin B). Such cleavable linkers can be used to prepare a ligand that can release a toxin moiety or toxin after the ligand is internalized.
A variety of methods for linking or conjugating a toxin moiety or toxin to a ligand can be used. The particular method selected will depend on the toxin moiety or toxin and ligand to be linked or conjugated. If desired, linkers that contain terminal functional groups can be used to link the ligand and toxin moiety or toxin. Generally, conjugation is accomplished by reacting toxin moiety or toxin that contains a reactive functional group (or is modified to contain a reactive functional group) with a linker or directly with a ligand. Covalent bonds formed by reacting an toxin moiety or toxin that contains (or is modified to contain) a chemical moiety or functional group that can, under appropriate conditions, react with a second chemical group thereby forming a covalent bond. If desired, a suitable reactive chemical group can be added to ligand or to a linker using any suitable method. (See, e.g., Hermanson, G. T., Bioconjugate Techniques, Academic Press: San Diego, CA (1996).) Many suitable reactive chemical group combinations are known in the art, for example an amine group can react with an electrophilic group such as tosylate, mesylate, halo (chloro, bromo, fluoro, iodo), N-hydroxysuccinimidyl ester (NHS), and the like. Thiols can react with maleimide, iodoacetyl, acrylolyl, pyridyl disulfides, 5-thiol-2-nitrobenzoic acid thiol (TNB-thiol), and the like. An aldehyde functional group can be coupled to amine- or hydrazide-containing molecules, and an azide group can react with a trivalent phosphorous group to form
phosphoramidate or phosphorimide linkages. Suitable methods to introduce activating groups into molecules are known in the art (see for example, Hermanson, G. T., Bioconjugate Techniques, Academic Press: San Diego, CA (1996)).
Suitable toxin moieties and toxins include, for example, a maytansinoid {e.g., maytansinol, e.g., DMl, DM4), a taxane, a calicheamicin, a duocarmycin, or derivatives thereof. The maytansinoid can be, for example, maytansinol or a maytansinol analogue. Examples of maytansinol analogues include those having a modified aromatic ring {e.g., C-19-decloro, C-20-demethoxy, C-20-acyloxy) and those having modifications at other positions {e.g., C-9-CH, C- 14-alkoxym ethyl, C- 14-hydroxymethyl or aceloxymethyl, C- 15-hydroxy/acyloxy, C- 15-methoxy, C-18- N-demethyl, 4,5-deoxy). Maytansinol and maytansinol analogues are described, for example, in U.S. Patent Nos 5,208,020 and 6,333,410, the contents of which are incorporated herein by reference. Maytansinol can be coupled to antibodies and antibody fragments using, e.g., an N-succinimidyl 3-(2-pyridyldithio) proprionate (also known as N-succinimidyl 4-(2-pyridyldithio)pentanoate or SPP), 4- succinimidyl-oxycarbonyl-a-(2-pyridyldithio)-toluene (SMPT), N-succinimidyl-3- (2-pyridyldithio)butyrate (SDPB), 2 iminothiolane, or S-acetylsuccinic anhydride. The taxane can be, for example, a taxol, taxotere, or novel taxane (see, e.g., WO 01/38318). The calicheamicin can be, for example, a bromo-complex calicheamicin (e.g., an alpha, beta or gamma bromo-complex), an iodo-complex calicheamicin (e.g., an alpha, beta or gamma iodo-complex), or analogs and mimics thereof.
Bromo-complex calicheamicins include Il -BR, I2-BR, I3-BR, I4-BR, Jl-BR, J2-BR and Kl-BR. Iodo-complex calicheamicins include H-I, I2-I, I3-I, Jl-I, J2-I, Ll-I and Kl-BR. Calicheamicin and mutants, analogs and mimics thereof are described, for example, in U.S. Patent Nos 4,970,198; 5,264,586; 5,550,246; 5,712,374, and 5,714,586, the contents of which are incorporated herein by reference. Duocarmycin analogs (e.g., KW-2189, DC88, DC89 CBI-TMI, and derivatives thereof) are described, for example, in U.S. Patent No. 5,070,092, U.S. Patent No. 5,187,186, U.S. Patent No. 5,641,780, U.S. Patent No. 5,641,780, U.S. Patent No. 4,923,990, and U.S. Patent No. 5,101,038, the contents of which are incorporated herein by reference.
Examples of other toxins include, but are not limited to antimetabolites (e.g. , methotrexate, 6-mercaptopurine, 6-thioguanine, cytarabine, 5-fiuorouracil decarbazine), alkylating agents (e.g., mechlorethamine, thioepa chlorambucil, CC- 1065 (see US Patent Nos. 5,475,092, 5,585,499, 5,846,545), melphalan, carmustine (BSNU) and lomustine (CCNU), cyclophosphamide, busulfan, dibromomannitol, streptozotocin, mitomycin C, and cis-dichlorodiamine platinum (II) (DDP) cisplatin), anthracyclines (e.g., daunorubicin (formerly daunomycin) and
doxorubicin), antibiotics (e.g., dactinomycin (formerly actinomycin), bleomycin, mithramycin, mitomycin, puromycin anthramycin (AMC)), duocarmycin and analogs or derivatives thereof, and anti-mitotic agents (e.g., vincristine, vinblastine, taxol, auristatins (e.g., auristatin E) and maytansinoids, and analogs or homologs thereof.
The toxin can also be a surface active toxin, such as a toxin that is a free radical generator (e.g. selenium containing toxin moieties), or radionuclide containing moiety. Suitable radionuclide containing moieties, include for example, moieties that contain radioactive iodine (131I or 125I), yttrium (90Y), lutetium (177Lu), actinium (225Ac), praseodymium, astatine (211At), rhenium (186Re), bismuth (212Bi or 213Bi), indium (111In), technetium ("mTc), phosphorus (32P), rhodium (188RIi), sulfur (35S), carbon (14C), tritium (3H), chromium (51Cr), chlorine (36Cl), cobalt (57Co or 58Co), iron (59Fe), selenium (75Se), or gallium (67Ga).
The toxin can be a protein, polypeptide or peptide, from bacterial sources, e.g., diphtheria toxin, pseudomonas exotoxin (PE) and plant proteins, e.g., the A chain of ricin (RTA), the ribosome inactivating proteins (RIPs) gelonin, pokeweed antiviral protein, saporin, and dodecandron are contemplated for use as toxins.
Antisense compounds of nucleic acids designed to bind, disable, promote degradation or prevent the production of the mRNA responsible for generating a particular target protein can also be used as a toxin. Antisense compounds include antisense RNA or DNA, single or double stranded, oligonucleotides, or their analogs, which can hybridize specifically to individual mRNA species and prevent transcription and/or RNA processing of the mRNA species and/or translation of the encoded polypeptide and thereby effect a reduction in the amount of the respective encoded polypeptide. Ching, et al., Proc. Natl. Acad. Sd. U.S.A. 86: 10006-10010 (1989); Broder, et al, Ann. Int. Med. 113: 604-618 (1990); Loreau, et al, FEBS
Letters 21 A: 53-56 (1990); Useful antisense therapeutics include for example: Veglin ™ (VasGene) and OGX-011 (Oncogenix).
Toxins can also be photoactive agents. Suitable photoactive agents include porphyrin-based materials such as porfimer sodium, the green porphyrins, chlorin E6, hematoporphyrin derivative itself, phthalocyanines, etiopurpurins, texaphrin, and the like.
The toxin can be an antibody or antibody fragment that binds an intracellular target {e.g., an intrabody), such as a dAb that binds an intracellular target. Such antibodies or antibody fragments (dAbs) can be directed to defined subcellular compartments or targets. For example, the antibodies or antibody fragments (dAbs) can bind an intracellular target selected from erbB2, EGFR, BCR-ABL, p21Ras, Caspase3, Caspase7, Bcl-2, p53, Cyclin E, ATF-1/CREB, HPV16 E7, HPl, Type IV collagenases, cathepsin L as well as others described in Kontermann, R.E., Methods, 34:163-170 (2004), incorporated herein by reference in its entirety.
Polypeptide Domains that Bind VEGF The invention provides polypeptide domains (e.g., immunoglobulin single variable domains, dAb monomers) that have a binding site with binding specificity for VEGF. In preferred embodiments, the polypeptide domain (e.g., dAb) binds to VEGF with an affinity (KD; KD=Koff (kd)/Kon (ka)) of 300 nM to 1 pM (i.e., 3 x 10"7 to 5 x 10"12M), preferably 50 nM to 1 pM, more preferably 5 nM to 1 pM and most preferably 1 nM to 1 pM, for example and KD of 1 X lO"7 M or less, preferably 1 x 10"8 M or less, more preferably 1 x 10"9 M or less, advantageously 1 x 10"10 M or less and most preferably 1 x 10"11 M or less; and/or a Koff rate constant of 5 x 10"1 s"1 to 1 x 10"7 S"1, preferably 1 x 10"2 s"1 to 1 x 10"6 s"1, more preferably 5 x 10"3 s"1 to 1 x 10"5 s"1, for example 5 X lO"1 s"1 or less, preferably 1 x 10"2 s"1 or less,
advantageously 1 x 10"3 s"1 or less, more preferably 1 x 10"4 s"1 or less, still more preferably 1 x 10"5 s"1 or less, and most preferably 1 x 10"6 s"1 or less as determined by surface plasmon resonance.
In some embodiments, the polypeptide domain that has a binding site with binding specificity for VEGF competes for binding to VEGF with a dAb selected from the group consisting of TAR15-1 (SEQ ID NO:100), TAR15-3 (SEQ ID NO:101), TAR15-4 (SEQ ID NO:102), TAR15-9 (SEQ ID NO:103), TAR15-10 (SEQ ID NO:104), TAR15-11 (SEQ ID NO:105), TAR15-12 (SEQ ID NO:106), TAR15-13 (SEQ ID NO:107), TAR15-14 (SEQ ID NO:108), TAR15-15 (SEQ ID NO:109), TARI S-IO (SEQ ID NOI I IO)5 TARI S-I V (SEQ ID NOI I I IX TARI S-I S (SEQ ID NO:112), TAR15-19 (SEQ ID NO:113), TAR15-20 (SEQ ID NO:114), TAR 15-22 (SEQ ID NO: 115), TARl 5-5 (SEQ ID NO: 116), TARl 5-6 (SEQ ID NO: 117), TARl 5-7 (SEQ ID NO: 118), TARl 5-8 (SEQ ID NO: 119), TARl 5-23 (SEQ ID NO:120), TAR15-24 (SEQ ID NO:121), TAR15-25 (SEQ ID NO:122), TAR15-26 (SEQ ID NO:123), TAR15-27 (SEQ ID NO:124), TAR15-29 (SEQ ID NO: 125), TARl 5-30 (SEQ ID NO: 126), TARl 5-6-500 (SEQ ID NO: 127), TARl 5- 6-501 (SEQ ID NO:128), TAR15-6-502 (SEQ ID NO:129), TAR15-6-503 (SEQ ID NO:130), TAR15-6-504 (SEQ ID NO:131), TAR15-6-505 (SEQ ID NO:132), TARl 5-6-506 (SEQ ID NO: 133), TARl 5-6-507 (SEQ ID NO: 134), TARl 5-6-508 (SEQ ID NO: 135), TARl 5-6-509 (SEQ ID NO: 136), TARl 5-6-510 (SEQ ID NO:137), TAR15-8-500 (SEQ ID NO:138), TAR15-8-501 (SEQ ID NO:139), TAR15-8-502 (SEQ ID NO:140), TAR15-8-503 (SEQ ID NO:141), TAR15-8-505 (SEQ ID NO:142), TAR15-8-506 (SEQ ID NO:143), TAR15-8-507 (SEQ ID NO:144), TAR15-8-508 (SEQ ID NO:145), TAR15-8-509 (SEQ ID NO:146), TARl 5-8-510 (SEQ ID NO: 147), TARl 5-8-511 (SEQ ID NO: 148), TARl 5-26-500 (SEQ ID NO: 149), TARl 5-26-501 (SEQ ID NO: 150), TARl 5-26-502 (SEQ ID NO:151), TAR15-26-503 (SEQ ID NO:152), TAR15-26-504 (SEQ ID NO:153), TARl 5-26-505 (SEQ ID NO: 154), TARl 5-26-506 (SEQ ID NO: 155), TARl 5-26- 507 (SEQ ID NO: 156), TARl 5-26-508 (SEQ ID NO: 157), TARl 5-26-509 (SEQ ID NO: 158), TARl 5-26-510 (SEQ ID NO: 159), TARl 5-26-511 (SEQ ID NO: 160), TAR15-26-512 (SEQ ID NO:161), TAR15-26-513 (SEQ ID NO:162), TAR15-26- 514 (SEQ ID NO:163), TAR15-26-515 (SEQ ID NO:164), TAR15-26-516 (SEQ ID NO: 165), TARl 5-26-517 (SEQ ID NO: 166), TARl 5-26-518 (SEQ ID NO: 167), TARl 5-26-519 (SEQ ID NO: 168), TARl 5-26-520 (SEQ ID NO: 169), TARl 5-26- 521 (SEQ ID NO:170), TAR15-26-522 (SEQ ID NO:171), TAR15-26-523 (SEQ ID NO: 172), TARl 5-26-524 (SEQ ID NO: 173), TARl 5-26-525 (SEQ ID NO: 174), TARl 5-26-526 (SEQ ID NO: 175), TARl 5-26-527 (SEQ ID NO: 176), TARl 5-26- 528 (SEQ ID NO: 177), TARl 5-26-529 (SEQ ID NO: 178), TARl 5-26-530 (SEQ ID NO: 179), TARl 5-26-531 (SEQ ID NO: 180), TARl 5-26-532 (SEQ ID NO: 181), TARl 5-26-533 (SEQ ID NO: 182), TARl 5-26-534 (SEQ ID NO: 183), TARl 5-26- 535 (SEQ ID NO:184), TAR15-26-536 (SEQ ID NO:185), TAR15-26-537 (SEQ ID NO: 186), TARl 5-26-538 (SEQ ID NO: 187), TARl 5-26-539 (SEQ ID NO: 188), TARl 5-26-540 (SEQ ID NO: 189), TARl 5-26-541 (SEQ ID NO: 190), TARl 5-26- 542 (SEQ ID NO:191), TAR15-26-543 (SEQ ID NO:192), TAR15-26-544 (SEQ ID NO: 193), TARl 5-26-545 (SEQ ID NO: 194), TARl 5-26-546 (SEQ ID NO: 195), TARl 5-26-547 (SEQ ID NO: 196), TARl 5-26-548 (SEQ ID NO: 197), TARl 5-26- 549 (SEQ ID NO:198), TAR15-26-550 (SEQ ID NO:539), and TAR15-26-551 (SEQ ID NO:540).
In some embodiments, the polypeptide domain that has a binding site with binding specificity for VEGF competes for binding to VEGF with TARl 5-26-555 (SEQ ID NO:704).
In some embodiments, the polypeptide domain that has a binding site with binding specificity for VEGF comprises an amino acid sequence that has at least about 80%, at least about 85%, at least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 94%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99% amino acid sequence identity with the amino acid sequence of a dAb selected from the group consisting of TAR15-1 (SEQ ID NO: 100), TAR15-3 (SEQ ID NO:101), TAR15-4 (SEQ ID NO:102), TAR15-9 (SEQ ID NO:103), TAR15-10 (SEQ ID NO: 104), TARl 5-11 (SEQ ID NO:105), TAR15-12 (SEQ ID NO:106), TAR15-13 (SEQ ID NO:107), TAR15-14 (SEQ ID NO:108), TAR15-15 (SEQ ID NO:109), TAR15-16 (SEQ ID NOrI lO), TAR15-17 (SEQ ID NOrI l 1), TAR15-18 (SEQ ID NOrI 12), TAR15-19 (SEQ ID NO:113), TARl 5-20 (SEQ ID NO:114), TAR 15-22 (SEQ ID NOrI 15), TARl 5-5 (SEQ ID NO: 116), TARl 5-6 (SEQ ID NO: 117), TARl 5-7 (SEQ ID
NOrI 18), TAR15-8 (SEQ ID NOrI 19), TAR15-23 (SEQ ID NO: 120), TAR15-24 (SEQ ID N0:121), TAR15-25 (SEQ ID NO:122), TAR15-26 (SEQ ID NO:123), TAR15-27 (SEQ ID NO:124), TAR15-29 (SEQ ID NO:125), TAR15-30 (SEQ ID NO:126), TAR15-6-500 (SEQ ID NO:127), TAR15-6-501 (SEQ ID NO: 128), TARl 5-6-502 (SEQ ID NO: 129), TARl 5-6-503 (SEQ ID NO: 130), TARl 5-6-504 (SEQ ID NO: 131), TARl 5-6-505 (SEQ ID NO: 132), TARl 5-6-506 (SEQ ID NO:133), TAR15-6-507 (SEQ ID NO:134), TAR15-6-508 (SEQ ID NO:135), TAR15-6-509 (SEQ ID NO:136), TAR15-6-510 (SEQ ID NO:137), TAR15-8-500 (SEQ ID NO:138), TAR15-8-501 (SEQ ID NO:139), TAR15-8-502 (SEQ ID NO:140), TAR15-8-503 (SEQ ID NO:141), TAR15-8-505 (SEQ ID NO: 142),
TAR15-8-506 (SEQ ID NO:143), TAR15-8-507 (SEQ ID NO: 144), TAR15-8-508 (SEQ ID NO:145), TAR15-8-509 (SEQ ID NO:146), TAR15-8-510 (SEQ ID NO:147), TARl 5-8-511 (SEQ ID NO:148), TAR15-26-500 (SEQ ID NO:149), TAR15-26-501 (SEQ ID NO:150), TAR15-26-502 (SEQ ID NO:151), TAR15-26- 503 (SEQ ID NO: 152), TARl 5-26-504 (SEQ ID NO: 153), TARl 5-26-505 (SEQ ID NO: 154), TARl 5-26-506 (SEQ ID NO: 155), TARl 5-26-507 (SEQ ID NO: 156), TARl 5-26-508 (SEQ ID NO: 157), TARl 5-26-509 (SEQ ID NO: 158), TARl 5-26- 510 (SEQ ID NO: 159), TARl 5-26-511 (SEQ ID NO: 160), TARl 5-26-512 (SEQ ID NO:161), TAR15-26-513 (SEQ ID NO:162), TAR15-26-514 (SEQ ID NO:163), TAR15-26-515 (SEQ ID NO: 164), TAR15-26-516 (SEQ ID NO:165), TAR15-26- 517 (SEQ ID NO: 166), TARl 5-26-518 (SEQ ID NO: 167), TARl 5-26-519 (SEQ ID NO:168), TAR15-26-520 (SEQ ID NO:169), TAR15-26-521 (SEQ ID NO:170), TAR15-26-522 (SEQ ID NO:171), TAR15-26-523 (SEQ ID NO:172), TAR15-26- 524 (SEQ ID NO: 173), TARl 5-26-525 (SEQ ID NO: 174), TARl 5-26-526 (SEQ ID NO:175), TAR15-26-527 (SEQ ID NO:176), TAR15-26-528 (SEQ ID NO:177), TAR15-26-529 (SEQ ID NO:178), TAR15-26-530 (SEQ ID NO:179), TAR15-26- 531 (SEQ ID NO:180), TAR15-26-532 (SEQ ID NO:181), TAR15-26-533 (SEQ ID NO: 182), TARl 5-26-534 (SEQ ID NO: 183), TARl 5-26-535 (SEQ ID NO: 184), TAR15-26-536 (SEQ ID NO:185), TAR15-26-537 (SEQ ID NO:186), TAR15-26- 538 (SEQ ID NO:187), TAR15-26-539 (SEQ ID NO:188), TAR15-26-540 (SEQ ID NO:189), TAR15-26-541 (SEQ ID NO:190), TAR15-26-542 (SEQ ID NO:191), TARl 5-26-543 (SEQ ID NO: 192), TARl 5-26-544 (SEQ ID NO: 193), TARl 5-26- 545 (SEQ ID NO: 194), TARl 5-26-546 (SEQ ID NO: 195), TARl 5-26-547 (SEQ ID NO:196), TAR15-26-548 (SEQ ID NO:197), TAR15-26-549 (SEQ ID NO:198), TARl 5-26-550 (SEQ ID NO:539), and TARl 5-26-551 (SEQ ID NO:540).
In some embodiments, the polypeptide domain that has a binding site with binding specificity for VEGF comprises an amino acid sequence that has at least about 80%, at least about 85%, at least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 94%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99% amino acid sequence identity with the amino acid sequence of TARl 5-26-555 (SEQ ID NO:704).
In preferred embodiments, the polypeptide domain that has a binding site with binding specificity for VEGF comprises an amino acid sequence that has at least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 94%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99% amino acid sequence identity with the amino acid sequence of a dAb selected from the group consisting of TARl 5-6 (SEQ ID
NO: 117), TARl 5-8 (SEQ ID NO:119), and TARl 5-26 (SEQ ID NO: 123). For example, the polypeptide domain that has a binding site with binding specificity for VEGF can comprise TARl 5-6 (SEQ ID NO: 117), TARl 5-8 (SEQ ID NO: 119), or TARl 5-26 (SEQ ID NO: 123).
In some embodiments, the polypeptide domain that has a binding site with binding specificity for VEGF competes with any of the dAbs disclosed herein for binding to VEGF. Preferably the polypeptide domain that has a binding site with binding specificity for VEGF is an immunoglobulin single variable domain. The polypeptide domain that has a binding site with binding specificity for VEGF can comprise any suitable immunoglobulin variable domain, and preferably comprises a human variable domain or a variable domain that comprises human framework regions. In certain embodiments, the polypeptide domain that has a binding site with binding specificity for VEGF comprises a universal framework, as described herein.
The universal framework can be a VL framework (Vλ or VK) , such as a framework that comprises the framework amino acid sequences encoded by the human germline DPKl, DPK2, DPK3, DPK4, DPK5, DPK6, DPK7, DPK8, DPK9, DPKlO, DPK12, DPK13, DPK15, DPKl 6, DPKl 8, DPK19, DPK20, DPK21, DPK22, DPK23, DPK24, DPK25, DPK26 or DPK 28 immunoglobulin gene segment. If desired, the VL framework can further comprise the framework amino acid sequence encoded by the human germline Jκl , JK2, JK3, JK4, or JK5
immunoglobulin gene segment.
In other embodiments the universal framework can be a VH framework, such as a framework that comprises the framework amino acid sequences encoded by the human germline DP4, DP7, DP8, DP9, DPlO, DP31, DP33, DP38, DP45, DP46, DP47, DP49, DP50, DP51, DP53, DP54, DP65, DP66, DP67, DP68 or DP69 immunoglobulin gene segment. If desired, the VH framework can further comprise the framework amino acid sequence encoded by the human germline JHI , JH2, JH3, JH4, Jii4b, JH5 and JH6 immunoglobulin gene segment.
In certain embodiments, the polypeptide domain that has a binding site with binding specificity for VEGF comprises one or more framework regions comprising an amino acid sequence that is the same as the amino acid sequence of a
corresponding framework region encoded by a human germline antibody gene segment, or the amino acid sequences of one or more of said framework regions collectively comprise up to 5 amino acid differences relative to the amino acid sequence of said corresponding framework region encoded by a human germline antibody gene segment. In other embodiments, the amino acid sequences of FWl, FW2, FW3 and FW4 of the polypeptide domain that has a binding site with binding specificity for VEGF are the same as the amino acid sequences of corresponding framework regions encoded by a human germline antibody gene segment, or the amino acid sequences of FWl, FW2, FW3 and FW4 collectively contain up to 10 amino acid differences relative to the amino acid sequences of corresponding framework regions encoded by said human germline antibody gene segment.
In other embodiments, the polypeptide domain that has a binding site with binding specificity for VEGF comprises FWl, FW2 and FW3 regions, and the amino acid sequence of said FWl, FW2 and FW3 regions are the same as the amino acid sequences of corresponding framework regions encoded by human germline antibody gene segments.
In particular embodiments, the polypeptide domain that has a binding site with binding specificity for VEGF comprises the DPK9 VL framework, or a VH framework selected from the group consisting of DP47, DP45 and DP38. The polypeptide domain that has a binding site with binding specificity for VEGF can comprise a binding site for a generic ligand, such as protein A, protein L and protein G.
In certain embodiments, the polypeptide domain that has a binding site with binding specificity for VEGF is substantially resistant to aggregation. For example, in some embodiments, less than about 10%, less than about 9%, less than about 8%, less than about 7%, less than about 6%, less than about 5%, less than about 4%, less than about 3%, less than about 2% or less than about 1% of the polypeptide domain that has a binding site with binding specificity for VEGF aggregates when a 1-5 mg/ml, 5-10 mg/ml, 10-20 mg/ml, 20-50 mg/ml, 50-100 mg/ml, 100-200 mg/ml or 200 -500 mg/ml solution of ligand or dAb in a solvent that is routinely used for drug formulation such as saline, buffered saline, citrate buffer saline, water, an emulsion, and, any of these solvents with an acceptable excipient such as those approved by the FDA, is maintained at about 220C, 22-250C, 25-3O0C, 30-370C, 37-4O0C, 40- 5O0C, 50-600C, 60-700C, 70-800C, 15-2O0C, 10-150C, 5-100C, 2-5°C, 0-20C, -1O0C to O0C, -2O0C to -1O0C, -4O0C to -2O0C, -6O0C to -4O0C, or -800C to -6O0C, for a period of about time, for example, 10 minutes, 1 hour, 8 hours, 24 hours, 2 days, 3 days, 4 days, 1 week, 2 weeks, 3 weeks, 1 month, 2 months, 3 months, 4 months, 6 months, 1 year, or 2 years.
Aggregation can be assessed using any suitable method, such as, by microscopy, assessing turbidity of a solution by visual inspection or spectroscopy or any other suitable method. Preferably, aggregation is assessed by dynamic light scattering. Polypeptide domains that have a binding site with binding specificity for VEGF that are resistant to aggregation provide several advantages. For example, such polypeptide domains that have a binding site with binding specificity for VEGF can readily be produced in high yield as soluble proteins by expression using a suitable biological production system, such as E. coli, and can be formulated and/or stored at higher concentrations than conventional polypeptides, and with less aggregation and loss of activity.
In addition, the polypeptide domain that has a binding site with binding specificity for VEGF that are resistant to aggregation can be produced more economically than other antigen- or epitope-binding polypeptides (e.g. , conventional antibodies). For example, generally, preparation of antigen- or epitope-binding polypeptides intended for in vivo applications includes processes (e.g., gel filtration) that remove aggregated polypeptides. Failure to remove such aggregates can result in a preparation that is not suitable for in vivo applications because, for example, aggregates of an antigen-binding polypeptide that is intended to act as an antagonist can function as an agonist by inducing cross-linking or clustering of the target antigen. Protein aggregates can also reduce the efficacy of therapeutic polypeptide by inducing an immune response in the subject to which they are administered.
In contrast, the aggregation resistant polypeptide domain that has a binding site with binding specificity for VEGF of the invention can be prepared for in vivo applications without the need to include process steps that remove aggregates, and can be used in in vivo applications without the aforementioned disadvantages caused by polypeptide aggregates.
In some embodiments, the polypeptide domain that has a binding site with binding specificity for VEGF unfolds reversibly when heated to a temperature (Ts) and cooled to a temperature (Tc), wherein Ts is greater than the melting temperature (Tm) of the polypeptide domain that has a binding site with binding specificity for VEGF, and Tc is lower than the melting temperature of the polypeptide domain that has a binding site with binding specificity for VEGF. For example, a polypeptide domain that has a binding site with binding specificity for VEGF can unfold reversibly when heated to 8O0C and cooled to about room temperature. A
polypeptide that unfolds reversibly loses function when unfolded but regains function upon refolding. Such polypeptides are distinguished from polypeptides that aggregate when unfolded or that improperly refold (misfolded polypeptides), i.e., do not regain function.
Polypeptide unfolding and refolding can be assessed, for example, by directly or indirectly detecting polypeptide structure using any suitable method. For example, polypeptide structure can be detected by circular dichroism (CD) (e.g., far- UV CD, near-UV CD), fluorescence (e.g., fluorescence of tryptophan side chains), susceptibility to proteolysis, nuclear magnetic resonance (NMR), or by detecting or measuring a polypeptide function that is dependent upon proper folding (e.g., binding to target ligand, binding to generic ligand). In one example, polypeptide unfolding is assessed using a functional assay in which loss of binding function (e.g., binding a generic and/or target ligand, binding a substrate) indicates that the polypeptide is unfolded.
The extent of unfolding and refolding of a polypeptide domain that has a binding site with binding specificity for VEGF can be determined using an unfolding or denaturation curve. An unfolding curve can be produced by plotting temperature as the ordinate and the relative concentration of folded polypeptide as the abscissa. The relative concentration of folded polypeptide domain that has a binding site with binding specificity for VEGF can be determined directly or indirectly using any suitable method (e.g., CD, fluorescence, binding assay). For example, a polypeptide domain that has a binding site with binding specificity for VEGF solution can be prepared and ellipticity of the solution determined by CD. The ellipticity value obtained represents a relative concentration of folded ligand (e.g., dAb monomer) of 100%. The polypeptide domain that has a binding site with binding specificity for VEGF in the solution is then unfolded by incrementally raising the temperature of the solution and ellipticity is determined at suitable increments (e.g., after each increase of one degree in temperature). The polypeptide domain that has a binding site with binding specificity for VEGF in solution is then refolded by incrementally reducing the temperature of the solution and ellipticity is determined at suitable increments. The data can be plotted to produce an unfolding curve and a refolding curve. The unfolding and refolding curves have a
characteristic sigmoidal shape that includes a portion in which the polypeptide domain that has a binding site with binding specificity for VEGF molecules are folded, an unfolding/refolding transition in which polypeptide domain that has a binding site with binding specificity for VEGF molecules are unfolded to various degrees, and a portion in which polypeptide domain that has a binding site with binding specificity for VEGF are unfolded. The y-axis intercept of the refolding curve is the relative amount of refolded polypeptide domain that has a binding site with binding specificity for VEGF recovered. A recovery of at least about 50%, or at least about 60%, or at least about 70%, or at least about 75%, or at least about 80%, or at least about 85%, or at least about 90%, or at least about 95% is indicative that the ligand or dAb monomer unfolds reversibly.
In a preferred embodiment, reversibility of unfolding of a polypeptide domain that has a binding site with binding specificity for VEGF is determined by preparing a polypeptide domain that has a binding site with binding specificity for VEGF solution and plotting heat unfolding and refolding curves. The polypeptide domain that has a binding site with binding specificity for VEGF solution can be prepared in any suitable solvent, such as an aqueous buffer that has a pH suitable to allow polypeptide domain that has a binding site with binding specificity for VEGF to dissolve (e.g., pH that is about 3 units above or below the isoelectric point (pi)). The polypeptide domain that has a binding site with binding specificity for VEGF solution is concentrated enough to allow unfolding/folding to be detected. For example, the ligand or dAb monomer solution can be about 0.1 μM to about 100 μM, or preferably about 1 μM to about 10 μM.
If the melting temperature (Tm) of a polypeptide domain that has a binding site with binding specificity for VEGF is known, the solution can be heated to about ten degrees below the Tm (Tm-10) and folding assessed by ellipticity or
fluorescence (e.g., far-UV CD scan from 200 ran to 250 ran, fixed wavelength CD at 235 ran or 225 nm; tryptophan fluorescent emission spectra at 300 to 450 nm with excitation at 298 nm) to provide 100% relative folded ligand or dAb monomer. The solution is then heated to at least ten degrees above Tm (Tm+ 10) in predetermined increments (e.g., increases of about 0.1 to about 1 degree), and ellipticity or fluorescence is determined at each increment. Then, the polypeptide domain that has a binding site with binding specificity for VEGF is refolded by cooling to at least Tm-10 in predetermined increments and ellipticity or fluorescence determined at each increment. If the melting temperature of a polypeptide domain that has a binding site with binding specificity for VEGF is not known, the solution can be unfolded by incrementally heating from about 250C to about 1000C and then refolded by incrementally cooling to at least about 250C, and ellipticity or fluorescence at each heating and cooling increment is determined. The data obtained can be plotted to produce an unfolding curve and a refolding curve, in which the y-axis intercept of the refolding curve is the relative amount of refolded protein recovered. In some embodiments, the polypeptide domain that has a binding site with binding specificity for VEGF does not comprise a Camelid
immunoglobulin variable domain, or one or more framework amino acids that are unique to immunoglobulin variable domains encoded by Camelid germline antibody gene segments.
Preferably, the polypeptide domain that has a binding site with binding specificity for VEGF is secreted in a quantity of at least about 0.5 mg/L when expressed in E. coli or in Pichia species (e.g., P. pastoris). In other preferred embodiments, a polypeptide domain that has a binding site with binding specificity for VEGF is secreted in a quantity of at least about 0.75 mg/L, at least about 1 mg/L, at least about 4 mg/L, at least about 5 mg/L, at least about 10 mg/L, at least about 15 mg/L, at least about 20 mg/L, at least about 25 mg/L, at least about 30 mg/L, at least about 35 mg/L, at least about 40 mg/L, at least about 45 mg/L, or at least about 50 mg/L, or at least about 100 mg/L, or at least about 200 mg/L, or at least about 300 mg/L, or at least about 400 mg/L, or at least about 500 mg/L, or at least about 600 mg/L, or at least about 700 mg/L, or at least about 800 mg/L, at least about 900 mg/L, or at least about lg/L when expressed in E. coli or in Pichia species (e.g., P. pastoris). In other preferred embodiments, a polypeptide domain that has a binding site with binding specificity for VEGF is secreted in a quantity of at least about 1 mg/L to at least about lg/L, at least about 1 mg/L to at least about 750 mg/L, at least about 100 mg/L to at least about 1 g/L, at least about 200 mg/L to at least about 1 g/L, at least about 300 mg/L to at least about 1 g/L, at least about 400 mg/L to at least about 1 g/L, at least about 500 mg/L to at least about lg/L, at least about 600 mg/L to at least about 1 g/L, at least about 700 mg/L to at least about 1 g/L, at least about 800 mg/L to at least about lg/L, or at least about 900 mg/L to at least about lg/L when expressed in E. coli or in Pichia species {e.g., P. pastoris). Although, a polypeptide domain that has a binding site with binding specificity for VEGF described herein can be secretable when expressed in E. coli or in Pichia species {e.g., P. pastoris), they can be produced using any suitable method, such as synthetic chemical methods or biological production methods that do not employ E. coli or Pichia species.
Polypeptide Domains that Bind EGFR
The invention provides polypeptide domains {e.g., dAb) that have a binding site with binding specificity for EGFR. In preferred embodiments, the polypeptide domain {e.g., dAb) binds to EGFR with an affinity (KD; KD=Koff (kd)/Kon (ka)) of 300 nM to 1 pM {i.e., 3 x 10"7to 5 x 10"12M), preferably 100 nM to 1 pM, or 50 nM to 10 pM, more preferably 10 nM to 100 pM and most preferably about 1 nM, for example and Kp of 1 x 10"7 M or less, preferably 1 x 10"8 M or less, more preferably about 1 x 10"9 M or less, 1 x 10"10 M or less or 1 x 10"11 M or less; and/or a Koff rate constant of 5 x 10"1 s"1 to 1 x 10"7 s"1, preferably 1 x 10"2 s"1 to 1 x 10"6 s"1, more preferably 5 x 10"3 s"1 to 1 x 10"5 s"1, for example 5 X lO"1 s"1 or less, preferably 1 x 10"2 s"1 or less, advantageously 1 x 10"3 s"1 or less, more preferably 1 x 10" s"1 or less, still more preferably 1 x 10"5 s"1 or less, and most preferably 1 x 10"6 s"1 or less as determined by surface plasmon resonance.
In some embodiments, the polypeptide domain that has a binding site with binding specificity for EGFR competes for binding to EGFR with a dAb selected from the group consisting of DOM16-17 (SEQ ID NO:325), DOM16-18 (SEQ ID NO:326), DOM16-19 (SEQ ID NO:327), DOM16-20 (SEQ ID NO:328), D0M16- 21 (SEQ ID NO:329), DOM16-22 (SEQ ID NO:330), DOM16-23 (SEQ ID
NO:331), DOM16-24 (SEQ ID NO:332), DOM16-25 (SEQ ID NO:333), DOM16- 26 (SEQ ID NO:334), DOM16-27 (SEQ ID NO:335), DOM16-28 (SEQ ID NO:336), DOM16-29 (SEQ ID NO:337), DOM16-30 (SEQ ID NO:338), DOM16- 31 (SEQ ID NO:339), DOM16-32 (SEQ ID NO:340), DOM16-33 (SEQ ID NO:341), DOM16-35 (SEQ ID NO:342), DOM16-37 (SEQ ID NO:343), DOM16- 38 (SEQ ID NO:344), DOM16-39 (SEQ ID NO:345), DOM16-40 (SEQ ID
NO:346), DOMl 6-41 (SEQ ID NO:347), DOMl 6-42 (SEQ ID NO:348), DOMl 6- 43 (SEQ ID NO:349), DOMl 6-44 (SEQ ID NO:350), DOMl 6-45 (SEQ ID NO:351), DOM16-46 (SEQ ID NO:352), DOM16-47 (SEQ ID NO:353), DOM16- 48 (SEQ ID NO:354), DOM16-49 (SEQ ID NO:355), DOM16-50 (SEQ ID NO:356), DOM16-59 (SEQ ID NO:357), DOM16-60 (SEQ ID NO:358), DOM16- 61 (SEQ ID NO:359), DOM16-62 (SEQ ID NO:360), DOM16-63 (SEQ ID NO:361), DOM16-64 (SEQ ID NO:362), DOM16-65 (SEQ ID NO:363), DOM16- 66 (SEQ ID NO:364), DOM16-67 (SEQ ID NO:365), DOM16-68 (SEQ ID NO:366), DOM16-69 (SEQ ID NO:367), DOM16-70 (SEQ ID NO:368), DOM16- 71 (SEQ ID NO:369), DOM16-72 (SEQ ID NO:370), DOM16-73 (SEQ ID
NO:371), DOM16-74 (SEQ ID NO:372), DOM16-75 (SEQ ID NO:373), DOM16- 76 (SEQ ID NO:374), DOM16-77 (SEQ ID NO:375), DOM16-78 (SEQ ID NO:376), DOM16-79 (SEQ ID NO:377), DOMl 6-80 (SEQ ID NO:378), DOM16- 81 (SEQ ID NO:379), DOMl 6-82 (SEQ ID NO:380), DOMl 6-83 (SEQ ID NO:381), DOM16-84 (SEQ ID NO:382), DOM16-85 (SEQ ID NO:383), DOM16- 87 (SEQ ID NO:384), DOMl 6-88 (SEQ ID NO:385), DOMl 6-89 (SEQ ID NO:386), DOM16-90 (SEQ ID NO:387), DOM16-91 (SEQ ID NO:388), DOM16- 92 (SEQ ID NO:389), DOMl 6-94 (SEQ ID NO:390), DOMl 6-95 (SEQ ID NO:391), DOM16-96 (SEQ ID NO:392), DOM16-97 (SEQ ID NO:393), DOM16- 98 (SEQ ID NO:394), DOMl 6-99 (SEQ ID NO:395), DOMl 6-100 (SEQ ID NO:396), DOM16-101 (SEQ ID NO:397), DOM16-102 (SEQ ID NO:398), DOM16-103 (SEQ ID NO:399), DOM16-104 (SEQ ID NO:400), DOM16-105 (SEQ ID NO:401), DOM16-106 (SEQ ID NO:402), DOM16-107 (SEQ ID
NO:403), DOM16-108 (SEQ ID NO:404), DOM16-109 (SEQ ID NO:405), DOM16-110 (SEQ ID NO:406), DOM16-111 (SEQ ID NO:407), DOMl 6-112 (SEQ ID NO:408), DOM16-113 (SEQ ID NO:409), DOMl 6-114 (SEQ ID
NO:410), DOM16-115 (SEQ ID NO:411), DOM16-116 (SEQ ID NO:412), DOM16-117 (SEQ ID NO:413), DOM16-118 (SEQ ID NO:414), DOM16-119 (SEQ ID NO:415), DOM16-39-6 (SEQ ID NO:416), DOM16-39-8 (SEQ ID NO:417), DOM16-39-34 (SEQ ID NO:418), DOM16-39-48 (SEQ ID NO:419), DOM16-39-87 (SEQ ID NO:420), DOM16-39-90 (SEQ ID NO:421), DOM16-39- 96 (SEQ ID NO:422), DOM16-39-100 (SEQ ID NO:423), DOMl 6-39-101 (SEQ ID NO:424), DOM16-39-102 (SEQ ID NO:425), DOM16-39-103 (SEQ ID NO:426), DOM16-39-104 (SEQ ID NO:427), DOM16-39-105 (SEQ ID NO:428), DOM16- 39-106 (SEQ ID NO:429), DOM16-39-107 (SEQ ID NO:430), DOM16-39-108 (SEQ ID NO:431), DOM16-39-109 (SEQ ID NO:432), DOM16-39-110 (SEQ ID NO:433), DOM16-39-111 (SEQ ID NO:434), DOM16-39-112 (SEQ ID NO:435), DOM16-39-113 (SEQ ID NO:436), DOM16-39-114 (SEQ ID NO:437), DOM16- 39-115 (SEQ ID NO:438), DOM16-39-116 (SEQ ID NO:439), DOM16-39-117 (SEQ ID NO:440), DOM16-39-200 (SEQ ID NO:441), DOM16-39-201 (SEQ ID NO:442), DOM16-39-202 (SEQ ID NO:443), DOM16-39-203 (SEQ ID NO:444), DOM16-39-204 (SEQ ID NO:445), DOM16-39-205 (SEQ ID NO:446), DOM16- 39-206 (SEQ ID NO:447), DOMl 6-39-207 (SEQ ID NO:448), DOMl 6-39-209 (SEQ ID NO:449), DOM16-52 (SEQ ID NO:450), NBl (SEQ ID NO:451), NB2 (SEQ ID NO:452), NB3 (SEQ ID NO:453), NB4 (SEQ ID NO:454), NB5 (SEQ ID NO:455), NB6 (SEQ ID NO:456), NB7 (SEQ ID NO:457), NB8 (SEQ ID NO:458), NB9 (SEQ ID NO:459), NBlO (SEQ ID NO:460), NBl 1 (SEQ ID NO:461), NB12 (SEQ ID NO:462), NB 13 (SEQ ID NO:463), NB 14 (SEQ ID NO:464), NB 15 (SEQ ID NO:465), NB 16 (SEQ ID NO:466), NB 17 (SEQ ID NO:467), NB 18 (SEQ ID NO:468), NB19 (SEQ ID NO:469), NB20 (SEQ ID NO:470), NB21 (SEQ ID NO:471), and NB22 (SEQ ID NO:472).
In some embodiments, the polypeptide domain that has a binding site with binding specificity for EGFR competes for binding to EGFR with a dAb selected from the group consisting of DOM 16-39-210 (SEQ ID NO:541), DOM16-39-211 (SEQ ID NO:542), DOM16-39-212 (SEQ ID NO:543), DOM16-39-213 (SEQ ID NO:544), DOM16-39-214 (SEQ ID NO:545), DOM16-39-215 (SEQ ID NO:546), DOM16-39-216 (SEQ ID NO:547), DOM16-39-217 (SEQ ID NO:548), DOM16- 39-218 (SEQ ID NO:549), DOM16-39-219 (SEQ ID NO:550), DOM16-39-220 (SEQ ID NO:551), DOM16-39-221 (SEQ ID NO:552), DOM16-39-222 (SEQ ID NO:553), DOMl 6-39-223 (SEQ ID NO:554), DOMl 6-39-224 (SEQ ID NO:555), DOM16-39-225 (SEQ ID NO:556), DOM16-39-226 (SEQ ID NO:557), DOM16- 39-227 (SEQ ID NO:558), DOM16-39-228 (SEQ ID NO:559), DOM16-39-229 (SEQ ID NO:560), DOM16-39-230 (SEQ ID NO:561), DOM16-39-231 (SEQ ID NO:562), DOM16-39-232 (SEQ ID NO:563), DOM16-39-233 (SEQ ID NO:564), DOMl 6-39-234 (SEQ ID NO:565), DOMl 6-39-235 (SEQ ID NO:566), DOMl 6- 39-500 (SEQ ID NO:725), DOMl 6-39-502(SEQ ID NO:726), DOMl 6-39-503 (SEQ ID NO:567), DOMl 6-39-504 (SEQ ID NO:568), DOMl 6-39-505 (SEQ ID NO:569), DOM16-39-506 (SEQ ID NO:570), DOM16-39-507 (SEQ ID NO:571), DOM16-39-508 (SEQ ID NO:572), DOM16-39-509 (SEQ ID NO:573), DOM16- 39-510 (SEQ ID NO:574), DOMl 6-39-511 (SEQ ID NO:575), DOMl 6-39-512 (SEQ ID NO:576), DOM16-39-521 (SEQ ID NO:577), DOM16-39-522 (SEQ ID NO:578), DOMl 6-39-523 (SEQ ID NO:579), DOMl 6-39-524 (SEQ ID NO:580), DOM16-39-527 (SEQ ID NO:581), DOM16-39-525 (SEQ ID NO:582), DOM16- 39-526 (SEQ ID NO:583), DOM16-39-540 (SEQ ID NO:584), DOM16-39-541 (SEQ ID NO:585), DOMl 6-39-542 (SEQ ID NO:586), DOMl 6-39-543 (SEQ ID NO:587), DOMl 6-39-544 (SEQ ID NO:588), DOMl 6-39-545 (SEQ ID NO:589), DOMl 6-39-550 (SEQ ID NO:590), DOMl 6-39-551 (SEQ ID NO:591), DOMl 6- 39-552 (SEQ ID NO:592), DOM16-39-553 (SEQ ID NO:593), DOM16-39-554 (SEQ ID NO:594), DOM16-39-555 (SEQ ID NO:595), DOM16-39-561 (SEQ ID NO:596), DOMl 6-39-562 (SEQ ID NO:597), DOMl 6-39-563 (SEQ ID NO:598), DOM16-39-564 (SEQ ID NO:599), DOM16-39-571 (SEQ ID NO:600), DOM16- 39-572 (SEQ ID NO:601), DOMl 6-39-573 (SEQ ID NO:602), DOMl 6-39-574 (SEQ ID NO:603), DOM16-39-580 (SEQ ID NO:604), DOM16-39-591 (SEQ ID NO:605), DOM16-39-592 (SEQ ID NO:606), DOM16-39-593 (SEQ ID NO:607), DOMl 6-39-601 (SEQ ID NO:608), DOMl 6-39-602 (SEQ ID NO:609), DOMl 6- 39-603 (SEQ ID NO:610), DOMl 6-39-604 (SEQ ID NO:611), DOMl 6-39-605 (SEQ ID NO:612), DOM16-39-607 (SEQ ID NO:613), DOM16-39-611 (SEQ ID NO:614), DOM16-39-612 (SEQ ID NO:615), DOM16-39-613 (SEQ ID NO:616), DOM16-39-614 (SEQ ID NO:617), DOM16-39-615 (SEQ ID NO:618), DOM16- 39-616 (SEQ ID NO:619), DOM16-39-617 (SEQ ID NO:620), DOM16-39-618 (SEQ ID NO:621), and DOM16-39-619 (SEQ ID NO:622). In some embodiments, the polypeptide domain that has a binding site with binding specificity for EGFR comprises an amino acid sequence that has at least about 80%, at least about 85%, at least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 94%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99% amino acid sequence identity with the amino acid sequence of a dAb selected from the group consisting of DOM16-17 (SEQ ID NO:325), DOM16-18 (SEQ ID NO:326), DOM16-19 (SEQ ID NO:327), DOM16-20 (SEQ ID NO:328), DOM16-21 (SEQ ID NO:329), DOM16- 22 (SEQ ID NO:330), DOM16-23 (SEQ ID NO:331), DOM16-24 (SEQ ID
NO:332), DOMl 6-25 (SEQ ID NO:333), DOMl 6-26 (SEQ ID NO:334), DOMl 6- 27 (SEQ ID NO:335), DOM16-28 (SEQ ID NO:336), DOM16-29 (SEQ ID
NO:337), DOM16-30 (SEQ ID NO:338), DOM16-31 (SEQ ID NO:339), D0M16- 32 (SEQ ID NO:340), DOMl 6-33 (SEQ ID NO:341), DOMl 6-35 (SEQ ID
NO:342), DOM16-37 (SEQ ID NO:343), DOM16-38 (SEQ ID NO:344), D0M16- 39 (SEQ ID NO:345), DOMl 6-40 (SEQ ID NO:346), DOMl 6-41 (SEQ ID
NO:347), DOM16-42 (SEQ ID NO:348), DOM16-43 (SEQ ID NO:349), D0M16- 44 (SEQ ID NO:350), DOM16-45 (SEQ ID NO:351), DOM16-46 (SEQ ID
NO:352), DOM16-47 (SEQ ID NO:353), DOM16-48 (SEQ ID NO:354), D0M16- 49 (SEQ ID NO:355), DOMl 6-50 (SEQ ID NO:356), DOMl 6-59 (SEQ ID
NO:357), DOM16-60 (SEQ ID NO:358), DOM16-61 (SEQ ID NO:359), D0M16- 62 (SEQ ID NO:360), DOM16-63 (SEQ ID NO:361), DOM16-64 (SEQ ID
NO:362), DOM16-65 (SEQ ID NO:363), DOM16-66 (SEQ ID NO:364), D0M16- 67 (SEQ ID NO:365), DOMl 6-68 (SEQ ID NO:366), DOMl 6-69 (SEQ ID
NO:367), DOM16-70 (SEQ ID NO:368), DOM16-71 (SEQ ID NO:369), D0M16- 72 (SEQ ID NO:370), DOM16-73 (SEQ ID NO:371), DOM16-74 (SEQ ID
NO:372), DOM16-75 (SEQ ID NO:373), DOM16-76 (SEQ ID NO:374), D0M16- 77 (SEQ ID NO:375), DOMl 6-78 (SEQ ID NO:376), DOMl 6-79 (SEQ ID
NO:377), DOM16-80 (SEQ ID NO:378), DOM16-81 (SEQ ID NO:379), D0M16- 82 (SEQ ID NO:380), DOM16-83 (SEQ ID NO:381), DOM16-84 (SEQ ID
NO:382), DOM16-85 (SEQ ID NO:383), DOM16-87 (SEQ ID NO:384), D0M16- 88 (SEQ ID NO:385), DOM16-89 (SEQ ID NO:386), DOM16-90 (SEQ ID
NO:387), DOM16-91 (SEQ ID NO:388), DOMl 6-92 (SEQ ID NO:389), D0M16- 94 (SEQ ID NO:390), DOM16-95 (SEQ ID NO:391), DOM16-96 (SEQ ID
NO:392), DOMl 6-97 (SEQ ID NO:393), DOMl 6-98 (SEQ ID NO:394), DOMl 6- 99 (SEQ ID NO:395), DOM16-100 (SEQ ID NO:396), DOMl 6-101 (SEQ ID NO:397), DOM16-102 (SEQ ID NO:398), DOM16-103 (SEQ ID NO:399), DOM16-104 (SEQ ID NO:400), DOM16-105 (SEQ ID NO:401), DOM16-106 (SEQ ID NO:402), DOMl 6-107 (SEQ ID NO:403), DOMl 6-108 (SEQ ID
NO:404), DOM16-109 (SEQ ID NO:405), DOM16-110 (SEQ ID NO:406), DOMl 6-111 (SEQ ID NO:407), DOMl 6-112 (SEQ ID NO:408), DOMl 6-113 (SEQ ID NO:409), DOM16-114 (SEQ ID NO:410), DOMl 6-115 (SEQ ID
NO:411), DOM16-116 (SEQ ID NO:412), DOM16-117 (SEQ ID NO:413),
DOM16-118 (SEQ ID NO:414), DOMl 6-119 (SEQ ID NO:415), DOMl 6-39-6 (SEQ ID NO:416), DOMl 6-39-8 (SEQ ID NO:417), DOMl 6-39-34 (SEQ ID NO:418), DOM16-39-48 (SEQ ID NO:419), DOM16-39-87 (SEQ ID NO:420), DOM16-39-90 (SEQ ID NO:421), DOM16-39-96 (SEQ ID NO:422), DOM16-39- 100 (SEQ ID NO:423), DOM16-39-101 (SEQ ID NO:424), DOM16-39-102 (SEQ ID NO:425), DOMl 6-39-103 (SEQ ID NO:426), DOMl 6-39-104 (SEQ ID
NO:427), DOM16-39-105 (SEQ ID NO:428), DOM16-39-106 (SEQ ID NO:429), DOM16-39-107 (SEQ ID NO:430), DOM16-39-108 (SEQ ID NO:431), DOM16- 39-109 (SEQ ID NO:432), DOM16-39-110 (SEQ ID NO:433), DOM16-39-111 (SEQ ID NO:434), DOM16-39-112 (SEQ ID NO:435), DOM16-39-113 (SEQ ID NO:436), DOM16-39-114 (SEQ ID NO:437), DOM16-39-115 (SEQ ID NO:438), DOM16-39-116 (SEQ ID NO:439), DOM16-39-117 (SEQ ID NO:440), DOM16- 39-200 (SEQ ID NO:441), DOM16-39-201 (SEQ ID NO:442), DOM16-39-202 (SEQ ID NO:443), DOMl 6-39-203 (SEQ ID NO:444), DOMl 6-39-204 (SEQ ID NO:445), DOMl 6-39-205 (SEQ ID NO:446), DOMl 6-39-206 (SEQ ID NO:447), DOM16-39-207 (SEQ ID NO:448), DOM16-39-209 (SEQ ID NO:449), DOM16-52 (SEQ ID NO:450), NBl (SEQ ID NO:451), NB2 (SEQ ID NO:452), NB3 (SEQ ID NO:453), NB4 (SEQ ID NO:454), NB5 (SEQ ID NO:455), NB6 (SEQ ID NO:456), NB7 (SEQ ID NO:457), NB8 (SEQ ID NO:458), NB9 (SEQ ID NO:459), NBlO (SEQ ID NO:460), NBI l (SEQ ID NO:461), NB12 (SEQ ID NO:462), NB13 (SEQ ID NO:463), NB14 (SEQ ID NO:464), NB15 (SEQ ID NO:465), NB16 (SEQ ID NO:466), NB17 (SEQ ID NO:467), NB18 (SEQ ID NO:468), NB19 (SEQ ID NO:469), NB20 (SEQ ID NO:470), NB21 (SEQ ID NO:471), and NB22 (SEQ ID NO:472).
In some embodiments, the polypeptide domain that has a binding site with binding specificity for EGFR comprises an amino acid sequence that has at least about 80%, at least about 85%, at least about 90%, at least about 91 %, at least about 92%, at least about 93%, at least about 94%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99% amino acid sequence identity with the amino acid sequence of a dAb selected from the group consisting of DOM16-39-210 (SEQ ID NO:541), DOM16-39-211 (SEQ ID NO.542), D0M16- 39-212 (SEQ ID NO:543), DOM16-39-213 (SEQ ID NO:544), DOM16-39-214 (SEQ ID NO:545), DOM16-39-215 (SEQ ID NO:546), DOM16-39-216 (SEQ ID NO:547), DOM16-39-217 (SEQ ID NO:548), DOM16-39-218 (SEQ ID NO:549), DOM16-39-219 (SEQ ID NO:550), DOMl 6-39-220 (SEQ ID NO:551), D0M16- 39-221 (SEQ ID NO:552), DOM16-39-222 (SEQ ID NO:553), DOM16-39-223 (SEQ ID NO:554), DOM16-39-224 (SEQ ID NO:555), DOM16-39-225 (SEQ ID NO:556), DOMl 6-39-226 (SEQ ID NO:557), DOMl 6-39-227 (SEQ ID NO:558), DOM16-39-228 (SEQ ID NO:559), DOM16-39-229 (SEQ ID NO:560), D0M16- 39-230 (SEQ ID NO:561), DOM16-39-231 (SEQ ID NO:562), DOM16-39-232 (SEQ ID NO:563), DOM16-39-233 (SEQ ID NO:564), DOM16-39-234 (SEQ ID NO:565), DOM16-39-235 (SEQ ID NO:566), DOM16-39-500 (SEQ ID NO:725), DOMl 6-39-502(SEQ ID NO:726), DOMl 6-39-503 (SEQ ID NO:567), DOMl 6-39- 504 (SEQ ID NO:568), DOMl 6-39-505 (SEQ ID NO:569), DOMl 6-39-506 (SEQ ID NO:570), DOM16-39-507 (SEQ ID NO:571), DOM16-39-508 (SEQ ID
NO:572), DOM16-39-509 (SEQ ID NO:573), DOM16-39-510 (SEQ ID NO:574), DOM16-39-511 (SEQ ID NO:575), DOM16-39-512 (SEQ ID NO:576), D0M16- 39-521 (SEQ ID NO:577), DOM16-39-522 (SEQ ID NO:578), DOM16-39-523 (SEQ ID NO:579), DOM16-39-524 (SEQ ID NO:580), DOM16-39-527 (SEQ ID NO:581), DOM16-39-525 (SEQ ID NO:582), DOM16-39-526 (SEQ ID NO:583), DOM16-39-540 (SEQ ID NO:584), DOM16-39-541 (SEQ ID NO:585), D0M16- 39-542 (SEQ ID NO:586), DOM16-39-543 (SEQ ID NO:587), DOM16-39-544 (SEQ ID NO:588), DOMl 6-39-545 (SEQ ID NO:589), DOMl 6-39-550 (SEQ ID NO:590), DOM16-39-551 (SEQ ID NO:591), DOM16-39-552 (SEQ ID NO:592), DOM16-39-553 (SEQ ID NO:593), DOM16-39-554 (SEQ ID NO:594), DOM16- 39-555 (SEQ ID NO:595), DOM16-39-561 (SEQ ID NO:596), DOM16-39-562 (SEQ ID NO:597), DOM16-39-563 (SEQ ID NO:598), DOMl 6-39-564 (SEQ ID NO:599), DOM16-39-571 (SEQ ID NO:600), DOM16-39-572 (SEQ ID NO:601), DOM16-39-573 (SEQ ID NO:602), DOM16-39-574 (SEQ ID NO:603), DOM16- 39-580 (SEQ ID NO:604), DOM16-39-591 (SEQ ID NO:605), DOM16-39-592 (SEQ ID NO:606), DOM16-39-593 (SEQ ID NO:607), DOM16-39-601 (SEQ ID NO:608), DOM16-39-602 (SEQ ID NO:609), DOM16-39-603 (SEQ ID NO:610), DOM16-39-604 (SEQ ID NO:611), DOM16-39-605 (SEQ ID NO:612), DOM16- 39-607 (SEQ ID NO:613), DOM16-39-611 (SEQ ID NO:614), DOM16-39-612 (SEQ ID NO:615), DOM16-39-613 (SEQ ID NO:616), DOM16-39-614 (SEQ ID NO:617), DOM16-39-615 (SEQ ID NO:618), DOM16-39-616 (SEQ ID NO:619), DOM16-39-617 (SEQ ID NO:620), DOM16-39-618 (SEQ ID NO:621), and
DOMl 6-39-619 (SEQ ID NO:622).
In preferred embodiments, the polypeptide domain that has a binding site with binding specificity for EGFR comprises an amino acid sequence that has at least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 94%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99% amino acid sequence identity with the amino acid sequence of DOM16-39 (SEQ ID NO:345). For example, the polypeptide domain that has a binding site with binding specificity for EGFR can comprise the amino acid sequence of DOMl 6-39-87 (SEQ ID NO:420), DOMl 6-39-100 (SEQ ID NO:423), DOM16-39-107 (SEQ ID NO:430), DOM16-39-109 (SEQ ID NO:432), DOM16-39-115 (SEQ ID NO:438), or DOM16-39-200 (SEQ ID NO:441).
In other preferred embodiments, the polypeptide domain that has a binding site with binding specificity for EGFR comprises an amino acid sequence that has at least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 94%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99% amino acid sequence identity with the amino acid sequence of DOM16-39-521 (SEQ ID NO:577), DOM16-39-541 (SEQ ID NO:585), DOM16-39-542 (SEQ ID NO:586), DOM16-39-551 (SEQ ID NO:591), D0M16- 39-601 (SEQ ID NO:608), DOM16-39-604 (SEQ ID NO:611), DOM16-39-618 (SEQ ID NO:621), and DOM 16-39-619 (SEQ ID NO:622).
In some embodiments, the polypeptide domain that has a binding site with binding specificity for EGFR competes with any of the dAbs disclosed herein for binding to EGFR.
Preferably, the polypeptide domain that has a binding site with binding specificity for EGFR is an immunoglobulin single variable domain. The
polypeptide domain that has a binding site with binding specificity for EGFR can comprise any suitable immunoglobulin variable domain, and preferably comprises a human variable domain or a variable domain that comprises human framework regions. In certain embodiments, the polypeptide domain that has a binding site with binding specificity for EGFR comprises a universal framework, as described herein.
In certain embodiments, the polypeptide domain that has a binding site with binding specificity for EGFR resists aggregation, unfolds reversibly, comprises a framework region and/or is secreted as described above for the polypeptide domain that has a binding site with binding specificity for VEGF. dAb Monomers that Bind Serum Albumin
The ligands of the invention can further comprise a dAb monomer that binds serum albumin (SA) with a K^ of InM to 500 μ M (i.e., x 10"9 to 5 x 10"4), preferably 100 nM to 10 μ M. Preferably, for a ligand comprising an anti-SA dAb, the binding (e.g. Kj and/or K0 ff as measured by surface plasmon resonance, e.g., using BiaCore) of the ligand to its target(s) is from 1 to 100000 times (preferably 100 to 100000, more preferably 1000 to 100000, or 10000 to 100000 times) stronger than for SA. Preferably, the serum albumin is human serum albumin (HSA). In one embodiment, the first dAb (or a dAb monomer) binds SA (e.g., HSA) with a K^ of approximately 50, preferably 70, and more preferably 100, 150 or 200 nM.
In certain embodiments, the dAb monomer that binds SA resists aggregation, unfolds reversibly and/or comprises a framework region as described above for dAb monomers that bind CD38. In particular embodiments, the antigen-binding fragment of an antibody that binds serum albumin is a dAb that binds human serum albumin, hi certain embodiments, the dAb binds human serum albumin and competes for binding to albumin with a dAb selected from the group consisting of DOM7m-16 (SEQ ID NO: 473), DOM7m-12 (SEQ ID NO: 474), DOM7m-26 (SEQ ID NO: 475), DOM7r-l (SEQ ID NO: 476), DOM7r-3 (SEQ ID NO: 477), DOM7r-4 (SEQ ID NO: 478), DOM7r-5 (SEQ ID NO: 479), DOM7r-7 (SEQ ID NO: 480), DOM7r-8 (SEQ ID NO: 481), DOM7h-2 (SEQ ID NO: 482), DOM7h-3 (SEQ ID NO: 483), DOM7h-4 (SEQ ID NO: 484), DOM7h-6 (SEQ ID NO: 485), DOM7h-l (SEQ ID ' NO: 486), DOM7h-7 (SEQ ID NO: 487), DOM7h-22 (SEQ ID NO: 489), DOM7h- 23 (SEQ ID NO: 490), DOM7h-24 (SEQ ID NO: 491), DOM7h-25 (SEQ ID NO: 492), DOM7h-26 (SEQ ID NO: 493), DOM7h-21 (SEQ ID NO: 494), DOM7h-27 (SEQ ID NO: 495), DOM7h-8 (SEQ ID NO: 496), DOM7r-13 (SEQ ID NO: 497), DOM7r-14 (SEQ ID NO: 498), DOM7r-15 (SEQ ID NO: 499), DOM7r-16 (SEQ ID NO: 500), DOM7r-17 (SEQ ID NO: 501), DOM7r-18 (SEQ ID NO: 502), DOM7r- 19 (SEQ ID NO: 503), DOM7r-20 (SEQ ID NO: 504), DOM7r-21 (SEQ ID NO: 505), DOM7r-22 (SEQ ID NO: 506), DOM7r-23 (SEQ ID NO: 507), DOM7r-24 (SEQ ID NO: 508), DOM7r-25 (SEQ ID NO: 509), DOM7r-26 (SEQ ID NO: 510), DOM7r-27 (SEQ ID NO: 511), DOM7r-28 (SEQ ID NO: 512), DOM7r-29 (SEQ ID NO: 513), DOM7r-30 (SEQ ID NO: 514), DOM7r-31 (SEQ ID NO: 515), DOM7r- 32 (SEQ ID NO: 516), and DOM7r-33 (SEQ ID NO: 517).
In certain embodiments, the dAb binds human serum albumin and comprises an amino acid sequence that has at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 96%, or at least about 97%, or at least about 98%, or at least about 99% amino acid sequence identity with the amino acid sequence of a dAb selected from the group consisting of DOM7m-16 (SEQ ID NO: 473), DOM7m-12 (SEQ ID NO: 474), DOM7m-26 (SEQ ID NO: 475), DOM7r-l (SEQ ID NO: 476), DOM7r-3 (SEQ ID NO: 477), DOM7r-4 (SEQ ID NO: 478), DOM7r-5 (SEQ ID NO: 479), DOM7r-7 (SEQ ID NO: 480), DOM7r-8 (SEQ ID NO: 481), DOM7h-2 (SEQ ID NO: 482), DOM7h-3 (SEQ ID NO: 483), DOM7h-4 (SEQ ID NO: 484), DOM7h-6 (SEQ ID NO: 485), DOM7h-l (SEQ ID NO: 486), DOM7h-7 (SEQ ID NO: 487), DOM7h-22 (SEQ ID NO: 489), DOM7h- 23 (SEQ ID NO: 490), DOM7h-24 (SEQ ID NO: 491), DOM7h-25 (SEQ ID NO: 492), DOM7h-26 (SEQ ID NO: 493), DOM7h-21 (SEQ ID NO: 494), DOM7h-27 (SEQ ID NO: 495), DOM7h-8 (SEQ ID NO: 496), DOM7r-13 (SEQ ID NO: 497), DOM7r-14 (SEQ ID NO: 498), DOM7r-15 (SEQ ID NO: 499), DOM7r-16 (SEQ ID NO: 500), DOM7r-17 (SEQ ID NO: 501), DOM7r-18 (SEQ ID NO: 502), DOM7r- 19 (SEQ ID NO: 503), DOM7r-20 (SEQ ID NO: 504), DOM7r-21 (SEQ ID NO: 505), DOM7r-22 (SEQ ID NO: 506), DOM7r-23 (SEQ ID NO: 507), DOM7r-24 (SEQ ID NO: 508), DOM7r-25 (SEQ ID NO: 509), DOM7r-26 (SEQ ID NO: 510), DOM7r-27 (SEQ ID NO: 511), DOM7r-28 (SEQ ID NO: 512), DOM7r-29 (SEQ ID NO: 513), DOM7r-30 (SEQ ID NO: 514), DOM7r-31 (SEQ ID NO: 515), DOM7r- 32 (SEQ ID NO: 516), and DOM7r-33 (SEQ ID NO: 517).
For example, the dAb that binds human serum albumin can comprise an amino acid sequence that has at least about 90%, or at least about 95%, or at least about 96%, or at least about 97%, or at least about 98%, or at least about 99% amino acid sequence identity with DOM7h-2 (SEQ ID NO:482), DOM7h-3 (SEQ ID NO:483), DOM7h-4 (SEQ ID NO:484), DOM7h-6 (SEQ ID NO:485), DOM7h-l (SEQ ID NO:486), DOM7h-7 (SEQ ID NO:487), DOM7h-8 (SEQ ID NO:496), DOM7r-13 (SEQ ID NO:497), DOM7r-14 (SEQ ID NO:498), DOM7h-22 (SEQ ID NO:489), DOM7h-23 (SEQ ID NO:490), DOM7h-24 (SEQ ID NO:491), DOM7h- 25 (SEQ ID NO:492), DOM7h-26 (SEQ ID NO:493), DOM7h-21 (SEQ ID
NO:494), and DOM7h-27 (SEQ ID NO:495).
Amino acid sequence identity is preferably determined using a suitable sequence alignment algorithm and default parameters, such as BLAST P (Karlin and Altschul, Proc. Natl. Acad. ScL USA S7(6):2264-2268 (1990)).
In more particular embodiments, the dAb is a Vκ dAb that binds human serum albumin and has an amino acid sequence selected from the group consisting of DOM7h-2 (SEQ ID NO:482), DOM7h-3 (SEQ ID NO:483), DOM7h-4 (SEQ ID NO:484), DOM7h-6 (SEQ ID NO:485), DOM7h-l (SEQ ID NO:486), DOM7h-7 (SEQ ID NO:487), DOM7h-8 (SEQ ID NO:496), DOM7r-13 (SEQ ID NO:497), and DOM7r-14 (SEQ ID NO:498) , or a VH dAb that has an amino acid sequence selected from the group consisting of: DOM7h-22 (SEQ ID NO:489), DOM7h-23 (SEQ ID NO:490), DOM7h-24 (SEQ ID NO:491), DOM7h-25 (SEQ ID NO:492), DOM7h-26 (SEQ ID NO:493), DOM7h-21 (SEQ ID NO:494), DOM7h-27 (SEQ ID NO:495). In other embodiments, the antigen-binding fragment of an antibody that binds serum albumin is a dAb that binds human serum albumin and comprises the CDRs of any of the foregoing amino acid sequences.
Suitable Camelid VjJiH tnat ^n& serurn albumin include those disclosed in
WO 2004/041862 (Ablynx N. V.) and herein, such as Sequence A (SEQ ID
NO:518), Sequence B (SEQ ID NO:519), Sequence C (SEQ ID NO:520), Sequence D (SEQ ID NO:521), Sequence E (SEQ ID NO:522), Sequence F (SEQ ID
NO:523), Sequence G (SEQ ID NO:524), Sequence H (SEQ ID NO:525), Sequence I (SEQ ID NO:526), Sequence J (SEQ ID NO:527), Sequence K (SEQ ID NO:528), Sequence L (SEQ ID NO:529), Sequence M (SEQ ID NO:530), Sequence N (SEQ ID NO:531), Sequence O (SEQ ID NO:532), Sequence P (SEQ ID NO:533), Sequence Q (SEQ ID NO:534). In certain embodiments, the Camelid VJJH binds human serum albumin and comprises an amino acid sequence that has at least about 80%, or at least about 85%, or at least about 90%, or at least about 95%, or at least about 96%, or at least about 97%, or at least about 98%, or at least about 99% amino acid sequence identity with any one of SEQ ID NOS :518-534.
Amino acid sequence identity is preferably determined using a suitable sequence alignment algorithm and default parameters, such as BLAST P (Karlin and Altschul, Proc. Natl. Acad. ScL USA 57(6):2264-2268 (1990)).
In some embodiments, the ligand comprises an anti-serum albumin dAb that competes with any anti-serum albumin dAb disclosed herein for binding to serum albumin {e.g., human serum albumin). Nucleic Acid Molecules, Vectors and Host Cells
The invention also provides isolated and/or recombinant nucleic acid molecules encoding ligands {e.g., dual-specific ligands and multispecific ligands) as described herein.
Nucleic acids referred to herein as "isolated" are nucleic acids which have been separated away from the nucleic acids of the genomic DNA or cellular RNA of their source of origin {e.g., as it exists in cells or in a mixture of nucleic acids such as a library), and include nucleic acids obtained by methods described herein or other suitable methods, including essentially pure nucleic acids, nucleic acids produced by chemical synthesis, by combinations of biological and chemical methods, and recombinant nucleic acids which are isolated (see e.g., Daugherty, B.L. et al., Nucleic Acids Res., 19(9): 2471-2476 (1991); Lewis, A.P. and J.S. Crowe, Gene, 101: 297-302 (1991)).
Nucleic acids referred to herein as "recombinant" are nucleic acids which have been produced by recombinant DNA methodology, including those nucleic acids that are generated by procedures which rely upon a method of artificial recombination, such as the polymerase chain reaction (PCR) and/or cloning into a vector using restriction enzymes.
In certain embodiments, the isolated and/or recombinant nucleic acid comprises a nucleotide sequence encoding a ligand, as described herein, wherein said ligand comprises an amino acid sequence that has at least about 80%, at least about 85%, at least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 94%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99% amino acid sequence identity with the amino acid sequence of a dAb that binds VEGF disclosed herein, or a dAb that binds EGFR disclosed herein.
For example, in some embodiments, the isolated and/or recombinant nucleic acid comprises a nucleotide sequence encoding a ligand that has binding specificity for VEGF, as described herein, wherein said ligand comprises an amino acid sequence that has at least about 80%, at least about 85%, at least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 94%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99% amino acid sequence identity with the amino acid sequence of a dAb selected . from the group consisting of TAR15-1 (SEQ ID NOrIOO), TAR15-3 (SEQ ID NO:101), TAR15-4 (SEQ ID NO: 102), TAR15-9 (SEQ ID NO:103), TAR15-10 (SEQ ID NO:104), TARl 5-11 (SEQ ID NO:105), TAR15-12 (SEQ ID NO:106), TAR15-13 (SEQ ID NO:107), TAR15-14 (SEQ ID NO: 108), TAR15-15 (SEQ ID NO:109), TAR15-16 (SEQ ID NO:110), TAR15-17 (SEQ ID NO:111), TAR15-18 (SEQ ID NO:112), TAR15-19 (SEQ ID NO:113), TAR15-20 (SEQ ID NO:114), TAR 15-22 (SEQ ID NO: 115), TARl 5-5 (SEQ ID NO: 116), TARl 5-6 (SEQ ID NO:117), TAR15-7 (SEQ ID NO:118), TAR15-8 (SEQ ID NO:119), TAR15-23 (SEQ ID NO:120), TAR15-24 (SEQ ID NO:121), TAR15-25 (SEQ ID NO:122), TAR15-26 (SEQ ID NO:123), TAR15-27 (SEQ ID NO: 124), TAR15-29 (SEQ ID NO:125), TAR15-30 (SEQ ID NO:126), TAR15-6-500 (SEQ ID NO: 127), TAR15- 6-501 (SEQ ID NO:128), TAR15-6-502 (SEQ ID NO:129), TAR15-6-503 (SEQ ID NO:130), TAR15-6-504 (SEQ ID NO:131), TAR15-6-505 (SEQ ID NO:132), TARl 5-6-506 (SEQ ID NO: 133), TARl 5-6-507 (SEQ ID NO: 134), TARl 5-6-508 (SEQ ID NO:135), TAR15-6-509 (SEQ ID NO:136), TAR15-6-510 (SEQ ID NO:137), TAR15-8-500 (SEQ ID NO:138), TAR15-8-501 (SEQ ID NO:139), TAR15-8-502 (SEQ ID NO: 140), TAR15-8-503 (SEQ ID NO:141), TAR15-8-505 (SEQ ID NO:142), TAR15-8-506 (SEQ ID NO:143), TAR15-8-507 (SEQ ID NO:144), TAR15-8-508 (SEQ ID NO:145), TAR15-8-509 (SEQ ID NO:146), TAR15-8-510 (SEQ ID NO: 147), TAR15-8-511 (SEQ E) NO: 148), TAR15-26-500 (SEQ ID NO: 149), TARl 5-26-501 (SEQ ID NO: 150), TARl 5-26-502 (SEQ ID NO:151), TARl 5-26-503 (SEQ ID NO: 152), TARl 5-26-504 (SEQ ID NO: 153), TARl 5-26-505 (SEQ ID NO: 154), TARl 5-26-506 (SEQ ID NO: 155), TARl 5-26- 507 (SEQ ID NO: 156), TARl 5-26-508 (SEQ ID NO: 157), TARl 5-26-509 (SEQ ID NO: 158), TARl 5-26-510 (SEQ ID NO: 159), TARl 5-26-511 (SEQ ID NO: 160), TAR15-26-512 (SEQ ID NO:161), TAR15-26-513 (SEQ ID NO: 162), TAR15-26- 514 (SEQ ID NO: 163), TARl 5-26-515 (SEQ ID NO: 164), TARl 5-26-516 (SEQ ID NO:165), TAR15-26-517 (SEQ E) NO: 166), TAR15-26-518 (SEQ ID NO: 167), TARl 5-26-519 (SEQ ID NO: 168), TARl 5-26-520 (SEQ ID NO: 169), TARl 5-26- 521 (SEQ ID NO:170), TAR15-26-522 (SEQ ID NO:171), TAR15-26-523 (SEQ ID NO: 172), TARl 5-26-524 (SEQ ID NO: 173), TARl 5-26-525 (SEQ ID NO: 174), TARl 5-26-526 (SEQ ID NO: 175), TARl 5-26-527 (SEQ ID NO: 176), TARl 5-26- 528 (SEQ ID NO:177), TAR15-26-529 (SEQ ID NO:178), TAR15-26-530 (SEQ ID NO: 179), TARl 5-26-531 (SEQ ID NO: 180), TARl 5-26-532 (SEQ ID NO: 181), TARl 5-26-533 (SEQ ID NO: 182), TARl 5-26-534 (SEQ ID NO: 183), TARl 5-26- 535 (SEQ ID NO:184), TAR15-26-536 (SEQ ID NO:185), TAR15-26-537 (SEQ ID NO: 186), TARl 5-26-538 (SEQ ID NO: 187), TARl 5-26-539 (SEQ ID NO: 188), TARl 5-26-540 (SEQ ID NO: 189), TARl 5-26-541 (SEQ ID NO: 190), TARl 5-26- 542 (SEQ ID NO:191), TAR15-26-543 (SEQ ID NO: 192), TAR15-26-544 (SEQ ID NO: 193), TARl 5-26-545 (SEQ ID NO: 194), TARl 5-26-546 (SEQ ID NO: 195), TARl 5-26-547 (SEQ ID NO: 196), TARl 5-26-548 (SEQ ID NO: 197), TARl 5-26- 549 (SEQ ID NO: 198), TARl 5-26-550 (SEQ ID NO:539), and TARl 5-26-551 (SEQ ID NO:540).
For example, in some embodiments, the isolated and/or recombinant nucleic acid comprises a nucleotide sequence encoding a ligand that has binding specificity for VEGFA, as described herein, wherein said ligand comprises an amino acid sequence that has at least about 80%, at least about 85%, at least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 94%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99% amino acid sequence identity with the amino acid sequence of SEQ ID NO:705 (TARl 5-26-555).
In other embodiments, the isolated and/or recombinant nucleic acid comprises a nucleotide sequence encoding a ligand that has binding specificity for EGFR, as described herein, wherein said ligand comprises an amino acid sequence that has at least about 80%, at least about 85%, at least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 94%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99% amino acid sequence identity with the amino acid sequence of a dAb selected from the group consisting of DOMl 6-17 (SEQ ID NO:325), DOMl 6-18 (SEQ ID
NO:326), DOMl 6-19 (SEQ ID NO:327), DOMl 6-20 (SEQ ID NO:328), DOMl 6- 21 (SEQ ID NO:329), DOM16-22 (SEQ ID NO:330), DOM16-23 (SEQ ID
NO:331), DOM16-24 (SEQ ID NO:332), DOM16-25 (SEQ ID NO:333), D0M16- 26 (SEQ ID NO:334), DOMl 6-27 (SEQ ID NO:335), DOMl 6-28 (SEQ ID
NO:336), DOM16-29 (SEQ ID NO:337), DOM16-30 (SEQ ID NO:338), DOMl 6- 31 (SEQ ID NO:339), DOM16-32 (SEQ ID NO:340), DOM16-33 (SEQ ID
NO:341), DOM16-35 (SEQ ID NO:342), DOM16-37 (SEQ ID NO:343), D0M16- 38 (SEQ ID NO:344), DOM16-39 (SEQ ID NO:345), DOM16-40 (SEQ ID
NO:346), DOMl 6-41 (SEQ ID NO:347), DOMl 6-42 (SEQ ID NO:348), DOMl 6- 43 (SEQ ID NO:349), DOMl 6-44 (SEQ ID NO:350), DOMl 6-45 (SEQ ID
NO:351), DOM16-46 (SEQ ID NO:352), DOM16-47 (SEQ ID NO:353), D0M16- 48 (SEQ ID NO:354), DOMl 6-49 (SEQ ID NO:355), DOMl 6-50 (SEQ ID NO:356), DOM16-59 (SEQ ID NO:357), DOM16-60 (SEQ ID NO:358), DOM16- 61 (SEQ ID NO:359), DOM16-62 (SEQ ID NO:360), DOM16-63 (SEQ ID
NO:361), DOM16-64 (SEQ ID NO:362), DOM16-65 (SEQ ID NO:363), DOM16- 66 (SEQ ID NO:364), DOMl 6-67 (SEQ ID NO:365), DOMl 6-68 (SEQ ID
NO:366), DOMl 6-69 (SEQ ID NO:367), DOMl 6-70 (SEQ ID NO:368), DOM16- 71 (SEQ ID NO:369), DOM16-72 (SEQ ID NO:370), DOM16-73 (SEQ ID
NO:371), DOM16-74 (SEQ ID NO:372), DOM16-75 (SEQ ID NO:373), DOM16- 76 (SEQ ID NO:374), DOMl 6-77 (SEQ ID NO:375), DOMl 6-78 (SEQ ID
NO:376), DOM16-79 (SEQ ID NO:377), DOM16-80 (SEQ ID NO:378), DOM16- 81 (SEQ ID NO:379), DOM16-82 (SEQ ID NO:380), DOM16-83 (SEQ ID
NO:381), DOM16-84 (SEQ ID NO:382), DOM16-85 (SEQ ID NO:383), DOM16- 87 (SEQ ID NO:384), DOM16-88 (SEQ ID NO:385), DOM16-89 (SEQ ID
NO:386), DOM16-90 (SEQ ID NO:387), DOM16-91 (SEQ ID NO:388), DOM16- 92 (SEQ ID NO:389), DOM16-94 (SEQ ID NO:390), DOM16-95 (SEQ ID
NO:391), DOM16-96 (SEQ ID NO:392), DOM16-97 (SEQ ID NO:393), DOM16- 98 (SEQ ID NO:394), DOMl 6-99 (SEQ ID NO:395), DOMl 6-100 (SEQ ID NO:396), DOM16-101 (SEQ ID NO:397), DOM16-102 (SEQ ID NO:398), DOM16-103 (SEQ ID NO:399), DOM16-104 (SEQ ID NO:400), DOM16-105 (SEQ ID NO:401), DOM16-106 (SEQ ID NO:402), DOM16-107 (SEQ ID
NO:403), DOM16-108 (SEQ ID NO:404), DOM16-109 (SEQ ID NO:405),
DOM16-110 (SEQ ID NO:406), DOMl 6-111 (SEQ ID NO:407), DOMl 6-112 (SEQ ID NO:408), DOMl 6-113 (SEQ ID NO:409), DOMl 6-114 (SEQ ID
NO:410), DOM16-115 (SEQ ID NO:411), DOM16-116 (SEQ ID NO:412), DOM16-117 (SEQ ID NO:413), DOM16-118 (SEQ ID NO:414), DOM16-119 (SEQ ID NO:415), DOM16-39-6 (SEQ ID NO:416), DOM16-39-8 (SEQ ID NO:417), DOMl 6-39-34 (SEQ ID NO:418), DOMl 6-39-48 (SEQ ID NO:419), DOM16-39-87 (SEQ ID NO:420), DOM16-39-90 (SEQ ID NO:421), DOM16-39- 96 (SEQ ID NO:422), DOM16-39-100 (SEQ ID NO:423), DOM16-39-101 (SEQ ID NO:424), DOM16-39-102 (SEQ ID NO:425), DOM16-39-103 (SEQ ID NO:426), DOM16-39-104 (SEQ ID NO:427), DOM16-39-105 (SEQ ID NO:428), DOM16- 39-106 (SEQ ID NO:429), DOM16-39-107 (SEQ ID NO:430), DOM16-39-108 (SEQ ID NO:431), DOM16-39-109 (SEQ ID NO:432), DOM16-39-110 (SEQ ID NO:433), DOM16-39-111 (SEQ ID NO:434), DOM16-39-112 (SEQ ID NO:435), DOM16-39-113 (SEQ ID NO:436), DOM16-39-114 (SEQ ID NO:437), DOM16- 39-115 (SEQ ID NO:438), DOM16-39-116 (SEQ ID NO:439), DOM16-39-117 (SEQ ID NO:440), DOMl 6-39-200 (SEQ ID NO:441), DOMl 6-39-201 (SEQ ID NO:442), DOMl 6-39-202 (SEQ ID NO:443), DOMl 6-39-203 (SEQ ID NO:444), DOM16-39-204 (SEQ ID NO:445), DOM16-39-205 (SEQ ID NO:446), DOM16- 39-206 (SEQ ID NO:447), DOMl 6-39-207 (SEQ ID NO:448), DOMl 6-39-209 (SEQ ID NO:449), DOM16-52 (SEQ ID NO:450), NBl (SEQ ID NO:451), NB2 (SEQ ID NO:452), NB3 (SEQ ID NO:453), NB4 (SEQ ID NO:454), NB5 (SEQ ID NO:455), NB6 (SEQ ID NO:456), NB7 (SEQ ID NO:457), NB8 (SEQ ID NO:458), NB9 (SEQ ID NO:459), NBlO (SEQ ID NO:460), NBl 1 (SEQ ID NO:461), NB12 (SEQ ID NO:462), NB 13 (SEQ ID NO:463), NB 14 (SEQ ID NO:464), NB 15 (SEQ ID NO:465), NB16 (SEQ ID NO:466), NB17 (SEQ ID NO:467), NBl 8 (SEQ ID NO:468), NB19 (SEQ ID NO:469), NB20 (SEQ ID NO:470), NB21 (SEQ ID NO:471), and NB22 (SEQ ID NO:472).
In other embodiments, the isolated and/or recombinant nucleic acid comprises a nucleotide sequence encoding a ligand that has binding specificity for EGFR, as described herein, wherein said ligand comprises an amino acid sequence that has at least about 80%, at least about 85%, at least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 94%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99% amino acid sequence identity with an amino acid sequence selected from the group consisting of SEQ ID NOS:623-703, 727 and 728.
In other embodiments, the isolated and/or recombinant nucleic acid encoding a ligand that has binding specificity for VEGF, as described herein, comprises a nucleotide sequence that has at least about 80%, at least about 85%, at least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 94%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99% nucleotide sequence identity with a nucleotide sequence encoding an anti-VEGF dAb selected from the group consisting of TARl 5-1 (SEQ ID NO: 1), TAR15-3 (SEQ ID NO:2), TAR15-4 (SEQ ID NO:3), TAR15-9 (SEQ ID NO:4), TARl 5-10 (SEQ ID NO:5), TARl 5-11 (SEQ ID NO:6), TARl 5-12 (SEQ ID NO:7), TAR15-13 (SEQ ID NO:8), TAR15-14 (SEQ ID NO:9), TAR15-15 (SEQ ID NO:10), TAR15-16 (SEQ ID NO:11), TAR15-17 (SEQ ID NO:12), TAR15-18 (SEQ ID NO:13), TAR15-19 (SEQ ID NO:14), TAR15-20 (SEQ ID NO:15), TAR 15-22 (SEQ ID NO: 16), TARl 5-5 (SEQ ID NO: 17), TARl 5-6 (SEQ ID NO: 18), TARl 5-7 (SEQ ID NO: 19), TARl 5-8 (SEQ ID NO:20), TARl 5-23 (SEQ ID NO:21), TAR15-24 (SEQ ID NO:22), TAR15-25 (SEQ ID NO:23), TAR15-26 (SEQ ID NO:24), TARl 5-27 (SEQ ID NO:25), TARl 5-29 (SEQ ID NO:26), TAR15-30 (SEQ ID NO:27), TAR15-6-500 (SEQ ID NO:28), TAR15-6-501 (SEQ ID NO:29), TARl 5-6-502 (SEQ ID NO:30), TARl 5-6-503 (SEQ ID NO:31), TARl 5-6-504 (SEQ ID NO:32), TARl 5-6-505 (SEQ ID NO:33), TARl 5-6-506
(SEQ ID NO:34), TARl 5-6-507 (SEQ ID NO:35), TARl 5-6-508 (SEQ ID NO:36), TAR15-6-509 (SEQ ID NO:37), TAR15-6-510 (SEQ ID NO:38), TAR15-8-500 (SEQ ID NO:39), TAR15-8-501 (SEQ ID NO:40), TAR15-8-502 (SEQ ID NO:41), TARl 5-8-503 (SEQ ID NO:42), TARl 5-8-505 (SEQ ID NO:43), TARl 5-8-506 (SEQ ID NO:44), TARl 5-8-507 (SEQ ID NO:45), TARl 5-8-508 (SEQ ID NO:46), TARl 5-8-509 (SEQ ID NO:47), Rl 5-8-510 (SEQ ID NO:48), TARl 5-8-511 (SEQ ID NO:49), TAR15-26-500 (SEQ ID NO:50), TAR15-26-501 (SEQ ID NO:51), TARl 5-26-502 (SEQ ID NO:52), TARl 5-26-503 (SEQ ID NO:53), TARl 5-26-504 (SEQ ID NO:54), TARl 5-26-505 (SEQ ID NO:55), TARl 5-26-506 (SEQ ID NO:56), TAR15-26-507 (SEQ ID NO:57), TAR15-26-508 (SEQ ID NO:58),
TARl 5-26-509 (SEQ ID NO:59), TARl 5-26-510 (SEQ ID NO:60), TARl 5-26-511 (SEQ ID NO:61), TAR15-26-512 (SEQ ID NO:62), TAR15-26-513 (SEQ ID NO:63), TARl 5-26-514 (SEQ ID NO:64), TARl 5-26-515 (SEQ ID NO:65), TARl 5-26-516 (SEQ ID NO:66), TARl 5-26-517 (SEQ ID NO:67), TARl 5-26-518 (SEQ ID NO:68), TARl 5-26-519 (SEQ ID NO:69), TARl 5-26-520 (SEQ ID NO:70), TAR15-26-521 (SEQ ID NO:71), TAR15-26-522 (SEQ ID NO:72), TARl 5-26-523 (SEQ ID NO:73), TARl 5-26-524 (SEQ ID NO:74), TARl 5-26-525 (SEQ ID NO:75), TARl 5-26-526 (SEQ ID NO:76), TARl 5-26-527 (SEQ ID NO:77), TARl 5-26-528 (SEQ ID NO:78), TARl 5-26-529 (SEQ ID NO:79), TAR15-26-530 (SEQ ID NO:80), TAR15-26-531 (SEQ ID NO:81), TAR15-26-532 (SEQ ID NO:82), TAR15-26-533 (SEQ ID NO:83), TAR15-26-534 (SEQ ID NO:84), TARl 5-26-535 (SEQ ID NO:85), TARl 5-26-536 (SEQ ID NO:86), TAR15-26-537 (SEQ ID NO:87), TAR15-26-538 (SEQ ID NO:88), TAR15-26-539 (SEQ ID NO:89), TARl 5-26-540 (SEQ ID NO:90), TARl 5-26-541 (SEQ ID NO:91), TAR15-26-542 (SEQ ID NO:92), TAR15-26-543 (SEQ ID NO:93), TARl 5-26-544 (SEQ ID NO:94), TARl 5-26-545 (SEQ ID NO:95), TARl 5-26-546 (SEQ ID NO:96), TARl 5-26-547 (SEQ ID NO:97), TARl 5-26-548 (SEQ ID
NO:98), TARl 5-26-549 (SEQ ID NO:99), TARl 5-21 (SEQ ID NO:535), TARl 5-2 (SEQ ID NO:536), TAR15-26-550 (SEQ ID NO:537), and TAR15-26-551 (SEQ ID NO:538). Preferably, nucleotide sequence identity is determined over the whole length of the nucleotide sequence that encodes the selected anti-VEGF dAb.
In other embodiments, the isolated and/or recombinant nucleic acid encoding a ligand that has binding specificity for VEGF, as described herein, comprises a nucleotide sequence that has at least about 80%, at least about 85%, at least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 94%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99% nucleotide sequence identity with a nucleotide sequence encoding TARl 5-26-555 (SEQ ID NO:705).
In other embodiments, the isolated and/or recombinant nucleic acid encoding a ligand that has binding specificity for EGFR, as described herein, comprises a nucleotide sequence that has at least about 80%, at least about 85%, at least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 94%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99% nucleotide sequence identity with a nucleotide sequence encoding an anti-VEGF dAb selected from the group consisting of DOM16-17 (SEQ ID NO:199), DOM16-18 (SEQ ID NO:200), DOM16-19 (SEQ ID NO:201), D0M16- 20 (SEQ ID NO:202), DOMl 6-21 (SEQ ID NO:203), DOMl 6-22 (SEQ ID
NO:204), DOMl 6-23 (SEQ ID NO:205), DOMl 6-24 (SEQ ID NO:206), DOMl 6- 25 (SEQ ID NO:207), DOMl 6-26 (SEQ ID NO:208), DOMl 6-27 (SEQ ID
NO:209), DOM16-28 (SEQ ID NO:210), DOM16-29 (SEQ ID N0:211), D0M16- 30 (SEQ ID NO:212), DOM16-31 (SEQ ID NO:213), DOM16-32 (SEQ ID
NO:214), DOM16-33 (SEQ ID NO:215), DOM16-35 (SEQ ID NO:216), D0M16- 37 (SEQ ID NO:217), DOM16-38 (SEQ ID NO:218), DOM16-39 (SEQ ID
NO:219), DOM16-40 (SEQ ID NO:220), DOM16-41 (SEQ ID NO:221), D0M16- 42 (SEQ ID NO:222), DOMl 6-43 (SEQ ID NO:223), DOMl 6-44 (SEQ ID NO:224), DOMl 6-45 (SEQ ID NO:225), DOMl 6-46 (SEQ ID NO:226), DOMl 6- 47 (SEQ ID NO:227), DOMl 6-48 (SEQ ID NO:228), DOMl 6-49 (SEQ ID NO:229), DOM16-50 (SEQ ID NO:230), DOM16-59 (SEQ ID NO:231), DOM16- 60 (SEQ ID NO:232), DOMl 6-61 (SEQ ID NO:233), DOMl 6-62 (SEQ ID
NO:234), DOMl 6-63 (SEQ ID NO:235), DOMl 6-64 (SEQ ID NO:236), DOMl 6- 65 (SEQ ID NO:237), DOMl 6-66 (SEQ ID NO:238), DOMl 6-67 (SEQ ID NO:239), DOM16-68 (SEQ ID NO:240), DOM16-69 (SEQ ID NO:241), D0M16- 70 (SEQ ID NO:242), DOM16-71 (SEQ ID NO:243), DOM16-72 (SEQ ID NO:244), DOMl 6-73 (SEQ ID NO:245), DOMl 6-74 (SEQ ID NO:246), DOMl 6- 75 (SEQ ID NO:247), DOMl 6-76 (SEQ ID NO:248), DOMl 6-77 (SEQ ID NO:249), DOM16-78 (SEQ ID NO:250), DOM16-79 (SEQ ID NO:251), DOM16- 80 (SEQ ID NO:252), DOMl 6-81 (SEQ ID NO:253), DOMl 6-82 (SEQ ID NO:254), DOMl 6-83 (SEQ ID NO:255), DOMl 6-84 (SEQ ID NO:256), DOMl 6- 85 (SEQ ID NO:257), DOMl 6-87 (SEQ ID NO:258), DOMl 6-88 (SEQ ID
NO:259), DOM16-89 (SEQ ID NO:260), DOM16-90 (SEQ ID NO:261), D0M16- 91 (SEQ ID NO:262), DOMl 6-92 (SEQ ID NO:263), DOMl 6-94 (SEQ ID NO:264), DOMl 6-95 (SEQ ID NO:265), DOMl 6-96 (SEQ ID NO:266), DOMl 6- 97 (SEQ ID NO:267), DOMl 6-98 (SEQ ID NO:268), DOMl 6-99 (SEQ ID NO:269), DOM16-100 (SEQ ID NO:270), DOM16-101 (SEQ ID NO:271),
DOM16-102 (SEQ ID NO:272), DOM16-103 (SEQ ID NO:273), DOM16-104 (SEQ ID NO:274), DOMl 6-105 (SEQ ID NO:275), DOMl 6-106 (SEQ ID
NO:276), DOM16-107 (SEQ ID NO:277), DOM16-108 (SEQ ID NO:278), DOM16-109 (SEQ ID NO:279), DOM16-110 (SEQ ID NO:280), D0M16-111 (SEQ ID NO:281), D0M16-112 (SEQ ID NO:282), DOM16-113 (SEQ ID
NO:283), DOM16-114 (SEQ ID NO:284), DOM16-115 (SEQ ID NO:285), DOMl 6-116 (SEQ ID NO:286), DOM16-117 (SEQ ID NO:287), DOMl 6-118 (SEQ ID NO:288), D0M16-119 (SEQ ID NO:289), DOM16-39-6 (SEQ ID NO:290), DOM16-39-8 (SEQ ID NO:291), DOM16-39-34 (SEQ ID NO:292), DOMl 6-39-48 (SEQ ID NO:293), DOMl 6-39-87 (SEQ ID NO:294), DOMl 6-39- 90 (SEQ ID NO:295), DOM16-39-96 (SEQ ID NO:296), DOM16-39-100 (SEQ ID NO:297), DOMl 6-39-101 (SEQ ID NO:298), DOMl 6-39-102 (SEQ ID NO:299), DOMl 6-39-103 (SEQ ID NO:300), DOMl 6-39-104 (SEQ ID NO:301), DOMl 6- 39-105 (SEQ ID NO:302), DOM16-39-106 (SEQ ID NO:303), DOM16-39-107 (SEQ ID NO:304), DOM16-39-108 (SEQ ID NO:305), DOM16-39-109 (SEQ ID NO:306), DOM16-39-110 (SEQ ID NO:307), DOM16-39-111 (SEQ ID NO:308), DOM16-39-112 (SEQ ID NO:309), DOMl 6-39-113 (SEQ ID NO:310), DOMl 6- 39-114 (SEQ ID NO:311), DOM16-39-115 (SEQ ID NO:312), DOM16-39-116 (SEQ ID NO:313), DOM16-39-117 (SEQ ID NO:314), DOM16-39-200 (SEQ ID NO:315), DOMl 6-39-201 (SEQ ID NO:316), DOMl 6-39-202 (SEQ ID NO:317), DOMl 6-39-203 (SEQ ID NO:318), DOMl 6-39-204 (SEQ ID NO:319), DOMl 6- 39-205 (SEQ ID NO:320), DOM16-39-206 (SEQ ID NO:321), DOM16-39-207 (SEQ ID NO:322), DOMl 6-39-209 (SEQ ID NO:323), and DOMl 6-52 (SEQ ID NO:324). Preferably, nucleotide sequence identity is determined over the whole length of the nucleotide sequence that encodes the selected anti-EGFR dAb.
In other embodiments, the isolated and/or recombinant nucleic acid encoding a ligand that has binding specificity for EGFR, as described herein, comprises a nucleotide sequence that has at least about 80%, at least about 85%, at least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 94%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99% nucleotide sequence identity with a nucleotide sequence encoding an anti-EGFR dAb selected from the group consisting of SEQ ID NOS:623-703, 727 and 728.
The invention also provides a vector comprising a recombinant nucleic acid molecule of the invention. In certain embodiments, the vector is an expression vector comprising one or more expression control elements or sequences that are operably linked to the recombinant nucleic acid of the invention. The invention also provides a recombinant host cell comprising a recombinant nucleic acid molecule or vector of the invention. Suitable vectors (e.g., plasmids, phagmids), expression control elements, host cells and methods for producing recombinant host cells of the invention are well-known in the art, and examples are further described herein.
Suitable expression vectors can contain a number of components, for example, an origin of replication, a selectable marker gene, one or more expression control elements, such as a transcription control element (e.g., promoter, enhancer, terminator) and/or one or more translation signals, a signal sequence or leader sequence, and the like. Expression control elements and a signal sequence, if present, can be provided by the vector or other source. For example, the
transcriptional and/or translational control sequences of a cloned nucleic acid encoding an antibody chain can be used to direct expression.
A promoter can be provided for expression in a desired host cell. Promoters can be constitutive or inducible. For example, a promoter can be operably linked to a nucleic acid encoding an antibody, antibody chain or portion thereof, such that it directs transcription of the nucleic acid. A variety of suitable promoters for prokaryotic (e.g., lac, tac, T3, T7 promoters for E. coli) and eukaryotic {e.g., simian virus 40 early or late promoter, Rous sarcoma virus long terminal repeat promoter, cytomegalovirus promoter, adenovirus late promoter) hosts are available.
In addition, expression vectors typically comprise a selectable marker for selection of host cells carrying the vector, and, in the case of a replicable expression vector, an origin or replication. Genes encoding products which confer antibiotic or drug resistance are common selectable markers and may be used in procaryotic (e.g. , lactamase gene (ampicillin resistance), Tet gene for tetracycline resistance) and eucaryotic cells (e.g., neomycin (G418 or geneticin), gpt (mycophenolic acid), ampicillin, or hygromycin resistance genes). Dihydrofolate reductase marker genes permit selection with methotrexate in a variety of hosts. Genes encoding the gene product of auxotrophic markers of the host (e.g., LEU2, URA3, HIS3) are often used as selectable markers in yeast. Use of viral (e.g., baculovirus) or phage vectors, and vectors which are capable of integrating into the genome of the host cell, such as retroviral vectors, are also contemplated. Suitable expression vectors for expression in mammalian cells and prokaryotic cells (E. coli), insect cells (Drosophila
Schnieder S2 cells, Sf9) and yeast (P. methanolica, P. pastoris, S. cerevisiae) are well-known in the art.
Suitable host cells can be prokaryotic, including bacterial cells such as E. coli, B. subtilis and/or other suitable bacteria; eukaryotic cells, such as fungal or yeast cells (e.g. , Pichia pastoris, Aspergillus sp. , Saccharomyces cerevisiae,
Schizosaccharomyces pombe, Neurospora crassa), or other lower eukaryotic cells, and cells of higher eukaryotes such as those from insects (e.g., Drosophila Schnieder S2 cells, Sf9 insect cells (WO 94/26087 (O'Connor)), mammals (e.g., COS cells, such as COS-I (ATCC Accession No. CRL-1650) and COS-7 (ATCC Accession No. CRL-1651), CHO (e.g., ATCC Accession No. CRL-9096, CHO DG44 (Urlaub, G. and Chasin, LA., Proc. Natl. Acac. ScL USA, 77(7) :4216-4220 (1980))), 293 (ATCC Accession No. CRL-1573), HeLa (ATCC Accession No. CCL-2), CVl (ATCC Accession No. CCL-70), WOP (Dailey, L., et al, J. Virol, 54:139-149 (1985), 3T3, 293T (Pear, W. S., et al, Proc. Natl Acad. Sci. U.S.A., 90:8392-8396 (1993)) NSO cells, SP2/0, HuT 78 cells and the like, or plants (e.g., tobacco). (See, for example, Ausubel, F.M. et al, eds. Current Protocols in Molecular Biology, Greene Publishing Associates and John Wiley & Sons hie. (1993).) In some embodiments, the host cell is an isolated host cell and is not part of a multicellular organism (e.g., plant or animal), hi preferred embodiments, the host cell is a non- human host cell.
The invention also provides a method for producing a ligand (e.g., dual- specific ligand, multispecific ligand) of the invention, comprising maintaining a recombinant host cell comprising a recombinant nucleic acid of the invention under conditions suitable for expression of the recombinant nucleic acid, whereby the recombinant nucleic acid is expressed and a ligand is produced. In some
embodiments, the method further comprises isolating the ligand.
Preparation of Immunoglobulin Based Ligands
Ligands (e.g., dual specific ligands, multispecific ligands) according to the invention can be prepared according to previously established techniques, used in the field of antibody engineering, for the preparation of scFv, "phage" antibodies and other engineered antibody molecules. Techniques for the preparation of antibodies are for example described in the following reviews and the references cited therein: Winter & Milstein, (1991) Nature 349:293-299; Pluckthun (1992) Immunological Reviews 13 0:151-188; Wright et al, (1992) CHt. Rev. Immunol 12:125-168; Holliger, P. & Winter, G. (1993) Curr. Opin. Biotechnol. 4, 446-449; Carter, et al (1995) J. Hematother. 4, 463-470; Chester, K.A. & Hawkins, R.E.
(1995) Trends Biotechnol. 13, 294-300; Hoogenboom, H.R. (1997) Nat. Biotechnol. 15, 125-126; Fearon, D. (1997) Nat. Biotechnol. 15, 618-619; Pluckthun, A. & Pack, P. (1997) lmmunotechnology 3, 83-105; Carter, P. & Merchant, A.M. (1997) Curr. Opin. Biotechnol. 8, 449-454; Holliger, P. & Winter, G. (1997) Cancer Immunol. Immunother. 45,128-130.
Suitable techniques employed for selection of antibody variable domains with a desired specificity employ libraries and selection procedures which are known in the art. Natural libraries (Marks et al (1991) J. MoI. Biol, 222: 581 ;
Vaughan et al. (1996) Nature Biotech., 14: 309) which use rearranged V genes harvested from human B cells are well known to those skilled in the art. Synthetic libraries (Hoogenboom & Winter (1992) J. MoI. Biol, 227: 381; Barbas et al. (1992) Proc. Natl. Acad. ScL USA, 89: 4457; Nissim et al. (1994) EMBO J, 13: 692;
Griffiths et al. (1994) EMBO J, 13: 3245; De Kruif et al. (1995) J. MoI. Biol, 248: 97) are prepared by cloning immunoglobulin V genes, usually using PCR. Errors in the PCR process can lead to a high degree of randomisation. Vjj and/or VL libraries may be selected against target antigens or epitopes separately, in which case single domain binding is directly selected for, or together.
Library vector systems
A variety of selection systems are known in the art which are suitable for use in the present invention. Examples of such systems are described below.
Bacteriophage lambda expression systems may be screened directly as bacteriophage plaques or as colonies of lysogens, both as previously described (Huse et al. (1989) Science, 246: 1275; Caton and Koprowski (1990) Proc. Natl Acad. ScL U.S.A., 87; Mullinax et al. (1990) Proc. Natl. Acad. ScL U.S.A., 87: 8095; Persson et al. (1991) Proc. Natl. Acad. ScL U.S.A., 88: 2432) and are of use in the invention. Whilst such expression systems can be used to screen up to 106 different members of a library, they are not really suited to screening of larger numbers (greater than 106 members). Of particular use in the construction of libraries are selection display systems, which enable a nucleic acid to be linked to the
polypeptide it expresses. As used herein, a selection display system is a system that permits the selection, by suitable display means, of the individual members of the library by binding the generic and/or target.
Selection protocols for isolating desired members of large libraries are known in the art, as typified by phage display techniques. Such systems, in which diverse peptide sequences are displayed on the surface of filamentous bacteriophage (Scott and Smith (1990) Science, 249: 386), have proven useful for creating libraries of antibody fragments (and the nucleotide sequences that encode them) for the in vitro selection and amplification of specific antibody fragments that bind a target antigen (McCafferty et al, WO 92/01047). The nucleotide sequences encoding the variable regions are linked to gene fragments which encode leader signals that direct them to the periplasmic space of E. coli and as a result the resultant antibody fragments are displayed on the surface of the bacteriophage, typically as fusions to bacteriophage coat proteins {e.g., pill or pVIII). Alternatively, antibody fragments are displayed externally on lambda phage capsids (phagebodies). An advantage of phage-based display systems is that, because they are biological systems, selected library members can be amplified simply by growing the phage containing the selected library member in bacterial cells. Furthermore, since the nucleotide sequence that encodes the polypeptide library member is contained on a phage or phagemid vector, sequencing, expression and subsequent genetic manipulation is relatively straightforward.
Methods for the construction of bacteriophage antibody display libraries and lambda phage expression libraries are well known in the art (McCafferty et al (1990) Nature, 348: 552; Rang et al (1991) Proc. Natl. Acad. Sci. U.S.A., 88: 4363; Clackson et al. (1991) Nature, 352: 624; Lowman et al. (1991) Biochemistry, 30:
10832; Burton et al. (1991) Proc. Natl. Acad. Sci U.S.A., 88: 10134; Hoogenboom et al. (1991) Nucleic Acids Res., 19: 4133; Chang et al. (1991) J. Immunol, 147:
3610; Breitling et al. (1991) Gene, 104: 147; Marks et al. (1991) supra; Barbas et al. (1992) supra; Hawkins and Winter (1992) J. Immunol, 22: 867; Marks et al, 1992, J. Biol. Chem., 267: 16007; Lerner et al (1992) Science, 258: 1313, incorporated herein by reference).
One particularly advantageous approach has been the use of scFv phage- libraries (Huston et al, 1988, Proc. Natl. Acad. Sci U.S.A., 85: 5879-5883;
Chaudhary et al (1990) Proc. Natl. Acad. Sci U.S.A., 87: 1066-1070; McCafferty et al. (1990) supra; Clackson et al (1991) Nature, 352: 624; Marks et al (1991) J.
MoI. Biol, 222: 581; Chiswell et al (1992) Trends Biotechnol, 10: 80; Marks et al (1992) J. Biol. Chem., 267). Various embodiments of scFv libraries displayed on bacteriophage coat proteins have been described. Refinements of phage display approaches are also known, for example as described in WO96/06213 and
WO92/01047 (Medical Research Council et al.) and WO97/08320 (Morphosys), which are incorporated herein by reference.
Other systems for generating libraries of polypeptides involve the use of cell- free enzymatic machinery for the in vitro synthesis of the library members, hi one method, RNA molecules are selected by alternate rounds of selection against a target and PCR amplification (Tuerk and Gold (1990) Science, 249: 505; Ellington and Szostak (1990) Nature, 346: 818). A similar technique may be used to identify DNA sequences which bind a predetermined human transcription factor (Thiesen and
Bach (1990) Nucleic 4cids Res., 18: 3203; Beaudry and Joyce (1992) Science, 257: 635; WO92/05258 and WO92/14843). In a similar way, in vitro translation can be used to synthesise polypeptides as a method for generating large libraries. These methods which generally comprise stabilised polysome complexes, are described further in WO88/08453, WO90/05785, WO90/07003, WO91/02076, WO91/05058, and WO92/02536. Alternative display systems which are not phage-based, such as those disclosed in WO95/22625 and WO95/11922 (Affymax) use the polysomes to display polypeptides for selection.
A still further category of techniques involves the selection of repertoires in artificial compartments, which allow the linkage of a gene with its gene product. For example, a selection system in which nucleic acids encoding desirable gene products may be selected in microcapsules formed by water-in-oil emulsions is described in WO99/02671, WO00/40712 and Tawfik & Griffiths (1998) Nature Biotechnol 16(7), 652-6. Genetic elements encoding a gene product having a desired activity are compartmentalised into microcapsules and then transcribed and/or translated to produce their respective gene products (RNA or protein) within the microcapsules. Genetic elements which produce gene product having desired activity are subsequently sorted. This approach selects gene products of interest by detecting the desired activity by a variety of means. Library Construction
Libraries intended for selection, may be constructed using techniques known in the art, for example as set forth above, or may be purchased from commercial sources. Libraries which are useful in the present invention are described, for example, in WO99/20749. Once a vector system is chosen and one or more nucleic acid sequences encoding polypeptides of interest are cloned into the library vector, one may generate diversity within the cloned molecules by undertaking mutagenesis prior to expression; alternatively, the encoded proteins may be expressed and selected, as described above, before mutagenesis and additional rounds of selection are performed. Mutagenesis of nucleic acid sequences encoding structurally optimized polypeptides is carried out by standard molecular methods. Of particular use is the polymerase chain reaction, or PCR, (Mullis and Faloona (1987) Methods EnzymoL, 155: 335, herein incorporated by reference). PCR, which uses multiple cycles of DNA replication catalyzed by a thermostable, DNA-dependent DNA polymerase to amplify the target sequence of interest, is well known in the art. The construction of various antibody libraries has been discussed in Winter et al. (1994) Ann. Rev. Immunology 12, 433-55, and references cited therein.
PCR is performed using template DNA (at least lfg; more usefully, 1-1000 ng) and at least 25 pmol of oligonucleotide primers; it maybe advantageous to use a larger amount of primer when the primer pool is heavily heterogeneous, as each sequence is represented by only a small fraction of the molecules of the pool, and amounts become limiting in the later amplification cycles. A typical reaction mixture includes: 2μl of DNA, 25 pmol of oligonucleotide primer, 2.5 μl of 1OX PCR buffer 1 (Perkin-Elmer, Foster City, CA), 0.4 μl of 1.25 μM dNTP, 0.15 μl (or 2.5 units) of Taq DNA polymerase (Perkin Elmer, Foster City, CA) and deionized water to a total volume of 25 μl. Mineral oil is overlaid and the PCR is performed using a programmable thermal cycler. The length and temperature of each step of a PCR cycle, as well as the number of cycles, is adjusted in accordance to the stringency requirements in effect. Annealing temperature and timing are determined both by the efficiency with which a primer is expected to anneal to a template and the degree of mismatch that is to be tolerated; obviously, when nucleic acid molecules are simultaneously amplified and mutagenised, mismatch is required, at least in the first round of synthesis. The ability to optimise the stringency of primer annealing conditions is well within the knowledge of one of moderate skill in the art. An annealing temperature of between 30 °C and 72 0C is used. Initial denaturation of the template molecules normally occurs at between 920C and 990C for 4 minutes, followed by 20-40 cycles consisting of denaturation (94-990C for 15 seconds to 1 minute), annealing (temperature determined as discussed above; 1 -2 minutes), and extension (720C for 1-5 minutes, depending on the length of the amplified product). Final extension is generally for 4 minutes at 720C, and may be followed by an indefinite (0-24 hour) step at 40C.
Combining Single Variable Domains
Domains useful in the invention, once selected, may be combined by a variety of methods known in the art, including covalent and non-covalent methods. Preferred methods include the use of polypeptide linkers, as described, for example, in connection with scFv molecules (Bird et al , (1988) Science 2A2-AΥh-A2€).
Discussion of suitable linkers is provided in Bird et al. Science 242, 423-426;
Hudson et al , Journal Immunol Methods 231 (1999) 177-189; Hudson et al, Proc Nat Acad Sci USA 85, 5879-5883. Linkers are preferably flexible, allowing the two single domains to interact. One linker example is a (GIy4 Ser)n linker, where n=l to 8, e.g., 2, 3, 4, 5 or 7. The linkers used in diabodies, which are less flexible, may also be employed (Holliger et al, (1993) Proc Nat Acad Sci USA 90:6444-6448). In one embodiment, the linker employed is not an immunoglobulin hinge region.
Variable domains may be combined using methods other than linkers. For example, the use of disulphide bridges, provided through naturally-occurring or engineered cysteine residues, may be exploited to stabilize VH~VH>VL'VL or VH-VL dimers (Reiter et al., (1994) Protein Eng. 7:697-704) or by remodelling the interface between the variable domains to improve the "fit" and thus the stability of interaction (Ridgeway et al, (1996) Protein Eng. 7:617-621; Zhu et al, (1997) Protein Science 6:781-788). Other techniques for joining or stabilizing variable domains of immunoglobulins, and in particular antibody VH domains, may be employed as appropriate. Characterisation of Ligands
The binding of a dual-specific ligand to the cell or the binding of each binding domain to each specific target can be tested by methods which will be familiar to those skilled in the art and include ELISA. In a preferred embodiment of the invention binding is tested using monoclonal phage ELISA. Phage ELISA may be performed according to any suitable procedure: an exemplary protocol is set forth below.
Populations of phage produced at each round of selection can be screened for binding by ELISA to the selected antigen or epitope, to identify "polyclonal" phage antibodies. Phage from single infected bacterial colonies from these populations can then be screened by ELISA to identify "monoclonal" phage antibodies. It is also desirable to screen soluble antibody fragments for binding to antigen or epitope, and this can also be undertaken by ELISA using reagents, for example, against a C- or N-terminal tag (see for example Winter et al. (1994) Ann. Rev. Immunology 12, 433- 55 and references cited therein.
The diversity of the selected phage monoclonal antibodies may also be assessed by gel electrophoresis of PCR products (Marks et al. 1991, supra; Nissim et al. 1994 supra), probing (Tomlinson et al, 1992) J. MoI. Biol. 227, 776) or by sequencing of the vector DNA.
Structure of Ligands
In the case that each variable domain is selected from V-gene repertoires, selected for instance using phage display technology as herein described, then these variable domains comprise a universal framework region, such that they may be recognized by a specific generic dual-specific ligand as herein defined. The use of universal frameworks, generic ligands and the like is described in WO99/20749.
Where V-gene repertoires are used variation in polypeptide sequence is preferably located within the structural loops of the variable domains. The polypeptide sequences of either variable domain may be altered by DNA shuffling or by mutation in order to enhance the interaction of each variable domain with its complementary pair. DNA shuffling is known in the art and taught, for example, by Stemmer, 1994, Nature 370: 389-391 and U.S. Patent No. 6,297,053, both of which are incorporated herein by reference. Other methods of mutagenesis are well known to those of skill in the art.
In general, nucleic acid molecules and vector constructs required for selection, preparation and formatting dual-specific ligands may be constructed and manipulated as set forth in standard laboratory manuals, such as Sambrook et at. (1989) Molecular Cloning: A Laboratory Manual, Cold Spring Harbor,. USA.
The manipulation of nucleic acids useful in the present invention is typically carried out in recombinant vectors. As used herein, vector refers to a discrete element that is used to introduce heterologous DNA into cells for the expression and/or replication thereof. Methods by which to select or construct and,
subsequently, use such vectors are well known to one of ordinary skill in the art. Numerous vectors are publicly available, including bacterial plasmids,
bacteriophage, artificial chromosomes and episomal vectors. Such vectors may be used for simple cloning and mutagenesis; alternatively a gene expression vector is employed. A vector of use according to the invention may be selected to
accommodate a polypeptide coding sequence of a desired size, typically from 0.25 kilobase (kb) to 40 kb or more in length A suitable host cell is transformed with the vector after in vitro cloning manipulations. Each vector contains various functional components, which generally include a cloning (or "polylinker") site, an origin of replication and at least one selectable marker gene. If the given vector is an expression vector, it additionally possesses one or more of the following: enhancer element, promoter, transcription termination and signal sequences, each positioned in the vicinity of the cloning site, such that they are operatively linked to the gene encoding a dual-specific ligand according to the invention.
Both cloning and expression vectors generally contain nucleic acid sequences that enable the vector to replicate in one or more selected host cells. Typically in cloning vectors, this sequence is one that enables the vector to replicate independently of the host chromosomal DNA and includes origins of replication or autonomously replicating sequences. Such sequences are well known for a variety of bacteria, yeast and viruses. The origin of replication from the plasmid pBR322 is suitable for most Gram-negative bacteria, the 2 micron plasmid origin is suitable for yeast, and various viral origins (e.g. SV 40, adenovirus) are useful for cloning vectors in mammalian cells. Generally, the origin of replication is not needed for mammalian expression vectors unless these are used in mammalian cells able to replicate high levels of DNA, such as COS cells.
Advantageously, a cloning or expression vector may contain a selection gene also referred to as a selectable marker. This gene encodes a protein necessary for the survival or growth of transformed host cells grown in a selective culture medium. Host cells not transformed with the vector containing the selection gene will therefore not survive in the culture medium. Typical selection genes encode proteins that confer resistance to antibiotics and other toxins, e.g. ampicillin, neomycin, methotrexate or tetracycline, complement auxotrophic deficiencies, or supply critical nutrients not available in the growth media.
Since the replication of vectors encoding a dual-specific ligand according to the present invention is most conveniently performed in E. coli, an E. co/z-selectable marker, for example, the β-lactamase gene that confers resistance to the antibiotic ampicillin, is of use. These can be obtained from E. coli plasmids, such as pBR322 or a pUC plasmid such as pUCl 8 or pUCl 9.
Expression vectors usually contain a promoter that is recognised by the host organism and is operably linked to the coding sequence of interest. Such a promoter may be inducible or constitutive. The term "operably linked" refers to a
juxtaposition wherein the components described are in a relationship permitting them to function in their intended manner. A control sequence "operably linked" to a coding sequence is ligated in such a way that expression of the coding sequence is achieved under conditions compatible with the control sequences.
Promoters suitable for use with prokaryotic hosts include, for example, the β- lactamase and lactose promoter systems, alkaline phosphatase, the tryptophan (trp) promoter system and hybrid promoters such as the tac promoter. Promoters for use in bacterial systems will also generally contain a Shine-Delgarno sequence operably linked to the coding sequence.
The preferred vectors are expression vectors that enables the expression of a nucleotide sequence corresponding to a polypeptide library member. Thus, selection with the first and/or second antigen or epitope can be performed by separate propagation and expression of a single clone expressing the polypeptide library member or by use of any selection display system. As described above, the preferred selection display system is bacteriophage display. Thus, phage or phagemid vectors may be used, (e.g., pITl or pIT2. Leader sequences useful in the invention include pelB, stll, ompA, phoA, bla and pelA. One example are phagemid vectors which have an E. coli. origin of replication (for double stranded replication) and also a phage origin of replication (for production of single-stranded DNA). The manipulation and expression of such vectors is well known in the art (Hoogenboom and Winter (1992) supra; Nissim et al. (1994) supra). Briefly, the vector contains a β-lactamase gene to confer selectivity on the phagemid and a lac promoter upstream of a expression cassette that consists (N to C terminal) of a pelB leader sequence (which directs the expressed polypeptide to the periplasmic space), a multiple cloning site (for cloning the nucleotide version of the library member), optionally, one or more peptide tag (for detection), optionally, one or more TAG stop codon and the phage protein pill. Thus, using various suppressor and non-suppressor strains of E. coli and with the addition of glucose, iso-propyl thio-β-D-galactoside (IPTG) or a helper phage, such as VCS M13, the vector is able to replicate as a plasmid with no expression, produce large quantities of the polypeptide library member only or produce phage, some of which contain at least one copy of the polypeptide-pIII fusion on their surface.
Construction of vectors encoding dual-specific ligands according to the invention employs conventional ligation techniques. Isolated vectors or DNA fragments are cleaved, tailored, and religated in the form desired to generate the required vector. If desired, analysis to confirm that the correct sequences are present in the constructed vector can be performed in a known fashion. Suitable methods for constructing expression vectors, preparing in vitro transcripts, introducing DNA into host cells, and performing analyses for assessing expression and function are known to those skilled in the art. The presence of a gene sequence in a sample is detected, or its amplification and/or expression quantified by conventional methods, such as Southern or Northern analysis, Western blotting, dot blotting of DNA, RNA or protein, in situ hybridisation, immunocytochemistry or sequence analysis of nucleic acid or protein molecules. Those skilled in the art will readily envisage how these methods may be modified, if desired. Skeletons
Skeletons may be based on immunoglobulin molecules or may be non- immunoglobulin in origin as set forth above. Each domain of the dual-specific ligand may be a different skeleton. Preferred immunoglobulin skeletons as herein defined includes any one or more of those selected from the following: an immunoglobulin molecule comprising at least (i) the CL (kappa or lambda subclass) domain of an antibody; or (ii) the CHl domain of an antibody heavy chain; an immunoglobulin molecule comprising the CHl and CH2 domains of an antibody heavy chain; an immunoglobulin molecule comprising the CHl, CH2 and CH3 domains of an antibody heavy chain; or any of the subset (ii) in conjunction with the CL (kappa or lambda subclass) domain of an antibody. A hinge region domain may also be included. Such combinations of domains may, for example, mimic natural antibodies, such as IgG or IgM, or fragments thereof, such as Fv, scFv, Fab or F(ab')2 molecules. Those skilled in the art will be aware that this list is not intended to be exhaustive.
Protein Scaffolds
Each binding domain comprises a protein scaffold and one or more CDRs which are involved in the specific interaction of the domain with one or more epitopes. Advantageously, an epitope binding domain according to the present invention comprises three CDRs. Suitable protein scaffolds include any of those selected from the group consisting of the following: those based on immunoglobulin domains, those based on fibronectin, those based on affibodies, those based on CTLA4, those based on chaperones such as GroEL, those based on lipocallin and those based on the bacterial Fc receptors SpA and SpD. Those skilled in the art will appreciate that this list is not intended to be exhaustive.
Scaffolds for Use in Constructing Ligands
Selection of the Main-chain Conformation
The members of the immunoglobulin superfamily all share a similar fold for their polypeptide chain. For example, although antibodies are highly diverse in terms of their primary sequence, comparison of sequences and crystallographic structures has revealed that, contrary to expectation, five of the six antigen binding loops of antibodies (Hl, H2, Ll, L2, L3) adopt a limited number of main-chain
conformations, or canonical structures (Chothia and Lesk (1987) J. MoI. Biol, 196: 901 ; Chothia et al (1989) Nature, 342: 877). Analysis of loop lengths and key residues has therefore enabled prediction of the main-chain conformations of Hl, H2, Ll, L2 and L3 found in the majority of human antibodies (Chothia et al (1992) J. MoI. Biol, 227: 799; Tomlinson et al. (1995) EMBO J, 14: 4628; Williams et al (1996) J. MoI Biol, 264: 220). Although the H3 region is much more diverse in terms of sequence, length and structure (due to the use of D segments), it also forms a limited number of main-chain conformations for short loop lengths which depend on the length and the presence of particular residues, or types of residue, at key positions in the loop and the antibody framework (Martin et al. (1996) J. MoI Biol, 263: 800; Shirai et al. (1996) FEBS Letters, 399: 1).
Libraries of ligands and/or binding domains can be designed in which certain loop lengths and key residues have been chosen to ensure that the main-chain conformation of the members is known. Advantageously, these are real
conformations of immunoglobulin superfamily molecules found in nature, to minimize the chances that they are non-functional, as discussed above. Germline V gene segments serve as one suitable basic framework for constructing antibody or T- cell receptor libraries; other sequences are also of use. Variations may occur at a low frequency, such that a small number of functional members may possess an altered main-chain conformation, which does not affect its function.
Canonical structure theory is also of use to assess the number of different main-chain conformations encoded by ligands, to predict the main-chain
conformation based on dual-specific ligand sequences and to choose residues for diversification which do not affect the canonical structure. It is known that, in the human Vκ domain, the Ll loop can adopt one of four canonical structures, the L2 loop has a single canonical structure and that 90% of human Vκ domains adopt one of four or five canonical structures for the L3 loop (Tomlinson et al. (1995) supra); thus, in the Vκ domain alone, different canonical structures can combine to create a range of different main-chain conformations. Given that the Vx domain encodes a different range of canonical structures for the Ll, L2 and L3 loops and that Vκ and Vλ domains can pair with any VH domain which can encode several canonical structures for the Hl and H2 loops, the number of canonical structure combinations observed for these five loops is very large. This implies that the generation of diversity in the main-chain conformation may be essential for the production of a wide range of binding specificities. However, by constructing an antibody library based on a single known main-chain conformation it has been found, contrary to expectation, that diversity in the main-chain conformation is not required to generate sufficient diversity to target substantially all antigens. Even more surprisingly, the single main-chain conformation need not be a consensus structure - a single naturally occurring conformation can be used as the basis for an entire library. Thus, in a preferred aspect, the ligands of the invention possess a single known main-chain conformation.
The single main-chain conformation that is chosen is preferably
commonplace among molecules of the immunoglobulin superfamily type in question. A conformation is commonplace when a significant number of naturally occurring molecules are observed to adopt it. Accordingly, in a preferred aspect of the invention, the natural occurrence of the different main-chain conformations for each binding loop of an immunoglobulin domain are considered and then a naturally occurring variable domain is chosen which possesses the desired combination of main-chain conformations for the different loops. If none is available, the nearest equivalent may be chosen. It is preferable that the desired combination of main- chain conformations for the different loops is created by selecting germline gene segments which encode the desired main-chain conformations. It is more preferable, that the selected germline gene segments are frequently expressed in nature, and most preferable that they are the most frequently expressed of all natural germline gene segments.
In designing ligands (e.g., ds-dAbs) or libraries thereof the incidence of the different main-chain conformations for each of the six antigen binding loops may be considered separately. For Hl, H2, Ll, L2 and L3, a given conformation that is adopted by between 20% and 100% of the antigen binding loops of naturally occurring molecules is chosen. Typically, its observed incidence is above 35% (i.e. between 35% and 100%) and, ideally, above 50% or even above 65%. Since the vast majority of H3 loops do not have canonical structures, it is preferable to select a main-chain conformation which is commonplace among those loops which do display canonical structures. For each of the loops, the conformation which is observed most often in the natural repertoire is therefore selected. In human antibodies, the most popular canonical structures (CS) for each loop are as follows: Hl - CS 1 (79% of the expressed repertoire), H2 - CS 3 (46%), Ll - CS 2 of Vκ (39%), L2 - CS 1 (100%), L3 - CS 1 of Vκ (36%) (calculation assumes a κ:λ ratio of 70:30, Hood et al. (1967) Cold Spring Harbor Symp. Quant. Biol, 48: 133). For H3 loops that have canonical structures, a CDR3 length (Kabat et al. (1991) Sequences of proteins of immunological interest, U.S. Department of Health and Human Services) of seven residues with a salt-bridge from residue 94 to residue 101 appears to be the most common. There are at least 16 human antibody sequences in the EMBL data library with the required H3 length and key residues to form this conformation and at least two crystallographic structures in the protein data bank which can be used as a basis for antibody modelling (2cgr and ltet). The most frequently expressed germline gene segments that this combination of canonical structures are the VH segment 3-23 (DP-47), the JH segment JH4b, the Vκ segment 02/012 (DPK9) and the Jκ segment Jκl. VH segments DP45 and DP38 are also suitable. These segments can therefore be used in combination as a basis to construct a library with the desired single main-chain conformation.
Alternatively, instead of choosing the single main-chain conformation based on the natural occurrence of the different main-chain conformations for each of the binding loops in isolation, the natural occurrence of combinations of main-chain conformations is used as the basis for choosing the single main-chain conformation. In the case of antibodies, for example, the natural occurrence of canonical structure combinations for any two, three, four, five or for all six of the antigen binding loops can be determined. Here, it is preferable that the chosen conformation is
commonplace in naturally occurring antibodies and most preferable that it is observed most frequently in the natural repertoire. Thus, in human antibodies, for example, when natural combinations of the five antigen binding loops, Hl, H2, Ll, L2 and L3, are considered, the most frequent combination of canonical structures is determined and then combined with the most popular conformation for the H3 loop, as a basis for choosing the single main-chain conformation.
Diversification of the Canonical Sequence
Having selected several known main-chain conformations or, preferably a single known main-chain conformation, dual-specific ligands (e.g., ds-dAbs) or libraries for use in the invention can be constructed by varying each binding site of the molecule in order to generate a repertoire with structural and/or functional diversity. This means that variants are generated such that they possess sufficient diversity in their structure and/or in their function so that they are capable of providing a range of activities.
The desired diversity is typically generated by varying the selected molecule at one or more positions. The positions to be changed can be chosen at random or are preferably selected. The variation can then be achieved either by randomisation, during which the resident amino acid is replaced by any amino acid or analogue thereof, natural or synthetic, producing a very large number of variants or by replacing the resident amino acid with one or more of a defined subset of amino acids, producing a more limited number of variants.
Various methods have been reported for introducing such diversity. Error- prone PCR (Hawkins et al. (1992) J. MoI. Biol, 226: 889), chemical mutagenesis (Deng et al. (1994) J. Biol. Chem., 269: 9533) or bacterial mutator strains (Low et al. (1996) J. MoI. Biol, 260: 359) can be used to introduce random mutations into the genes that encode the molecule. Methods for mutating selected positions are also well known in the art and include the use of mismatched oligonucleotides or degenerate oligonucleotides, with or without the use of PCR. For example, several synthetic antibody libraries have been created by targeting mutations to the antigen binding loops. The H3 region of a human tetanus toxoid-binding Fab has been randomised to create a range of new binding specificities (Barbas et al (1992) Proc. Natl. Acad. ScL USA, 89: 4457). Random or semi-random H3 and L3 regions have been appended to germline V gene segments to produce large libraries with unmutated framework regions (Hoogenboom & Winter (1992) J. MoI. Biol, 227: 381; Barbas et al (1992) Proc. Natl Acad. Sci. USA, 89: 4457; Nissim et al (1994) EMBO J., 13: 692; Griffiths et al. (1994) EMBO J., 13: 3245; De Kruif et al. (1995) J. MoI. Biol, 248: 97). Such diversification has been extended to include some or all of the other antigen binding loops (Crameri et al. (1996) Nature Med., 2: 100;
Riechmann et al. (1995) Bio/Technology, 13: 475; Morphosys, WO97/08320, supra).
Since loop randomization has the potential to create approximately more than 1015 structures for H3 alone and a similarly large number of variants for the other five loops, it is not feasible using current transformation technology or even by using cell free systems to produce a library representing all possible combinations. For example, in one of the largest libraries constructed to date, 6 x 101 different antibodies, which is only a fraction of the potential diversity for a library of this design, were generated (Griffiths et al. (1994) supra).
Preferably, only the residues that are directly involved in creating or modifying the desired function of each domain of the dual-specific ligand molecule are diversified. For many molecules, the function of each domain will be to bind a target and therefore diversity should be concentrated in the target binding site, while avoiding changing residues which are crucial to the overall packing of the molecule or to maintaining the chosen main-chain conformation. Diversification of the Canonical Sequence as it Applies to Antibody Domains
In the case of antibody based ligands {e.g., ds-dAbs), the binding site for each target is most often the antigen binding site. Thus, preferably only those residues in the antigen binding site are varied. These residues are extremely diverse in the human antibody repertoire and are known to make contacts in high-resolution antibody/antigen complexes. For example, in L2 it is known that positions 50 and 53 are diverse in naturally occurring antibodies and are observed to make contact with the antigen. In contrast, the conventional approach would have been to diversify all the residues in the corresponding Complementarity Determining Region (CDRl) as defined by Kabat et al. (1991, supra), some seven residues compared to the two diversified in the library for use according to the invention. This represents a significant improvement in terms of the functional diversity required to create a range of antigen binding specificities. In nature, antibody diversity is the result of two processes: somatic recombination of germline V, D and J gene segments to create a naive primary repertoire (so called germline and junctional diversity) and somatic hypermutation of the resulting rearranged V genes. Analysis of human antibody sequences has shown that diversity in the primary repertoire is focused at the centre of the antigen binding site whereas somatic hypermutation spreads diversity to regions at the periphery of the antigen binding site that are highly conserved in the primary repertoire (see Tomlinson et al. (1996) J. MoI. Biol, 256: 813). This
complementarity has probably evolved as an efficient strategy for searching sequence space and, although apparently unique to antibodies, it can easily be applied to other polypeptide repertoires. The residues which are varied are a subset of those that form the binding site for the target. Different (including overlapping) subsets of residues in the target binding site are diversified at different stages during selection, if desired.
In the case of an antibody repertoire, an initial 'naive' repertoire can be created where some, but not all, of the residues in the antigen binding site are diversified. As used herein in this context, the term "naive" refers to antibody molecules that have no pre-determined target. These molecules resemble those which are encoded by the immunoglobulin genes of an individual who has not undergone immune diversification, as is the case with fetal and newborn individuals, whose immune systems have not yet been challenged by a wide variety of antigenic stimuli. This repertoire is then selected against a range of antigens or epitopes. If required, further diversity can then be introduced outside the region diversified in the initial repertoire. This matured repertoire can be selected for modified function, specificity or affinity.
Naive repertoires of binding domains for the construction of dual-specific ligands in which some or all of the residues in the antigen binding site are varied are known in the art. (See, WO 2004/058821, WO 2004/003019, and WO 03/002609). The "primary" library mimics the natural primary repertoire, with diversity restricted to residues at the centre of the antigen binding site that are diverse in the germline V gene segments (germline diversity) or diversified during the recombination process (junctional diversity). Those residues which are diversified include, but are not limited to, H50, H52, H52a, H53, H55, H56, H58, H95, H96, H97, H98, L50, L53, L91, L92, L93, L94 and L96. In the "somatic" library, diversity is restricted to residues that are diversified during the recombination process (junctional diversity) or are highly somatically mutated. Those residues which are diversified include, but are not limited to: H31, H33, H35, H95, H96, H97, H98, L30, L31, L32, L34 and L96. All the residues listed above as suitable for diversification in these libraries are known to make contacts in one or more antibody-antigen complexes. Since in both libraries, not all of the residues in the antigen binding site are varied, additional diversity is incorporated during selection by varying the remaining residues, if it is desired to do so. It shall be apparent to one skilled in the art that any subset of any of these residues (or additional residues which comprise the antigen binding site) can be used for the initial and/or subsequent diversification of the antigen binding site.
In the construction of libraries for use in the invention, diversification of chosen positions is typically achieved at the nucleic acid level, by altering the coding sequence which specifies the sequence of the polypeptide such that a number of possible amino acids (all 20 or a subset thereof) can be incorporated at that position. Using the IUPAC nomenclature, the most versatile codon is NNK, which encodes all amino acids as well as the TAG stop codon. The NNK codon is preferably used in order to introduce the required diversity. Other codons which achieve the same ends are also of use, including the NNN codon, which leads to the production of the additional stop codons TGA and TAA.
A feature of side-chain diversity in the antigen binding site of human antibodies is a pronounced bias which favors certain amino acid residues. If the amino acid composition of the ten most diverse positions in each of the VH, VK and V^ regions are summed, more than 76% of the side-chain diversity comes from only seven different residues, these being, serine (24%), tyrosine (14%), asparagine (11%), glycine (9%), alanine (7%), aspartate (6%) and threonine (6%). This bias towards hydrophilic residues and small residues which can provide main-chain flexibility probably reflects the evolution of surfaces which are predisposed to binding a wide range of antigens or epitopes and may help to explain the required promiscuity of antibodies in the primary repertoire. Since it is preferable to mimic this distribution of amino acids, the distribution of amino acids at the positions to be varied preferably mimics that seen in the antigen binding site of antibodies. Such bias in the substitution of amino acids that permits selection of certain polypeptides (not just antibody polypeptides) against a range of target antigens is easily applied to any polypeptide repertoire. There are various methods for biasing the amino acid distribution at the position to be varied (including the use of tri-nucleotide mutagenesis, see WO97/08320), of which the preferred method, due to ease of synthesis, is the use of conventional degenerate codons. By comparing the amino acid profile encoded by all
combinations of degenerate codons (with single, double, triple and quadruple degeneracy in equal ratios at each position) with the natural amino acid use it is possible to calculate the most representative codon. The codons (AGT)(AGC)T, (AGT)(AGC)C and (AGT)(AGC)(CT) - that is, DVT, DVC and DVY, respectively using IUPAC nomenclature - are those closest to the desired amino acid profile: they encode 22% serine and 11% tyrosine, asparagine, glycine, alanine, aspartate, threonine and cysteine. Preferably, therefore, libraries are constructed using either the DVT, DVC or DVY codon at each of the diversified positions.
Therapeutic and diagnostic compositions and uses
The invention provides compositions comprising the ligands of the invention and a pharmaceutically acceptable carrier, diluent or excipient, and therapeutic and diagnostic methods that employ the ligands or compositions of the invention. The ligands according to the method of the present invention may be employed in in vivo therapeutic and prophylactic applications, in vivo diagnostic applications and the like.
Therapeutic and prophylactic uses of ligands of the invention involve the administration of ligands according to the invention to a recipient mammal, such as a human. The ligands bind to targets with high affinity and/or avidity. In some embodiments, such as IgG-like ligands, the ligands can allow recruitment of cytotoxic cells to mediate killing of camcer cells, for example by antibody dependent cellular cytoxicity.
Substantially pure ligands of at least 90 to 95% homogeneity are preferred for administration to a mammal, and 98 to 99% or more homogeneity is most preferred for pharmaceutical uses, especially when the mammal is a human. Once purified, partially or to homogeneity as desired, the ligands may be used
diagnostically or therapeutically (including extracorporeally) or in developing and performing assay procedures, immunofluorescent stainings and the like (Lefkovite and Pernis, (1979 and 1981) Immunological Methods, Volumes I and II, Academic Press, NY).
For example, the ligands of the present invention will typically find use in preventing, suppressing or treating disease states. For example, ligands can be administered to treat, suppress or prevent a chronic inflammatory disease, allergic hypersensitivity, cancer, bacterial or viral infection, autoimmune disorders (which include, but are not limited to, Type I diabetes, asthma, multiple sclerosis, rheumatoid arthritis, juvenile rheumatoid arthritis, psoriatic arthritis,
spondylarthropathy {e.g., ankylosing spondylitis), systemic lupus erythematosus, inflammatory bowel disease {e.g., Crohn's disease, ulcerative colitis), myasthenia gravis and Behcet's syndrome, psoriasis, endometriosis, and abdominal adhesions {e.g., post abdominal surgery).
The ligands are useful for treating infectious diseases in which cells infected with an infectious agent contain higer levels of cell surface EGFR than uninfected cells or that contain one or more cell surface targets that are not present on ininfected cells, such as a protein that is encoded by the infectious agent {e.g., bacteria, virus).
Ligands according to the invention that are able to bind to EGFR can be internalized by cells that express EGFR {e.g., endocytosed), and can deliver therapeutic agents {e.g., a toxin) intracellular^ {e.g., deliver a dAb that binds an intracellular target). In addition, ligands provide a means by which a binding domain {e.g., a dAb monomer) that is specifically able to bind to an intracellular target can be delivered to an intracellular environment. This strategy requires, for example, a binding domain with physical properties that enable it to remain functional inside the cell. Alternatively, if the final destination intracellular compartment is oxidising, a well folding ligand may not need to be disulphide free.
In the instant application, the term "prevention" involves administration of the protective composition prior to the induction of the disease. "Suppression" refers to administration of the composition after an inductive event, but prior to the clinical appearance of the disease. "Treatment" involves administration of the protective composition after disease symptoms become manifest. Treatment includes ameliorating symptoms associated with the disease, and also preventing or delaying the onset of the disease and also lessening the severity or frequency of symptoms of the disease.
The term "cancer" refers to the pathological condition in mammals that is typically characterized by dysregulated cellular proliferation or survival. Examples of cancer include, but are not limited to, carcinoma, lymphoma, blastoma, sarcoma, and leukemia and lymphoid malignancies. More particular examples of cancers include squamous cell cancer (e.g. epithelial squamous cell cancer), lung cancer (e.g., small-cell lung carcinoma, non-small cell lung carcinoma, adenocarcinoma of the lung, squamous carcinoma of the lung), cancer of the peritoneum, hepatocellular cancer, gastric or stomach cancer including gastrointestinal cancer, pancreatic cancer, glioblastoma, cervical cancer, ovarian cancer, liver cancer, bladder cancer, gall bladder cancer, hepatoma, breast cancer, colon cancer, rectal cancer, colorectal cancer, multiple myeloma, chronic myelogenous leukemia, acute myelogenous leukemia, endometrial or uterine carcinoma, salivary gland carcinoma, kidney or renal cancer, prostate cancer, vulval cancer, thyroid cancer, hepatic carcinoma, anal carcinoma, penile carcinoma, head and neck cancer, and the like. Cancers characterized by expression of EGFR on the surface of cancerous cells (EGFR- expressing cancers) include, for example, bladder cancer, ovarian cancer, colorectal cancer, breast cancer, lung cancer (e.g., non-small cell lung carcinoma), gastric cancer, pancreatic cancer, prostate cancer, head and neck cancer, renal cancer and gall bladder cancer.
Animal model systems which can be used to assess efficacy of the ligands of the inventon in preventing treating or suppressing disease (e.g., cancer) are available. Suitable models of cancer include, for example, xenograft and orthotopic models of human cancers in animal models, such as the SCID-hu myeloma model (Epstein J, and Yaccoby, S., Methods MoI Med. 773:183-90 (2005), Tassone P, et al, Clin Cancer Res. 11(11):4251-8 (2005)), mouse models of human lung cancer (e.g., Meuwissen R and Berns A, Genes Dev.l P(6):643-64 (2005)), and mouse models of metastatic cancers (e.g., Kubota T '., J Cell Biochem. 56(l):4-8 (1994)).
Generally, the present ligands will be utilized in purified form together with pharmacologically appropriate carriers. Typically, these carriers include aqueous or alcoholic/aqueous solutions, emulsions or suspensions, including saline and/or buffered media. Parenteral vehicles include sodium chloride solution, Ringer's dextrose, dextrose and sodium chloride and lactated Ringer's. Suitable
physiologically-acceptable adjuvants, if necessary to keep a polypeptide complex in suspension, may be chosen from thickeners such as carboxymethylcellulose, polyvinylpyrrolidone, gelatin and alginates.
Intravenous vehicles include fluid and nutrient replenishers and electrolyte replenishers, such as those based on Ringer's dextrose. Preservatives and other additives, such as antimicrobials, antioxidants, chelating agents and inert gases, may also be present (Mack (1982) Remington's Pharmaceutical Sciences, 16th Edition). A variety of suitable formulations can be used, including extended release formulations.
The ligands of the present invention may be used as separately administered compositions or in conjunction with other agents. The ligands can be administered and or formulated together with one or more additional therapeutic or active agents. When a ligand is administered with an additional therapeutic agent, the ligand can be administered before, simultaneously with or subsequent to administration of the additional agent. Generally, the ligand and additional agent are administered in a manner that provides an overlap of therapeutic effect. Additional agents that can be administered or formulated with the ligand of the invention include, for example, various immunotherapeutic drugs, such as cylcosporine, methotrexate, adriamycin or cisplatinum, antibiotics, antimycotics, anti-viral agents and immunotoxins. For example, when the antagonist is administered to prevent, suppress or treat lung inflammation or a respiratory disease, it can be administered in conjuction with phosphodiesterase inhibitors (e.g., inhibitors of phosphodiesterase 4),
bronchodilators (e.g., beta2-agonists, anticholinergerics, theophylline), short-acting beta-agonists (e.g., albuterol, salbutamol, bambuterol, fenoterol, isoetherine, isoproterenol, levalbuterol, metaproterenol, pirbuterol, terbutaline and tornlate), long-acting beta-agonists (e.g., formoterol and salmeterol), short acting
anticholinergics (e.g., ipratropium bromide and oxitropium bromide), long-acting anticholinergics (e.g., tiotropium), theophylline (e.g. short acting formulation, long acting formulation), inhaled steroids (e.g., beclomethasone, beclometasone, budesonide, fiunisolide, fluticasone propionate and triamcinolone), oral steroids (e.g., methylprednisolone, prednisolone, prednisolon and prednisone), combined short-acting beta-agonists with anticholinergics (e.g.,
albuterol/salbutamol/ipratopium, and fenoterol/ipratopium), combined long-acting beta-agonists with inhaled steroids (e.g., salmeterol/fluticasone, and
formoterol/budesonide) and mucolytic agents (e.g., erdosteine, acetylcysteine, bromheksin, carbocysteine, guiafenesin and iodinated glycerol).
The ligands of the invention can be coadministered (e.g., to treat cancer, an inflammatory disease or other disease) with a variety of suitable co-therapeutic agents, including cytokines, analgesics/antipyretics, antiemetics, and
chemotherapeutics. Further suitable co-therapeutic agents include
immunosuppressive agents selected from the group consisting of cyclosporine, azathioprine, mycophenolic acid, mycophenolate mofetil, corticosteroids, methotrexate, gold salts, sulfasalazine, antimalarials, brequinar, leflunomide, mizoribine, 15-deoxyspergualine, 6-mercaptopurine, cyclophosphamide, rapamycin, tacrolimus (FK-506), OKT3, and anti-thymocyte globulin, anti-inflammatory agents selected from the group consisting of aspirin, other salicylates, steroidal drugs, NSAIDs (nonsteroidal anti-inflammatory drugs), Cox-2 inhibitors, and DMARDs (disease modifying antirheumatic drugs); anti-psoriasis agents selected from the group consisting of coal tar, A vitamin, anthralin, calcipotrien, tarazotene, corticosteroids, methotrexate, retinoids, cyclosporine, etanercept, alefacept, efaluzimab, 6-thioguanine, mycophenolate mofetil, tacrolimus (FK-506), and hydroxyurea.
Cytokines include, without limitation, a lymphokine, tumor necrosis factors, tumor necrosis factor-like cytokine, lymphotoxin, interferon, macrophage inflammatory protein, granulocyte monocyte colony stimulating factor, interleukin (including, without limitation, interleukin- 1, interleukin-2, interleukin-6, interleukin- 12, interleukin-15, interleukin- 18), growth factors, which include, without limitation, {e.g., growth hormone, insulin-like growth factor 1 and 2 (IGF-I and IGF-2), granulocyte colony stimulating factor (GCSF), platelet derived growth factor (PGDF), epidermal growth factor (EGF), and agents for erythropoiesis stimulation, e.g., recombinant human erythropoietin (Epoetin alfa), EPO, a hormonal agonist, hormonal antagonists {e.g., flutamide, tamoxifen, leuprolide acetate
(LUPRON)), and steroids {e.g., dexamethasone, retinoid, betamethasone, Cortisol, cortisone, prednisone, dehydrotestosterone, glucocorticoid, mineralocorticoid, estrogen, testosterone, progestin).
Analgesics/antipyretics can include, without limitation, e.g., aspirin, acetaminophen, ibuprofen, naproxen sodium, buprenorphine hydrochloride, propoxyphene hydrochloride, propoxyphene napsylate, meperidine hydrochloride, hydromorphone hydrochloride, morphine sulfate, oxycodone hydrochloride, codeine phosphate, dihydrocodeine bitartrate, pentazocine hydrochloride, hydrocodone bitartrate, levorphanol tartrate, diflunisal, trolamine salicylate, nalbuphine hydrochloride, mefenamic acid, butorphanol tartrate, choline salicylate, butalbital, phenyltoloxamine citrate, diphenhydramine citrate, methotrimeprazine,
cinnamedrine hydrochloride, meprobamate, and the like.
Antiemetics can also be coadministered to prevent or treat nausea and vomiting e.g., suitable antiemetics include meclizine hydrochloride, nabilone, prochlorperazine, dimenhydrinate, promethazine hydrochloride, thiethylperazine, scopolamine, and the like.
Chemotherapeutic agents, as that term is used herein, include, but are not limited to, for example antimicrotubule agents, e.g., taxol (paclitaxel), taxotere (docetaxel); alkylating agents, e.g., cyclophosphamide, carmustine, lomustine, and chlorambucil; cytotoxic antibiotics, e.g., dactinomycin, doxorubicin, mitomycin-C, and bleomycin; antimetabolites, e.g., cytarabine, gemcitatin, methotrexate, and 5- fluorouracil; antimiotics, e.g., vincristine vinca alkaloids, e.g., etoposide, vinblastine, and vincristine; and others such as cisplatin, dacarbazine, procarbazine, and hydroxyurea; and combinations thereof.
The ligands of the invention can be used to treat cancer in combination with another therapeutic agent. For example, a ligand of the invention can be
administered in combination with a chemotherapeutic agent or an antineoplastic compositon comprising a (at least one) chemotherapeutic agent. Advantageously, in such a therapeutic approach, the amount of chemotherapeutic agent that must be administered to be effective can be reduced. Thus the invention provides a method of treating cancer comprising administering to a patient in need thereof a
therapeutically effective amount of a ligand of the invention and a chemotherapeutic agent, wherein the chemotherapeutic agent is administered at a low dose. Generally the amount of chemotherapeutic agent that is coadministered with a ligand of the invention is about 80%, or about 70%, or about 60%, or about 50%, or about 40%, or about 30%, or about 20%, or about 10% or less, of the dose of chemotherapeutic agent alone that is normally administered to a patient. Thus, cotherapy is particularly advantageous when the chemotherapeutic agent causes deleterious or undesirable side effects that may be reduced or eliminated at a lower doses.
Pharmaceutical compositions can include "cocktails" of various cytotoxic or other agents in conjunction with ligands of the present invention, or even combinations of ligands according to the present invention having different specificities, such as ligands selected using different target antigens or epitopes, whether or not they are pooled prior to administration.
The route of administration of pharmaceutical compositions according to the invention may be any suitable route, such as any of those commonly known to those of ordinary skill in the art. For therapy, including without limitation
immunotherapy, the ligands of the invention can be administered to any patient in accordance with standard techniques. The administration can be by any appropriate mode, including parenterally, intravenously, intramuscularly, intraperitoneally, transdermally, intrathecally, intraarticularly, via the pulmonary route, or also, appropriately, by direct infusion (e.g., with a catheter). The dosage and frequency of administration will depend on the age, sex and condition of the patient, concurrent administration of other drugs, counterindications and other parameters to be taken into account by the clinician. Administration can be local (e.g., local delivery to the lung by pulmonary administration,(e.g. , intranasal administration) or local injection directly into a tumor) or systemic as indicated.
The ligands of this invention can be lyophilised for storage and reconstituted in a suitable carrier prior to use. This technique has been shown to be effective with conventional immunoglobulins and art-known lyophilisation and reconstitution techniques can be employed. It will be appreciated by those skilled in the art that lyophilisation and reconstitution can lead to varying degrees of antibody activity loss (e.g. with conventional immunoglobulins, IgM antibodies tend to have greater activity loss than IgG antibodies) and that use levels may have to be adjusted upward to compensate.
The compositions containing the ligands can be administered for
prophylactic and/or therapeutic treatments. In certain therapeutic applications, an adequate amount to accomplish at least partial inhibition, suppression, modulation, killing, or some other measurable parameter, of a population of selected cells is defined as a "therapeutically-effective dose". Amounts needed to achieve this dosage will depend upon the severity of the disease and the general state of the patient's health, but generally range from 0.005 to 5.0 mg of ligand per kilogram of body weight, with doses of 0.05 to 2.0 mg/kg/dose being more commonly used. For prophylactic applications, compositions containing the present ligands or cocktails thereof may also be administered in similar or slightly lower dosages, to prevent, inhibit or delay onset of disease {e.g., to sustain remission or quiescence, or to prevent acute phase). The skilled clinician will be able to determine the appropriate dosing interval to treat, suppress or prevent disease. When a ligand is administered to treat, suppress or prevent a disease, it can be administered up to four times per day, twice weekly, once weekly, once every two weeks, once a month, or once every two months, at a dose of, for example, about 10 μg/kg to about 80 mg/kg, about 100 μg/kg to about 80 mg/kg, about 1 mg/kg to about 80 mg/kg, about 1 mg/kg to about 70 mg/kg, about 1 mg/kg to about 60 mg/kg, about 1 mg/kg to about 50 mg/kg, about 1 mg/kg to about 40 mg/kg, about 1 mg/kg to about 30 mg/kg, about 1 mg/kg to about 20 mg/kg , about 1 mg/kg to about 10 mg/kg, about 10 μg/kg to about 10 mg/kg, about 10 μg/kg to about 5 mg/kg, about 10 μg/kg to about 2.5 mg/kg, about 1 mg/kg, about 2 mg/kg, about 3 mg/kg, about 4 mg/kg, about 5 mg/kg, about 6 mg/kg, about 7 mg/kg, about 8 mg/kg, about 9 mg/kg or about 10 mg/kg. In particular embodiments, the dual-specific ligand is administered to treat, suppress or prevent a chronic inflammatory disease once every two weeks or once a month at a dose of about 10 μg/kg to about 10 mg/kg (e.g., about 10 μg/kg, about 100 μg/kg, about 1 mg/kg, about 2 mg/kg, about 3 mg/kg, about 4 mg/kg, about 5 mg/kg, about 6 mg/kg, about 7 mg/kg, about 8 mg/kg, about 9 mg/kg or about 10 mg/kg.)
In particular embodiments, the ligand of the invention is administered at a dose that provides saturation of EGFR or a desired serum concentration in vivo. The skilled physician can determine appropriate dosing to achieve saturation, for example by titrating ligand and monitoring the amount of free binding sites on EGFR expressing cells or the serum concentration of ligand. Therapeutic regiments that involve administering a therapeutic agent to achieve target saturation or a desired serum concentration of agent are common in the art, particularly in the field of oncology.
Treatment or therapy performed using the compositions described herein is considered "effective" if one or more symptoms are reduced {e.g., by at least 10% or at least one point on a clinical assessment scale), relative to such symptoms present before treatment, or relative to such symptoms in an individual (human or model animal) not treated with such composition or other suitable control. Symptoms will obviously vary depending upon the disease or disorder targeted, but can be measured by an ordinarily skilled clinician or technician. Such symptoms can be measured, for example, by monitoring the level of one or more biochemical indicators of the disease or disorder {e.g., levels of an enzyme or metabolite correlated with the disease, affected cell numbers, etc.), by monitoring physical manifestations {e.g., inflammation, tumor size, etc.), or by an accepted clinical assessment scale, for example, the Expanded Disability Status Scale (for multiple sclerosis), the Irvine Inflammatory Bowel Disease Questionnaire (32 point assessment evaluates quality of life with respect to bowel function, systemic symptoms, social function and emotional status - score ranges from 32 to 224, with higher scores indicating a better quality of life), the Quality of Life Rheumatoid Arthritis Scale, or other accepted clinical assessment scale as known in the field. A sustained {e.g., one day or more, preferably longer) reduction in disease or disorder symptoms by at least 10% or by one or more points on a given clinical scale is indicative of "effective" treatment. Similarly, prophylaxis performed using a composition as described herein is
"effective" if the onset or severity of one or more symptoms is delayed, reduced or abolished relative to such symptoms in a similar individual (human or animal model) not treated with the composition.
A composition containing ligands according to the present invention may be utilized in prophylactic and therapeutic settings to aid in the alteration, inactivation, killing or removal of a select target cell population in a mammal. In addition, the ligands and selected repertoires of polypeptides described herein may be used extracorporeally or in vitro selectively to kill, deplete or otherwise effectively remove a target cell population from a heterogeneous collection of cells. Blood from a mammal may be combined extracorporeally with the ligands, e.g. antibodies, cell- surface receptors or binding proteins thereof whereby the undesired cells are killed or otherwise removed from the blood for return to the mammal in accordance with standard techniques.
In one embodiment, the invention relates to a method for delivering anti- angiogenic therapy (anti-VEGF therapy) to a site containing cells that express or overexpress EGFR, comprising administereing an effective amount of a ligand that has binding specificity for VEGF and for EGFR to a subject in need thereof.
The invention also relates to use of a ligand that has binding specificity for VEGF and for EGFR for delivering anti-angiogenic therapy (anti-VEGF therapy) to a site containing cells that express or overexpress EGFR. The invention also relates to use of a ligand that has binding specificity for VEGF and for EGFR for the manufacture of a medicament for delivering anti-angiogenic therapy (anti-VEGF therapy) to a site containing cells that express or overexpress EGFR, or for inhibiting angiogenesis at a site containing cells that express of overexpress EGFR.
In particular embodiments, the invention relates to a method of treating cancer comprising administering to a subject in need thereof a therapeutically effective amount of a ligand, as described herein, that has binding specificity for VEGF and for EGFR. In particular embodiments, the patient has an EGFR- expressing cancer, such as, bladder cancer, ovarian cancer, colorectal cancer, breast cancer, lung cancer {e.g., non-small cell lung carcinoma), gastric cancer, pancreatic cancer, prostate cancer, head and neck cancer, renal cancer and gall bladder cancer.
In other embodiments, the invention relates to a method for treating cancer, comprising administering to a subject in need thereof a therapeutically effective amount of ligand, as described herein, {e.g., a ligand that has binding specificity for VEGF, a ligand that has binding specificity for EGFR, a ligand that has binding specificity for VEGF and EGFR) and an anti-neoplastic composition, wherein said anti-neoplastic composition comprises at least one chemotherapeutic agent selected from the group consisting of alkylating agents, antimetabolites, folic acid analogs, pyrimidine analogs, purine analogs and related inhibitors, vinca alkaloids, epipodopyyllotoxins, antibiotics, L-Asparaginase, topoisomerase inhibitor, interferons, platinum cooridnation complexes, anthracenedione substituted urea, methyl hydrazine derivatives, adrenocortical suppressant, adrenocorticosteroides, progestins, estrogens, antiestrogen, androgens, antiandrogen, and gonadotropin- releasing hormone analog. In some embodiments, the chemotherapeutic agent is selected from the group consisting of cisplatin, dicarbazine, dactinomycin, mechlorethamine, streptozocin, cyclophosphamide, capecitabine, carmustine, lomustine, doxorubicin, daunorubicin, procarbazine, mitomycin, cytarabine, etoposide, methotrexate, 5-fluorouracil, vinblastine, vincristine, bleomycin, paclitaxel, docetaxel, doxetaxe, aldesleukin, asparaginase, busulfan, carboplatin, cladribine, dacarbazine, floxuridine, fludarabine, hydroxyurea, ifosfamide, interferon alpha, irinotecan, leuprolide, leucovorin, megestrol, melphalan, mercaptopurine, oxaliplatin, plicamycin, mitotane, pegaspargase, pentostatin, pipobroman, plicamycin, streptozocin, tamoxifen, teniposide, testolactone, thioguanine, thiotepa, uracil mustard, vinorelbine, chlorambucil, taxol, an additional growth factor receptor antagonist, and a combination of any of the foregoing.
Assays for evaluating ligands
The ligands of the invention can be assayed using any suitable in vitro or in vivo assay. For example, using the receptor binding assays or bioassays described herein.
Bioassay for VEGF activity:
This bioassay measures the ability of ligands {e.g., dAbs) to neutralise the
VEGF induced proliferation of HUVE (human vascular endothelial) cells. HUVE cells plated in 96-well plates are incubated for 72 hours with pre-equilibrated VEGF and dAb protein. Cell number is then measured using a cell viability dye.
The assay is performed as follows. HUVE cells are trypsinized from a sub- confluent 175cm2 flask. Medium is aspirated off, the cells are washed with 5ml trypsin and then incubated with 2ml trypsin at room temperature for 5min. The cells are gently dislodged from the base of the flask by knocking against your hand. 8ml of induction medium are then added to the flask, pipetting the cells to disperse any clumps. Viable cells are counted using trypan blue stain.
Cells are spun down and washed 2X in induction medium, spinning cells down and aspirating the medium after each wash. After the final aspiration the cells are diluted to 105 cells/ml (in induction medium) and plated at 100 μl per well into a 96 well plate (10,000 cells/well). The plate is incubated for >2h @ 37C to allow attachment of cells.
60μl dAb protein and 60μl induction media containing 40ng/ml VEGF165 (for a final concentration of 1 Ong/ml) is added to a v-bottom 96-well plate and sealed with film. The dAb/VEGF mixture is then incubated at 37C for 0.5-1 hour.
The dAb/VEGF plate is removed from the incubator and lOOμl of solution added to each well of the HUVEC containing plate (final volume of 200μl). This plate is then returned to the 370C incubator for a period of at least 72 hours.
Control wells include the following: wells containing cells, but no VEGF; wells containing cells, a positive control neutralising anti-VEGF antibody and VEGF; and control wells containing cells and VEGF only.
Cell viability is assessed by adding 20μl per well Celltiter96 reagent, and the plate is incubated at 370C for 2-4h until a brown colour develops. The reaction is stopped by the addition of 20μl per well of 10% (w/v) SDS. The absorbance is then read at 490nm using a Wallac microplate reader.
The absorbance of the no VEGF control wells is subtracted from all other values. Absorbance is proportional to cell number. The control wells containing control anti-VEGF antibodies should also exhibit minimum cell proliferation. The wells containing VEGF only should exhibit maximum cell proliferation.
EXAMPLES EXAMPLE 1. VEGF Receptor Binding Assays
VEGF is a specific mitogen for endothelial cells in vitro and a potent angiogenic factor in vivo, with high levels of the protein being expressed in various types of tumours. It is a 45kDa glycoprotein that is active as a homodimer. Several isoforms have been described which occur through alternative mRNA splicing. Of these isoforms VEGF-121 and VEGF-165 appear to be the most abundant.
The specific action of VEGF on endothelial cells is mainly regulated by two types of receptor tyrosine kinases (RTK), VEGF Rl (FIt-I), and VEGF R2
(KDR/Flk-1). However, it appears that the VEGF activities such as mitogenicity, chemotaxis, and induction of morphological changes are mediated by VEGF R2, even though both receptors undergo phosphorylation upon binding of VEGF.
VEGF Receptor 2 binding assay
This method describes a VEGF receptor binding assay for measuring the ability of ligands (e.g., dAbs) to prevent binding of VEGF- 165 to VEGF Receptor 2. A recombinant human VEGF R2/Fc chimera was used in this assay, comprising the extracellular domain of human VEGF R2 fused to the Fc region of human IgG1. Briefly, the receptor was captured on an ELISA plate, then the plate was blocked to prevent nonspecific binding. A mixture of VEGF- 165 and ligand was then added, the plate was washed and receptor bound VEGF-165 detected using a biotinylated anti-VEGF antibody and an Horse-radish Peroxidase (HRP) conjugated anti-biotin antibody. The plate was developed using a colorimetric substrate and the OD read at 450nm. If the dAb blocked VEGF binding to the receptor then no colour was detected.
The assay was performed as follows. A 96 well Nunc Maxisorp assay plate was coated overnight at 4°C with 100 μl per well of recombinant human VEGF R2/Fc (R&D Systems, Cat. No: 357-KD-050) at a concentration of 0.5 μg/ml in carbonate buffer. Wells were washed 3 times with 0.05%Tween/PBS and 3 times with PBS. 200μl per well of 2% BSA in PBS was added to block the plate and the plate was incubated for a minimum of 1 hour at room temperature.
Wells were washed (as above), then 50μl per well of ligand was added to each well. 50μl of VEGF, at a concentration of 6ng/ml in diluent (for a final concentration of 3ng/ml), was then added to each well and the plate was incubated for 2 hours at room temperature (for assay of supernatants; 80μl of supernatant was added to each well then 20μl of VEGF at a concentration of 15ng/ml).
The following controls were included: Ong/ml VEGF (diluent only); 3ng/ml VEGF (R&D Systems, Cat No: 293-VE-050); 3ng/ml VEGF with 0.1 μg/ml anti- VEGF neutralizing antibody (R&D Systems cat#MAB293).
The plate was washed (as above) and then lOOμl biotinylated anti-VEGF antibody (R&D Systems, Cat No: BAF293), 0.5μg/ml in diluent, was added and incubated for 2 hours at room temperature.
Wells were washed (as above) then lOOμl HRP conjugated anti-biotin antibody (1:5000 dilution in diluent; Stratech, Cat No: 200-032-096) was added. The plate was then incubated for 1 hour at room temperature.
The plate was washed (as above) ensuring any traces of Tween-20 were removed to limit background in the subsequent peroxidase assay and to help the prevention of bubbles in the assay plate wells that might give inaccurate OD readings.
lOOμl of SureBlue 1 -Component TMB Micro Well Peroxidase solution was added to each well, and the plate was left at room temperature for up to 20 minutes. A deep blue soluble product developed as bound HRP labelled conjugate reacted with the substrate. The reaction was stopped by the addition of lOOμl IM
hydrochloric acid (the blue colour turned yellow). The OD, at 450nm, of the plate was read in a 96-well plate reader within 30 minutes of acid addition. The OD450nm is proportional to the amount of bound streptavidin-HRP conjugate.
For some assays protein L was added. Protein L cross links two dAb monomers.
Expected results from the controls are as follows: 0ng/ml VEGF should give a low signal of <0.15 OD; 3ng/ml VEGF should give a signal of >0.5 OD; and 3ng/ml VEGF pre-incubated with 0.1 μg/ml neutralising antibody should give a signal <0.2 OD.
VEGF Receptor 1 binding assay This assay measures the binding of VEGF-165 to VEGF Rl and the ability of ligands to block this interaction. A recombinant human VEGF Rl/Fc chimera was used here, comprising the extracellular domain of human VEGF Rl fused to the Fc region of human IgG1. The receptor was captured on an ELISA plate then the plate was blocked to prevent non-specific binding. A mixture of VEGF-165 and ligand was then added, the plate was washed and receptor bound VEGF-165 detected using a biotinylated anti-VEGF antibody and an HRP conjugated anti- biotin antibody. The plate was developed using a colorimetric substrate and the OD read at 450nm.
The assay was performed as follows. A 96-well Nunc Maxisorp assay plate was coated overnight at 40C with lOOμl per well of recombinant human VEGF Rl/Fc (R&D Systems, Cat No: 321-FL-050) @ 0.1 μg/ml in carbonate buffer.
Wells were washed 3 times with 0.05%Tween/PBS and 3 times with PBS.
200μl per well of 2% BSA in PBS was added to block the plate and the plate was incubated for a minimum of 1 hour at room temperature.
Wells were washed (as above), then 50μl per well of purified dAb protein was added to each well. 50μl of VEGF, at a concentration of lng/ml in diluent (for a final concentration of 500pg/ml), was then added to each well and the plate incubated for 1 hour at room temperature (assay of supernatants; 80μl of supernatant was added to each well then 20μl of VEGF @ 2.5ng/ml).
The following controls were included: Ong/ml VEGF (diluent only);
500ρg/ml VEGF; and 500pg/ml VEGF with 1 μg/ml anti-VEGF antibody (R&D Systems cat#MAB293).
The plate was washed (as above) and then lOOμl biotinylated anti-VEGF antibody, 50ng/ml in diluent, was added and incubated for 1 hour at room
temperature.
Wells were washed (as above) then lOOμl HRP conjugated anti-biotin antibody was added (1 :5000 dilution in diluent). The plate was then incubated for 1 hour at room temperature.
The plate was washed (as above), ensuring any traces of Tween-20 were removed to limit background in the subsequent peroxidase assay and to help the prevention of bubbles in the assay plate wells that might give inaccurate OD readings.
lOOμl of SureBlue 1 -Component TMB Micro Well Peroxidase solution was added to each well, and the plate was left at room temperature for up to 20 minutes. A deep blue soluble product developed as bound HRP labelled conjugate reacted with the substrate. The reaction was stopped by the addition of lOOμl IM
hydrochloric acid. The OD, at 450nm, of the plate was read in a 96-well plate reader within 30 minutes of acid addition. The OD450 nm is proportional to the amount of bound streptavidin-HRP conjugate.
Expected result from the controls: 0 ng/ml VEGF should give a low signal of <0.15 OD; 500pg/ml VEGF should give a signal of >0.8 OD; and 500pg/ml VEGF pre-incubated with lμg/ml neutralising antibody should give a signal <0.3 OD
Table 1
Figure imgf000165_0001
The TARl 5-1 had a Kd of 50-80 nM when tested at various concentrations on a low density BIAcore chip. Other VK dAbs were passed over the low density chip at one concentration (5OnM). Different dAbs showed different binding kinetics. Table 2
Figure imgf000166_0001
* dAb was assayed at 5OnM
VH dAbs were passed over the low density VEGF chip on a BIAcore at one concentration (5OnM). Different dAbs showed different binding kinetics.
Example 2. EGFR binding
EGFR binding assay
25ul of ligand (e.g., dAb) were plated into a 96- well plate and then 25ul streptavidin-Alexa Fluor (lug/ml) (Molecular Probes) and 25 ul A431 cells (ATCC No. CRL-1555) (8xlO5/ml) were added. All reagents were prepared in PBS/1% BSA. The plate was incubated for 30 minutes at room temperature.
Without disturbing the cells, 25ul biotinylated EGF (Invitrogen) at 40ng/ml was added to each well, and the plate was incubated for three hours at room temperature. Fluoresecence was measured using the AB 8200 Cellular Detection System (Applied Biosystems).
Ligands (e.g., dAbs) that inhibited the binding of biotinylated EGF to EGFR expressed on A431 cells resulted in lower fluorescence counts. Wells without ligand provided a reference of the maximum fluorescence (i.e., biotinylated EGF binding) and wells without ligand or biotinylated EGF provided a reference of the
background level of fluorescence. These controls were included in all assays.
Results obtained in this assay using certain anti-EGFR dAbs are presented in the Table 3.
EGFR kinase assay
In a 96-well plate, 5xlO4 A431 cells (ATCC No. CRL-1555) were plated per well in RPMI- 1640 supplemented with 10% foetal calf serum. The plate was incubated overnight at 37°C/ 5% CO2 to allow the cells to adhere, then the medium was replaced with RPMI-1640. The plate was incubated for 4 hours at 37°C / 5% CO2. The ligand (prepared in RPMI-1640) was added to the wells and the plate was incubated for 45 minutes at 37°C / 5% CO2. EGF (Invitrogen) was added to the wells to give a final concentration of 100ng/ml and the plate was incubated for 10 minutes at room temperature. The wells were washed twice with ice cold PBS. Cold lysis buffer (1 % NP-40, 2OmM Tris, 137mM NaCl, 10% glycerol, 2mM EDTA, ImM sodium ortho vanadate, lOug/ml aprotinin, lOug/ml leupeptin) was added and the plate was incubated on ice for 10 minutes.
The supernatants were transfered to an ELISA plate which had been coated overnight with anti-EGFR antibody (R&D Systems) at lug/ml in carbonate buffer. The ELISA plate was incubated for 2 hours at room temperature. The plate was washed three times with PBS/0.05% Tween 20. Anti-phosphotyrosine antibody conjugated to horse-radish peroxidase (Upstate Biotechnology) at lug/ml was added and the plate was incubated for 1 hour at room temperature. The plate was washed three times with PBS/Tween and three times with PBS. The reaction was developed with SureBlue TMB 1 -component microwell peroxidase substrate (KPL) and the reaction was stopped with IM HCl after 25 minutes. The absorbance was read using a Wallac plate reader.
Results obtained in this assay using certain anti-EGFR dAbs are presented in the Table 3.
Table 3
Figure imgf000168_0001
* the data presented are the lowest to highest values obtained and the (average)
Example 3. IgG-like foπnats that have binding specificity for VEGF and EGFR. Vectors
The pBudCE4.1 backbone (Invitrogen) was used for cloning
immunoglobulin constant regions, such as the IgGl heavy chain constant region and light chain kappa constant region (see FIG. 16 for overview). An Ig Kappa chain leader was used to facilitate secretion of the expressed protein. Ig constant regions (human IgGl and CK) were produced by GeneArt (Germany).
The heavy chain constant region and signal peptide were cloned into pBudCE4.1 as a Hind III/Bglll fragment into the Hindlll/BamH I restriction sites.
The light chain constant region and signal peptide were cloned into pBudCE4.1 as a Notl/MluI fragment. Cloning of dAb in IgG vectors and production of IgG-like format
VK dAb (specific to VEGF or EGFR) was cloned into IgG vector as a Sall/BsiWI fragment. VH dAb (specific to VEGF or EGFR was cloned into IgG vector as a BamHI/XhoI fragment.
The plasmid was then transfected into HEK293T cells (ATCC CRL-11268) and IgG was expressed transiently for five days. The IgG produced was purified using streamline Protein A.
Purified IgG was checked on a reducing and non-reducing SDS gel and bands of expected size were observed.
Several dAbs that bind VEGF or EGFR were formatted into IgG-like formats that have binding specificity for VEGF and EGFR. The IgG-like formats were prepared by producing constructs that encoded an IgG heavy chain wherein VH is a dAb, and a Kappa light chain wherein VK is a dAb. The IgG-like formats that were prepared are shown in Table 4, and the results obtained for some of the IgG-like formats in assays are presented in Table 5. (Dummy VH and Dummy VK are germ line sequences that do not bind VEGF or EGFR).
Figure imgf000170_0001
Figure imgf000171_0001
Example 4. Dual Specific Inline Formats
Domain antibodies that bind VEGF or EGFR were incorporated into fusion polypeptides that contained an anti- VEGFR dAb and an anti-EGFR dAb in a single polypeptide chain. Some of the fusion polypeptides also included an antibody Fc region (-CH2-CH3 of human IgGl). Specific examples of the fusion polypeptides that were cloned and expressed include TAR15-10 fused to DOM16-39-206 and to Fc (SEQ ID NO:715); DOM16-39-206 fused to TAR15-10 and to Fc (SEQ ID NO:716); DOM16-39-206 fused to TAR15-26-501 and to Fc (SEQ ID NO:717); TAR15-26-501 fused to DOM16-39-206 and to Fc (SEQ ID NO:718); TAR15-10 fused to DOMl 6-39-206 (SEQ ID NO:719); DOMl 6-39-206 fused to TARl 5-10 (SEQ ID NO:720); DOMl 6-39-206 fused to TARl 5-26-501 (SEQ ID NO:721); and TAR15-26-501 fused to DOM16-39-206 (SEQ ID NO:722). The positions of the foregoing fusions are listed as they appear in the fusion proteins from amino terminus to carboxy terminus.
DNA encoding dAbs was PCR amplified and cloned into expression vectors using standard methods. Inline fusion polypeptides were produced by expressing the expression vectors in Pichia (fusion that did not contain an Fc region) or in HEK 293T cells (Fc region containing fusions). Inline fusions were batch bound and affinity purified on streamline protein A and streamline protein L resins for HEK 293T cells (Fc-tagged) and Pichia expressed constructs respectively.
The portions of several fusions that contain Fc are listed in Table 6 as they appear in the fusion proteins, from amino terminus to carboxy terminus.
Accordingly, the structure of the fusion proteins can be appreciated by reading the table from left to right. The first fusion protein presented in Table 6 has the structure, from amino terminus to carboxy terminus, DOM15-10— Linker 1—
DOMl 6-39-206— Linker 2— Fc. Binding activities of the fusions were assessed using the EGFR binding assay described in Example 2, and the VEGF receptor 2 binding assay described in Example 1.
General robustness and resistance to degradation were tested by subjecting the inline fusions to proteolysis with trypsin. A solution of dual specific ligand and trypsin (1/25 (w/w) trypsin to ligand) was prepared and incubated at 3O0C. Samples were taken at 0 minutes (i.e., before addition of trypsin), 60 minutes, 180 minutes, and 24 hours. At the given time points, the reaction was stopped by the addition of complete protease inhibitor cocktail at 2X final concentration (Roche code: 11 836 145 001) with PAGE loading dye, followed by flash freezing the samples in liquid nitrogen. Samples were analyzed by SDS-PAGE, and protein bands were visualized to reveal a time course for the protease degradation of the fusions. These experiments showed that inline fusions having a "natural" linker (KVEIKRTVAAPS (SEQ ID NO:706), which contains the carboxy-terminal amino acids of Vk and amino-terminal amino acids of Ck, were susceptible to rapid proteolysis, with degradation evident as soon as the 10 minute time point. SDS- PAGE analysis revealed that degredation occurred at the linkers between dAbs and at the linkers between dAb and Fc.
New linkers were designed that contain fewer Lys and Arg residues, which are cleavage points for trypsin and are abundant in the natural linker. Fusions that contained the engineered linkers (LVTVSSAST (SEQ ID NO:707)) or
(LVTVSSGGGGSGGGS (SEQ ID NO:708)) showed much improved resistance to trypsin proteolysis.
Additional binding assays were performed to assess the potency of the inline fusions that contained the engineered linkers. The results revealed engineered linkers did not have any substantial adverse effect on potency.
Figure imgf000174_0001
Example 5. Additional Engineered Linkers
Several designed mutations were introduced to the C-terminal region of Vk dAbs expressed on the light chain of IgG-like formats to reduce protease sensitivity. The "natural linker" was GQGTKVEIKRTVAAPS (SEQ ID NO:709 which contains the carboxy- terminal amino acids of Vk and amino-terminal amino acids of Ck). Variant linkers 1-3 were designed with amino acid replacements that replaced some or all of the positively charged residues in the natural linker with the most conservative substitutions that are not positively charged at physiological pH. It is likely that the arginine residue in the natural linker is less amenable to alteration due to ionic interactions it forms within the CL domain.
Variant linker 1 (GQGTNVEINRTVAAPS (SEQ ID NO:710)) substitutes both lysines in the natural linker with asparagines. Variant linker 1, and variant linker 2 (GQGTNVEINQTVAAPS (SEQ ID NO:711)), which additionally changes the arginine in the natural linker to glutamine, introduce an N-glycosylation site (NxT) into the linker. SDS-PAGE analysis of IgG-like formats containing variant linker 1 or variant linker 2 showed that the light chain had a higher molecular weight, consistent with an N-glycosylation event. Variant linker 3
(GQGTNVEIQRTVAAPS (SEQ ID NO:712) removes the N-glycosylation site while leaving the arginine in the natural linker in place. Variant linker 4
(GQGTLVTVSSTVAAPS (SEQ ID NO:713)) replaces the six C-terminal amino acids of the Vk domain with the corresponding residues from a VH domain, and is devoid of positive charges.
Protease resistance (trypsin resistance assessed as described in Example 4) of
IgG-like formats that contain variant linkers 1-4 revealed that IgG-like formats that contained engineered variant linkers were more protease resistant than an IgG-like format that contained the natural linker. Example 6. DOM 16 dAb-anti-serum albumin d Ab fusions DOMl 6 dAb- anti-serum albumin dAb fusions were designed and expressed as a fusion of an anti-EGFR dAb to an anti-serum albumin dAb (a D0M7 dAb). The portions of the fusions are listed in Table 7 as they appear in the fusion proteins, from amino terminus to carboxy terminus. Accordingly, the structure of the fusion proteins can be appreciated by reading the table from left to right. The first fusion protein presented in Table 7 has the structure, from amino terminus to carboxy terminus, DOM16-39-618(S12P)— Linker— DOM7h-14.
DOMl 6-39-618 contains a Serine to Proline mutation at position 12, which stops binding to protein L and prevents light chain aggregation. The iD0M7 dAbs are mutated so as to abrogate binding to albumin, and thus are inactivated.
Table 7
Figure imgf000175_0001
Example 7. EGFR Epitope Mapping
Epitope mapping was conducted in competitive binding assays using anti- EGFR dAbs, EGF and ERBITUX (cetuximab; Imclone Systems). The binding studies were performed using a BIAcore biosensor. DOMl 6-39-200 was used as a reference in this study. DOM16-39-200, and other dAbs designated DOM-16-39-X, are affinity matured variants of DOM16-39. Accordingly, all dAbs in the D0M-16- 39 series will have substantially the same epitopic specificity, because affinity maturation produces dAbs that bind with higher affinity, but does not alter the specificity of the dAb. The results showed that DOM16-72, DOM16-79 and DOM16-112 competed with DOMl 6-39-200 for binding to EGFR, indicating that these dAbs bind overlapping epitopes. However, DOMl 6-32, DOMl 6-52 and DOMl 6-80 were shown to bind to a different epitope. ERBITUX (cetuximab; Imclone Systems) is known to inhibit binding of EGF to EGFR (cetuximab and EGF bind overlapping epitopes on EGFR). The results of this study also demonstrated that DOMl 6-39- 200 and cetuximab competed for binding to EGFR, indicating that the DOMl 6-39- 200 epitope overlaps with the cetuximab epitope, and with the binding site for EGF. While this invention has been particularly shown and described with references to preferred embodiments thereof, it will be understood by those skilled in the art that various changes in form and details may be made therein without departing from the scope of the invention encompassed by the appended claims.

Claims

CLAIMS What is claimed is: 1. A ligand that has binding specificity for vascular endothelial growth factor (VEGF) and epidermal growth factor receptor (EGFR), comprising at least one protein moiety that has a binding site with binding specificity for VEGF and at least one protein moiety that has a binding site with binding specificity for EGFR.
2. The ligand of claim 1 , wherein each said protein moiety that has a binding site with binding specificity for EGFR competes for binding to EGFR with an anti- EGFR domain antibody (dAb) selected from the group consisting of DOM16-39 (SEQ ID NO:345), DOM16-39-87 (SEQ ID NO:420), DOM16-39-100 (SEQ ID NO:423), DOM16-39-107 (SEQ ID NO:430), DOM16-39-109 (SEQ ID NO:432), DOM16-39-115 (SEQ ID NO:438), and DOMl 6-39-200 (SEQ ID NO:441).
3. The ligand of claim 1 , wherein each said protein moiety that has a binding site with binding specificity for EGFR competes for binding to EGFR with an anti- EGFR domain antibody (dAb) selected from the group consisting of DOM 16-39- 521 (SEQ ID NO:577), DOM16-39-541 (SEQ ID NO:585), DOM16-39-542 (SEQ ID NO:586), DOM16-39-551 (SEQ ID NO:591), DOM16-39-601 (SEQ ID
NO:608), DOM16-39-604 (SEQ ID NO:611), DOM16-39-618 (SEQ ID NO:621), and DOM16-39-619 (SEQ ID NO:622).
4. The ligand of claim 1 , wherein each said protein moiety that has a binding site with binding specificity for VEGF competes for binding to VEGF with an anti- VEGF domain antibody (dAb) selected from the group consisting of TARl 5-6 (SEQ ID NO: 117), TARl 5-8 (SEQ ID NO: 119), and TARl 5-26 (SEQ ID NO: 123); and wherein each said protein moiety that has a binding site with binding specificity for EGFR competes for binding to EGFR with an anti-EGFR domain antibody (dAb) selected from the group consisting of DOMl 6-39 (SEQ ID NO:345), DOMl 6-39-87 (SEQ ID NO:420), DOM16-39-100 (SEQ ID NO:423), DOM16-39-107 (SEQ ID NO:430), DOM16-39-109 (SEQ ID NO:432), DOM16-39-115 (SEQ ID NO:438), and DOMl 6-39-200 (SEQ ID NO:441).
5. The ligand of claim 1 , wherein each said protein moiety that has a binding site with binding specificity for VEGF competes for binding to VEGF with an anti- VEGF domain antibody (dAb) selected from the group consisting of TARl 5-6 (SEQ ID NO: 117), TARl 5-8 (SEQ ID NO:119), and TARl 5-26 (SEQ ID NO: 123); and wherein each said protein moiety that has a binding site with binding specificity for EGFR competes for binding to EGFR with an anti-EGFR domain antibody (dAb) selected from the group consisting of DOM 16-39-521 (SEQ ID NO:577), D0M16- 39-541 (SEQ ID NO:585), DOM16-39-542 (SEQ ID NO:586), DOM16-39-551 (SEQ ID NO:591), DOMl 6-39-601 (SEQ ID NO:608), DOMl 6-39-604 (SEQ ID NO:611), DOM16-39-618 (SEQ ID NO: 621), and DOM16-39-619 (SEQ ID
NO:622).
6. The ligand of claim 1 , wherein each said protein moiety that has a binding site with binding specificity for VEGF competes for binding to VEGF with bevacizumab.
7. The ligand of claim 1 or claim 4, wherein each said protein moiety that has a binding site with binding specificity for EGFR competes for binding to EGFR with cetuximab.
8. The ligand of any one of claims 1 -7, wherein each said protein moiety that has a binding site with binding specificity for VEGF competes for binding to VEGF with bevacizumab; and wherein each said protein moiety that has a binding site with binding specificity for EGFR competes for binding to EGFR with cetuximab.
9. The ligand of any one of claims 1-8, wherein each protein moiety that has a binding site with binding specificity for VEGF and each protein moiety that has a binding site with binding specificity for EGFR is provided by an antibody fragment.
10. The ligand of claim 9, wherein said antibody fragment is an immunoglobulin single variable domain.
11. A ligand that has binding specificity for vascular endothelial growth factor (VEGF) and epidermal growth factor receptor (EGFR), comprising at least one immunoglobulin single variable domain with binding specificity for VEGF and at least one immunoglobulin single variable domain with binding specificity for EGFR, wherein each said immunoglobulin single variable domain with binding specificity for EGFR competes for binding to EGFR with an anti-EGFR domain antibody (dAb) selected from the group consisting of DOM16-39 (SEQ ID NO:345), DOM16-39-87 (SEQ ID NO:420), DOM16-39-100 (SEQ ID NO:423), DOM16-39- 107 (SEQ ID NO:430), DOM16-39-109 (SEQ ID NO:432), DOM16-39-115 (SEQ ID NO:438), and DOM16-39-200 (SEQ ID NO:441).
12. A ligand that has binding specificity for vascular endothelial growth factor (VEGF) and epidermal growth factor receptor (EGFR), comprising at least one immunoglobulin single variable domain with binding specificity for VEGF and at least one immunoglobulin single variable domain with binding specificity for EGFR, wherein each said immunoglobulin single variable domain with binding specificity for EGFR competes for binding to EGFR with an anti-EGFR domain antibody (dAb) selected from the group consisting of DOMl 6-39-521 (SEQ ID NO:577), DOMl 6-39-541 (SEQ ID NO:585), DOMl 6-39-542 (SEQ ID NO:586), DOM16-39-551 (SEQ ID NO:591), DOM16-39-601 (SEQ ID NO:608), D0M16- 39-604 (SEQ ID NO:611), DOM16-39-618 (SEQ ID NO:621), and DOM16-39-619 (SEQ ID NO:622).
13. The ligand of claim 11 or claim 12, wherein each said immunoglobulin single variable domain with binding specificity for EGFR is fused to an Fc region of an antibody.
14. The ligand of claim 11 or claim 12, wherein each said immunoglobulin single variable domain with binding specificity for VEGF is fused to an Fc region of an antibody.
15. The ligand of claim 11 , wherein each said immunoglobulin single variable domain with binding specificity for EGFR comprises an amino acid sequence that has at least about 85% amino acid sequence identity with the amino acid sequence of a dAb selected from the group consisting of DOMl 6-39 (SEQ ID NO:345),
DOM16-39-87 (SEQ ID NO:420), DOMl 6-39-100 (SEQ ID NO:423), DOM16-39- 107 (SEQ ID NO:430), DOM16-39-109 (SEQ ID NO:432), DOM16-39-115 (SEQ ID NO:438), and DOMl 6-39-200 (SEQ ID NO:441).
16. The ligand of claim 12, wherein each said immunoglobulin single variable domain with binding specificity for EGFR comprises an amino acid sequence that has at least about 85% amino acid sequence identity with the amino acid sequence of a dAb selected from the group consisting of DOM16-39-521 (SEQ ID NO:577), DOMl 6-39-541 (SEQ ID NO:585), DOMl 6-39-542 (SEQ ID NO:586), DOMl 6- 39-551 (SEQ ID NO:591), DOM16-39-601 (SEQ ID NO:608), DOM16-39-604 (SEQ ID NO:611), DOMl 6-39-618 (SEQ ID NO:621), and DOM16-39-619 (SEQ ID NO:622).
17. A ligand that has binding specificity for vascular endothelial growth factor (VEGF) and epidermal growth factor receptor (EGFR), comprising at least one immunoglobulin single variable domain with binding specificity for VEGF and at least one immunoglobulin single variable domain with binding specificity for EGFR, wherein
each said immunoglobulin single variable domain with binding specificity for VEGF competes for binding to VEGF with an anti-VEGF domain antibody (dAb) selected from the group consisting of TARl 5-6 (SEQ ID NO:117), TARl 5-8 (SEQ ED NOrI 19), and TAR15-26 (SEQ ID NO: 123); and
each said immunoglobulin single variable domain with binding specificity for EGFR competes for binding to EGFR with an anti-EGFR domain antibody (dAb) selected from the group consisting of DOM16-39 (SEQ ID NO:345),
DOMl 6-39-87 (SEQ ID NO:420), DOMl 6-39-100 (SEQ ID NO:423), DOMl 6-39- 107 (SEQ ID NO:430), DOM16-39-109 (SEQ ID NO:432), DOM16-39-115 (SEQ ID NO:438), and DOM16-39-200 (SEQ ID NO:441).
18. A ligand that has binding specificity for vascular endothelial growth factor (VEGF) and epidermal growth factor receptor (EGFR), comprising at least one immunoglobulin single variable domain with binding specificity for VEGF and at least one immunoglobulin single variable domain with binding specificity for EGFR, wherein
each said immunoglobulin single variable domain with binding specificity for VEGF competes for binding to VEGF with an anti-VEGF domain antibody (dAb) selected from the group consisting of TAR15-6 (SEQ ID NO:117), TAR15-8 (SEQ ID NO: 119), and TARl 5-26 (SEQ ID NO: 123); and
each said immunoglobulin single variable domain with binding specificity for EGFR competes for binding to EGFR with an anti-EGFR domain antibody (dAb) selected from the group consisting of DOMl 6-39-521 (SEQ ID NO:577), DOM16-39-541 (SEQ ID NO:585), DOM16-39-542 (SEQ ID NO:586), D0M16- 39-551 (SEQ ID NO:591), DOMl 6-39-601 (SEQ ID NO:608), DOMl 6-39-604 (SEQ ID NO:611), DOMl 6-39-618 (SEQ ID NO:621), and DOMl 6-39-619 (SEQ ID NO:622).
19. The ligand of claim 17, wherein each said immunoglobulin single variable domain with binding specificity for VEGF comprises an amino acid sequence that has at least about 85% amino acid sequence identity with the amino acid sequence of a dAb selected from the group consisting of TAR15-6 (SEQ ID NO:117), TAR15-8
(SEQ ID N0:119), and TAR15-26 (SEQ ID NO:123); and
each said immunoglobulin single variable domain with binding specificity for EGFR competes for binding to EGFR with an anti-EGFR domain antibody (dAb) selected from the group consisting of DOMl 6-39 (SEQ ID NO:345),
DOM16-39-87 (SEQ ID NO:420), DOM16-39-100 (SEQ ID NO:423), DOM16-39- 107 (SEQ ID NO:430), DOM16-39-109 (SEQ ID NO:432), DOM16-39-115 (SEQ ID NO:438), and DOM16-39-200 (SEQ ID NO:441).
20. The ligand of claim 18, wherein each said immunoglobulin single variable domain with binding specificity for VEGF comprises an amino acid sequence that has at least about 85% amino acid sequence identity with the amino acid sequence of a dAb selected from the group consisting of TAR15-6 (SEQ ID NO:117), TAR15-8 (SEQ ID NO:119), and TARl 5-26 (SEQ ID NO: 123); and
each said immunoglobulin single variable domain with binding specificity for EGFR competes for binding to EGFR with an anti-EGFR domain antibody (dAb) selected from the group consisting of DOM16-39-521 (SEQ ID NO:577), DOMl 6-39-541 (SEQ ID NO:585), DOMl 6-39-542 (SEQ ID NO:586), DOMl 6- 39-551 (SEQ ID NO:591), DOM16-39-601 (SEQ ID NO:608), DOM16-39-604 (SEQ ID NO:611), DOM16-39-618 (SEQ ID NO:621), and DOM16-39-619 (SEQ ID NO:622).
21. The ligand of any one of claims 1-19, wherein said ligand inhibits binding of epidermal growth factor (EGF) and/or transforming growth factor alpha (TGFalpha) to EGFR.
22. The ligand of any one of claims 1-19, wherein said ligand inhibits the activity of EGFR.
23. The ligand of any one of claims 1-19, wherein said ligand inhibits the activity of EGFR without substantially inhibiting binding of epidermal growth factor (EGF) and/or transforming growth factor alpha (TGFalpha) to EGFR.
24. The ligand of any one of claims 1 -23, wherein said ligand inhibits binding of VEGF to vascular endothelial growth factor receptor 1 (VEGFRl) and/or vascular endothelial growth factor receptor 2 (VEGFR2).
25. The ligand of any one of claims 1-23, wherein said ligand inhibits the activity of VEGF.
26. The ligand of any one of claims 1-23, wherein said ligand inhibits the activity of VEGF without substantially inhibiting binding of VEGF to vascular endothelial growth factor receptor 1 (VEGFRl) and/or vascular endothelial growth factor receptor 2 (VEGFR2).
27. The ligand of any one of claims 10-26 wherein each said immunoglobulin single variable domain with binding specificity for VEGF binds VEGF with an affinity (KD) that is between about 100 nM and about 1 pM, as determined by surface plasmon resonance.
28. The ligand of any one of claims 10-26 wherein each said immunoglobulin single variable domain with binding specificity for EGFR binds EGFR with an affinity (KD) that is between about 100 nM and about 1 pM, as determined by surface plasmon resonance.
29. The ligand of claim 28 wherein each said immunoglobulin single variable domain with binding specificity for EGFR binds EGFR with an affinity (KD) that is between about 10 nM and about 100 pM, as determined by surface plasmon resonance.
30. The ligand of any one of claims 1-26 wherein said ligand binds VEGF with an affinity (KD) that is between about 100 nM and about 1 pM, as determined by surface plasmon resonance.
31. The ligand of any one of claims 1 -26 and 30 wherein said ligand binds EGFR with an affinity (KD) that is between about 100 nM and about 1 pM, as determined by surface plasmon resonance.
32. The ligand of claim 31 wherein said ligand binds EGFR with an affinity (KD) that is between about 10 nM and about 100 pM, as determined by surface plasmon resonance.
33. The ligand of any one of claims 10-32 wherein said ligand comprises an immunoglobulin single variable domain with binding specificity for VEGF that is a VHH and/or an immunoglobulin single variable domain with binding specificity for EGFR that is a VHH.
34. The ligand of any one of claims 10-32, wherein each said immunoglobulin single variable domain with binding specificity for VEGF and each said immunoglobulin single variable domain with binding specificity for EGFR are independently selected from the group consisting of a human VH and a human VL.
35. The ligand of any one of claims 1-34, wherein said ligand is an IgG-like format comprising two immunoglobulin single variable domains with binding specificity for VEGF, and two immunoglobulin single variable domains with binding specificity for EGFR.
36. The ligand of any one of claims 1-35, wherein said ligand comprises an antibody Fc region.
37. A ligand that has binding specificity for vascular endothelial growth factor
(VEGF) and epidermal growth factor receptor (EGFR), comprising at least one immunoglobulin single variable domain with binding specificity for VEGF and at least one immunoglobulin single variable domain with binding specificity for
EGFR, wherein
each said immunoglobulin single variable domain with binding specificity for VEGF competes for binding to VEGF with an anti-VEGF domain antibody (dAb) selected from the group consisting of TARl 5-6 (SEQ ID NO: 117), TARl 5-8
(SEQ ID NO:119), and TARl 5-26 (SEQ ID NO: 123); and each said immunoglobulin single variable domain with binding specificity for EGFR competes for binding to EGFR with cetuximab.
38. The ligand of claim 37, wherein each said immunoglobulin single variable domain with binding specificity for VEGF comprises an amino acid sequence that has at least about 85% amino acid sequence identity with the amino acid sequence of a dAb selected from the group consisting of TAR15-6 (SEQ ID NO:117), TAR15-8 (SEQ ID NO: 119), and TAR15-26 (SEQ ID NO:123) 39. A ligand that has binding specificity for vascular endothelial growth factor (VEGF) and epidermal growth factor receptor (EGFR), comprising at least one immunoglobulin single variable domain with binding specificity for VEGF and at least one immunoglobulin single variable domain with binding specificity for EGFR, wherein
each said immunoglobulin single variable domain with binding specificity for VEGF competes for binding to VEGF with bevacizumab; and
each said immunoglobulin single variable domain with binding specificity for EGFR competes for binding to EGFR with an anti-EGFR domain antibody (dAb) selected from the group consisting of DOMl 6-39 (SEQ ID NO:345), DOMl 6-39-87 (SEQ ID NO:420), DOMl 6-39-100 (SEQ ID NO:423), DOMl 6-39-107 (SEQ ID
NO:430), DOM16-39-109 (SEQ ID NO:432), DOM16-39-115 (SEQ ID NO:438), and DOMl 6-39-200 (SEQ ID NO:441).
40. A ligand that has binding specificity for vascular endothelial growth factor (VEGF) and epidermal growth factor receptor (EGFR), comprising at least one immunoglobulin single variable domain with binding specificity for VEGF and at least one immunoglobulin single variable domain with binding specificity for EGFR, wherein
each said immunoglobulin single variable domain with binding specificity for VEGF competes for binding to VEGF with bevacizumab; and
each said immunoglobulin single variable domain with binding specificity for EGFR competes for binding to EGFR with an anti-EGFR domain antibody (dAb) selected from the group consisting of DOM16-39-521 (SEQ ID NO:577), DOM16-39-541 (SEQ ID NO:585), DOM16-39-542 (SEQ ID NO:586), DOM16-39-551 (SEQ ID NO:591), DOMl 6-39-601 (SEQ ID NO:608), DOMl 6-39-604 (SEQ ID NO:611), DOM16-39-618 (SEQ ID NO:621), and DOM16-39-619 (SEQ ID NO:622).
41. The ligand of claim 39, wherein each said immunoglobulin single variable domain with binding specificity for EGFR comprises an amino acid sequence that has at least about 85% amino acid sequence identity with the amino acid sequence of a dAb selected from the group consisting of DOM16-39 (SEQ ID NO:345),
DOM16-39-87 (SEQ ID NO:420), DOM16-39-100 (SEQ ID NO:423), DOM16-39- 107 (SEQ ID NO:430), DOMl 6-39-109 (SEQ ID NO:432), DOMl 6-39-115 (SEQ ID NO:438), and DOM16-39-200 (SEQ ID NO:441).
42. The ligand of claim 40, wherein each said immunoglobulin single variable domain with binding specificity for EGFR comprises an amino acid sequence that has at least about 85% amino acid sequence identity with the amino acid sequence of a dAb selected from the group consisting of DOM16-39-521 (SEQ ID NO:577), DOMl 6-39-541 (SEQ ID NO:585), DOMl 6-39-542 (SEQ ID NO:586), DOMl 6- 39-551 (SEQ ID NO:591), DOM16-39-601 (SEQ ID NO:608), DOM16-39-604 (SEQ ID NO:611), DOM16-39-618 (SEQ ID NO:621), and DOM16-39-619 (SEQ ID NO:622).
43. A ligand that has binding specificity for vascular endothelial growth factor (VEGF) and epidermal growth factor receptor (EGFR), comprising a first immunoglobulin single variable domain with binding specificity for VEGF and a second immunoglobulin single variable domain with binding specificity for EGFR, wherein
said first immunoglobulin single variable domain competes for binding to
VEGF with bevacizumab; and
said second immunoglobulin single variable domain competes for binding to
EGFR with cetuximab.
44. The ligand of any one of claims 37-43, wherein said ligand inhibits binding of epidermal growth factor (EGF) and/or transforming growth factor alpha
(TGFalpha) to EGFR.
45. The ligand of any one of claims 37-43, wherein said ligand inhibits the activity of EGFR.
46. The ligand of any one of claims 37-43, wherein said ligand inhibits the activity of EGFR without substantially inhibiting binding of epidermal growth factor (EGF) and/or transforming growth factor alpha (TGFalpha) to EGFR.
47. The ligand of any one of claims 37-46, wherein said ligand inhibits binding of VEGF to vascular endothelial growth factor receptor 1 (VEGFRl) and/or vascular endothelial growth factor receptor 2 (VEGFR2).
48. The ligand of any one of claims 37-46, wherein said ligand inhibits the activity of VEGF.
49. The ligand of any one of claims 37-46, wherein said ligand inhibits the activity of VEGF without substantially inhibiting binding of VEGF to vascular endothelial growth factor receptor 1 (VEGFRl) and/or vascular endothelial growth factor receptor 2 (VEGFR2).
50. The ligand of any one of claims 37-49, wherein each said immunoglobulin single variable domain with binding specificity for VEGF binds VEGF with an affinity (KD) that is between about 100 nM and about 1 pM, as determined by surface plasmon resonance.
51. The ligand of any one of claims 37-50, wherein each said immunoglobulin single variable domain with binding specificity for EGFR binds EGFR with an affinity (KD) that is between about 100 nM and about 1 pM, as determined by surface plasmon resonance.
52. The ligand of claim 51 , wherein each said immunoglobulin single variable domain with binding specificity for EGFR binds EGFR with an affinity (KD) that is between about 10 nM and about 100 pM, as determined by surface plasmon resonance.
53. The ligand of any one of claims 37-49, wherein said ligand binds VEGF with an affinity (KD) that is between about 100 nM and about 1 pM, as determined by surface plasmon resonance.
54. The ligand of any one of claims 37-49 and 53, wherein said ligand binds EGFR with an affinity (KD) that is between about 100 nM and about 1 pM, as determined by surface plasmon resonance.
55. The ligand of claim 54, wherein said ligand binds EGFR with an affinity (KD) that is between about 10 nM and about 100 pM, as determined by surface plasmon resonance.
56. The ligand of any one of claims 37-55, wherein said ligand comprises an immunoglobulin single variable domain with binding specificity for VEGF that is a
VHH and/or an immunoglobulin single variable domain with binding specificity for EGFR that is a VHH.
57. The ligand of any one of claims 37-55, wherein each said immunoglobulin single variable domain with binding specificity for VEGF and each said
immunoglobulin single variable domain with binding specificity for EGFR are selected from the group consisting of a human VH and a human VL-
58. The ligand of any one of claims 37-57, wherein said ligand is an IgG-like format comprising two immunoglobulin single variable domains with binding specificity for VEGF, and two immunoglobulin single variable domains with binding specificity for EGFR.
59. The ligand of any one of claims 37-58, wherein said ligand comprises an antibody Fc region.
60. A ligand that has binding specificity for epidermal growth factor receptor (EGFR), comprising at least one immunoglobulin single variable domain with binding specificity for EGFR, wherein each said immunoglobulin single variable domain with binding specificity for EGFR competes for binding to EGFR with an anti-EGFR domain antibody (dAb) selected from the group consisting of DOM 16- 39 (SEQ ID NO:345), DOMl 6-39-87 (SEQ ID NO:420), DOMl 6-39-100 (SEQ ID NO:423), DOM16-39-107 (SEQ ID NO:430), DOM16-39-109 (SEQ ID NO:432), DOM16-39-115 (SEQ ID NO:438), and DOMl 6-39-200 (SEQ ID NO:441).
61. A ligand that has binding specificity for epidermal growth factor receptor (EGFR), comprising at least one immunoglobulin single variable domain with binding specificity for EGFR, wherein each said immunoglobulin single variable domain with binding specificity for EGFR competes for binding to EGFR with an anti-EGFR domain antibody (dAb) selected from the group consisting of DOM 16- 39-521 (SEQ ID NO:577), DOM16-39-541 (SEQ ID NO:585), DOM16-39-542 (SEQ ID NO:586), DOM16-39-551 (SEQ ID NO:591), DOM16-39-601 (SEQ ID NO:608), DOMl 6-39-604 (SEQ ID NO:611), DOMl 6-39-618 (SEQ ID NO:621), and DOM16-39-619 (SEQ ID NO:622).
62. The ligand of claim 60 or claim 61, wherein said ligand inhibits binding of epidermal growth factor (EGF) and/or transforming growth factor alpha (TGFalpha) to EGFR.
63. The ligand of claim 60 or claim 61, wherein said ligand inhibits the activity of EGFR.
64. The ligand of claim 60 or claim 61, wherein said ligand inhibits the activity of EGFR without substantially inhibiting binding of epidermal growth factor (EGF) and/or transforming growth factor alpha (TGFalpha) to EGFR.
65. The ligand of any one of claims 60-64, wherein each said immunoglobulin single variable domain with binding specificity for EGFR binds EGFR with an affinity (KD) that is between about 100 nM and about 1 pM, as determined by surface plasmon resonance.
66. The ligand of claim 65, wherein each said immunoglobulin single variable domain with binding specificity for EGFR binds EGFR with an affinity (KD) that is between about 10 nM and about 100 pM, as determined by surface plasmon resonance.
67. The ligand of any one of claims 60-64, wherein said ligand binds EGFR with an affinity (KD) that is between about 100 nM and about 1 pM, as determined by surface plasmon resonance.
68. The ligand of claim 67, wherein said ligand binds EGFR with an affinity (KD) that is between about 10 nM and about 100 pM, as determined by surface plasmon resonance.
69. The ligand of any one of claims 60-68 wherein said ligand comprises an immunoglobulin single variable domain with binding specificity for EGFR that is a VHH.
70. The ligand of any one of claims 60-68, wherein each said immunoglobulin single variable domain with binding specificity for EGFR is independently selected from the group consisting of a human VH and a human VL.
71. The ligand of any one of claims 1 -70, wherein said ligand further comprises a toxin.
72. The ligand of claim 71 , wherein said toxin is a surface active toxin.
73. The ligand of claim 72, wherein said surface active toxin comprises a free radical generator or a radionucleotide.
74. The ligand of claim 73, wherein said toxin is a cytotoxin, free radical generator, antimetabolite, protein, polypeptide, peptide, photoactive agent, antisense compound, chemotherapeutic, radionuclide or intrabody.
75. The ligand of any one of claims 1-74, wherein said ligand further comprises a half-life extending moiety.
76. The ligand of claim 75, wherein said half-life extending moiety is a polyalkylene glycol moiety, serum albumin or a fragment thereof, transferrin receptor or a transferrin-binding portion thereof, or a moiety comprising a binding site for a polypeptide that enhances half-life in vivo.
11. The ligand of claim 76, wherein said half-life extending moiety is a moiety comprising a binding site for a polypeptide that enhances half-life in vivo selected from the group consisting of an affibody, a SpA domain, an LDL receptor class A domain, an EGF domain, and an avimer.
78. The ligand of claim 76, wherein said half-life extending moiety is a polyethylene glycol moiety.
19. The ligand of claim 76, wherein said half-life extending moiety is an antibody or antibody fragment comprising a binding site for serum albumin or neonatal Fc receptor.
80. The ligand of claim 79, wherein said antibody or antibody fragment comprising a binding site for serum albumin or neonatal Fc receptor is an antibody fragment, and said antibody fragment is an immunoglobulin single variable domain comprising a binding site for serum albumin.
81. The ligand of claim 80, wherein said immunoglobulin single variable domain comprising a binding site for serum albumin competes for binding to human serum albumin with a dAb selected from the group consisting of DOM7m-16 (SEQ ID NO: 473), DOM7m-12 (SEQ ID NO: 474), DOM7m-26 (SEQ ID NO: 475), DOM7r-l (SEQ ID NO: 476), DOM7r-3 (SEQ ID NO: 477), DOM7r-4 (SEQ ID NO: 478), DOM7r-5 (SEQ ID NO: 479), DOM7r-7 (SEQ ID NO: 480), DOM7r-8 (SEQ ID NO: 481), DOM7h-2 (SEQ ID NO: 482), DOM7h-3 (SEQ ID NO: 483), DOM7h-4 (SEQ ID NO: 484), DOM7h-6 (SEQ ID NO: 485), DOM7h-l (SEQ ID NO: 486), DOM7h-7 (SEQ ID NO: 487), DOM7h-22 (SEQ ID NO: 489), DOM7h- 23 (SEQ ID NO: 490), DOM7h-24 (SEQ ID NO: 491), DOM7h-25 (SEQ ID NO: 492), DOM7h-26 (SEQ ID NO: 493), DOM7h-21 (SEQ ID NO: 494), DOM7h-27 (SEQ ID NO: 495), DOM7h-8 (SEQ ID NO: 496), DOM7r-13 (SEQ ID NO: 497), DOM7r-14 (SEQ ID NO: 498), DOM7r-15 (SEQ ID NO: 499), DOM7r-16 (SEQ ID NO: 500), DOM7r-17 (SEQ ID NO: 501), DOM7r-18 (SEQ ID NO: 502), DOM7r- 19 (SEQ ID NO: 503), DOM7r-20 (SEQ ID NO: 504), DOM7r-21 (SEQ ID NO: 505), DOM7r-22 (SEQ ID NO: 506), DOM7r-23 (SEQ ID NO: 507), DOM7r-24 (SEQ ID NO: 508), DOM7r-25 (SEQ ID NO: 509), DOM7r-26 (SEQ ID NO: 510), DOM7r-27 (SEQ ID NO: 511), DOM7r-28 (SEQ ID NO: 512), DOM7r-29 (SEQ ID NO: 513), DOM7r-30 (SEQ ID NO: 514), DOM7r-31 (SEQ ID NO: 515), DOM7r- 32 (SEQ ID NO: 516), and DOM7r-33 (SEQ ID NO: 517).
82. The ligand of claim 80, wherein said immunoglobulin single variable domain comprising a binding site for serum albumin comprises an amino acid sequence that has at least 85% amino acid sequence identity with the amino acid sequence of a dAb selected from the group consisting of DOM7m-16 (SEQ ID NO: 473), DOM7m-12 (SEQ ID NO: 474), DOM7m-26 (SEQ ID NO: 475), DOM7r-l (SEQ ID NO: 476), DOM7r-3 (SEQ ID NO: 477), DOM7r-4 (SEQ ID NO: 478), DOM7r- 5 (SEQ ID NO: 479), DOM7r-7 (SEQ ID NO: 480), DOM7r-8 (SEQ ID NO: 481), DOM7h-2 (SEQ ID NO: 482), DOM7h-3 (SEQ ID NO: 483), DOM7h-4 (SEQ ID NO: 484), DOM7h-6 (SEQ ID NO: 485), DOM7h-l (SEQ ID NO: 486), DOM7h-7 (SEQ ID NO: 487), DOM7h-22 (SEQ ID NO: 489), DOM7h-23 (SEQ ID NO: 490), DOM7h-24 (SEQ ID NO: 491), DOM7h-25 (SEQ ID NO: 492), DOM7h-26 (SEQ ID NO: 493), DOM7h-21 (SEQ ID NO: 494), DOM7h-27 (SEQ ID NO: 495), DOM7h-8 (SEQ ID NO: 496), DOM7r-13 (SEQ ID NO: 497), DOM7r-14 (SEQ ID NO: 498), DOM7r-15 (SEQ ID NO: 499), DOM7r-16 (SEQ ID NO: 500), DOM7r- 17 (SEQ ID NO: 501), DOM7r-18 (SEQ ID NO: 502), DOM7r-19 (SEQ ID NO: 503), DOM7r-20 (SEQ ID NO: 504), DOM7r-21 (SEQ ID NO: 505), DOM7r-22 (SEQ ID NO: 506), DOM7r-23 (SEQ ID NO: 507), DOM7r-24 (SEQ ID NO: 508), DOM7r-25 (SEQ ID NO: 509), DOM7r-26 (SEQ ID NO: 510), DOM7r-27 (SEQ ID NO: 511), DOM7r-28 (SEQ ID NO: 512), DOM7r-29 (SEQ ID NO: 513), DOM7r- 30 (SEQ ID NO: 514), DOM7r-31 (SEQ ID NO: 515), DOM7r-32 (SEQ ID NO: 516), and DOM7r-33 (SEQ ID NO: 517).
83. A ligand of any one of Claims 1 -70 for use in therapy or diagnosis.
84. A ligand of any one of Claims 1 -70 for use in treating cancer.
85. A ligand of any one of Claims 1-70 for treating cancer cells that overexpress EGFR and/or VEGF.
86. Use of the ligand of any one of Claims 1 -70 for the manufacture of a medicament for treating cancer.
87. Use of the ligand of any one of Claims 1 -70 for the manufacture of a medicament for treating cancer cells that overexpress EGFR and/or VEGF.
88. A method for treating cancer comprising administering to a subject in need thereof a therapeutically effective amount of ligand of any one of claims 1-70.
89. A method for treating cancer, comprising administering to a subject in need thereof a therapeutically effective amount of ligand of any one of claims 1-70 and a chemotherapeutic agent.
90. A method for treating cancer, comprising administering to a subject in need thereof a therapeutically effective amount of ligand of any one of claims 1-70 and an anti-neoplastic composition, wherein said anti-neoplastic composition comprises at least one chemotherapeutic agent selected from the group consisting of alkylating agents, antimetabolites, folic acid analogs, pyrimidine analogs, purine analogs and related inhibitors, vinca alkaloids, epipodopyyllotoxins, antibiotics, L-Asparaginase, topoisomerase inhibitor, interferons, platinum cooridnation complexes,
anthracenedione substituted urea, methyl hydrazine derivatives, adrenocortical suppressant, adrenocorticosteroides, progestins, estrogens, antiestrogen, androgens, antiandrogen, and gonadotropin-releasing hormone analog.
91. The method of claim 90, wherein the chemotherapeutic agent is selected from the group consisting of cisplatin, dicarbazine, dactinomycin, mechlorethamine, streptozocin, cyclophosphamide, capecitabine, carmustine, lomustine, doxorubicin, daunorubicin, procarbazine, mitomycin, cytarabine, etoposide, methotrexate, 5- fluorouracil, vinblastine, vincristine, bleomycin, paclitaxel, docetaxel, doxetaxe, aldesleukin, asparaginase, busulfan, carboplatin, cladribine, dacarbazine, floxuridine, fmdarabine, hydroxyurea, ifosfamide, interferon alpha, irinotecan, leuprolide, leucovorin, megestrol, melphalan, mercaptopurine, oxaliplatin, plicamycin, mitotane, pegaspargase, pentostatin, pipobroman, plicamycin, streptozocin, tamoxifen, teniposide, testolactone, thioguanine, thiotepa, uracil mustard, vinorelbine, chlorambucil, taxol, an additional growth factor receptor . antagonist, and a combination of any of the foregoing.
92. The method of any one of claims 88-91, wherein the cancer is bladder cancer, ovarian cancer, colorectal cancer, breast cancer, lung cancer, gastric cancer, pancreatic cancer, prostate cancer, head and neck cancer, renal cancer and gall bladder cancer.
93. The method of any one of claims 88-91, wherein the cancer is non-small cell lung carcinoma or colorectal carcinoma.
94. A method of administering to a subject anti-VEGF treatment and anti-EGFR treatment, the method comprising simultaneous administration of an anti-VEGF treatment and an anti-EGFR treatment by administering to said subject a
therapeutically effective amount of a ligand of any one of claims 1-59.
95. A composition comprising a ligand of any one of Claims 1-70 and a physiologically acceptable carrier.
96. The composition of claim 95, wherein said composition comprises a vehicle for intravenous, intramuscular, intraperitoneal, intraarterial, intrathecal,
intraarticular, or subcutaneous administration.
97. The composition of claim 95, wherein said composition comprises a vehicle is for pulmonary, intranasal, vaginal, or rectal administration.
98. A drug delivery device comprising the composition of claim 95.
99. A drug delivery device for simultaneously administering to a subject anti- VEGF treatment and anti-EGFR treatment, the device comprising a ligand of any one of claims 1-59.
100. The drug delivery device of claim 98 or 99, wherein said device comprises a plurality of therapeutically effective doses of ligand.
101. The drug delivery device of any one of claims 98-100, wherein said drug delivery device is selected from the group consisting of a parenteral delivery device, intravenous delivery device, intramuscular delivery device, intraperitoneal delivery device, transdermal delivery device, pulmonary delivery device, intraarterial delivery device, intrathecal delivery device, intraarticular delivery device, subcutaneous delivery device, intranasal delivery device, vaginal delivery device, and rectal delivery device.
102. The drug delivery device of claim 101, wherein said device is selected from the group consisting of a syringe, a transdermal delivery device, a capsule, a tablet, a nebulizer, an inhaler, an atomizer, an aerosolizer, a mister, a dry powder inhaler, a metered dose inhaler, a metered dose sprayer, a metered dose mister, a metered dose atomizer, a catheter.
103. Use of the ligand of any one of Claims 1 -70, for the manufacture of a medicament for selectively killing cancer cells over normal cells.
104. Use of the ligand of any one of Claims 1 -70, for the manufacture of a medicament for killing cells
105. The use of claim 104, wherein said ligand comprises an antibody Fc region.
106. An isolated or recombinant nucleic acid encoding a ligand of any one of claims 1-70.
107. A vector comprising the recombinant nucleic acid of claim 106.
108. A host cell comprising the recombinant nucleic acid of claim 106 or the vector of claim 107.
109. A method for producing a ligand comprising maintaining the host cell of claim 108 under conditions suitable for expression of said nucleic acid or vector, whereby a ligand is produced.
110. The method of claim 109, further comprising isolating the ligand.
111. A composition comprising a single domain antibody polypeptide construct that antagonizes human EGFR binding to a receptor, wherein said single domain antibody polypeptide construct comprises a CDR3 sequence that is the same as the sequence of CDR3 of a dAb selected from the group consisting of DOM 16-39 (SEQ ID NO:345), DOM16-39-87 (SEQ ID NO:420), DOM16-39-100 (SEQ ID NO:423), DOM16-39-107 (SEQ ID NO:430), DOM16-39-109 (SEQ ID NO:432), D0M16- 39-115 (SEQ ID NO:438), and DOMl 6-39-200 (SEQ ID NO:441).
112. A composition comprising a single domain antibody polypeptide construct that antagonizes human EGFR binding to a receptor, wherein said single domain antibody polypeptide construct comprises a CDR3 sequence that is the same as the sequence of CDR3 of a dAb selected from the group consisting of DOMl 6-39-521 (SEQ ID NO:577), DOM16-39-541 (SEQ ID NO:585), DOM16-39-542 (SEQ ID NO:586), DOM16-39-551 (SEQ ID NO:591), DOM16-39-601 (SEQ ID NO:608), DOM16-39-604 (SEQ ID NO:611), DOM16-39-618 (SEQ ID NO:621), and DOM16-39-619 (SEQ ID NO:622).
113. A composition comprising a single domain antibody polypeptide construct that antagonizes human EGFR binding to a receptor, wherein said single domain antibody polypeptide construct comprises an amino acid sequence selected from the group consisting of DOM16-39 (SEQ ID NO:345), DOM16-39-87 (SEQ ID
NO:420), DOM16-39-100 (SEQ ID NO:423), DOM16-39-107 (SEQ ID NO:430), DOM16-39-109 (SEQ ID NO:432), DOM16-39-115 (SEQ ID NO:438), DOM16- 39-200 (SEQ ID NO:441), and a sequence at least 85% identical to any one of the foregoing.
114. A composition comprising a single domain antibody polypeptide construct that antagonizes human EGFR binding to a receptor, wherein said single domain antibody polypeptide construct comprises an amino acid sequence selected from the group consisting of DOMl 6-39-521 (SEQ ID NO:577), DOMl 6-39-541 (SEQ ID NO:585), DOM16-39-542 (SEQ ID NO:586), DOM16-39-551 (SEQ ID NO:591), DOMl 6-39-601 (SEQ ID NO:608), DOMl 6-39-604 (SEQ ID NO:611), DOMl 6- 39-618 (SEQ ID NO:621), DOM16-39-619 (SEQ ID NO:622), and a sequence at least 85% identical to any one of the foregoing.
115. A composition comprising a single domain antibody polypeptide construct that antagonizes human EGFR binding to a receptor, wherein said single domain antibody polypeptide construct comprises an amino acid sequence selected from the group consisting of DOMl 6-39 (SEQ ID NO:345), DOMl 6-39-87 (SEQ ID NO:420), DOM16-39-100 (SEQ ID NO:423), DOM16-39-107 (SEQ ID NO:430), DOM16-39-109 (SEQ ID NO:432), DOM16-39-115 (SEQ ID NO:438), D0M16- 39-200 (SEQ ID NO:441), and a sequence at least 85% identical to any one of the foregoing.
116. A composition comprising a single domain antibody polypeptide construct that antagonizes human EGFR binding to a receptor, wherein said single domain antibody polypeptide construct comprises an amino acid sequence selected from the group consisting of DOMl 6-39-521 (SEQ ID NO:577), DOMl 6-39-541 (SEQ ID NO:585), DOMl 6-39-542 (SEQ ID NO:586), DOMl 6-39-551 (SEQ ID NO:591), DOM16-39-601 (SEQ ID NO:608), DOM16-39-604 (SEQ ID NO:611), D0M16- 39-618 (SEQ ID NO:621), DOM16-39-619 (SEQ ID NO:622), and a sequence at least 85% identical to any one of the foregoing.
117. A composition comprising a single domain antibody polypeptide construct that antagonizes human EGFR binding to a receptor, wherein said single domain antibody polypeptide construct comprises an amino acid sequence selected from the group consisting of DOMl 6-39 (SEQ ID NO:345), DOMl 6-39-87 (SEQ ID NO:420), DOMl 6-39-100 (SEQ ID NO:423), DOMl 6-39-107 (SEQ ID NO:430), DOM16-39-109 (SEQ ID NO:432), DOM16-39-115 (SEQ ID NO:438), D0M16- 39-200 (SEQ ID NO:441), and a sequence at least 92% identical to any one of the foregoing.
118. A composition comprising a single domain antibody polypeptide construct that antagonizes human EGFR binding to a receptor, wherein said single domain antibody polypeptide construct comprises an amino acid sequence selected from the group consisting of DOMl 6-39-521 (SEQ ID NO:577), DOMl 6-39-541 (SEQ ID NO:585), DOM16-39-542 (SEQ ID NO:586), DOM16-39-551 (SEQ ID NO:591), DOM16-39-601 (SEQ ID NO:608), DOM16-39-604 (SEQ ID NO:611), DOM16- 39-618 (SEQ ID NO:621), DOM16-39-619 (SEQ ID NO:622), and a sequence at least 92% identical to any one of the foregoing.
119. A composition comprising a single domain antibody polypeptide construct that antagonizes human EGFR binding to a receptor, wherein said single domain antibody polypeptide construct comprises an amino acid sequence selected from the group consisting of DOM16-39 (SEQ ID NO:345), DOM16-39-87 (SEQ ID NO:420), DOM16-39-100 (SEQ ID NO:423), DOM16-39-107 (SEQ ID NO:430), DOM16-39-109 (SEQ ID NO:432), DOM16-39-115 (SEQ ID NO:438), D0M16- 39-200 (SEQ ID NO:441), and a sequence at least 94% identical to any one of the foregoing.
120. A composition comprising a single domain antibody polypeptide construct that antagonizes human EGFR binding to a receptor, wherein said single domain antibody polypeptide construct comprises an amino acid sequence selected from the group consisting of DOM16-39-521 (SEQ ID NO:577), DOM16-39-541 (SEQ ID NO:585), DOM16-39-542 (SEQ ID NO:586), DOM16-39-551 (SEQ ID NO:591), DOM16-39-601 (SEQ ID NO:608), DOM16-39-604 (SEQ ID NO:611), D0M16- 39-618 (SEQ ID NO:621), DOM16-39-619 (SEQ ID NO:622), and a sequence at least 94% identical to any one of the foregoing.
121. A composition comprising a single domain antibody polypeptide construct that antagonizes human EGFR binding to a receptor, wherein said single domain antibody polypeptide construct comprises an amino acid sequence selected from the group consisting of DOMl 6-39 (SEQ ID NO:345), DOMl 6-39-87 (SEQ ID NO:420), DOM16-39-100 (SEQ ID NO:423), DOM16-39-107 (SEQ ID NO:430), DOM16-39-109 (SEQ ID NO:432), DOM16-39-115 (SEQ ID NO:438), D0M16- 39-200 (SEQ ID NO:441), and a sequence at least 96% identical to any one of the foregoing.
122. A composition comprising a single domain antibody polypeptide construct that antagonizes human EGFR binding to a receptor, wherein said single domain antibody polypeptide construct comprises an amino acid sequence selected from the group consisting of DOM16-39-521 (SEQ ID NO:577), DOM16-39-541 (SEQ ID NO:585), DOM16-39-542 (SEQ ID NO:586), DOM16-39-551 (SEQ ID NO:591), DOM16-39-601 (SEQ ID NO:608), DOM16-39-604 (SEQ ID NO:611), D0M16- 39-618 (SEQ ID NO:621), DOM16-39-619 (SEQ ID NO:622) and a sequence at least 96% identical to any one of the foregoing.
123. A composition comprising a single domain antibody polypeptide construct that antagonizes human EGFR binding to a receptor, wherein said single domain antibody polypeptide construct comprises an amino acid sequence selected from the group consisting of DOM16-39 (SEQ ID NO:345), DOM16-39-87 (SEQ ID NO:420), DOM16-39-100 (SEQ ID NO:423), DOM16-39-107 (SEQ ID NO:430), DOMl 6-39-109 (SEQ ID NO:432), DOMl 6-39-115 (SEQ ID NO:438), DOM16- 39-200 (SEQ ID NO:441), and a sequence at least 98% identical to any one of the foregoing.
124. A composition comprising a single domain antibody polypeptide construct that antagonizes human EGFR binding to a receptor, wherein said single domain antibody polypeptide construct comprises an amino acid sequence selected from the group consisting of DOMl 6-39-521 (SEQ ID NO:577), DOM16-39-541 (SEQ ID NO:585), DOM16-39-542 (SEQ ID NO:586), DOM16-39-551 (SEQ ID NO:591), DOM16-39-601 (SEQ ID NO:608), DOM16-39-604 (SEQ ID NO:611), D0M16- 39-618 (SEQ ID NO:621), DOM16-39-619 (SEQ ID NO:622), and a sequence at least 98% identical to any one of the foregoing.
125. A composition comprising a single domain antibody polypeptide construct that antagonizes human EGFR binding to a receptor, wherein said single domain antibody polypeptide construct comprises an amino acid sequence selected from the group consisting of DOMl 6-39 (SEQ ID NO:345), DOMl 6-39-87 (SEQ ID NO:420), DOM16-39-100 (SEQ ID NO:423), DOM16-39-107 (SEQ ID NO:430), DOMl 6-39-109 (SEQ ID NO:432), DOMl 6-39-115 (SEQ ID NO:438), DOMl 6- 39-200 (SEQ ID NO:441), and a sequence at least 99% identical to any one of the foregoing.
126. A composition comprising a single domain antibody polypeptide construct that antagonizes human EGFR binding to a receptor, wherein said single domain antibody polypeptide construct comprises an amino acid sequence selected from the group consisting of DOM16-39-521 (SEQ ID NO:577), DOM16-39-541 (SEQ ID NO:585), DOM16-39-542 (SEQ ID NO:586), DOM16-39-551 (SEQ ID NO:591), DOM16-39-601 (SEQ ID NO:608), DOM16-39-604 (SEQ ID NO:611), DOM16- 39-618 (SEQ ID NO:621), DOM16-39-619 (SEQ ID NO:622), and a sequence at least 99% identical to any one of the foregoing.
127. A composition of any one of claims 111-125 wherein said single domain antibody polypeptide construct comprises a tetravalent, dual-specific antibody polypeptide construct comprising:
a) a first copy of a first fusion protein comprising a single domain antibody polypeptide that binds a first epitope, fused to an IgG heavy chain constant domain; b) a second copy of said first fusion protein;
c) a first copy of a second fusion protein comprising a single domain antibody polypeptide that binds a second epitope, fused to a light chain constant domain;
d) a second copy of said second fusion protein;
wherein said first and said second copies of said first fusion protein are disulfide bonded to each other via their respective IgG heavy chain constant domains, and
wherein said light chain constant domain of said first copy of said second fusion protein is disulfide bonded to the IgG heavy chain constant domain of said first copy of said first fusion protein, and
wherein said light chain constant domain of said second copy of second fusion protein is disulfide bonded to the IgG heavy chain constant domain of said second copy of said first fusion protein, and
wherein said polypeptide construct binds said first and said second epitopes.
128. The composition of claim 111 wherein said first and/or said second epitope is a EGFR epitope.
129. An immunoglobulin single variable domain polypeptide that binds EGFR, wherein the polypeptide has an amino acid sequence that is identical to the amino acid sequence of DOM16-39 (SEQ ID NO:345), DOM16-39-87 (SEQ ID NO:420), DOM16-39-100 (SEQ ID NO:423), DOM16-39-107 (SEQ ID NO:430), D0M16- 39-109 (SEQ ID NO:432), DOM16-39-115 (SEQ ID NO:438), and DOMl 6-39-200 (SEQ ID NO:441), or differs from the amino acid sequence of DOM16-39 (SEQ ID NO:345), DOM16-39-87 (SEQ ID NO:420), DOM16-39-100 (SEQ ID NO:423), DOMl 6-39-107 (SEQ ID NO:430), DOM16-39-109 (SEQ ID NO:432), D0M16- 39-115 (SEQ ID NO:438), and DOM16-39-200 (SEQ ID NO:441) at no more than 25 amino acid positions and has a CDRl sequence that has at least 50% identity to the CDRl sequence of DOMl 6-39 (SEQ ID NO:345), DOMl 6-39-87 (SEQ ID NO:420), DOM16-39-100 (SEQ ID NO:423), DOM16-39-107 (SEQ ID NO:430), DOM16-39-109 (SEQ ID NO:432), DOM16-39-115 (SEQ ID NO:438), and DOMl 6-39-200 (SEQ ID NO:441).
130. An immunoglobulin single variable domain polypeptide that binds EGFR, wherein the polypeptide has an amino acid sequence that is identical to the amino acid sequence of DOMl 6-39-521 (SEQ ID NO:577), DOMl 6-39-541 (SEQ ID NO:585), DOM16-39-542 (SEQ ID NO:586), DOM16-39-551 (SEQ ID NO:591), DOM16-39-601 (SEQ ID NO:608), DOM16-39-604 (SEQ ID NO:611), D0M16- 39-618 (SEQ ID NO:621), and DOM16-39-619 (SEQ ID NO:622), or differs from the amino acid sequence of DOM16-39-521 (SEQ ID NO:577), DOM16-39-541 (SEQ ID NO:585), DOM16-39-542 (SEQ ID NO:586), DOM16-39-551 (SEQ ID NO:591), DOM16-39-601 (SEQ ID NO:608), DOM16-39-604 (SEQ ID NO:611), DOM16-39-618 (SEQ ID NO:621), and DOM 16-39-619 (SEQ ID NO:622) at no more than 25 amino acid positions and has a CDRl sequence that has at least 50% identity to the CDRl sequence of DOMl 6-39-521 (SEQ ID NO:577), DOM16-39- 541 (SEQ ID NO:585), DOM16-39-542 (SEQ ID NO:586), DOM16-39-551 (SEQ ID NO:591), DOM16-39-601 (SEQ ID NO:608), DOM16-39-604 (SEQ ID NO:611), DOM16-39-618 (SEQ ID NO:621), and DOM 16-39-619 (SEQ ID
NO:622).
131. An immunoglobulin single variable domain polypeptide that binds to EGFR, wherein the polypeptide has an amino acid sequence that is identical to the amino acid sequence of DOMl 6-39 (SEQ ID NO:345), DOMl 6-39-87 (SEQ ID NO:420), DOM16-39-100 (SEQ ID NO:423), DOM16-39-107 (SEQ ID NO:430), D0M16- 39-109 (SEQ ID NO:432), DOMl 6-39-115 (SEQ ID NO:438), and DOMl 6-39-200 (SEQ ID NO:441), or differs from the amino acid sequence of DOM16-39 (SEQ ID NO:345), DOM16-39-87 (SEQ ID NO:420), DOM16-39-100 (SEQ ID NO:423), DOM16-39-107 (SEQ ID NO:430), DOM16-39-109 (SEQ ID NO:432), DOM16- 39-115 (SEQ ID NO:438), and DOM16-39-200 (SEQ ID NO:441) at no more than 25 amino acid positions and has a CDR2 sequence that has at least 50% identity to the CDR2 sequence of DOM16-39 (SEQ ID NO:345), DOM16-39-87 (SEQ ID NO:420), DOM16-39-100 (SEQ ID NO:423), DOM16-39-107 (SEQ ID NO:430), DOMl 6-39-109 (SEQ ID NO:432), DOMl 6-39-115 (SEQ ID NO:438), and DOMl 6-39-200 (SEQ ID NO:441).
132. An immunoglobulin single variable domain polypeptide that binds EGFR, wherein the polypeptide has an amino acid sequence that is identical to the amino acid sequence of DOMl 6-39-521 (SEQ ID NO:577), DOMl 6-39-541 (SEQ ID NO:585), DOM16-39-542 (SEQ ID NO:586), DOM16-39-551 (SEQ ID NO:591), DOMl 6-39-601 (SEQ ID NO:608), DOMl 6-39-604 (SEQ ID N0:611), DOMl 6- 39-618 (SEQ ID NO:621), and DOM16-39-619 (SEQ ID NO:622), or differs from the amino acid sequence of DOM16-39-521 (SEQ ID NO:577), DOM16-39-541 (SEQ ID NO:585), DOM16-39-542 (SEQ ID NO:586), DOM16-39-551 (SEQ ID NO:591), DOM16-39-601 (SEQ ID NO:608), DOM16-39-604 (SEQ ID N0:611), DOM16-39-618 (SEQ ID NO:621), and DOM16-39-619 (SEQ ID NO:622) at no more than 25 amino acid positions and has a CDR2 sequence that has at least 50% identity to the CDR2 sequence of DOM16-39-521 (SEQ ID NO:577), DOM16-39- 541 (SEQ ID NO:585), DOM16-39-542 (SEQ ID NO:586), DOM16-39-551 (SEQ ID NO:591), DOM16-39-601 (SEQ ID NO:608), DOM16-39-604 (SEQ ID NO:611), DOMl 6-39-618 (SEQ ID NO:621), and DOM16-39-619 (SEQ ID NO:622).
133. An immunoglobulin single variable domain polypeptide that binds EGFR, wherein the polypeptide has an amino acid sequence that is identical to the amino acid sequence of DOM16-39 (SEQ ID NO:345), DOM16-39-87 (SEQ ID NO:420), DOM16-39-100 (SEQ ID NO:423), DOM16-39-107 (SEQ ID NO:430), D0M16- 39-109 (SEQ ID NO:432), DOM16-39-115 (SEQ ID NO:438), and DOM16-39-200 (SEQ ID NO:441), or differs from the amino acid sequence of DOMl 6-39 (SEQ ID NO:345), DOM16-39-87 (SEQ ID NO:420), DOM16-39-100 (SEQ ID NO:423), DOM16-39-107 (SEQ ID NO:430), DOM16-39-109 (SEQ ID NO:432), DOM16- 39-115 (SEQ ID NO:438), and DOM16-39-200 (SEQ ID NO:441) at no more than 25 amino acid positions and has a CDR3 sequence that has at least 50% identity to the CDR3 sequence of DOM16-39 (SEQ ID NO:345), DOM16-39-87 (SEQ ID NO:420), DOM16-39-100 (SEQ ID NO:423), DOM16-39-107 (SEQ ID NO:430), DOM16-39-109 (SEQ ID NO:432), DOM16-39-115 (SEQ ID NO:438), and DOM16-39-200 (SEQ ID NO:441).
134. An immunoglobulin single variable domain polypeptide that binds EGFR, wherein the polypeptide has an amino acid sequence that is identical to the amino acid sequence of DOM16-39-521 (SEQ ID NO:577), DOM16-39-541 (SEQ ID NO:585), DOM16-39-542 (SEQ ID NO:586), DOM16-39-551 (SEQ ID NO:591), DOMl 6-39-601 (SEQ ID NO:608), DOMl 6-39-604 (SEQ ID NO:611), DOMl 6- 39-618 (SEQ ID NO:621), and DOM16-39-619 (SEQ ID NO:622), or differs from the amino acid sequence of DOM16-39-521 (SEQ ID NO:577), DOM16-39-541 (SEQ ID NO:585), DOM16-39-542 (SEQ ID NO:586), DOM16-39-551 (SEQ ID NO:591), DOM16-39-601 (SEQ ID NO:608), DOM16-39-604 (SEQ ID NO:611), DOM16-39-618 (SEQ ID NO:621), and DOM16-39-619 (SEQ ID NO:622) at no more than 25 amino acid positions and has a CDR3 sequence that has at least 50% identity to the CDR3 sequence of DOM16-39-521 (SEQ ID NO:577), DOM16-39- 541 (SEQ ID NO:585), DOMl 6-39-542 (SEQ ID NO:586), DOMl 6-39-551 (SEQ ID NO:591), DOM16-39-601 (SEQ ID NO:608), DOM16-39-604 (SEQ ID NO.-611), DOM16-39-618 (SEQ ID NO:621), and DOM16-39-619 (SEQ ID NO:622).
135. An immunoglobulin single variable domain polypeptide that binds EGFR, wherein the polypeptide has an amino acid sequence that is identical to the amino acid sequence of DOM16-39 (SEQ ID NO:345), DOM16-39-87 (SEQ ID NO:420), DOM16-39-100 (SEQ ID NO:423), DOM16-39-107 (SEQ ID NO:430), DOM16- 39-109 (SEQ ID NO:432), DOM16-39-115 (SEQ ID NO:438), and DOM16-39-200 (SEQ ID NO:441), or differs from the amino acid sequence of DOM16-39 (SEQ ID NO:345), DOM16-39-87 (SEQ ID NO:420), DOM16-39-100 (SEQ ID NO:423), DOM16-39-107 (SEQ ID NO:430), DOM16-39-109 (SEQ ID NO:432), DOM16- 39-115 (SEQ ID NO:438), and DOM16-39-200 (SEQ ID NO:441) at no more than 25 amino acid positions and has a CDRl sequence that has at least 50% identity to the CDRl sequence of DOM16-39 (SEQ ID NO:345), DOM16-39-87 (SEQ ID NO:420), DOM16-39-100 (SEQ ID NO:423), DOM16-39-107 (SEQ ID NO:430), DOM16-39-109 (SEQ ID NO:432), DOM16-39-115 (SEQ ID NO:438), and DOM16-39-200 (SEQ ID NO:441) and has a CDR2 sequence has at least 50% identity to the CDR2 sequence of DOM16-39 (SEQ ID NO:345), DOM16-39-87 (SEQ ID NO:420), DOM16-39-100 (SEQ ID NO:423), DOM16-39-107 (SEQ ID NO:430), DOMl 6-39-109 (SEQ ID NO:432), DOM16-39-115 (SEQ ID NO:438), and DOM16-39-200 (SEQ ID NO:441).
136. An immunoglobulin single variable domain polypeptide that binds EGFR, wherein the polypeptide has an amino acid sequence that is identical to the amino acid sequence of DOM16-39-521 (SEQ ID NO:577), DOM16-39-541 (SEQ ID NO:585), DOM16-39-542 (SEQ ID NO:586), DOM16-39-551 (SEQ ID NO:591), DOM16-39-601 (SEQ ID NO:608), DOM16-39-604 (SEQ ID NO:611), D0M16- 39-618 (SEQ ID NO:621), and DOM16-39-619 (SEQ ID NO:622), or differs from the amino acid sequence of DOM16-39-521 (SEQ ID NO:577), DOM16-39-541 (SEQ ID NO:585), DOM16-39-542 (SEQ ID NO:586), DOM16-39-551 (SEQ ID NO:591), DOM16-39-601 (SEQ ID NO:608), DOM16-39-604 (SEQ ID NO:611), DOMl 6-39-618 (SEQ ID NO:621), and DOMl 6-39-619 (SEQ ID NO:622) at no more than 25 amino acid positions and has a CDRl sequence that has at least 50% identity to the CDRl sequence of DOM16-39-521 (SEQ ID NO:577), DOM16-39- 541 (SEQ ID NO:585), DOM16-39-542 (SEQ ID NO:586), DOM16-39-551 (SEQ ID NO:591), DOM16-39-601 (SEQ ID NO:608), DOM16-39-604 (SEQ ID
NO:611), DOM16-39-618 (SEQ ID NO:621), and DOM 16-39-619 (SEQ ID
NO:622) and has a CDR2 sequence has at least 50% identity to the CDR2 sequence of DOM16-39-521 (SEQ ID NO:577), DOM16-39-541 (SEQ ID NO:585), D0M16- 39-542 (SEQ ID NO:586), DOM16-39-551 (SEQ ID NO:591), DOM16-39-601 (SEQ ID NO:608), DOM16-39-604 (SEQ ID NO:611), DOM16-39-618 (SEQ ID NO:621), and DOM16-39-619 (SEQ ID NO:622).
137. An immunoglobulin single variable domain polypeptide that binds EGFR, wherein the polypeptide has an amino acid sequence that is identical to the amino acid sequence of DOMl 6-39 (SEQ ID NO:345), DOMl 6-39-87 (SEQ ID NO:420), DOMl 6-39-100 (SEQ ID NO:423), DOM16-39-107 (SEQ ID NO:430), DOM16- 39-109 (SEQ ID NO:432), DOM16-39-115 (SEQ ID NO:438), and DOM16-39-200 (SEQ ID NO:441), or differs from the amino acid sequence of DOMl 6-39 (SEQ ID NO:345), DOMl 6-39-87 (SEQ ID NO:420), DOMl 6-39-100 (SEQ ID NO:423), DOM16-39-107 (SEQ ID NO:430), DOM16-39-109 (SEQ ID NO:432), D0M16- 39-115 (SEQ ID NO:438), and DOM16-39-200 (SEQ ID NO:441) at no more than 25 amino acid positions and has a CDR2 sequence that has at least 50% identity to the CDR2 sequence of DOM16-39 (SEQ ID NO:345), DOM16-39-87 (SEQ ID NO:420), DOMl 6-39-100 (SEQ ID NO:423), DOMl 6-39-107 (SEQ ID NO:430), DOM16-39-109 (SEQ ID NO:432), DOM16-39-115 (SEQ ID NO:438), and DOM16-39-200 (SEQ ID NO:441) and has a CDR3 sequence that has at least 50% identity to the CDR3 sequence of DOM16-39 (SEQ ID NO:345), DOM16-39-87 (SEQ ID NO:420), DOMl 6-39-100 (SEQ ID NO:423), DOMl 6-39-107 (SEQ ID NO:430), DOM16-39-109 (SEQ ID NO:432), DOM16-39-115 (SEQ ID NO:438), and DOMl 6-39-200 (SEQ ID NO:441).
138. An immunoglobulin single variable domain polypeptide that binds EGFR, wherein the polypeptide has an amino acid sequence that is identical to the amino acid sequence of DOM 16-39-521 (SEQ ID NO:577), DOM16-39-541 (SEQ ID NO:585), DOM16-39-542 (SEQ ID NO:586), DOM16-39-551 (SEQ ID NO:591), DOM16-39-601 (SEQ ID NO:608), DOM16-39-604 (SEQ ID NO:611), D0M16- 39-618 (SEQ ID NO:621), and DOM16-39-619 (SEQ ID NO:622), or differs from the amino acid sequence of DOM16-39-521 (SEQ ID NO:577), DOM16-39-541 (SEQ ID NO:585), DOM16-39-542 (SEQ ID NO:586), DOM16-39-551 (SEQ ID NO:591), DOM16-39-601 (SEQ ID NO:608), DOM16-39-604 (SEQ ID NO:611), DOM16-39-618 (SEQ ID NO:621), and DOM16-39-619 (SEQ ID NO:622) at no more than 25 amino acid positions and has a CDR2 sequence that has at least 50% identity to the CDR2 sequence of DOM16-39-521 (SEQ ID NO:577), DOM16-39- 541 (SEQ ID NO:585), DOM16-39-542 (SEQ ID NO:586), DOM16-39-551 (SEQ ID NO:591), DOM16-39-601 (SEQ ID NO:608), DOMl 6-39-604 (SEQ ID
NO:611), DOM16-39-618 (SEQ ID NO:621), and DOM16-39-619 (SEQ ID
NO:622) and has a CDR3 sequence has at least 50% identity to the CDR3 sequence of DOMl 6-39-521 (SEQ ID NO:577), DOM16-39-541 (SEQ ID NO:585), D0M16- 39-542 (SEQ ID NO:586), DOM16-39-551 (SEQ ID NO:591), DOM16-39-601 (SEQ ID NO:608), DOM16-39-604 (SEQ ID NO:611), DOM16-39-618 (SEQ ID NO:621), and DOM16-39-619 (SEQ ID NO:622).
139. An immunoglobulin single variable domain polypeptide that binds EGFR, wherein the polypeptide has an amino acid sequence that is identical to the amino acid sequence of DOM16-39 (SEQ ID NO:345), DOM16-39-87 (SEQ ID NO:420), DOM16-39-100 (SEQ ID NO:423), DOM16-39-107 (SEQ ID NO:430), D0M16- 39-109 (SEQ ID NO:432), DOM16-39-115 (SEQ ID NO:438), and DOM16-39-200 (SEQ ID NO:441), or differs from the amino acid sequence of DOMl 6-39 (SEQ ID NO:345), DOM16-39-87 (SEQ ID NO:420), DOM16-39-100 (SEQ ID NO:423), DOM16-39-107 (SEQ ID NO:430), DOM16-39-109 (SEQ ID NO:432), D0M16- 39-115 (SEQ ID NO:438), and DOM16-39-200 (SEQ ID NO:441) at no more than 25 amino acid positions and has a CDRl sequence that has at least 50% identity to the CDRl sequence of DOM16-39 (SEQ ID NO:345), DOM16-39-87 (SEQ ID NO:420), DOMl 6-39-100 (SEQ ID NO:423), DOMl 6-39-107 (SEQ ID NO:430), DOM16-39-109 (SEQ ID NO:432), DOM16-39-115 (SEQ ID NO:438), and DOMl 6-39-200 (SEQ ID NO:441) and has a CDR3 sequence that has at least 50% identity to the CDR3 sequence of DOM16-39 (SEQ ID NO:345), DOM16-39-87 (SEQ ID NO:420), DOM16-39-100 (SEQ ID NO:423), DOMl 6-39-107 (SEQ ID NO:430), DOM16-39-109 (SEQ ID NO:432), DOM16-39-115 (SEQ ID NO:438), and DOM16-39-200 (SEQ ID NO:441).
140. An immunoglobulin single variable domain polypeptide that binds EGFR, wherein the polypeptide has an amino acid sequence that is identical to the amino acid sequence of DOMl 6-39-521 (SEQ ID NO:577), DOM16-39-541 (SEQ ID NO:585), DOM16-39-542 (SEQ ID NO:586), DOM16-39-551 (SEQ ID NO:591), DOM16-39-601 (SEQ ID NO:608), DOM16-39-604 (SEQ ID NO:611), DOM16- 39-618 (SEQ ID NO:621), and DOM16-39-619 (SEQ ID NO:622), or differs from the amino acid sequence of DOM16-39-521 (SEQ ID NO:577), DOM16-39-541 (SEQ ID NO:585), DOM16-39-542 (SEQ ID NO:586), DOM16-39-551 (SEQ ID NO:591), DOMl 6-39-601 (SEQ ID NO:608), DOMl 6-39-604 (SEQ ID NO:611), DOM16-39-618 (SEQ ID NO:621), and DOM 16-39-619 (SEQ ID NO:622) at no more than 25 amino acid positions and has a CDRl sequence that has at least 50% identity to the CDRl sequence of DOM16-39-521 (SEQ ID NO:577), DOM16-39- 541 (SEQ ID NO:585), DOM16-39-542 (SEQ ID NO:586), DOM16-39-551 (SEQ ID NO:591), DOMl 6-39-601 (SEQ ID NO:608), DOMl 6-39-604 (SEQ ID
NO:611), DOM16-39-618 (SEQ ID NO:621), and DOM16-39-619 (SEQ ID
NO:622) and has a CDR3 sequence has at least 50% identity to the CDR3 sequence of DOMl 6-39-521 (SEQ ID NO:577), DOMl 6-39-541 (SEQ ID NO:585), DOMl 6- 39-542 (SEQ ID NO:586), DOM16-39-551 (SEQ ID NO:591), DOM16-39-601 (SEQ ID NO:608), DOM16-39-604 (SEQ ID NO:611), DOM16-39-618 (SEQ ID NO:621), and DOM16-39-619 (SEQ ID NO:622).
141. An immunoglobulin single variable domain polypeptide that binds EGFR, wherein the polypeptide has an amino acid sequence that is identical to the amino acid sequence of DOM16-39 (SEQ ID NO:345), DOM16-39-87 (SEQ ID NO:420), DOM16-39-100 (SEQ ID NO:423), DOM16-39-107 (SEQ ID NO:430), D0M16- 39-109 (SEQ ID NO:432), DOMl 6-39-115 (SEQ ID NO:438), and DOMl 6-39-200 (SEQ ID NO:441), or differs from the amino acid sequence of DOM16-39 (SEQ ID NO:345), DOMl 6-39-87 (SEQ ID NO:420), DOMl 6-39-100 (SEQ ID NO:423), DOM16-39-107 (SEQ ID NO:430), DOM16-39-109 (SEQ ID NO:432), D0M16- 39-115 (SEQ ID NO:438), and DOM16-39-200 (SEQ ID NO:441) at no more than 25 amino acid positions and has a CDRl sequence that has at least 50% identity to the CDRl sequence of DOMl 6-39 (SEQ ID NO:345), DOMl 6-39-87 (SEQ ID NO:420), DOMl 6-39-1 OO (SEQ ID NO:423), DOMl 6-39-107 (SEQ ID NO:430), DOM16-39-109 (SEQ ID NO:432), DOM16-39-115 (SEQ ID NO:438), and DOMl 6-39-200 (SEQ ID NO:441) and has a CDR2 sequence that has at least 50% identity to the CDR2 sequence of DOMl 6-39 (SEQ ID NO:345), DOMl 6-39-87 (SEQ ID NO:420), DOMl 6-39-1 OO (SEQ ID NO:423), DOMl 6-39-107 (SEQ ID NO:430), DOM16-39-109 (SEQ ID NO:432), DOM16-39-115 (SEQ ID NO:438), and DOMl 6-39-200 (SEQ ID NO:441) and has a CDR3 sequence that has at least 50% identity to the CDR3 sequence of DOM16-39 (SEQ ID NO:345), DOM16-39- 87 (SEQ ID NO:420), DOM16-39-100 (SEQ ID NO:423), DOM16-39-107 (SEQ ID NO:430), DOM16-39-109 (SEQ ID NO:432), DOMl 6-39-115 (SEQ ID NO:438), and DOM16-39-200 (SEQ ID NO:441).
142. An immunoglobulin single variable domain polypeptide that binds EGFR, wherein the polypeptide has an amino acid sequence that is identical to the amino acid sequence of DOM16-39-521 (SEQ ID NO:577), DOM16-39-541 (SEQ ID NO:585), DOM16-39-542 (SEQ ID NO:586), DOM16-39-551 (SEQ ID NO:591), DOM16-39-601 (SEQ ID NO:608), DOM16-39-604 (SEQ ID NO:611), D0M16- 39-618 (SEQ ID NO:621), and DOM16-39-619 (SEQ ID NO:622), or differs from the amino acid sequence of DOM16-39-521 (SEQ ID NO:577), DOM16-39-541 (SEQ ID NO:585), DOMl 6-39-542 (SEQ ID NO:586), DOMl 6-39-551 (SEQ ID NO:591), DOMl 6-39-601 (SEQ ID NO:608), DOMl 6-39-604 (SEQ ID N0:611), DOM16-39-618 (SEQ ID NO:621), and DOMl 6-39-619 (SEQ ID NO:622) at no more than 25 amino acid positions and has a CDRl sequence that has at least 50% identity to the CDRl sequence of DOM16-39-521 (SEQ ID NO:577), DOM16-39- 541 (SEQ ID NO:585), DOM16-39-542 (SEQ ID NO:586), DOM16-39-551 (SEQ ID NO:591), DOM16-39-601 (SEQ ID NO:608), DOM16-39-604 (SEQ ID NO:611), DOM16-39-618 (SEQ ID NO:621), and DOM16-39-619 (SEQ ID
NO:622) and has a CDR2 sequence that has at least 50% identity to the CDR2 sequence of DOM16-39-521 (SEQ ID NO:577), DOM16-39-541 (SEQ ID NO:585), DOM16-39-542 (SEQ ID NO:586), DOM16-39-551 (SEQ ID NO:591), D0M16- 39-601 (SEQ ID NO:608), DOM16-39-604 (SEQ ID NO:611), DOM16-39-618 (SEQ ID NO:621), and DOMl 6-39-619 (SEQ ID NO:622) and has a CDR3 sequence that has at least 50% identity to the CDR3 sequence of DOMl 6-39-521 (SEQ ID NO:577), DOMl 6-39-541 (SEQ ID NO:585), DOMl 6-39-542 (SEQ ID NO:586), DOM16-39-551 (SEQ ID NO:591), DOM16-39-601 (SEQ ID NO:608), DOM16-39-604 (SEQ ID NO:611), DOM16-39-618 (SEQ ID NO:621), and DOMl 6-39-619 (SEQ ID NO:622).
143. An EGFR antagonist having a CDRl sequence that has at least 50% identity to the CDRl sequence of DOMl 6-39 (SEQ ID NO:345), DOMl 6-39-87 (SEQ ID NO:420), DOM16-39-100 (SEQ ID NO:423), DOM16-39-107 (SEQ ID NO:430), DOM16-39-109 (SEQ ID NO:432), DOM16-39-115 (SEQ ID NO:438), and DOM16-39-200 (SEQ ID NO:441).
144. An EGFR antagonist having a CDRl sequence that has at least 50% identity to the CDRl sequence of DOM16-39-521 (SEQ ID NO:577), DOM16-39-541 (SEQ
ID NO:585), DOM16-39-542 (SEQ ID NO:586), DOM16-39-551 (SEQ ID
NO:591), DOM16-39-601 (SEQ ID NO:608), DOM16-39-604 (SEQ ID NO:611), DOM16-39-618 (SEQ ID NO:621), and DOM16-39-619 (SEQ ID NO:622).
145. An EGFR antagonist having a CDR2 sequence that has at least 50% identity to the CDR2 sequence of DOMl 6-39 (SEQ ID NO:345), DOMl 6-39-87 (SEQ ID NO:420), DOM16-39-100 (SEQ ID NO:423), DOM16-39-107 (SEQ ID NO:430), DOM16-39-109 (SEQ ID NO:432), DOM16-39-115 (SEQ ID NO:438), and DOM16-39-200 (SEQ ID NO:441).
146. An EGFR antagonist having a CDR2 sequence that has at least 50% identity to the CDR2 sequence of DOM16-39-521 (SEQ ID NO:577), DOM16-39-541 (SEQ ID NO:585), DOM16-39-542 (SEQ ID NO:586), DOM16-39-551 (SEQ ID
NO:591), DOM16-39-601 (SEQ ID NO:608), DOM16-39-604 (SEQ ID NO:611), DOM16-39-618 (SEQ ID NO:621), and DOM16-39-619 (SEQ ID NO:622).
147. An EGFR antagonist having a CDR3 sequence that has at least 50% identity to the CDR3 sequence of DOMl 6-39 (SEQ ID NO:345), DOMl 6-39-87 (SEQ ID NO:420), DOM16-39-100 (SEQ ID NO:423), DOM16-39-107 (SEQ ID NO:430), DOM16-39-109 (SEQ ID NO:432), DOM16-39-115 (SEQ ID NO:438), and DOM16-39-200 (SEQ ID NO:441).
148. An EGFR antagonist having a CDR3 sequence that has at least 50% identity to the CDR3 sequence of DOM16-39-521 (SEQ ID NO:577), DOM16-39-541 (SEQ ID NO:585), DOM16-39-542 (SEQ ID NO:586), DOM16-39-551 (SEQ ID
NO:591), DOM16-39-601 (SEQ ID NO:608), DOM16-39-604 (SEQ ID NO:611), DOMl 6-39-618 (SEQ ID NO:621), and DOMl 6-39-619 (SEQ ID NO:622).
149. An EGFR antagonist having a CDRl sequence that has at least 50% identity to the CDRl sequence of DOM16-39 (SEQ ID NO:345), DOM16-39-87 (SEQ ID NO:420), DOM16-39-100 (SEQ ID NO:423), DOM16-39-107 (SEQ ID NO:430), DOM16-39-109 (SEQ ID NO:432), DOM16-39-115 (SEQ ID NO:438), and DOM16-39-200 (SEQ ID NO:441) and a CDR2 sequence that has at least 50% identity to the CDR2 sequence of DOM16-39 (SEQ ID NO:345), DOM16-39-87 (SEQ ID NO:420), DOM16-39-100 (SEQ ID NO:423), DOM16-39-107 (SEQ ID NO:430), DOM16-39-109 (SEQ ID NO:432), DOM16-39-115 (SEQ ID NO:438), and DOM16-39-200 (SEQ ID NO:441).
150. An EGFR antagonist having a CDRl sequence that has at least 50% identity to the CDRl sequence of DOMl 6-39-521 (SEQ ID NO:577), DOM16-39-541 (SEQ ID NO:585), DOM16-39-542 (SEQ ID NO:586), DOM16-39-551 (SEQ ID
NO:591), DOM16-39-601 (SEQ ID NO:608), DOM16-39-604 (SEQ ID NO:611), DOM16-39-618 (SEQ ID NO:621), and DOM16-39-619 (SEQ ID NO:622) and a CDR2 sequence that has at least 50% identity to the CDR2 sequence of DOM16-39- 521 (SEQ ID NO:577), DOM16-39-541 (SEQ ID NO:585), DOM16-39-542 (SEQ ID NO:586), DOM16-39-551 (SEQ ID NO:591), DOM16-39-601 (SEQ ID
NO:608), DOM16-39-604 (SEQ ID NO:611), DOM16-39-618 (SEQ ID NO:621), and DOM16-39-619 (SEQ ID NO:622).
151. An EGFR antagonist having a CDR2 sequence that has at least 50% identity to the CDR2 sequence of DOM16-39 (SEQ ID NO:345), DOMl 6-39-87 (SEQ ID NO:420), DOM16-39-100 (SEQ ID NO:423), DOM16-39-107 (SEQ ID NO:430), DOMl 6-39-109 (SEQ ID NO:432), DOM16-39-115 (SEQ ID NO:438), and DOM16-39-200 (SEQ ID NO:441) and a CDR3 sequence that has at least 50% identity to the CDR3 sequence of DOM16-39 (SEQ ID NO:345), DOM16-39-87 (SEQ ID NO:420), DOM16-39-100 (SEQ ID NO:423), DOM16-39-107 (SEQ ID NO:430), DOM16-39-109 (SEQ ID NO:432), DOM16-39-115 (SEQ ID NO:438), and DOMl 6-39-200 (SEQ ID NO:441).
152. An EGFR antagonist having a CDR2 sequence that has at least 50% identity to the CDR2 sequence of DOM16-39-521 (SEQ ID NO:577), DOM16-39-541 (SEQ ID NO:585), DOMl 6-39-542 (SEQ ID NO:586), DOMl 6-39-551 (SEQ ID
NO:591), DOM16-39-601 (SEQ ID NO:608), DOM16-39-604 (SEQ ID NO:611), DOM16-39-618 (SEQ ID NO:621), and DOM16-39-619 (SEQ ID NO:622) and a CDR3 sequence that has at least 50% identity to the CDR3 sequence of DOMl 6-39- 521 (SEQ ID NO:577), DOMl 6-39-541 (SEQ ID NO:585), DOMl 6-39-542 (SEQ ID NO:586), DOM16-39-551 (SEQ ID NO:591), DOM16-39-601 (SEQ ID
NO:608), DOM16-39-604 (SEQ ID NO:611), DOM16-39-618 (SEQ ID NO:621), and DOMl 6-39-619 (SEQ ID NO:622).
153. An EGFR antagonist having a CDRl sequence that has at least 50% identity to the CDRl sequence of DOM16-39 (SEQ ID NO:345), DOM16-39-87 (SEQ ID NO:420), DOM16-39-100 (SEQ ID NO:423), DOM16-39-107 (SEQ ID NO:430), DOM16-39-109 (SEQ ID NO:432), DOM16-39-115 (SEQ ID NO:438), and DOM16-39-200 (SEQ ID NO:441) and a CDR3 sequence that has at least 50% identity to the CDR3 sequence of DOM16-39 (SEQ ID NO:345), DOM16-39-87 (SEQ ID NO:420), DOM16-39-100 (SEQ ID NO:423), DOM16-39-107 (SEQ ID NO:430), DOM16-39-109 (SEQ ID NO:432), DOM16-39-115 (SEQ ID NO:438), and DOMl 6-39-200 (SEQ ID NO:441).
154. An EGFR antagonist having a CDRl sequence that has at least 50% identity to the CDRl sequence of DOMl 6-39-521 (SEQ ID NO:577), DOM16-39-541 (SEQ ID NO:585), DOM16-39-542 (SEQ ID NO:586), DOM16-39-551 (SEQ ID
NO:591), DOM16-39-601 (SEQ ID NO:608), DOM16-39-604 (SEQ ID NO:611), DOM16-39-618 (SEQ ID NO:621), and DOM16-39-619 (SEQ ID NO:622) and a CDR3 sequence that has at least 50% identity to the CDR3 sequence of DOMl 6-39- 521 (SEQ ID NO:577), DOMl 6-39-541 (SEQ ID NO:585), DOMl 6-39-542 (SEQ ID NO:586), DOM16-39-551 (SEQ ID NO:591), DOM16-39-601 (SEQ ID
NO:608), DOM16-39-604 (SEQ ID NO:611), DOM16-39-618 (SEQ ID NO:621), and DOM16-39-619 (SEQ ID NO:622).
155. An EGFR antagonist having a CDRl sequence that has at least 50% identity to the CDRl sequence of DOMl 6-39 (SEQ ID NO:345), DOMl 6-39-87 (SEQ ID NO:420), DOM16-39-100 (SEQ ID NO:423), DOM16-39-107 (SEQ ID NO:430), DOM16-39-109 (SEQ ID NO:432), DOM16-39-115 (SEQ ID NO:438), and DOM16-39-200 (SEQ ID NO:441) and a CDR2 sequence that has at least 50% identity to the CDR2 sequence of DOM16-39 (SEQ ID NO:345), DOM16-39-87 (SEQ ID NO:420), DOM16-39-100 (SEQ ID NO:423), DOM16-39-107 (SEQ ID NO:430), DOMl 6-39-109 (SEQ ID NO:432), DOMl 6-39-115 (SEQ ID NO:438), and DOM16-39-200 (SEQ ID NO:441) and a CDR3 sequence that has at least 50% identity to the CDR3 sequence of DOM16-39 (SEQ ID NO:345), DOM16-39-87 (SEQ ID NO:420), DOM16-39-100 (SEQ ID NO:423), DOM16-39-107 (SEQ ID NO:430), DOM16-39-109 (SEQ ID NO:432), DOM16-39-115 (SEQ ID NO:438), and DOMl 6-39-200 (SEQ ID NO:441).
156. An EGFR antagonist having a CDRl sequence that has at least 50% identity to the CDRl sequence of DOM16-39-521 (SEQ ID NO:577), DOM16-39-541 (SEQ ID NO:585), DOM16-39-542 (SEQ ID NO:586), DOM16-39-551 (SEQ ID NO:591), DOM16-39-601 (SEQ ID NO:608), DOM16-39-604 (SEQ ID NO:611), DOM16-39-618 (SEQ ID NO:621), and DOM16-39-619 (SEQ ID NO:622) and a CDR2 sequence that has at least 50% identity to the CDR2 sequence of DOMl 6-39- 521 (SEQ ID NO:577), DOM16-39-541 (SEQ ID NO:585), DOMl 6-39-542 (SEQ ID NO:586), DOM16-39-551 (SEQ ID NO:591), DOM16-39-601 (SEQ ID
NO:608), DOM16-39-604 (SEQ ID NO:611), DOM16-39-618 (SEQ ID NO:621), and DOMl 6-39-619 (SEQ ID NO:622) and a CDR3 sequence that has at least 50% identity to the CDR3 sequence of DOMl 6-39-521 (SEQ ID NO:577), DOM16-39- 541 (SEQ ID NO:585), DOM16-39-542 (SEQ ID NO:586), DOM16-39-551 (SEQ ID NO:591), DOM16-39-601 (SEQ ID NO:608), DOMl 6-39-604 (SEQ ID
NO:611), DOM16-39-618 (SEQ ID NO:621), and DOMl 6-39-619 (SEQ ID NO:622).
157. A ligand that has binding specificity for vascular endothelial growth factor (VEGF) and epidermal growth factor receptor (EGFR), comprising at least one protein moiety that has a binding site with binding specificity for VEGF, at least one protein moiety that has a binding site with binding specificity for EGFR, and an Fc region of an antibody.
158. The ligand of claim 157, wherein said protein moiety that has a binding site with binding specificity for VEGF is a domain antibody (dAb) that binds VEGF.
159. The ligand of claim 158, wherein said dAb that binds VEGF competes for binding to VEGF with an anti-VEGF dAb selected from the group consisting of TARl 5-6 (SEQ ID NO: 117), TARl 5-8 (SEQ ID NO: 119), and TARl 5-26 (SEQ ID NO:123).
160. The ligand of claim 158, wherein said dAb that binds VEGF competes for binding to VEGF with TARl 5-26-555 (SEQ ID NO:704).
161. The ligand of any one of claims 157-160, wherein said protein moiety that has a binding site with binding specificity for EGFR is a domain antibody (dAb) that binds EGFR.
162. The ligand of claim 161, wherein said dAb that binds EGFR competes for binding to EGFR with an anti-EGFR dAb selected from the group consisting of DOM16-39 (SEQ ID NO:345), DOM16-39-87 (SEQ ID NO:420), DOM16-39-100 (SEQ ID NO:423), DOM16-39-107 (SEQ ID NO:430), DOM16-39-109 (SEQ ID NO:432), DOM16-39-115 (SEQ ID NO:438), and DOM16-39-200 (SEQ ID NO:441).
163. The ligand of claim 161, wherein said dAb that binds EGFR competes for binding to EGFR with an anti-EGFR dAb selected from the group consisting of DOMl 6-39-521 (SEQ ID NO:577), DOMl 6-39-541 (SEQ ID NO:585), DOMl 6- 39-542 (SEQ ID NO:586), DOM16-39-551 (SEQ ID NO:591), DOM16-39-601 (SEQ ID NO:608), DOM16-39-604 (SEQ ID NO:611), DOM16-39-618 (SEQ ID NO:621), and DOM16-39-619 (SEQ ID NO:622).
164. A ligand that has binding specificity for vascular endothelial growth factor (VEGF), comprising at least one protein moiety that has a binding site with binding specificity for VEGF, and an Fc region of an antibody.
165. The ligand of claim 164, wherein said protein moiety that has a binding site with binding specificity for VEGF is a domain antibody (dAb).
166. The ligand of claim 165, wherein said dAb competes for binding to VEGF with an anti-VEGF dAb selected from the group consisting of TARl 5-6 (SEQ ID NO:117), TARl 5-8 (SEQ ID NO:119), and TARl 5-26 (SEQ ID NO: 123).
167. The ligand of claim 165, wherein said dAb competes for binding to VEGF with TARl 5-26-555 (SEQ ID NO:704).
168. A ligand that has binding specificity for epidermal growth factor receptor (EGFR), comprising at least one protein moiety that has a binding site with binding specificity for EGFR, and an Fc region of an antibody.
169. The ligand of claim 168, wherein said protein moiety that has a binding site with binding specificity for EGFR is a domain antibody (dAb).
170. The ligand of claim 169, wherein said dAb competes for binding to EGFR with an anti-EGFR dAb selected from the group consisting of DOMl 6-39 (SEQ ID NO:345), DOM16-39-87 (SEQ ID NO:420), DOM16-39-100 (SEQ ID NO:423), DOM16-39-107 (SEQ ID NO:430), DOM16-39-109 (SEQ ID NO:432), DOM16- 39-115 (SEQ ID NO:438), and DOM16-39-200 (SEQ ID NO:441).
171. The ligand of claim 169, wherein said dAb competes for binding to EGFR with an anti-EGFR dAb selected from the group consisting of DOM 16-39-521 (SEQ ID NO:577), DOMl 6-39-541 (SEQ ID NO:585), DOMl 6-39-542 (SEQ ID
NO:586), DOMl 6-39-551 (SEQ ID NO:591), DOMl 6-39-601 (SEQ ID NO:608), DOM16-39-604 (SEQ ID NO:611), DOM16-39-618 (SEQ ID NO:621), and DOMl 6-39-619 (SEQ ID NO:622).
172. The ligand of claim 168, wherein said ligand comprises at least two protein moieties that have binding sites with binding specificity for EGFR and an Fc region of an antibody.
173. A ligand that has binding specificity for vascular endothelial growth factor (VEGF) and epidermal growth factor receptor (EGFR), comprising at least one immunoglobulin single variable domain with binding specificity for VEGF, at least one immunoglobulin single variable domain with binding specificity for EGFR, and a linker, wherein said immunoglobulin single variable domain with binding specificity for EGFR is bonded via said linker to said immunoglobulin single variable domain with binding specificity for VEGF.
174. A ligand that has binding specificity for vascular endothelial growth factor (VEGF) and epidermal growth factor receptor (EGFR), comprising at least one immunoglobulin single variable domain with binding specificity for VEGF, at least one immunoglobulin single variable domain with binding specificity for EGFR, wherein said immunoglobulin single variable domain with binding specificity for EGFR is directly fused to said immunoglobulin single variable domain with binding specificity for VEGF.
175. The ligand of claim 173 wherein said linker is selected from the group consisting of SEQ ID NO:706, SEQ ID NO:707, SEQ ID NO:708, SEQ ID NO:709, SEQ ID NO:710, SEQ ID NO:711, SEQ ID NO:712, SEQ ID NO:713, SEQ ID NO:714, SEQ ID NO:723 and SEQ ID NO:724.
176. The ligand of claim 173 or 175, wherein said ligand further comprises an Fc region of an antibody.
177. The ligand of any one of claims 176, wherein said ligand further comprises a second linker and wherein one of the immunoglobulin single variable domains is bonded via said second linker to said Fc region.
178. The ligand of claim 177, wherein said second linker is selected from the group consisting of SEQ ID NO:706, SEQ ID NO:707, SEQ ID NO:708, SEQ ID NO:709, SEQ ID NO:710, SEQ ID NO:711, SEQ ID NO:712, SEQ ID NO:713, SEQ ID NO:714, SEQ ID NO:723 and SEQ ID NO:724.
179. The ligand of claim 174, wherein said ligand further comprises a linker and an Fc region of an antibody, and wherein one of said immunoglobulin single variable domains is bonded via said linker to said Fc region of an antibody.
180. The ligand of claim 179, wherein said linker is selected from the group consisting of SEQ ID NO:706, SEQ ID NO:707, SEQ ID NO:708, SEQ ID NO:709, SEQ ID NO:710, SEQ ID NO:711, SEQ ID NO:712, SEQ ID NO:713, SEQ ID NO:714, SEQ ID NO:723 and SEQ ID NO:724.
181. The ligand of any one of claims 173-180, wherein
a, said immunoglobulin single variable domain with binding specificity for VEGF is a heavy chain variable domain, and said immunoglobulin single variable domain with binding specificity for EGFR is a light chain variable domain;
b, said immunoglobulin single variable domain with binding specificity for VEGF is a light chain variable domain, and said immunoglobulin single variable domain with binding specificity for EGFR is a heavy chain variable domain;
c, said immunoglobulin single variable domain with binding specificity for VEGF is a heavy chain variable domain, and said immunoglobulin single variable domain with binding specificity for EGFR is a heavy chain variable domain; or
d, said immunoglobulin single variable domain with binding specificity for VEGF is a light chain variable domain, and said immunoglobulin single variable domain with binding specificity for EGFR is a light chain variable domain.
182. The ligand of claim 181, wherein said heavy chain variable domain is a VH or VHH-
183. The ligand of claim 181, wherein said heavy chain variable domain is a human VH.
184. The ligand of claim 181, wherein said light chain variable domain is a VK-
185. A ligand that has binding specificity for vascular endothelial growth factor (VEGF) and/or epidermal growth factor receptor (EGFR), comprising a first immunoglobulin single variable domain, a second single immunoglobulin variable domain and an Fc region of an antibody.
186. The ligand of claim 185, wherein said ligand has the formula, from amino- terminal to carboxy-terminal, dAbl-dAb2-Fc, wherein dAbl is said first
immunoglobulin single variable domain, dAb2 is said second immunoglobulin single variable domain, and Fc is said Fc region of an antibody, wherein dAbl is bonded to dAb 2 directly or through a first linker, and dAb2 is bonded to Fc directly or through a second linker.
187. The ligand of claim 186 wherein dAb 1 and dAb2 are the same.
188. The ligand of claim 186 or 187 wherein dAbl and dAb2 are each a light chain single variable domain.
189. The ligand of claim 188 wherein dAb 1 and dAb2 are each Vk.
190. The ligand of claim 186 or 187 wherein dAbl and dAb2 are each a heavy chain single variable domain.
191. The ligand of any one of claims 185-190 wherein said first immunoglobulin single variable domain and said second immunoglobulin single variable domain each have binding specificity for EGFR.
192. The ligand of any one of claims 185-190 wherein said first immunoglobulin single variable domain and said second immunoglobulin single variable domain each have binding specificity for VEGF.
193. The ligand of any one of claims 186 and 188-190, wherein dAbl has binding specificity for EGFR and dAb2 has binding specificity for VEGF.
194. The ligand of any one of claims 186 and 188-190, wherein dAbl has binding specificity for VEGF and dAb2 has binding specificity for EGFR.
195. A ligand, wherein said ligand is a dimer comprising a first ligand and a second ligand, wherein said first ligand and said second ligand are each as defined in any one of claims 185-194.
196. The ligand of claim 195, wherein said dimer comprises a disulfide bond between said first ligand and said second ligand.
PCT/GB2006/004559 2002-06-28 2006-12-05 Ligands that have binding specificity for egfr and/or vegf and methods of use therefor WO2007066106A1 (en)

Priority Applications (27)

Application Number Priority Date Filing Date Title
US12/086,020 US20100056439A1 (en) 2005-12-06 2006-12-05 Ligands that have binding specificity for egfr and/or vegf and methods of use therefor
BRPI0619463-0A BRPI0619463A2 (en) 2005-12-06 2006-12-05 binding, use thereof, methods for treating cancer and for administering anti-vegf and anti-egfr treatment to an individual, composition, drug dispensing device, isolated or recombinant nucleic acid, vector, host cell, method for producing a ligand, immunoglobulin single variable domain polypeptide, and egfr antagonist
EA200801172A EA013878B1 (en) 2005-12-06 2006-12-05 Ligands that have binding specificity for egfr and/or vegf and methods of use therefor
CA002632417A CA2632417A1 (en) 2005-12-06 2006-12-05 Ligands that have binding specificity for egfr and/or vegf and methods of use therefor
EP06820440A EP1966242A1 (en) 2005-12-06 2006-12-05 Ligands that have binding specificity for egfr and/or vegf and methods of use therefor
AU2006323412A AU2006323412A1 (en) 2005-12-06 2006-12-05 Ligands that have binding specificity for EGFR and/or VEGF and methods of use therefor
JP2008543892A JP2009518024A (en) 2005-12-06 2006-12-05 Ligands having binding specificity for EGFR and / or VEGF and methods of use thereof
TW096102778A TW200804593A (en) 2006-01-24 2007-01-24 Fusion proteins that contain natural junctions
AU2007209201A AU2007209201A1 (en) 2006-01-24 2007-01-24 Fusion proteins that contain natural junctions
EP07705002A EP1976991A1 (en) 2006-01-24 2007-01-24 Fusion proteins that contain natural junctions
CA002640066A CA2640066A1 (en) 2006-01-24 2007-01-24 Fusion proteins that contain natural junctions
US12/087,924 US20100047171A1 (en) 2006-01-24 2007-01-24 Fusion Proteins That Contain Natural Junctions
JP2008551863A JP2009523459A (en) 2006-01-24 2007-01-24 Fusion proteins containing natural linkages
PCT/GB2007/000227 WO2007085814A1 (en) 2006-01-24 2007-01-24 Fusion proteins that contain natural junctions
EP11157601.3A EP2441838A3 (en) 2006-01-24 2007-01-24 Fusion proteins that contain natural junctions
CA002667141A CA2667141A1 (en) 2006-10-30 2007-10-26 Novel polypeptides and uses thereof
ARP070104757A AR063416A1 (en) 2006-10-30 2007-10-26 IMMUNOGLOBULIN CHAIN AGGREGATION INHIBITOR AND ITS USE
CL200703101A CL2007003101A1 (en) 2006-10-30 2007-10-26 INHIBITOR OF THE AGGREGATION OF IMMUNOGLOBULINS; NUCLEIC ACID OF SUCH INHIBITOR; VECTOR AND CELL GUEST WITH SUCH NUCLEIC ACID; PROCEDURE TO MAKE SUCH INHIBITOR; PHARMACEUTICAL FORMULATION THAT UNDERSTANDS IT; USE OF THE INHIBITOR TO TREAT IN
JP2009533859A JP2010508016A (en) 2006-10-30 2007-10-26 Novel polypeptides and their uses
EP07821877A EP2079761A2 (en) 2006-10-30 2007-10-26 Prevention of aggregation of immunoglobulin light or heavy chains
US12/447,908 US8236931B2 (en) 2006-10-30 2007-10-26 Prevention of aggregation of immunoglobulin light or heavy chains
TW096140428A TW200825103A (en) 2006-10-30 2007-10-26 Novel polypeptides and uses thereof
PCT/EP2007/061514 WO2008052933A2 (en) 2006-10-30 2007-10-26 Prevention of aggregation of immunoglobulin light or heavy chains
PE2007001465A PE20081141A1 (en) 2006-10-30 2007-10-26 NEW POLYEPTIDES AND USES OF THEM
US12/006,933 US20080241166A1 (en) 2002-06-28 2008-01-07 Ligands that bind a receptor
NO20082386A NO20082386L (en) 2005-12-06 2008-05-26 Ligands having binding specificity for EGFR and / or VEGF and methods for their use
US13/644,414 US20130041136A1 (en) 2005-12-06 2012-10-26 Ligands that have binding specificity for egfr and/or vegf and methods of use therefor

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US74299205P 2005-12-06 2005-12-06
US60/742,992 2005-12-06
US75835506P 2006-01-11 2006-01-11
US60/758,355 2006-01-11

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US13/644,414 Continuation US20130041136A1 (en) 2005-12-06 2012-10-26 Ligands that have binding specificity for egfr and/or vegf and methods of use therefor

Publications (1)

Publication Number Publication Date
WO2007066106A1 true WO2007066106A1 (en) 2007-06-14

Family

ID=37820653

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB2006/004559 WO2007066106A1 (en) 2002-06-28 2006-12-05 Ligands that have binding specificity for egfr and/or vegf and methods of use therefor

Country Status (13)

Country Link
US (2) US20100056439A1 (en)
EP (1) EP1966242A1 (en)
JP (1) JP2009518024A (en)
KR (1) KR20080077261A (en)
AU (1) AU2006323412A1 (en)
BR (1) BRPI0619463A2 (en)
CA (1) CA2632417A1 (en)
CR (1) CR10024A (en)
EA (1) EA013878B1 (en)
MA (1) MA30021B1 (en)
NO (1) NO20082386L (en)
TW (1) TW200804425A (en)
WO (1) WO2007066106A1 (en)

Cited By (26)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007080392A2 (en) * 2006-01-11 2007-07-19 Domantis Limited Ligands that have binding specificity for vegf and/or egfr and methods of use therefor
WO2008052933A2 (en) * 2006-10-30 2008-05-08 Domantis Limited Prevention of aggregation of immunoglobulin light or heavy chains
WO2008149146A2 (en) * 2007-06-06 2008-12-11 Domantis Limited Polypeptides, antibody variable domains and antagonists
WO2009068649A2 (en) 2007-11-30 2009-06-04 Glaxo Group Limited Antigen-binding constructs
WO2009074634A2 (en) * 2007-12-13 2009-06-18 Glaxo Group Limited Compositions for pulmonary delivery
WO2009121804A1 (en) 2008-03-31 2009-10-08 Glaxo Group Limited Drug fusions and conjugates
WO2010033249A2 (en) * 2008-09-22 2010-03-25 Massachusetts Institute Of Technology Compositions of and methods using ligand dimers
WO2010060768A1 (en) * 2008-11-26 2010-06-03 Glaxo Group Limited Polypeptides, antibody variable domains & antagonists
WO2010136483A3 (en) * 2009-05-28 2011-01-20 Glaxo Group Limited Antigen-binding proteins
WO2011095545A1 (en) 2010-02-05 2011-08-11 Ablynx Nv Peptides capable of binding to serum albumin and compounds, constructs and polypeptides comprising the same
JP2012506237A (en) * 2008-10-21 2012-03-15 ドマンティス リミテッド Ligand with binding specificity for DC-SIGN
CN102405236A (en) * 2009-02-19 2012-04-04 葛兰素集团有限公司 Improved anti-tnfr1 polypeptides, antibody variable domains & antagonists
WO2012022703A3 (en) * 2010-08-20 2012-04-26 Glaxo Group Limited Improved anti-serum albumin binding variants
US8217140B2 (en) 2008-04-17 2012-07-10 Ablynx N.V. Peptides capable of binding to serum proteins and compounds, constructs and polypeptides comprising the same
JP2012532620A (en) * 2009-07-16 2012-12-20 グラクソ グループ リミテッド Improved antiserum albumin binding single variable domain
US8399219B2 (en) 2009-02-23 2013-03-19 Cytomx Therapeutics, Inc. Protease activatable interferon alpha proprotein
US8513390B2 (en) 2009-01-12 2013-08-20 Cytomx Therapeutics, Inc. Modified antibody compositions, methods of making and using thereof
AU2008212682B2 (en) * 2007-02-08 2013-09-26 Domantis Limited Antibody single variable domains against serum albumin
EP2853542A1 (en) * 2010-11-24 2015-04-01 Glaxo Group Limited Multispecific antigen binding proteins targeting HGF
WO2015044386A1 (en) * 2013-09-26 2015-04-02 Ablynx Nv Bispecific nanobodies
US9029328B2 (en) 2010-03-24 2015-05-12 The Brigham And Women's Hospital, Inc. Methods for cardioprotection and cardioregeneration with dimers of EGF family ligands
US9169321B2 (en) 2007-08-22 2015-10-27 The Regents Of The University Of California Activatable binding polypeptides and methods of identification and use thereof
US10308720B2 (en) 2015-07-16 2019-06-04 Inhibrx, Inc. Multivalent and multispecific DR5-binding fusion proteins
US10526397B2 (en) 2015-01-21 2020-01-07 Inhibrx, Inc. Non-immunogenic single domain antibodies
EP3768722A4 (en) * 2018-03-19 2021-12-29 Wuxi Biologics Ireland Limited. Novel anti-egfr antibody polypeptide
WO2022047169A1 (en) * 2020-08-28 2022-03-03 Trustees Of Boston University Engineered extracellular receptor constructs and uses thereof

Families Citing this family (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
SG10201605629VA (en) 2008-01-03 2016-08-30 Scripps Research Inst Antibody targeting through a modular recognition domain
US8454960B2 (en) * 2008-01-03 2013-06-04 The Scripps Research Institute Multispecific antibody targeting and multivalency through modular recognition domains
US20120100166A1 (en) 2010-07-15 2012-04-26 Zyngenia, Inc. Ang-2 Binding Complexes and Uses Thereof
CN103857699B (en) 2011-05-24 2016-08-31 泽恩格尼亚股份有限公司 Multivalence and unit price polyspecific complex and application thereof
US20140255471A1 (en) 2013-03-11 2014-09-11 Wake Forest University Health Sciences Method of treating brain tumors
CA2907181C (en) 2013-03-15 2023-10-17 Viktor Roschke Multivalent and monovalent multispecific complexes and their uses
CA2974720A1 (en) 2015-02-06 2016-08-11 University Of Maryland, Baltimore Tetra-specific, octameric binding agents and antibodies against clostridium difficile toxin a and toxin b for treatment of c. difficile infection
KR101685532B1 (en) * 2016-04-26 2016-12-13 한국프라임제약주식회사 A VEGFR fusion protein
KR20210121274A (en) * 2019-03-29 2021-10-07 주식회사 녹십자 Fusion proteins comprising anti-mesothelin antibodies, anti-CD3 antibodies or anti-EGFR antibodies and bispecific or trispecific antibodies comprising same and uses thereof
CN115724968B (en) * 2021-08-27 2023-08-08 三优生物医药(上海)有限公司 VEGF binding molecules and uses thereof

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20020058033A1 (en) * 2000-10-13 2002-05-16 Raisch Kevin Paul Human anti-epidermal growth factor receptor single-chain antibodies
WO2004041867A2 (en) * 2002-11-08 2004-05-21 Ablynx N.V. Camelidae antibodies against imminoglobulin e and use thereof for the treatment of allergic disorders
WO2004081026A2 (en) * 2003-06-30 2004-09-23 Domantis Limited Polypeptides
WO2005044858A1 (en) * 2003-11-07 2005-05-19 Ablynx N.V. Camelidae single domain antibodies vhh directed against epidermal growth factor receptor and uses therefor
WO2006003388A2 (en) * 2004-06-30 2006-01-12 Domantis Limited Compositions and methods for treating inflammatory disorders

Family Cites Families (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5844093A (en) * 1994-03-17 1998-12-01 Merck Patent Gesellschaft Mit Beschrankter Haftung Anti-EGFR single-chain Fvs and anti-EGFR antibodies
IL129497A0 (en) * 1996-10-25 2000-02-29 Nexstar Pharmaceuticals Inc Vascular endothelial growth factor (vegf) nucleic acid ligand complexes
DE69829891T2 (en) * 1997-04-07 2005-10-06 Genentech, Inc., South San Francisco Anti-VEGF antibody
RU2295537C2 (en) * 2000-10-20 2007-03-20 Тугаи Сейяку Кабусики Кайся Modified antagonistic antibody
US7667004B2 (en) * 2001-04-17 2010-02-23 Abmaxis, Inc. Humanized antibodies against vascular endothelial growth factor
US20050271663A1 (en) * 2001-06-28 2005-12-08 Domantis Limited Compositions and methods for treating inflammatory disorders
WO2004003019A2 (en) * 2002-06-28 2004-01-08 Domantis Limited Immunoglobin single variant antigen-binding domains and dual-specific constructs
JP2006523090A (en) * 2002-12-27 2006-10-12 ドマンティス リミテッド Bispecific single domain antibody specific for ligand and for ligand receptor
US20050106667A1 (en) * 2003-08-01 2005-05-19 Genentech, Inc Binding polypeptides with restricted diversity sequences
US20090252681A1 (en) * 2005-10-11 2009-10-08 Ablynx N.V. Nanobodies and Polypeptides Against EGFR and IGF-IR

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20020058033A1 (en) * 2000-10-13 2002-05-16 Raisch Kevin Paul Human anti-epidermal growth factor receptor single-chain antibodies
WO2004041867A2 (en) * 2002-11-08 2004-05-21 Ablynx N.V. Camelidae antibodies against imminoglobulin e and use thereof for the treatment of allergic disorders
WO2004081026A2 (en) * 2003-06-30 2004-09-23 Domantis Limited Polypeptides
WO2005044858A1 (en) * 2003-11-07 2005-05-19 Ablynx N.V. Camelidae single domain antibodies vhh directed against epidermal growth factor receptor and uses therefor
WO2006003388A2 (en) * 2004-06-30 2006-01-12 Domantis Limited Compositions and methods for treating inflammatory disorders

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
HOANG ET AL: "PD-144 Tumor response augmentation with combination cetuximab(Erbitux(R)) and bevacizumab (Avastin(R))", LUNG CANCER, ELSEVIER, AMSTERDAM, NL, vol. 49, July 2005 (2005-07-01), pages S108 - S109, XP005048925, ISSN: 0169-5002 *
US NATIONAL INSTITUTE OF HEALTH: "Bevacizumab adn Cetuximab with or without Irinotecan in treating patients with Irinotecan-refractory metastatic colon cancer", CLINICALTRIALS, no. NCT00077298, 10 February 2004 (2004-02-10) - 10 February 2004 (2004-02-10), pages 1 - 6, XP002424711, Retrieved from the Internet <URL:http://clinicaltrials.gov> [retrieved on 20070313] *

Cited By (59)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9321832B2 (en) 2002-06-28 2016-04-26 Domantis Limited Ligand
US7696320B2 (en) 2004-08-24 2010-04-13 Domantis Limited Ligands that have binding specificity for VEGF and/or EGFR and methods of use therefor
JP2009523162A (en) * 2006-01-11 2009-06-18 ドマンティス リミテッド Ligands having binding specificity for VEGF and / or EGFR and methods of use thereof
WO2007080392A3 (en) * 2006-01-11 2007-11-29 Domantis Ltd Ligands that have binding specificity for vegf and/or egfr and methods of use therefor
WO2007080392A2 (en) * 2006-01-11 2007-07-19 Domantis Limited Ligands that have binding specificity for vegf and/or egfr and methods of use therefor
WO2008052933A3 (en) * 2006-10-30 2008-11-13 Domantis Ltd Prevention of aggregation of immunoglobulin light or heavy chains
US8236931B2 (en) 2006-10-30 2012-08-07 Glaxo Group Limited Prevention of aggregation of immunoglobulin light or heavy chains
WO2008052933A2 (en) * 2006-10-30 2008-05-08 Domantis Limited Prevention of aggregation of immunoglobulin light or heavy chains
AU2008212682B2 (en) * 2007-02-08 2013-09-26 Domantis Limited Antibody single variable domains against serum albumin
WO2008149146A3 (en) * 2007-06-06 2009-06-11 Domantis Ltd Polypeptides, antibody variable domains and antagonists
WO2008149146A2 (en) * 2007-06-06 2008-12-11 Domantis Limited Polypeptides, antibody variable domains and antagonists
US10077300B2 (en) 2007-08-22 2018-09-18 The Regents Of The University Of California Activatable binding polypeptides and methods of identification and use thereof
US11028162B2 (en) 2007-08-22 2021-06-08 The Regents Of The University Of California Methods for manufacturing activatable binding polypeptides comprising matrix metalloprotease cleavable moieties
US9169321B2 (en) 2007-08-22 2015-10-27 The Regents Of The University Of California Activatable binding polypeptides and methods of identification and use thereof
EA023031B1 (en) * 2007-11-30 2016-04-29 Глаксо Груп Лимитед Antigen-binding construct and use thereof
GB2468232A (en) * 2007-11-30 2010-09-01 Glaxo Group Ltd Antigen-bindng constructs
AU2008328726B2 (en) * 2007-11-30 2014-06-12 Glaxo Group Limited Antigen-binding constructs
WO2009068649A2 (en) 2007-11-30 2009-06-04 Glaxo Group Limited Antigen-binding constructs
JP2011504742A (en) * 2007-11-30 2011-02-17 グラクソ グループ リミテッド Antigen binding construct
GB2468232B (en) * 2007-11-30 2012-10-24 Glaxo Group Ltd Antigen-bindng constructs
WO2009068649A3 (en) * 2007-11-30 2010-02-04 Glaxo Group Limited Antigen-binding constructs
WO2009074634A2 (en) * 2007-12-13 2009-06-18 Glaxo Group Limited Compositions for pulmonary delivery
JP2011506396A (en) * 2007-12-13 2011-03-03 グラクソ グループ リミテッド Pulmonary delivery composition
WO2009074634A3 (en) * 2007-12-13 2010-09-23 Glaxo Group Limited Polypeptides, antibody variable domains & antagonists
WO2009121804A1 (en) 2008-03-31 2009-10-08 Glaxo Group Limited Drug fusions and conjugates
JP2011517561A (en) * 2008-03-31 2011-06-16 グラクソ グループ リミテッド Drug fusions and conjugates
EA018471B1 (en) * 2008-03-31 2013-08-30 Глаксо Груп Лимитед Drug fusions and conjugates
US8217140B2 (en) 2008-04-17 2012-07-10 Ablynx N.V. Peptides capable of binding to serum proteins and compounds, constructs and polypeptides comprising the same
WO2010033249A3 (en) * 2008-09-22 2010-08-19 Massachusetts Institute Of Technology Compositions of and methods of using ligand dimers
US9198952B2 (en) 2008-09-22 2015-12-01 The Brigham And Women's Hospital, Inc. Compositions of and methods of using ligand dimers
WO2010033249A2 (en) * 2008-09-22 2010-03-25 Massachusetts Institute Of Technology Compositions of and methods using ligand dimers
JP2012506237A (en) * 2008-10-21 2012-03-15 ドマンティス リミテッド Ligand with binding specificity for DC-SIGN
WO2010060768A1 (en) * 2008-11-26 2010-06-03 Glaxo Group Limited Polypeptides, antibody variable domains & antagonists
US10118961B2 (en) 2009-01-12 2018-11-06 Cytomx Therapeutics, Inc. Modified antibody containing the cleavable peptide with the amino acid sequence TGRGPSWV
US10875913B2 (en) 2009-01-12 2020-12-29 Cytomx Therapeutics, Inc. Methods of treatment using activatable anti-EGFR antibodies
US8513390B2 (en) 2009-01-12 2013-08-20 Cytomx Therapeutics, Inc. Modified antibody compositions, methods of making and using thereof
US8563269B2 (en) 2009-01-12 2013-10-22 Cytomx Therapeutics, Inc. Modified antibody compositions, methods of making and using thereof
US10059762B2 (en) 2009-01-12 2018-08-28 Cytomx Therapeutics, Inc. Anti-EGFR activatable antibodies
US9453078B2 (en) 2009-01-12 2016-09-27 Cytomx Therapeutics, Inc. Modified antibody compositions, methods of making and using thereof
CN102405236A (en) * 2009-02-19 2012-04-04 葛兰素集团有限公司 Improved anti-tnfr1 polypeptides, antibody variable domains & antagonists
US9644016B2 (en) 2009-02-23 2017-05-09 Cytomx Therapeutics, Inc. Soluble notch receptor proproteins and methods of use thereof
US8399219B2 (en) 2009-02-23 2013-03-19 Cytomx Therapeutics, Inc. Protease activatable interferon alpha proprotein
US10513549B2 (en) 2009-02-23 2019-12-24 Cytomx Therapeutics, Inc. Cleavage-activatable interferon-alpha proprotein
US9394374B2 (en) 2009-05-28 2016-07-19 Glaxo Group Limited Antigen-binding proteins
WO2010136483A3 (en) * 2009-05-28 2011-01-20 Glaxo Group Limited Antigen-binding proteins
US20120070439A1 (en) * 2009-05-28 2012-03-22 Paul Andrew Hamblin Antigen-binding proteins
JP2012532620A (en) * 2009-07-16 2012-12-20 グラクソ グループ リミテッド Improved antiserum albumin binding single variable domain
WO2011095545A1 (en) 2010-02-05 2011-08-11 Ablynx Nv Peptides capable of binding to serum albumin and compounds, constructs and polypeptides comprising the same
US9029328B2 (en) 2010-03-24 2015-05-12 The Brigham And Women's Hospital, Inc. Methods for cardioprotection and cardioregeneration with dimers of EGF family ligands
WO2012022703A3 (en) * 2010-08-20 2012-04-26 Glaxo Group Limited Improved anti-serum albumin binding variants
EP2853542A1 (en) * 2010-11-24 2015-04-01 Glaxo Group Limited Multispecific antigen binding proteins targeting HGF
WO2015044386A1 (en) * 2013-09-26 2015-04-02 Ablynx Nv Bispecific nanobodies
US10526397B2 (en) 2015-01-21 2020-01-07 Inhibrx, Inc. Non-immunogenic single domain antibodies
US10308720B2 (en) 2015-07-16 2019-06-04 Inhibrx, Inc. Multivalent and multispecific DR5-binding fusion proteins
US11117973B2 (en) 2015-07-16 2021-09-14 Inhibrx, Inc. Multivalent and multispecific DR5-binding fusion proteins and methods of treating neoplasms
US11976126B2 (en) 2015-07-16 2024-05-07 Inhibrx, Inc. Multivalent and multispecific DR5-binding fusion proteins and methods of modulating immune cells
EP3768722A4 (en) * 2018-03-19 2021-12-29 Wuxi Biologics Ireland Limited. Novel anti-egfr antibody polypeptide
US11773172B2 (en) 2018-03-19 2023-10-03 WuXi Biologics Ireland Limited Anti-EGFR antibody polypeptide
WO2022047169A1 (en) * 2020-08-28 2022-03-03 Trustees Of Boston University Engineered extracellular receptor constructs and uses thereof

Also Published As

Publication number Publication date
TW200804425A (en) 2008-01-16
KR20080077261A (en) 2008-08-21
JP2009518024A (en) 2009-05-07
EA013878B1 (en) 2010-08-30
CA2632417A1 (en) 2007-06-14
AU2006323412A1 (en) 2007-06-14
CR10024A (en) 2008-09-22
NO20082386L (en) 2008-08-27
MA30021B1 (en) 2008-12-01
US20130041136A1 (en) 2013-02-14
EP1966242A1 (en) 2008-09-10
EA200801172A1 (en) 2008-12-30
BRPI0619463A2 (en) 2013-01-08
US20100056439A1 (en) 2010-03-04

Similar Documents

Publication Publication Date Title
US7696320B2 (en) Ligands that have binding specificity for VEGF and/or EGFR and methods of use therefor
US20130041136A1 (en) Ligands that have binding specificity for egfr and/or vegf and methods of use therefor
US20100021473A1 (en) Bispecific Ligands With Binding Specificity to Cell Surface Targets and Methods of Use Therefor
US20110159003A1 (en) Ligands That Bind Il-4 and/or Il-13
EP1841452B1 (en) Single domain antibodies against tnfr1 and methods of use therefor
US20080241166A1 (en) Ligands that bind a receptor
US20150087813A1 (en) Ligand
AU2005311103A1 (en) PLAD domain peptides with increased serum half life due to conjugation to domain antibodies
US20080233129A1 (en) Ligand that has binding specificity for IL-4 and/or lL-13
US9028822B2 (en) Antagonists against TNFR1 and methods of use therefor
MX2008008854A (en) Ligands that have binding specificity for vegf and/or egfr and methods of use therefor
MX2008007351A (en) Ligands that have binding specificity for egfr and/or vegf and methods of use therefor
US20110223168A1 (en) Ligand that has binding specificity for il-4 and/or il-13
MX2008009528A (en) Ligands that bind il-4 and/or il-13
MX2008007369A (en) Bispecific ligands with binding specificity to cell surface targets and methods of use therefor

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 191462

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: 568375

Country of ref document: NZ

WWE Wipo information: entry into national phase

Ref document number: 2006323412

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 200801172

Country of ref document: EA

WWE Wipo information: entry into national phase

Ref document number: 2008543892

Country of ref document: JP

WWE Wipo information: entry into national phase

Ref document number: 2008060943

Country of ref document: EG

Ref document number: 12008501340

Country of ref document: PH

Ref document number: 2632417

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: MX/a/2008/007351

Country of ref document: MX

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2343/KOLNP/2008

Country of ref document: IN

ENP Entry into the national phase

Ref document number: 2006323412

Country of ref document: AU

Date of ref document: 20061205

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 08060267

Country of ref document: CO

WWP Wipo information: published in national office

Ref document number: 2006323412

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2006820440

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 1020087016537

Country of ref document: KR

WWE Wipo information: entry into national phase

Ref document number: 200680052392.X

Country of ref document: CN

WWP Wipo information: published in national office

Ref document number: 2006820440

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 12086020

Country of ref document: US

ENP Entry into the national phase

Ref document number: PI0619463

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20080606