NZ623724B2 - Vegf/dll4 binding agents and uses thereof - Google Patents

Vegf/dll4 binding agents and uses thereof Download PDF

Info

Publication number
NZ623724B2
NZ623724B2 NZ623724A NZ62372412A NZ623724B2 NZ 623724 B2 NZ623724 B2 NZ 623724B2 NZ 623724 A NZ623724 A NZ 623724A NZ 62372412 A NZ62372412 A NZ 62372412A NZ 623724 B2 NZ623724 B2 NZ 623724B2
Authority
NZ
New Zealand
Prior art keywords
seq
antibody
dll4
vegf
heavy chain
Prior art date
Application number
NZ623724A
Other versions
NZ623724A (en
Inventor
Christopher John Bond
Austin L Gurney
Aaron Ken Sato
Original Assignee
Oncomed Pharmaceuticals Inc
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 Oncomed Pharmaceuticals Inc filed Critical Oncomed Pharmaceuticals Inc
Priority claimed from PCT/US2012/056886 external-priority patent/WO2013044215A1/en
Publication of NZ623724A publication Critical patent/NZ623724A/en
Publication of NZ623724B2 publication Critical patent/NZ623724B2/en

Links

Classifications

    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2300/00Mixtures or combinations of active ingredients, wherein at least one active ingredient is fully defined in groups A61K31/00 - A61K41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/4738Quinolines; Isoquinolines ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/4745Quinolines; Isoquinolines ortho- or peri-condensed with heterocyclic ring systems condensed with ring systems having nitrogen as a ring hetero atom, e.g. phenantrolines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7052Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides
    • A61K31/706Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom
    • A61K31/7064Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines
    • A61K31/7068Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines having oxo groups directly attached to the pyrimidine ring, e.g. cytidine, cytidylic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • 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
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/3955Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against proteinaceous materials, e.g. enzymes, hormones, lymphokines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • 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
    • 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
    • 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
    • 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
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • 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
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • C07K16/303Liver or Pancreas
    • 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
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • C07K16/3046Stomach, Intestines
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/565Complementarity determining region [CDR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value

Abstract

Disclosed is a bispecific antibody comprising: a) a first antigen-binding site that specifically binds human vascular endothelial growth factor (VEGF), and b) a second antigen-binding site that specifically binds human delta-like 4 ligand (DLL4), comprising the CDRs of the sequences as defined in the specification. Also disclosed is its use in treating cancer. the specification. Also disclosed is its use in treating cancer.

Description

VEGF/DLL4 BINDING AGENTS AND USES THEREOF FIELD OF THE INVENTION The t invention generally relates to antibodies and other agents that bind VEGF, DLL4, or both VEGF and DLL4, ularly anti—VEGF/anti—DLL4 bispecific antibodies, as well as to methods of using the antibodies or other agents for the treatment of diseases such as cancer.
BACKGROUND OF THE INVENTION Angiogenesis plays an important role in the pathogenesis of a number of disorders, including solid tumors and metastasis. The production ofnew blood vessels is essential for providing oxygen and nutrients for the growth and spread of a tumor, and therefore angiogenesis is a good target for cancer therapeutics.
Angiogenesis involves a family ofproteins acting as angiogenic activators, including ar endothelial growth factor (VEGF-A), , VEGF—C, VEGF-E, and their respective receptors - 1, VEGFR-Z, and VEGFR-3). VEGF—A, also referred to as VEGF or vascular permeability factor (VPF), exists in several isoforms that arise from alternative splicing ofmRNA of a single VEGF gene, with VEGF165 being the most biologically relevant isoform.
Anti-VEGF dies have been shown to suppress the growth oftumor cells in vitro and in vivo.
A humanized anti-VEGF monoclonal antibody, bevacizumab IN) has been developed and approved in the United States as a cancer therapeutic.
The Notch signaling pathway is a universally conserved signal transduction system. It is involved in cell fate determination during development ing embryonic n formation and post-embryonic tissue nance. In addition, Notch signaling has been identified as a critical factor in the maintenance of hematopoietic stem cells.
The Notch pathway has been linked to the pathogenesis of both hematologic and solid tumors and cancers. us cellular functions and microenvironmental cues associated with tumorigenesis have been shown to be ted by Notch pathway signaling, including cell proliferation, apoptosis, adhesion, and angiogenesis (Leong et al., 2006, Blood, 107:2223-2233). In addition, Notch receptors and/or Notch ligands have been shown to play potential oncogenic roles in a number of human cancers, including acute myelogenous leukemia, B cell c lymphocytic leukemia, Hodgkin lymphoma, multiple myeloma, T—cell acute lymphoblastic leukemia, brain cancer, breast cancer, cervical cancer, colon cancer, lung cancer, atic cancer, prostate cancer, and skin cancer. (Leong et al., 2006, Blood, 107:2223-2233).
Delta-like 4 ligand (DLL4) is an important component of the Notch pathway and has been identified as a target for cancer therapy. DLL4 is a Notch ligand, characterized by an N-terminal domain, a Delta/Serrate/Lag-2 (DSL) domain and tandem EGF-like repeats within the extracellular domain. It has been reported that DLL4 is induced by VEGF and that DLL4 may act as a negative feedback tor for vascular proliferation.
Anti-DLL4 antibodies have been shown to enhance angiogenic sprouting and branching which leads to non-productive enesis and decreased tumor growth ra-Troise et al., 2006, Nature, 444:1032-1037). In addition, an anti-DLL4 antibody, 21M18, has been shown to inhibit tumor growth and reduce the frequency of cancer stem cells in xenograft tumor models (Hoey et al., 2009, Cell Stem Cell, 5:168-177; U.S. Patent No. 7,750,124).
Although there have been icant strides in development of monoclonal antibodies for use in cancer treatments, there is still great potential for further improvements. One class of antibody molecules with the promise of enhanced y and/or reduced side effects (e.g., toxicity) is bispecific antibodies.
Early bispecific molecules were mainly generated using chemical cross-linking of two antibodies, or were hybrid hybridomas or “quadromas”. One success of the quadroma format is triomabs, which are mouse/rat combinations that demonstrate a preferential species-specific heavy/light chain pairing. More recently, advances in dy engineering have provided a wide variety of new antibody formats, including, but not limited to, tandem scFv (bi-scFv), diabodies, tandem ies (tetra-bodies), single chain diabodies, and dual variable domain antibodies.
It is one of the objectives of the present invention to provide improved molecules for cancer treatment, particularly bispecific antibodies that specifically bind human VEGF and human DLL4.
SUMMARY OF THE ION [0011A] In a first , the t invention es a bispecific antibody comprising: a) a first antigen-binding site that specifically binds human vascular endothelial growth factor (VEGF), and b) a second antigen-binding site that specifically binds human delta-like 4 ligand (DLL4), n the first antigen-binding site comprises a heavy chain CDR1 comprising NYWMH (SEQ ID NO:17), a heavy chain CDR2 comprising DINPSNGRTSYKEKFKR (SEQ ID NO:18), and a heavy chain CDR3 sing HYDDKYYPLMDY (SEQ ID NO:19); wherein the second antigen-binding site comprises a heavy chain CDR1 comprising TAYYIH (SEQ ID NO:13) or AYYIH (SEQ ID NO:79), a heavy chain CDR2 comprising YIX1X2YX3X4ATNYNQKFKG (SEQ ID NO:80), wherein X1 is serine or alanine, X2 is serine, 11283960_1 asparagine, or glycine, X3 is asparagine or lysine, and X4 is glycine, arginine, or aspartic acid , and a heavy chain CDR3 comprising RDYDYDVGMDY (SEQ ID NO:16); and wherein both the first and second antigen-binding sites comprise a light chain CDR1 comprising RASESVDNYGISFMK (SEQ ID NO:20), a light chain CDR2 comprising AASNQGS (SEQ ID NO:21), and a light chain CDR3 comprising QQSKEVPWTFGG (SEQ ID NO:22). [0011B] In a second aspect, the present invention provides a ific antibody that specifically binds human VEGF and human DLL4, which comprises: (a) a heavy chain of SEQ ID NO:7; (b) a heavy chain of SEQ ID NO:5, SEQ ID NO:6, SEQ ID NO:56, or SEQ ID NO:62; and (c) two light chains of SEQ ID NO:8. [0011C] In a third , the present invention provides a bispecific antibody that specifically binds human VEGF and human DLL4, which comprises: (a) a heavy chain encoded by the DNA comprising SEQ ID NO:75, a heavy chain encoded by the DNA comprising SEQ ID NO:33, and a light chain encoded by the DNA comprising SEQ ID NO:34; (b) a heavy chain encoded by the DNA comprising SEQ ID NO:31, a heavy chain encoded by the DNA comprising SEQ ID NO:33, and a light chain encoded by the DNA comprising SEQ ID NO:34; (c) a heavy chain encoded by the DNA sing SEQ ID NO:72, a heavy chain encoded by the DNA comprising SEQ ID NO:33, and a light chain encoded by the DNA sing SEQ ID NO:34; or (d) a heavy chain encoded by the DNA comprising SEQ ID NO:74, a heavy chain encoded by the DNA comprising SEQ ID NO:33, and a light chain d by the DNA comprising SEQ ID NO:34. [0011D] In a fourth aspect, the t invention provides an isolated antibody that specifically binds human VEGF, which ses: (a) a heavy chain CDR1 comprising NYWMH (SEQ ID NO:17), a heavy chain CDR2 comprising DINPSNGRTSYKEKFKR (SEQ ID NO:18), and a heavy chain CDR3 comprising HYDDKYYPLMDY (SEQ ID NO:19); and 11283960_1 (b) a light chain CDR1 comprising RASESVDNYGISFMK (SEQ ID NO:20), a light chain CDR2 comprising AASNQGS (SEQ ID NO:21), and a light chain CDR3 comprising QQSKEVPWTFGG (SEQ ID NO:22). [0011E] In a fifth aspect, the present invention provides a pharmaceutical composition comprising the antibody of any of the first to fourth aspects and a pharmaceutically acceptable carrier. [0011F] In a sixth aspect, the present invention provides a cell comprising or producing the antibody of any of the first to fourth aspects, wherein the cell is not within a human body. [0011G] In a seventh aspect, the present invention provides an isolated polynucleotide molecule comprising a nucleotide sequence that encodes the antibody of any of the first to fourth aspects. [0011H] In an eighth aspect, the present invention provides a vector comprising the polynucleotide of the seventh aspect. [0011I] In a ninth aspect, the present invention provides a cell comprising the polynucleotide of the seventh aspect, wherein the cell is not within a human body. [0011J] In a tenth aspect, the present invention provides use of the dy of any of the first to fourth aspects in the manufacture of a medicament for treatment of cancer. [0011K] In an th aspect, the present invention provides use of the antibody of any of the first to fourth aspects in the manufacture of a ment for ting growth of a tumor. [0011L] In an twelfth aspect, the t invention provides a method for the production of an dy, comprising expressing at least one cleotide of the seventh aspect in a cell, wherein the antibody is not produced within a human body.
The present ion provides binding agents, such as antibodies, that bind VEGF, DLL4, or both VEGF and DLL4 (VEGF/DLL4-binding agents), as well as itions, such as pharmaceutical compositions, comprising the binding agents. Binding agents that bind VEGF or DLL4, as well as at least one additional antigen or target, and ceutical compositions of such binding agents, are also ed. In certain ments, the binding agents are novel polypeptides, such as antibodies, antibody nts, and other polypeptides related to such antibodies. In certain embodiments, the binding agents are antibodies that specifically bind human VEGF. In some embodiments, the binding agents are antibodies that specifically bind human DLL4. In some ments, the binding agents are bispecific antibodies that specifically bind human VEGF and human DLL4. The invention r provides methods of inhibiting the growth of a tumor by administering the binding agents to a subject with a tumor. 11283960_1 The invention further provides methods of treating cancer by administering the g agents to a subject in need thereof. In some embodiments, the methods of treating cancer or inhibiting tumor growth comprise targeting cancer stem cells with the binding . In certain embodiments, the methods comprise reducing the frequency of cancer stem cells in a tumor, reducing the number of cancer stem cells in a 11283960_1 tumor, reducing the tumorigenicity of a tumor, and/or reducing the tumorigenicity of a tumor by reducing the number or ncy of cancer stem cells in the tumor.
In one aspect, the invention provides a binding agent, such as an dy, that cally binds human VEGF. In some embodiments, the binding agent inhibits binding ofVEGF to at least one VEGF receptor. In some embodiments, the binding agent inhibits binding ofVEGF to VEGFR-l and/or VEGFR-Z. In some embodiments, the binding agent tes angiogenesis. In certain embodiments, the antibody or other binding agent further specifically binds to and/or inhibits human DLL4 in addition to human VEGF.
In some embodiments, the binding agent is an antibody which comprises a heavy chain CDRl comprising NYWMH (SEQ ID NO: 17), a heavy chain CDR2 comprising DINPSNGRTSYKEKFKR (SEQ ID NO: 18), and a heavy chain CDR3 comprising HYDDKYYPLMDY (SEQ ID NO: 19); and a light chain CDRl comprising RASESVDNYGISFMK (SEQ ID NO:20), a light chain CDR2 comprising AASNQGS (SEQ ID N021), and a light chain CDR3 comprising QQSKEVPWTFGG (SEQ ID NO:22).
In certain embodiments, the binding agent is an antibody that ses a heavy chain variable region having at least 80% sequence identity to SEQ ID NO:11; and/or a light chain variable region having at least 80% sequence identity to SEQ ID NO:12. In certain embodiments, the binding agent comprises a heavy chain variable region having at least 90% sequence ty to SEQ ID NO:11; and/or a light chain variable region having at least 90% sequence identity to SEQ ID NO: 12. In certain ments, the g agent comprises a heavy chain variable region having at least 95% ce identity to SEQ ID NO:11; and/or a light chain variable region having at least 95% sequence ty to SEQ ID NO:12. In certain embodiments, the binding agent is an antibody that comprises a heavy chain variable region of SEQ ID NO:11; and/or a light chain variable region of SEQ ID NO:12.
In some embodiments, the g agent is antibody 219R45, 219R45-MB-21M18, 219R45-MB- 21R79, 219R45-MB-21R75, or 219R45-MB—21R83.
In another aspect, the invention provides a binding agent, such as an antibody, that specifically binds human DLL4. In some embodiments, the binding agent inhibits binding of DLL4 to at least one Notch receptor. In some embodiments, the binding agent ts binding of DLL4 to Notchl, , Notch3, and/or Notch4. In some embodiments, the binding agent inhibits Notch signaling. In some embodiments, the binding agent promotes unproductive angiogenesis. In certain embodiments, the antibody or other g agent further cally binds to and/or inhibits human VEGF in addition to human DLL4.
In some embodiments, the binding agent is an antibody that binds human DLL4 and comprises a heavy chain CDRl comprising TAYYIH (SEQ ID NO:13) or AYYIH (SEQ ID NO:79), a heavy chain CDR2 comprising YIXleYX3X4ATNYNQKFKG (SEQ ID NO:80), wherein X1 is serine or alanine, X2 is serine, asparagine, or glycine, X3 is asparagine or lysine, and X4 is glysine, arginine,or aspartic acid and a heavy chain CDR3 comprising RDYDYDVGMDY (SEQ ID ; and a light chain CDRl comprising RASESVDNYGISFMK (SEQ ID NO:20), a light chain CDR2 comprising AASNQGS (SEQ ID NO:21), and a light chain CDR3 comprising QQSKEVPWTFGG (SEQ ID . In some embodiments, the antibody comprises a heavy chain CDRl comprising TAYYIH (SEQ ID NO:13) or AYYIH (SEQ ID NO:79), a heavy chain CDR2 comprising YIANYNRATNYNQKFKG (SEQ ID NO:14), and a heavy chain CDR3 comprising RDYDYDVGMDY (SEQ ID NO:16); and a light chain CDRl comprising RASESVDNYGISFMK (SEQ ID NO:20), a light chain CDR2 comprising AASNQGS (SEQ ID NO:21), and a light chain CDR3 comprising QQSKEVPWTFGG (SEQ ID NO:22).
In certain embodiments, the binding agent is an dy that comprises a heavy chain variable region having at least 90% or at least 95% sequence identity to SEQ ID NO: 10; and/or a light chain variable region having at least 90% or at least 95% sequence identity to SEQ ID NO: 12. In n embodiments, the binding agent is an antibody that comprises a heavy chain variable region of SEQ ID NO: 10; and a light chain variable region of SEQ ID NO: 12.
In some embodiments, the binding agent is antibody 21R79 or antibody 219R45-MB-21R79.
In some embodiments, the binding agent is an antibody which comprises a heavy chain CDRl comprising TAYYIH (SEQ ID NO: 13) or AYYIH (SEQ ID NO:79), a heavy chain CDR2 comprising YIAGYKDATNYNQKFKG (SEQ ID NO:59), and a heavy chain CDR3 comprising RDYDYDVGMDY (SEQ ID NO:16); and a light chain CDRl comprising RASESVDNYGISFMK (SEQ ID , a light chain CDR2 sing S (SEQ ID NO:21), and a light chain CDR3 comprising QQSKEVPWTFGG (SEQ ID NO:22).
In certain embodiments, the g agent is an antibody that comprises a heavy chain variable region having at least 90% or at least 95% sequence identity to SEQ ID NOS 8; and/or a light chain variable region having at least 90% or at least 95% sequence ty to SEQ ID NO: 12. In certain embodiments, the binding agent is an antibody that comprises a heavy chain variable region of SEQ ID NOS8; and a light chain variable region of SEQ ID NO:12.
In some embodiments, the binding agent is antibody 21R75 or antibody 219R45-MB-21R75.
In some embodiments, the binding agent is an antibody which comprises a heavy chain CDRl comprising TAYYIH (SEQ ID NO:13) or AYYIH (SEQ ID NO:79), a heavy chain CDR2 comprising YISNYNRATNYNQKFKG (SEQ ID , and a heavy chain CDR3 comprising RDYDYDVGMDY (SEQ ID NO: 16); and a light chain CDRl comprising RASESVDNYGISFMK (SEQ ID NO:20), a light chain CDR2 comprising AASNQGS (SEQ ID NO:21), and a light chain CDR3 comprising QQSKEVPWTFGG (SEQ ID .
In certain embodiments, the g agent is an antibody that comprises a heavy chain variable region having at least 90% or at least 95% sequence identity to SEQ ID NO:64; and/or a light chain variable region having at least 90% or at least 95% ce identity to SEQ ID NO:12. In certain embodiments, the binding agent is an antibody that comprises a heavy chain variable region of SEQ ID NO:64; and a light chain variable region of SEQ ID NO: 12.
In some embodiments, the binding agent is antibody 21R83 or dy 219R45-MB-21R83.
In certain embodiments of each of the aforementioned aspects or embodiments, as well as other s and/or embodiments described elsewhere herein, the binding agent is a ific antibody. In some embodiments, the bispecific antibody specifically binds human VEGF and a second target. In some embodiments, the bispecific antibody specifically binds human DLL4 and a second . In some embodiments, the bispecific antibody specifically binds both human VEGF and human DLL4. In some embodiments, the ific antibody modulates angiogenesis. In certain embodiments, the bispecific antibody ts Notch signaling. In some embodiments, the bispecific antibody modulates angiogenesis and ts Notch ing. In some embodiments, the bispecific antibody reduces the number of frequency of cancer stem cells. In certain embodiments, the bispecific dy comprises two identical light chains. In certain embodiments the bispecific antibody is an IgG antibody (e.g., IgG2).
In some embodiments, the bispecific antibody comprises: a first antigen-binding site that specifically binds human VEGF, wherein the first antigen—binding site comprises a heavy chain CDRl sing NYWMH (SEQ ID NO: 17), a heavy chain CDR2 comprising DINPSNGRTSYKEKFKR (SEQ ID NO:18), and a heavy chain CDR3 comprising HYDDKYYPLMDY (SEQ ID NO:19), In some embodiments, the bispecific antibody further comprises: a light chain CDRl comprising RASESVDNYGISFMK (SEQ ID NO:20), a light chain CDR2 comprising AASNQGS (SEQ ID NO:21), and a light chain CDR3 comprising QQSKEVPWTFGG (SEQ ID NO:22). In some embodiments, the ific antibody comprises: a first antigen—binding site that specifically binds human VEGF, wherein the first antigen-binding site comprises (a) a heavy chain CDRl comprising NYWMH (SEQ ID NO:17), a heavy chain CDR2 comprising DINPSNGRTSYKEKFKR (SEQ ID NO:18), and a heavy chain CDR3 comprising HYDDKYYPLMDY (SEQ ID NO:19), and (b) a light chain CDRl comprising RASESVDNYGISFMK (SEQ ID NO:20), a light chain CDR2 comprising S (SEQ ID NO:21), and a light chain CDR3 sing QQSKEVPWTFGG (SEQ ID NO:22).
In certain embodiments, the bispecific antibody ses: a first antigen-binding site that cally binds human DLL4, wherein the first antigen—binding site comprises a heavy chain CDRl comprising TAYYIH (SEQ ID NO:13) or AYYIH (SEQ ID NO:79), a heavy chain CDR2 comprising YIX1X2YX3X4ATNYNQKFKG (SEQ ID NO:80), wherein X1 is serine or alanine, X2 is serine, asparagine, or glycine, X3 is asparagine or lysine, and X4 is glysine, arginine,or aspartic acid and a heavy chain CDR3 comprising RDYDYDVGMDY (SEQ ID NO:16); and a light chain CDRl comprising RASESVDNYGISFMK (SEQ ID NO:20), a light chain CDR2 comprising AASNQGS (SEQ ID NO:21), and a light chain CDR3 comprising QQSKEVPWTFGG (SEQ ID NO:22). In some ments, the bispecific antibody comprises: a first antigen—binding site that specifically binds human DLL4, wherein the first antigen-binding site comprises a heavy chain CDR1 comprising TAYYIH (SEQ ID NO: 13), a heavy chain CDR2 comprising YIANYNRATNYNQKFKG (SEQ ID NO:14), YISSYNGATNYNQKFKG (SEQ ID NO:15), YIAGYKDATNYNQKFKG (SEQ ID NO:59), or YISNYNRATNYNQKFKG (SEQ ID NO:65), and a heavy chain CDR3 comprising RDYDYDVGMDY (SEQ ID NO: 16). In some embodiments, the bispecific antibody fiirther comprises: a light chain CDR1 comprising RASESVDNYGISFMK (SEQ ID NO:20), a light chain CDR2 comprising AASNQGS (SEQ ID NO:21), and a light chain CDR3 comprising QQSKEVPWTFGG (SEQ ID NO:22). In some embodiments, the the bispecific antibody comprises: a first antigen-binding site that specifically binds human DLL4, wherein the first antigen-binding site comprises (a) a heavy chain CDR1 comprising TAYYIH (SEQ ID NO: 13), a heavy chain CDR2 comprising YIANYNRATNYNQKFKG (SEQ ID NO: 14), YISSYNGATNYNQKFKG (SEQ ID NO: 15), YIAGYKDATNYNQKFKG (SEQ ID NO:59), or YISNYNRATNYNQKFKG (SEQ ID NO:65), and a heavy chain CDR3 comprising RDYDYDVGMDY (SEQ ID NO: 16), and (b) a light chain CDR1 comprising RASESVDNYGISFMK (SEQ ID NO:20), a light chain CDR2 comprising S (SEQ ID NO:21), and a light chain CDR3 comprising QQSKEVPWTFGG (SEQ ID NO:22).
In some embodiments, the ific antibody comprises: a) a first antigen-binding site that specifically binds human VEGF, and b) a second antigen—binding site that specifically binds human DLL4, wherein the first antigen-binding site comprises a heavy chain CDR1 sing NYWMH (SEQ ID NO: 17), a heavy chain CDR2 comprising DINPSNGRTSYKEKFKR (SEQ ID NO: 18), and a heavy chain CDR3 comprising HYDDKYYPLMDY (SEQ ID NO: 19); wherein the second antigen-binding site comprises a heavy chain CDR1 comprising TAYYIH (SEQ ID NO:13) or AYYIH (SEQ ID NO:79), a heavy chain CDR2 comprising YIXlXZYX3X4ATNYNQKFKG (SEQ ID NO:80), wherein X1 is serine or alanine, X2 is serine, asparagine, or e, X3 is asparagine or lysine, and X4 is glysine, arginine,or ic acid and a heavy chain CDR3 sing RDYDYDVGMDY (SEQ ID NO: 16); and a light chain CDR1 comprising RASESVDNYGISFMK (SEQ ID NO:20), a light chain CDR2 comprising AASNQGS (SEQ ID NO:21), and a light chain CDR3 comprising QQSKEVPWTFGG (SEQ ID NO:22).
In some ments, the bispecific antibody comprises: a) a first antigen—binding site that specifically binds human VEGF, and b) a second antigen—binding site that specifically binds human DLL4, wherein the first antigen-binding site ses a heavy chain CDR1 comprising NYWMH (SEQ ID , a heavy chain CDR2 sing DINPSNGRTSYKEKFKR (SEQ ID NO:18), and a heavy chain CDR3 comprising HYDDKYYPLMDY (SEQ ID NO:19); wherein the second antigen-binding site ses a heavy chain CDR1 comprising TAYYIH (SEQ ID NO:13), a heavy chain CDR2 comprising YIANYNRATNYNQKFKG (SEQ ID NO:14), and a heavy chain CDR3 comprising RDYDYDVGMDY (SEQ ID NO:16); and wherein both the first and second antigen—binding sites se a light chain CDR1 sing RASESVDNYGISFMK (SEQ ID NO:20), a light chain CDR2 comprising AASNQGS (SEQ ID NO:21), and a light chain CDR3 comprising QQSKEVPWTFGG (SEQ ID NO:22). In some embodiments, the bispecific antibody comprises: a) a first antigen-binding site that specifically binds human VEGF, and b) a second antigen-binding site that specifically binds human DLL4, wherein the first antigen-binding site comprises a heavy chain CDRl comprising NYWMH (SEQ ID NO:17), a heavy chain CDR2 comprising DINPSNGRTSYKEKFKR (SEQ ID NO:18), and a heavy chain CDR3 comprising HYDDKYYPLMDY (SEQ ID NO: 19); n the second antigen-binding site comprises a heavy chain CDRl comprising TAYYIH (SEQ ID NO:13), a heavy chain CDR2 comprising YISSYNGATNYNQKFKG (SEQ ID NO: 15), and a heavy chain CDR3 comprising RDYDYDVGMDY (SEQ ID NO: 16); and wherein both the first and second antigen-binding sites se a light chain CDRl comprising RASESVDNYGISFMK (SEQ ID NO:20), a light chain CDR2 comprising AASNQGS (SEQ ID NO:21), and a light chain CDR3 sing QQSKEVPWTFGG (SEQ ID NO:22). In some embodiments, the a bispecific dy ses: a) a first n-binding site that specifically binds human VEGF, and b) a second n—binding site that specifically binds human DLL4, wherein the first antigen-binding site comprises a heavy chain CDRl comprising NYWMH (SEQ ID NO: 17), a heavy chain CDR2 comprising GRTSYKEKFKR (SEQ ID NO: 18), and a heavy chain CDR3 comprising HYDDKYYPLMDY (SEQ ID ; wherein the second antigen-binding site comprises a heavy chain CDRl comprising TAYYIH (SEQ ID NO: 13), a heavy chain CDR2 comprising YIAGYKDATNYNQKFKG (SEQ ID NO:59), and a heavy chain CDR3 comprising RDYDYDVGMDY (SEQ ID NO: 16); and n both the first and second antigen-binding sites comprise a light chain CDRl comprising RASESVDNYGISFMK (SEQ ID NO:20), a light chain CDR2 comprising S (SEQ ID NO:21), and a light chain CDR3 comprising QQSKEVPWTFGG (SEQ ID NO:22). In some embodiments, the bispecific antibody comprises: a) a first antigen-binding site that specifically binds human VEGF, and b) a second antigen-binding site that specifically binds human DLL4, wherein the first antigen-binding site comprises a heavy chain CDRl comprising NYWMH (SEQ ID NO: 17), a heavy chain CDR2 comprising DINPSNGRTSYKEKFKR (SEQ ID NO:18), and a heavy chain CDR3 sing HYDDKYYPLMDY (SEQ ID NO: 19); wherein the second antigen-binding site comprises a heavy chain CDRl comprising TAYYIH (SEQ ID NO:13), a heavy chain CDR2 comprising YISNYNRATNYNQKFKG (SEQ ID NO:65), and a heavy chain CDR3 comprising RDYDYDVGMDY (SEQ ID NO: 16); and n both the first and second antigen-binding sites comprise a light chain CDRl comprising RASESVDNYGISFMK (SEQ ID NO:20), a light chain CDR2 comprising AASNQGS (SEQ ID NO:21), and a light chain CDR3 comprising QQSKEVPWTFGG (SEQ ID NO:22).
In some embodiments, the bispecific antibody that specifically binds human VEGF, and comprises: a heavy chain variable region having at least 90% sequence identity to SEQ ID NO:11, and/or a light chain variable region having at least 90% sequence identity to SEQ ID NO: 12. In some embodiments, the bispecific antibody specifically binds human VEGF, and comprises: a heavy chain variable region having at least 95% ce identity to SEQ ID NO:11, and/or a light chain variable region having at least 95% sequence identity to SEQ ID NO: 12.
In some embodiments, the bispecific antibody specifically binds human DLL4, and comprises: a heavy chain variable region having at least 90% sequence ty to SEQ ID NO:9, SEQ ID NO: 10, SEQ ID N025 8, or SEQ ID NO:64; and/or a light chain variable region having at least 90% ce identity to SEQ ID NO:12. In some embodiments, the bispecific dy specifically binds human DLL4, and comprises: a heavy chain variable region having at least 95% sequence identity to SEQ ID NO:9, SEQ ID NO: 10, SEQ ID N0258, or SEQ ID NO:64; and/or a light chain variable region having at least 95% ce identity to SEQ ID NO:12.
In some embodiments, the bispecific dy specifically binds human VEGF and human DLL4, and comprises: (a) a first heavy chain variable region having at least 90% sequence identity to SEQ ID NO:11; (b) a second heavy chain variable region having at least 90% sequence identity to SEQ ID NO:9, SEQ ID NO: 10, SEQ ID N025 8, or SEQ ID NO:64; and (c) a first and a second light chain variable region having at least 90% sequence identity to SEQ ID NO:12. In some embodiments, the VEGF/DLL4 bispecific antibody comprises (a) a first heavy chain variable region having at least 95% sequence identity to SEQ ID NO:11; (b) a second heavy chain variable region having at least 95% sequence identity to SEQ ID NO:9; and (c) a first and a second light chain variable region having at least 95% sequence identity to SEQ ID NO:12. In some embodiments, the VEGF/DLL4 ific antibody comprises (a) a first heavy chain variable region having at least 95% sequence identity to SEQ ID NO:11; (b) a second heavy chain variable region having at least 95% sequence identity to SEQ ID NO: 10; and (c) a first and a second light chain variable region having at least 95% sequence identity to SEQ ID NO:12. In some embodiments, the VEGF/DLL4 bispecific antibody comprises (a) a first heavy chain variable region having at least 95% sequence identity to SEQ ID NO:11; (b) a second heavy chain variable region having at least 95% sequence identity to SEQ ID NO:58; and (c) a first and a second light chain variable region having at least 95% sequence identity to SEQ ID NO:12. In some embodiments, the VEGF/DLL4 bispecific antibody comprises (a) a first heavy chain variable region having at least 95% sequence identity to SEQ ID NO:11; (b) a second heavy chain variable region having at least 95% sequence identity to SEQ ID NO:64; and (c) a first and a second light chain variable region having at least 95% sequence identity to SEQ ID NO:12.
In some embodiments, the VEGF/DLL4-binding agent is a bispecific antibody comprising (a) a first antigen-binding site that binds human VEGF with a KD between about 0.1 nM and about 1.0 nM and (b) a second antigen-binding site that cally binds human DLL4 with a KD between about 0.1 nM and about 20 nM. In certain ments, the ific dy ses two identical light chains.
In some embodiments, the VEGF/DLL4—binding agent is a bispecific antibody selected from the group consisting of 219R45-MB-21M18, 219R45—MB—21R79, 2 1 9R45-MB-2 1 R75, and 219R45-MB- 21R83.
In certain embodiments of each of the aforementioned aspects, as well as other aspects and/or embodiments described elsewhere herein, the binding agent or antibody is ed.
In another aspect, the invention provides a polypeptide selected from the group consisting of: SEQ ID NO: 1, SEQ ID N022, SEQ ID NO:3, SEQ ID NO:4, SEQ ID N0:5, SEQ ID N0:6, SEQ ID N027, SEQ ID N028, SEQ ID N029, SEQ ID N0:10, SEQ ID N0:l l, SEQ ID N0:12, SEQ ID N0:46, SEQ ID N0:47, SEQ ID N0248, SEQ ID NO:49, SEQ ID N0:56, SEQ ID N0:57, SEQ ID N0:58, SEQ ID N0:62, SEQ ID N0:63, and SEQ ID N0:64. In some embodiments, the polypeptide is isolated. In certain embodiments, the polypeptide is substantially pure. In n ments, the polypeptide is an antibody or part of an antibody, such as an dy fragment.
In another aspect, the invention provides ed polynucleotide molecules comprising a polynucleotide that encodes the binding agents and/or polypeptides of each of the entioned aspects, as well as other aspects and/or embodiments described herein. In some embodiments, the polynucleotide comprises a sequence selected from the group consisting of: SEQ ID N0:29, SEQ ID N0:30, SEQ ID N0:31, SEQ ID N0:32, SEQ ID N0:33, SEQ ID N0:34, SEQ ID N0:35, SEQ ID N0:36, SEQ ID N0:37, SEQ ID N0:38, SEQ ID N0:39, SEQ ID N0:40, SEQ ID N0:50, SEQ ID N0:51, SEQ ID N0:52, SEQ ID N0:53, SEQ ID N0:54, SEQ ID N0:55, SEQ ID N0:60, SEQ ID N0:61, SEQ ID N0:66, SEQ ID N0:67, SEQ ID N0:68, SEQ ID N0:69, SEQ ID N0:70, SEQ ID N0:71, SEQ ID N0:72, SEQ ID N0:73, and SEQ ID NO:74. The invention fiirther provides expression vectors that comprise the polynucleotides, as well as cells that comprise the expression s and/or the polynucleotides. In some embodiments, the cell is a prokaryotic cell or a eukaryotic cell.
In other aspects, the ion provides s of inhibiting growth of a tumor, comprising ting the tumor with an effective amount of an antibody (or other binding agent) that binds VEGF, DLL4, or both VEGF and DLL4, including each of those antibodies (or other binding agents) described herein.
In another aspect, the invention provides a method of inhibiting the growth of a tumor in a subject, comprising administering to the subject a therapeutically ive amount of an antibody (or other binding agent) that binds VEGF, DLL4, or both VEGF and DLL4, including each of those antibodies (or other binding agents) described herein.
In another aspect, the invention provides a method of modulating angiogenesis in a subject, comprising administering to the subject a therapeutically effective amount of an antibody (or other binding agent) that binds VEGF, DLL4, or both VEGF and DLL4, including each of those antibodies (or other g agents) bed herein.
In another aspect, the invention provides a method of reducing the tumorigenicity of a tumor in a subject, comprising administering to the subject a therapeutically effective amount of an antibody (or other binding agent) that binds VEGF, DLL4, or both VEGF and DLL4, including each of those antibodies (or other binding agents) described herein.
In another aspect, the invention provides a method of reducing the tumorigenicity of a tumor in a subject by reducing the frequency of cancer stem cells in the tumor, comprising administering to the subject a therapeutically effective amount of an antibody (or other binding agent) that binds VEGF, DLL4, or both VEGF and DLL4, including each of those antibodies (or other binding agents) bed In other aspects, the invention provides methods of treating cancer in a subject, comprising administering to the subject a therapeutically effective amount of an antibody (or other g agent) that binds VEGF, DLL4, or both VEGF and DLL4, including each of those dies (or other binding agents) described herein. ceutical compositions comprising a binding agent (e. g., antibody) described herein and a ceutically acceptable carrier are further provided, as are cell lines that express and/or produce the binding agents. Methods of treating cancer and/or inhibiting tumor growth in a t (e. g., a human) comprising administering to the subject an effective amount of a composition comprising the binding agents are also provided.
Where aspects or embodiments of the invention are described in terms of a Markush group or other grouping of alternatives, the present invention encompasses not only the entire group listed as a whole, but also each member of the group individually and all possible subgroups of the main group, and also the main group absent one or more of the group members. The present invention also envisages the explicit exclusion of one or more of any of the group members in the claimed ion.
BRIEF DESCRIPTIONS OF THE DRAWINGS Figure 1. 1A) Heavy chain and light chain CDRs of anti-VEGF/anti-DLL4 bispecific antibodies 219R45-MB-21M18, 219R45-MB-21M79, -MB-21M?5, and -MB-21M83; 1B) Heavy chain and light chain variable region SEQ ID NOs; 1C) Heavy chain and light chain SEQ ID NOs.
Figure 2. HTRF assay for simultaneous binding of bispecific antibodies to human VEGF and human DLL4. Results are reported in Relative Fluorescence Units (RFU), which represent the ratio of the relative fluorescence intensity at 665nm to the relative fluorescence intensity at 620nm. 219R45-MB- 21M18 (); 219R45-MB-21R79 (-I-); 219R45 plus 21M18 (-A-); 219R45 plus 21R79 (43-); 219R45 (- V-); 21M18 (-0—); 21R79 (—0—); control antibody LZ—l (—A—).
Figure 3. tion of VEGF—induced HUVEC proliferation by anti-VEGF/anti-DLL4 ific antibodies. Fluorescence intensity is read using an excitation ngth of 530nm and an emission wavelength of 590. 219R45-MB-21M18 (—0—); 219R45—MB—21R79 (-A-); 219R45 (-l-); Medium with no VEGF ().
Figure 4. Inhibition of nduced Notch signaling by anti-VEGF/anti-DLL4 bispecific antibodies. Luciferase activity was measured using a dual luciferase assay kit with firefly luciferase activity normalized to Renilla luciferase activity. 219R45—MB-21M18 (); 219R45-MB-21R79 (-l-); 21M18 (); 21R79 (-E-).
Figure 5. Inhibition of colon tumor growth in vivo by an EGF/anti-DLL4 bispeciflc antibody. OMP-C8 colon tumor cells were injected subcutaneously into a human skin graft in ID mice. Mice were treated with control antibody (—I—), anti—hDLL4 dy 21M18 (-A -), anti-VEGF antibody bevacizumab (), or anti-VEGF/anti-DLL4 ific antibody 219R45-MB-21M18 (- V-).
Data is shown as tumor volume (photons/sec) over days post-treatment. Antibodies were administered intraperitoneally at a dose of 25mg/kg once a week.
Figure 6. Tumorigenicity of pancreatic tumor cells after treatment with anti-VEGF/anti-DLL4 bispecific antibodies. 8 tumor cells from mice treated with control antibody, anti-hDLL4 antibody 21M18, anti-VEGF antibody bevacizumab, or anti—VEGF/anti-DLL4 bispecific antibodies 219R45-MB-21M18 or 219R45-MB-21R79 with or without gemcitabine were processed to single cell suspensions, and serially transplanted into mice. 90 cells from each treatment group were injected subcutaneously into NOD/SCID mice. Tumors were allowed to grow with no ent. Data is shown as tumor volume (mm3) on day 55. Tumor frequency is shown as number of tumors over total number of mice injected in each group.
Figure 7. Bispecific antibody ELISA. Bispecific antibodies 219R45-MB-21M18, 219R45-MB- 2 1 R79, 2 1 9R45-MB-2 1 R75, and 219R45-MB-21R83 were diluted in blocking buffer (1x PBS, 0.1% gelatin, 0.1% Polysorbate-20, pH 7.4) containing 2ug/ml biotin-DLL4-hFc. The antibodies were serially diluted 3-fold from SOOng/ml to 0.008ng/ml. The antibody samples were incubated for 2 hours in blocking buffer containing the biotin-DLL4—hFc. After incubation, the antibody samples were transferred to a VEGF-coated assay plate (100 l) and incubated for 2 hours. Streptavidin—HRP was added to each well and incubated for 1 hr. TMB substrate was added to the wells with a 10 minute color development and the reaction was stopped with 2M sulfuric acid. Absorbance was read at 450—65Onm and the data analyzed using the 4-parameter fit within the Softmax Pro analysis program.
Figure 8. Imaged capillary isoelectric focusing of anti—VEGF/anti—DLL4 bispecific antibodies.
Figure 9. Inhibition of colon tumor growth by EGF/anti-DLL4 bispecific antibodies in tumor recurrence model. OMP-C8 colon tumor cells were injected subcutaneously in NOD/SCID mice.
Mice were d with control antibody (-I-), anti-hDLL4 antibody 21M18 (), anti-VEGF antibody bevacizumab (— A-), a combination of 21M1 8 and bevacizumab (— V -), anti-VEGF/anti-DLL4 ific antibody 219R45-MB-21M18 (—0—), or anti—VEGF/anti—DLL4 bispecific antibody 219R45-MB-21R79 (-o- ), all in combination with irinotecan. Antibodies 21M18 and bevacizumab were stered intraperitoneally at a dose of kg once a week, ific antibodies 219R45-MB-21M18 and 219R45-MB-21R79 were administered intraperitoneally at a dose of lSmg/kg once a week, and irinotecan was administered for the first 4 weeks at a dose of 45mg/kg. Data are shown as tumor volume (mm3) over days post-treatment.
Figure 10. Tumorigenicity of OMP-C3 colon tumor cells after treatment with anti-VEGF/anti- DLL4 bispecific dies. Tumors from mice treated with control antibody, anti-hDLL4 antibody 21Ml8, anti-VEGF antibody bevacizumab, a combination of 21M18 and bevacizumab, or anti- VEGF/anti-DLL4 bispecific antibodies —MB—21M1 8 or 219R45—MB—21R79 with or without irinotecan were processed to single cell suspensions, and serially transplanted into mice. 150 cells from each treatment group were injected aneously into NOD/SCID mice. Tumors were allowed to grow with no treatment. Data are shown as tumor volume (mm3) on day 68.
Figure 11. Inhibition of colon tumor growth in vivo by EGF/anti-DLL4 ific antibodies. OMP-C8 colon tumor cells were ed subcutaneously into ID mice. Mice were treated with control antibody (-l—), anti—VEGF antibody bevacizumab (- A -), or anti-VEGF/anti-DLL4 bispecific antibodies 219R45-MB-21M18 (—<>—), 219R45—MB-21R75 (4-), 219R45-MB-21R79 (-o-), or 219R45-MB-21R83 (- V-). Mice were d with antibodies as single agents (Fig. 10A) or in combination with irinotecan (Fig. 10B). Antibodies were administered intraperitoneally at a dose of 15mg/kg once a week and irinotecan at a dose of 7.5mg/kg one a week. Data are shown as tumor volume (mm3) over days post-treatment.
DETAILED DESCRIPTION OF THE INVENTION The present invention provides novel binding agents, including but not d to polypeptides such as dies, that bind VEGF and/or DLL4 (e.g., a VEGF/DLL4 binding agent). Related polypeptides and polynucleotides, compositions comprising the VEGF/DLL4-binding agents, and methods of making the VEGF/DLL4-binding agents are also provided. Methods of using the novel VEGF/DLL4-binding agents, such as methods of inhibiting tumor growth, methods of treating cancer, methods of reducing tumorigenicity of a tumor, methods of reducing the frequency of cancer stem cells in a tumor, and/or methods of modulating angiogenesis, are fiirther provided.
A monoclonal dy that specifically binds human VEGF has been identified, 219R45. This antibody has a binding affinity for human VEGF of about 0.67nM, and a binding affinity for mouse VEGF of about 23nM. Several monoclonal dies that specifically bind human DLL4 have been identified, 21R79, 21R75 and 21R83. Antibody 21R79 has a g affinity for human DLL4 of less than 0.1nM. Bispecific antibodies that specifically bind human VEGF and human DLL4 have been ed, 219R45-MB-21M18, 219R45—MB-21R79, 219R45—MB-21R75, and 219R45-MB-21R83 (CDR ces in Figure 1). As used herein, the “MB” within an antibody name refers to “monovalent/bispecific”. Bispecific antibody 219R45—MB—21M18 has a binding affinity for human VEGF of less than 1.0nM and a binding affinity for human DLL4 of about 16nM. Bispecific antibody 219R45-MB-21R79 has a binding affinity for human VEGF of less than 1.0nM and a binding affinity for human DLL4 of less than 1.0nM. Bispecific antibody 219R45-MB-21R75 has a binding affinity for human DLL4 of about SnM, while ific antibody —MB-21R83 has a binding affinity for human DLL4 of about lnM. Bispecific antibodies 219R45—MB—21Ml 8 and 219R45-MB-21R79 bind mouse VEGF le 1, Table 3). Anti—VEGF/anti—DLL4 bispecific antibodies bind human VEGF and human DLL4 simultaneously (Example 2, Figure 2). Anti—VEGF/anti—DLL4 bispecific dies inhibit VEGF-induced proliferation of HUVEC cells (Example 3, Figure 3). Anti-VEGF/anti-DLL4 bispecific dies inhibit nduced Notch signaling (Example 4, Figure 4). Anti-VEGF/anti-DLL4 bispecific antibodies inhibit tumor growth (Examples 5, 9, 11 and Figures 5, 9, 11). Anti-VEGF/anti- DLL4 bispecific antibodies inhibit tumorigenicity (Examples 6 and 10 and Figures 6, 10). Anti- VEGF/anti-DLL4 bispecific antibodies bind both VEGF and DLL4 in a bispecific ELISA (Example 7, Figure 7). Anti-VEGF/anti-DLL4 bispecific antibodies are isolated and purified to a product comprising at least 90% heterodimeric antibody (Example 8, Table 7). 1. Definitions To facilitate an understanding of the present invention, a number of terms and phrases are defined below.
The term “antibody” as used herein refers to an immunoglobulin le that recognizes and cally binds a target, such as a protein, polypeptide, peptide, carbohydrate, polynucleotide, lipid, or combinations of the foregoing, through at least one antigen recognition site within the variable region of the immunoglobulin molecule. As used herein, the term encompasses intact polyclonal antibodies, intact monoclonal antibodies, single chain antibodies, antibody fragments (such as Fab, Fab', F(ab')2, and Fv fragments), single chain Fv (scFv) antibodies, multispecific antibodies such as bispecific antibodies, monospecific dies, lent antibodies, ic antibodies, humanized antibodies, human antibodies, fusion proteins comprising an antigen—binding site of an antibody, and any other modified immunoglobulin molecule comprising an antigen recognition site (i.e., antigen-binding site) as long as the antibodies exhibit the desired ical activity. An antibody can be any of the five major classes of immunoglobulins: IgA, IgD, IgE, IgG, and IgM, or subclasses (isotypes) thereof (e.g., IgGl, IgGZ, IgG3, IgG4, IgAl, and IgA2), based on the identity of their heavy chain nt domains referred to as alpha, delta, epsilon, gamma, and mu, respectively. The ent classes of immunoglobulins have different and well-known subunit structures and three—dimensional configurations. Antibodies can be naked or conjugated to other molecules, including but not limited to, toxins and radioisotopes.
The term “antibody fragment” refers to a portion of an intact antibody and refers to the antigenic determining variable regions of an intact antibody. Examples of dy fragments include, but are not limited to, Fab, Fab', F(ab')2, and Fv fragments, linear antibodies, single chain antibodies, and pecific antibodies formed from antibody fragments. “Antibody fragment” as used herein comprises an antigen-binding site or epitope-binding site.
The term “variable region” of an antibody refers to the variable region of an antibody light chain, or the le region of an antibody heavy chain, either alone or in combination. The variable regions of the heavy and light chains each t of four framework regions (FR) connected by three complementarity ining regions (CDRs), also known as “hypervariable regions”. The CDRs in each chain are held together in close proximity by the framework regions and, with the CDRs from the other chain, contribute to the formation of the antigen-binding site of the antibody. There are at least two techniques for determining CDRs: (1) an approach based on cross-species ce variability (i.e., Kabat et al., 1991, ces ofProteins 0fImmunoiogical Interest, 5th Edition, National Institutes of Health, Bethesda, MD), and (2) an approach based on crystallographic studies of antigen-antibody complexes (Al- Lazikani et al., 1997, J. M0]. Biol, 273:927—948). In addition, combinations of these two approaches are sometimes used in the art to determine CDRs.
The term “monoclonal antibody” as used herein refers to a homogeneous antibody population involved in the highly specific recognition and binding of a single antigenic determinant or epitope. This is in contrast to polyclonal antibodies that typically include a mixture of ent antibodies directed against a y of different antigenic determinants. The term “monoclonal antibody” encompasses both intact and full-length monoclonal antibodies as well as antibody fragments (e.g., Fab, Fab', F(ab')2, Fv), single chain (scFv) antibodies, fusion proteins comprising an antibody portion, and any other modified immunoglobulin molecule comprising an n recognition site (antigen-binding site). Furthermore, “monoclonal antibody” refers to such antibodies made by any number of techniques, including but not limited to, hybridoma production, phage selection, inant expression, and enic animals.
The term “humanized antibody” as used herein refers to forms of non-human (e. g., murine) antibodies that are specific immunoglobulin chains, chimeric immunoglobulins, or fragments thereof that contain minimal non-human sequences. Typically, humanized antibodies are human immunoglobulins in which residues of the CDRs are replaced by residues from the CDRs of a man species (e.g., mouse, rat, , or hamster) that have the desired city, affinity, and/or binding capability (Jones et al., 1986, Nature, 321:522-525; ann et al., 1988, Nature, 332:323-327; Verhoeyen et al., 1988, Science, 239: 1534-1536). In some instances, the Fv framework region residues of a human immunoglobulin are replaced with the corresponding residues in an antibody from a non-human species that has the desired specificity, y, and/or binding lity. The humanized antibody can be further modified by the substitution of additional residues either in the Fv framework region and/or within the replaced non-human residues to refine and optimize antibody specificity, affinity, and/or binding lity. In general, the zed dy will comprise substantially all of at least one, and typically two or three, variable domains containing all or substantially all of the CDRs that correspond to the non- human immunoglobulin whereas all or ntially all of the framework regions are those of a human immunoglobulin consensus sequence. The humanized antibody can also comprise at least a portion of an immunoglobulin constant region or domain (Fc), typically that of a human immunoglobulin. Examples of methods used to generate humanized antibodies are described in, for example, US. Patent 5,225,539.
The term “human antibody” as used herein refers to an antibody produced by a human or an dy having an amino acid sequence corresponding to an antibody produced by a human. A human antibody may be made using any of the techniques known in the art. This definition of a human antibody specifically excludes a humanized antibody comprising non-human CDRs.
The term “chimeric antibody” as used herein refers to an antibody wherein the amino acid sequence of the immunoglobulin le is derived from two or more species. Typically, the variable region of both light and heavy chains corresponds to the variable region of antibodies derived from one species of mammals (e. g., mouse, rat, rabbit, etc.) with the desired specificity, affinity, and/or binding capability, while the constant regions correspond to sequences in antibodies derived from another species (usually human).
The phrase “affinity-matured antibody” as used herein refers to an antibody with one or more alterations in one or more CDRs thereof that result in an improvement in the affinity of the dy for antigen, compared to a parent antibody that does not possess those alterations(s). The definition also includes alterations in non-CDR residues made in conjunction with tions to CDR es. Preferred affinity-matured antibodies will have nanomolar or even lar affinities for the target antigen. y-matured antibodies are produced by ures known in the art. For example, Marks et al., 1992, Bio/Technology 10:779-783, describes affinity maturation by VH and VL domain shuffling.
Random mutagenesis of CDR and/or framework residues is described by Barbas et al., 1994, PNAS, 91 :3809-3813; Schier et al., 1995, Gene, 169:147—155; Yelton et al., 1995, J. Immunol. 155: 1994-2004; Jackson et al., 1995, J. Immunol, 154:3310—9; and Hawkins et al., 1992, J. Mol. Biol, 226:889-896. Site- ed mutagenesis may also be used to obtain affinity—matured antibodies.
The terms “epitope” and “antigenic determinant” are used interchangeably herein and refer to that portion of an antigen capable of being recognized and specifically bound by a particular antibody. When the n is a polypeptide, epitopes can be formed both from contiguous amino acids and noncontiguous amino acids juxtaposed by tertiary folding of a n. es formed from contiguous amino acids (also referred to as linear epitopes) are lly retained upon protein ring, whereas epitopes formed by tertiary folding (also referred to as conformational epitopes) are lly lost upon protein ring. An epitope typically includes at least 3, and more usually, at least 5 or 8-10 amino acids in a unique spatial conformation.
The terms omultimeric molecule” or omultimer” or “heteromultimeric complex” or “heteromultimeric polypeptide” are used interchangeably herein to refer to a molecule comprising at least a first polypeptide and a second polypeptide, wherein the second polypeptide differs in amino acid sequence from the first polypeptide by at least one amino acid residue. The heteromultimeric molecule can comprise a “heterodimer” formed by the first and second polypeptide or can form higher order tertiary structures where onal polypeptides are present.
The terms onist” and “antagonistic” as used herein refer to any molecule that partially or fully blocks, inhibits, reduces, or neutralizes a biological activity of a target and/or signaling pathway (e.g., the Notch pathway). The term “antagonist” is used herein to include any molecule that partially or fully blocks, inhibits, reduces, or neutralizes the activity of a protein. Suitable antagonist les specifically include, but are not d to, antagonist antibodies or antibody fragments.
The terms “modulation” and “modulate” as used herein refer to a change or an tion in a biological activity. Modulation includes, but is not limited to, stimulating or ting an activity.
Modulation may be an increase or a decrease in activity (e.g., a decrease in angiogenesis or an increase in angiogenesis), a change in binding teristics, or any other change in the biological, functional, or immunological properties ated with the activity of a protein, pathway, or other biological point of interest.
The terms “selectively binds” or “specifically binds” mean that a binding agent or an antibody reacts or associates more frequently, more rapidly, with greater duration, with greater affinity, or with some combination of the above to the epitope, protein, or target le than with alternative substances, including unrelated proteins. In certain embodiments “specifically binds” means, for instance, that an antibody binds a n with a KB of about 0.1mM or less, but more y less than about 1 uM. In certain embodiments, “specifically binds” means that an antibody binds a target at times with a KD of at least about 0.1uM or less, at other times at least about 0.01pM or less, and at other times at least about 1nM or less. Because of the sequence identity between homologous proteins in different species, specific binding can include an antibody that recognizes a protein in more than one species (e.g., human VEGF and mouse VEGF). Likewise, because of homology within n regions of polypeptide sequences of different proteins, specific binding can include an antibody (or other polypeptide or binding agent) that recognizes more than one n (e.g., human VEGF-A and human VEGF-B). It is understood that, in certain embodiments, an antibody or binding moiety that specifically binds a first target may or may not specifically bind a second . As such, “specific binding” does not necessarily require (although it can include) exclusive binding, i.e. binding to a single target. Thus, an antibody may, in n embodiments, specifically bind more than one target. In certain ments, multiple targets may be bound by the same antigen-binding site on the antibody. For example, an antibody may, in certain instances, comprise two identical antigen-binding sites, each of which specifically binds the same epitope on two or more proteins. In certain alternative embodiments, an antibody may be multispecific and comprise at least two antigen-binding sites with differing specificities. By way of non-limiting example, a bispecific antibody may comprise one antigen-binding site that recognizes an epitope on one protein (e.g., human VEGF) and further comprise a second, different antigen—binding site that recognizes a different epitope on a second protein (e. g., human DLL4). Generally, but not necessarily, reference to binding means c binding.
The terms “polypeptide” and “peptide” and “protein” are used interchangeably herein and refer to polymers of amino acids of any length. The polymer may be linear or branched, it may comprise modified amino acids, and it may be interrupted by non-amino acids. The terms also encompass an amino acid polymer that has been modified naturally or by ention; for example, disulfide bond formation, ylation, lipidation, acetylation, phosphorylation, or any other manipulation or modification, such as conjugation with a labeling ent. Also included within the definition are, for example, polypeptides containing one or more s of an amino acid (including, for e, ral amino acids), as well as other modifications known in the art. It is understood that, because the polypeptides of this invention may be based upon antibodies, in certain embodiments, the polypeptides can occur as single chains or associated chains.
The terms “polynucleotide” and “nucleic acid” are used interchangeably herein and refer to polymers of nucleotides of any length, and e DNA and RNA. The nucleotides can be deoxyribonucleotides, ribonucleotides, modified nucleotides or bases, and/or their analogs, or any substrate that can be incorporated into a polymer by DNA or RNA polymerase.
“Conditions of high ency” may be identified by those that: (1) employ low ionic th and high temperature for washing, for example 15mM sodium chloride/1.5mM sodium citrate/0. 1% sodium dodecyl sulfate at 50°C; (2) employ during hybridization a ring agent, such as formamide, for example, 50% (v/v) formamide with 0.1% bovine serum n/0. 1% Ficoll/0. 1% polyvinylpyrrolidone/50mM sodium phosphate buffer at pH 6.5 in 5x SSC (0.75M NaCl, 75mM sodium citrate) at 42°C; or (3) employ during hybridization 50% formamide in 5x SSC, 50mM sodium phosphate (pH 6.8), 0.1% sodium pyrophosphate, 5x Denhardt's on, sonicated salmon sperm DNA (50ug/ml), 0.1% SDS, and 10% dextran e at 42°C, with washes at 42°C in 0.2x SSC and 50% formamide, followed by a high-stringency wash consisting of 0. 1x SSC containing EDTA at 55°C.
The terms “identical” or percent “identity” in the context of two or more nucleic acids or polypeptides, refer to two or more sequences or subsequences that are the same or have a specified percentage of nucleotides or amino acid residues that are the same, when compared and aligned (introducing gaps, if necessary) for maximum correspondence, not considering any conservative amino acid substitutions as part of the sequence identity. The percent identity may be measured using sequence comparison software or algorithms or by Visual inspection. Various algorithms and software that may be used to obtain alignments of amino acid or nucleotide sequences are well-known in the art. These include, but are not limited to, BLAST, ALIGN, Megalign, BestFit, GCG Wisconsin Package, and variations thereof. In some embodiments, two nucleic acids or polypeptides of the invention are substantially identical, meaning they have at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, and in some embodiments at least 95%, 96%, 97%, 98%, 99% nucleotide or amino acid residue identity, when compared and aligned for maximum correspondence, as measured using a sequence comparison algorithm or by visual inspection. In some embodiments, identity exists over a region of the sequences that is at least about 10, at least about 20, at least about 40—60 residues, at least about 60-80 es in length or any integral value therebetween. In some embodiments, identity exists over a longer region than 60-80 residues, such as at least about 80-100 residues, and in some embodiments the sequences are substantially identical over the full length of the sequences being compared, such as the coding region of a nucleotide sequence.
A “conservative amino acid tution” is one in which one amino acid residue is replaced with another amino acid residue having a similar side chain. Families of amino acid residues having similar side chains have been defined in the art, including basic side chains (e.g., , arginine, histidine), acidic side chains (e.g., aspartic acid, glutamic acid), uncharged polar side chains (e.g., glycine, asparagine, glutamine, serine, threonine, tyrosine, cysteine), ar side chains (e. g., alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan), beta-branched side chains (e.g., threonine, valine, cine) and aromatic side chains (e.g., ne, phenylalanine, tryptophan, histidine). For example, substitution of a phenylalanine for a tyrosine is a vative substitution. ably, conservative substitutions in the sequences of the ptides and antibodies of the invention do not abrogate the binding of the polypeptide or antibody containing the amino acid sequence, to the n to which the polypeptide or antibody binds. Methods of identifying nucleotide and amino acid conservative substitutions which do not eliminate antigen binding are well-known in the art.
The term “vector” as used herein means a uct, which is capable of delivering, and usually expressing, one or more gene(s) or sequence(s) of interest in a host cell. Examples of vectors include, but are not limited to, viral vectors, naked DNA or RNA expression vectors, plasmid, cosmid, or phage vectors, DNA or RNA expression vectors associated with cationic condensing agents, and DNA or RNA expression vectors ulated in liposomes.
A ptide, antibody, cleotide, vector, cell, or composition which is “isolated” is a ptide, dy, polynucleotide, vector, cell, or composition which is in a form not found in nature.
Isolated polypeptides, antibodies, polynucleotides, vectors, cells, or compositions include those which have been purified to a degree that they are no longer in a form in which they are found in nature. In some embodiments, a polypeptide, antibody, cleotide, vector, cell, or composition which is isolated is substantially pure.
The term “substantially pure” as used herein refers to al which is at least 50% pure (i.e., free from contaminants), at least 90% pure, at least 95% pure, at least 98% pure, or at least 99% pure.
The terms “cancer” and “cancerous” as used herein refer to or describe the physiological condition in mammals in which a population of cells are characterized by unregulated cell growth.
Examples of cancer include, but are not limited to, carcinoma, blastoma, sarcoma, and hematologic cancers such as lymphoma and ia.
The terms “tumor” and “neoplasm” as used herein refer to any mass of tissue that results from excessive cell growth or proliferation, either benign (noncancerous) or malignant (cancerous) including pre-cancerous lesions.
The term “metastasis” as used herein refers to the process by which a cancer spreads or transfers from the site of origin to other s of the body with the development of a r cancerous lesion at a new location. A “metastatic” or “metastasizing” cell is one that loses adhesive contacts with neighboring cells and migrates via the bloodstream or lymph from the y site of disease to invade neighboring body structures.
The terms “cancer stem cell” and “CSC” and “tumor stem cell” and “tumor initiating cell” are used interchangeably herein and refer to cells from a cancer or tumor that: (1) have extensive proliferative capacity; 2) are capable of asymmetric cell division to generate one or more types of differentiated cell progeny wherein the entiated cells have d proliferative or developmental ial; and (3) are capable of ric cell divisions for self-renewal or self-maintenance. These ties confer on the cancer stem cells the ability to form or establish a tumor or cancer upon serial transplantation into an immunocompromised host (e.g., a mouse) compared to the majority oftumor cells that fail to form tumors. Cancer stem cells undergo enewal versus differentiation in a chaotic manner to form tumors with abnormal cell types that can change over time as ons occur.
The terms “cancer cell” and “tumor cell” refer to the total population of cells derived from a cancer or tumor or pre-cancerous lesion, including both non—tumorigenic cells, which comprise the bulk of the cancer cell population, and tumorigenic stem cells (cancer stem cells). As used herein, the terms “cancer cell” or “tumor cell” will be modified by the term “non—tumorigenic” when referring solely to those cells lacking the capacity to renew and differentiate to distinguish those tumor cells from cancer stem cells.
The term “tumorigenic” as used herein refers to the functional features of a cancer stem cell including the properties of self-renewal (giving rise to additional tumorigenic cancer stem cells) and proliferation to generate all other tumor cells (giving rise to differentiated and thus non-tumorigenic tumor cells).
The term “tumorigenicity” as used herein refers to the ability of a random sample of cells from the tumor to form palpable tumors upon serial transplantation into immunocompromised hosts (e.g., mice).
This definition also includes enriched and/or isolated populations of cancer stem cells that form palpable tumors upon serial transplantation into immunocompromised hosts (e. g., mice).
The term “subject” refers to any animal (e.g., a ), including, but not limited to, humans, non-human primates, canines, felines, rodents, and the like, which is to be the ent of a particular treatment. Typically, the terms “subject” and “patient” are used interchangeably herein in reference to a human subject.
The term “pharmaceutically acceptable” refers to a product or compound approved (or approvable) by a regulatory agency of the Federal government or a state government or listed in the US.
Pharmacopeia or other generally recognized pharmacopeia for use in animals, including humans.
The terms “pharmaceutically acceptable excipient, carrier or adjuvant” or “acceptable pharmaceutical carrier” refer to an excipient, carrier or adjuvant that can be administered to a subject, together with at least one binding agent (e.g., an antibody) of the present disclosure, and which does not destroy the activity of the g agent. The excipient, carrier or adjuvant should be nontoxic when administered with a binding agent in doses sufficient to deliver a therapeutic effect.
The terms tive amount” or “therapeutically ive amount” or “therapeutic effect” refer to an amount of a binding agent, an antibody, polypeptide, polynucleotide, small organic molecule, or other drug effective to ” a e or disorder in a subject or mammal. In the case of cancer, the therapeutically ive amount of a drug (e.g., an dy) has a eutic effect and as such can reduce the number of cancer cells; se tumorigenicity, tumorigenic frequency or tumorigenic capacity; reduce the number or frequency of cancer stem cells; reduce the tumor size; reduce the cancer cell population; inhibit and/or stop cancer cell infiltration into peripheral organs including, for example, the spread of cancer into soft tissue and bone; inhibit and/or stop tumor or cancer cell metastasis; inhibit and/or stop tumor or cancer cell growth; relieve to some extent one or more of the symptoms associated with the cancer; reduce morbidity and mortality; improve quality of life; or a combination of such effects.
To the extent the agent, for example an antibody, prevents growth and/or kills existing cancer cells, it can be referred to as cytostatic and/or xic.
The terms “treating” or “treatment” or “to treat” or “alleviating” or “to alleviate” refer to both 1) therapeutic measures that cure, slow down, lessen symptoms of, and/or halt progression of a diagnosed pathologic condition or disorder and 2) lactic or preventative measures that t or slow the development of a targeted pathologic condition or disorder. Thus those in need of treatment include those y with the disorder; those prone to have the disorder; and those in whom the disorder is to be prevented. In some embodiments, a subject is successfully “treated” according to the methods of the present invention if the t shows one or more of the following: a reduction in the number of or complete absence of cancer cells; a reduction in the tumor size; inhibition of or an absence of cancer cell ration into peripheral organs including the spread of cancer cells into soft tissue and bone; inhibition of or an absence of tumor or cancer cell metastasis; inhibition or an absence of cancer ; relief of one or more symptoms associated with the specific cancer; reduced morbidity and mortality; ement in quality of life; reduction in genicity; reduction in the number or frequency of cancer stem cells; or some combination of effects.
As used in the present disclosure and claims, the singular forms EL 99 L: a an” and “the” include plural forms unless the context clearly dictates otherwise.
It is understood that wherever embodiments are bed herein with the language “comprising” otherwise analogous embodiments described in terms of “consisting of” and/or “consisting essentially of” are also provided. It is also understood that wherever embodiments are described herein with the language “consisting essentially of” otherwise analogous embodiments described in terms of “consisting of” are also provided.
The term “and/or” as used in a phrase such as “A and/or B” herein is intended to include both A and B; A or B; A (alone); and B (alone). Likewise, the term “and/or” as used in a phrase such as “A, B, and/or C” is intended to encompass each of the following embodiments: A, B, and C; A, B, or C; A or C; A or B; B or C; A and C; A and B; B and C; A (alone); B (alone); and C (alone).
II. Antibodies The present invention provides agents that specifically bind human VEGF proteins and/or human DLL4 proteins. These agents are referred to herein as “VEGF/DLL4-binding agents”. The phrase DLL4-binding agent” encompasses agents that bind only VEGF, agents that bind only DLL4, and bispecific agents that bind both VEGF and DLL4. In certain embodiments, in addition to specifically binding VEGF and/or DLL4, the VEGF/DLL4—binding agents further cally bind at least one onal target or antigen. In some embodiments, the VEGF/DLL4-binding agent is an antibody. In some embodiments, the VEGF/DLL4-binding agent is a polypeptide. In certain embodiments, the VEGF/DLL4-binding agent specifically binds human VEGF. In n ments, the VEGF/DLL4- binding agent specifically binds human DLL4. In certain embodiments, the LL4-binding agent is a bispecific antibody. In certain embodiments, the VEGF/DLL4—binding agent is a bispecific dy that specifically binds human VEGF and human DLL4. The full—length amino acid (aa) sequences for human VEGF (VEGF-A) and human DLL4 are known in the art and are provided herein as SEQ ID NO:27 (VEGF) and SEQ ID NO:23 (DLL4).
In certain embodiments, the VEGF/DLL4-binding agent or antibody binds VEGF and/or DLL4 with a dissociation constant (KD) of about luM or less, about 100nM or less, about 40nM or less, about 20nM or less, about 10nM or less, about lnM or less, or about 0.1nM or less. In some embodiments, a VEGF/DLL4-binding agent or dy binds VEGF and/or DLL4 with a KD of about 20nM or less. In some embodiments, a LL4-binding agent or antibody binds VEGF and/or DLL4 with a KD of about 10nM or less. In some embodiments, a VEGF/DLL4—binding agent or antibody binds VEGF and/or DLL4 with a KD of about lnM or less. In some embodiments, a VEGF/DLL4-binding agent or antibody binds VEGF and/or DLL4 with a KD of about 0.1nM or less. In some embodiments, the VEGF/DLL4- binding agent binds both human VEGF and mouse VEGF with a KD of about 100nM or less. In some embodiments, the VEGF/DLL4-binding agent binds both human VEGF and mouse VEGF with a KD of about SOnM or less. In some ments, a LL4—binding agent binds both human DLL4 and mouse DLL4 with a KB of about 100nM or less. In some embodiments, a VEGF/DLL4-binding agent binds both human DLL4 and mouse DLL4 with a KD of about SOnM or less. In some embodiments, the iation nt of the binding agent (e.g., an antibody) to VEGF is the iation constant determined using a VEGF fusion protein comprising at least a n ofVEGF lized on a Biacore chip. In some embodiments, the dissociation constant of the binding agent (e. g., an antibody) to DLL4 is the dissociation constant determined using a DLL4—fusion protein comprising at least a portion of DLL4 immobilized on a Biacore chip.
In some embodiments, the VEGF/DLL4—binding agent is a bispecific antibody which comprises a first antigen-binding site that specifically binds VEGF and a second antigen-binding site that specifically binds DLL4. In some embodiments, a VEGF/DLL4—binding agent or antibody binds both VEGF and DLL4 with a KD of about 100nM or less. In some embodiments, a VEGF/DLL4-binding agent or antibody binds both VEGF and DLL4 with a KD of about SOnM or less. In some embodiments, a VEGF/DLL4-binding agent or antibody binds both VEGF and DLL4 with a KD of about 20nM or less. In some embodiments, a VEGF/DLL4-binding agent or antibody binds both VEGF and DLL4 with a KD of about 10nM or less. In some embodiments, a VEGF/DLL4-binding agent or antibody binds both VEGF and DLL4 with a KD of about lnM or less. In some embodiments, the affinity of one of the antigen- binding sites may be weaker than the affinity of the other antigen-binding site. For example, the KD of one antigen binding site may be about lnM and the KD of the second antigen-binding site may be about 10nM. In some embodiments, the difference in affinity between the two antigen-binding sites may be about 2-fold or more, about 3-fold or more, about 5—fold or more, about 8-fold or more, about 10-fold or more, about d or more, about 20-fold or more, about 30—fold or more, about 50-fold or more, or about 100-fold or more. Modulation of the ies of the two n—binding sites may affect the biological activity of the bispecific antibody. For example, decreasing the affinity of the antigen-binding site for DLL4 or VEGF, may have a desirable effect, for example decreased toxicity of the binding agent or increased therapeutic index.
By way of non-limiting example, the bispecific antibody may comprise (a) a first antigen-binding site that binds human VEGF with a KD between about 0.1 nM and about 1.0 nM, and (b) a second antigen- binding site that specifically binds human DLL4 with a KD n about 0.1 nM and about 20 nM, between about 0.5nM and about 20nM, between about 1.0 nM and 10nM. In n embodiments, the bispecific antibody comprises two identical light chains.
In certain embodiments, the VEGF/DLL4—binding agent (e.g., an dy) binds VEGF and/or DLL4 with a half maximal effective concentration (EC50) of about lnM or less, about lOOnM or less, about 40nM or less, about 20nM or less, about lOnM or less, about lnM or less, or about 0.1nM or less.
In certain embodiments, a VEGF/DLL4—binding agent (e.g., an antibody) binds VEGF and/or DLL4 with a half maximal effective concentration (EC50) of about 1 uM or less, about lOOnM or less, about 40nM or less, about 20nM or less, about lOnM or less, about lnM or less, or about 0.1nM or less.
In certain embodiments, the VEGFXDLL4—binding agent is an antibody. In some embodiments, the antibody is a recombinant antibody. In some embodiments, the antibody is a monoclonal antibody. In some embodiments, the antibody is a chimeric antibody. In some embodiments, the antibody is a humanized antibody. In some embodiments, the antibody is a human antibody. In certain embodiments, the antibody is an IgA, IgD, IgE, IgG, or IgM antibody. In certain embodiments, the antibody is an IgGl dy. In certain embodiments, the antibody is an IgG2 antibody. In certain embodiments, the antibody is an antibody fragment comprising an n—binding site. In some embodiments, the antibody is a bispeciflc dy. In some embodiments, the antibody is monovalent, eciflc, bivalent, or multispeciflc. In some embodiments, the antibody is conjugated to a cytotoxic moiety. In some embodiments, the antibody is isolated. In some embodiments, the antibody is substantially pure.
The VEGF/DLL4-binding agents (e.g., antibodies) of the present invention can be assayed for specific binding by any method known in the art. The immunoassays which can be used include, but are not limited to, competitive and non-competitive assay systems using techniques such as e analysis, FACS analysis, fluorescence, immunocytochemistry, n blot analysis, radioimmunoassay, ELISA, “sandwich” immunoassay, immunoprecipitation assay, precipitation reaction, gel diffusion precipitin reaction, immunodiffusion assay, agglutination assay, complement-fixation assay, immunoradiometric assay, fluorescent immunoassay, homogeneous time-resolved fluorescence assay (HTRF), and protein A assay. Such assays are routine and well-known in the art (see, e.g., Ausubel et al., Editors, l994-present, Current Protocols in Molecular Biology, John Wiley & Sons, Inc., New York, NY).
For example, the specific binding of an dy to human VEGF and/or human DLL4 may be determined using ELISA. An ELISA assay comprises preparing antigen, coating wells of a 96 well microtiter plate with antigen, adding the antibody or other binding agent conjugated to a detectable compound such as an enzymatic substrate (e.g. adish peroxidase or alkaline phosphatase) to the well, incubating for a period of time, and detecting the presence of the binding agent bound to the antigen.
In some embodiments, the g agent or dy is not ated to a detectable nd, but instead a second antibody that izes the binding agent or antibody (e.g., an anti-Fc antibody) and is conjugated to a detectable compound is added to the well. In some embodiments, instead of coating the well with the antigen, the binding agent or antibody can be coated to the well and a second antibody conjugated to a detectable compound can be added following the addition of the antigen to the coated well. One of skill in the art would be knowledgeable as to the parameters that can be modified to increase the signal detected as well as other variations of ELISAs known in the art.
In another example, the specific binding of an antibody to human VEGF and/or human DLL4 may be determined using FACS. A FACS screening assay may comprise generating a cDNA construct that expresses an antigen as a fusion protein, ecting the construct into cells, expressing the antigen on the e of the cells, mixing the binding agent or antibody with the transfected cells, and incubating for a period of time. The cells bound by the binding agent or antibody may be identified by using a secondary antibody conjugated to a detectable compound (e.g., jugated anti-Fe antibody) and a flow cytometer. One of skill in the art would be knowledgeable as to the parameters that can be modified to ze the signal detected as well as other variations of FACS that may enhance screening (e.g., screening for blocking antibodies).
The binding affinity of an antibody or other binding—agent to an n (e.g., VEGF or DLL4) and the off-rate of an antibody-antigen interaction can be determined by competitive binding assays. One example of a competitive binding assay is a radioimmunoassay comprising the incubation of labeled antigen (e.g., 3H or 125I), or fragment or variant thereof, with the antibody of interest in the presence of increasing amounts of unlabeled n followed by the detection of the antibody bound to the labeled antigen. The affinity of the antibody for the n and the binding off-rates can be determined from the data by Scatchard plot analysis. In some embodiments, Biacore kinetic analysis is used to determine the binding on and off rates of antibodies or agents that bind an antigen (e.g., VEGF or DLL4). Biacore kinetic analysis comprises analyzing the binding and iation of dies from chips with immobilized antigen (e.g., VEGF or DLL4) on their surface.
In certain embodiments, the ion provides a VEGF-binding agent (e. g., an antibody) that specifically binds human VEGF, wherein the VEGF-binding agent (e.g., an dy) comprises one, two, three, four, five, and/or six of the CDRs of dy 219R45 (see Table 1). In some embodiments, the VEGF-binding agent comprises one or more of the CDRs of 219R45, two or more of the CDRs of 219R45, three or more of the CDRs of 2 l9R45, four or more of the CDRs of 219R45, five or more of the CDRs of 219R45, or all six of the CDRs of 219R45. In some ments, the VEGF-binding agent binds human VEGF and mouse VEGF.
Table 1 NYWMH HC CDRI (SEQ ID NO:17) DINPSNGRTSYKEKFKR HC CDRZ (SEQ ID NO: 18) HYDDKYYPLMDY HC CDR3 (SEQ ID NO: 19) RASESVDNYGISFMK LC CDR] (SEQ ID NO:20) AASN GSQ LC CDR2 (SEQ ID NO:21) QQSKEVPWTFGG LC CDR3 (SEQ ID NO:22) In certain embodiments, the invention provides a VEGF-binding agent (e.g., an antibody) that cally binds human VEGF, wherein the VEGF-binding agent comprises a heavy chain CDRI sing NYWMH (SEQ ID , a heavy chain CDR2 comprising DINPSNGRTSYKEKFKR (SEQ ID NO:18), and a heavy chain CDR3 comprising HYDDKYYPLMDY (SEQ ID NO:19). In some embodiments, the VEGF-binding agent further comprises a light chain CDRl comprising DNYGISFMK (SEQ ID NO:20), a light chain CDR2 comprising AASNQGS (SEQ ID NO:21), and a light chain CDR3 comprising QQSKEVPWTFGG (SEQ ID NO:22). In certain embodiments, the VEGF-binding agent comprises: (a) a heavy chain CDRl comprising NYWMH (SEQ ID NO: 17), a heavy chain CDR2 sing DINPSNGRTSYKEKFKR (SEQ ID NO: 18), and a heavy chain CDR3 comprising HYDDKYYPLMDY (SEQ ID NO:19), and (b) a light chain CDRl comprising RASESVDNYGISFMK (SEQ ID NO:20), a light chain CDR2 comprising AASNQGS (SEQ ID NO:21), and a light chain CDR3 comprising QQSKEVPWTFGG (SEQ ID NO:22).
In certain embodiments, the invention provides a VEGF-binding agent (e.g., an antibody) that specifically binds human VEGF, wherein the VEGF—binding agent comprises: (a) a heavy chain CDRl comprising NYWMH (SEQ ID NO:17), or a variant thereof comprising 1, 2, 3, or 4 amino acid substitutions; (b) a heavy chain CDR2 comprising DINPSNGRTSYKEKFKR (SEQ ID NO: 18), or a t f comprising 1, 2, 3, or 4 amino acid substitutions; (c) a heavy chain CDR3 sing HYDDKYYPLMDY (SEQ ID NO:19), or a variant thereof comprising 1, 2, 3, or 4 amino acid substitutions; (d) a light chain CDRl sing RASESVDNYGISFMK (SEQ ID NO:20), or a t thereof comprising 1, 2, 3, or 4 amino acid substitutions; (e) a light chain CDR2 comprising AASNQGS (SEQ ID NO:21), or a variant thereof comprising 1, 2, 3, or 4 amino acid substitutions; and (l) a light chain CDR3 comprising QQSKEVPWTFGG (SEQ ID NO:22), or a variant thereof comprising 1, 2, 3, or 4 amino acid substitutions. In certain embodiments, the amino acid substitutions are conservative substitutions.
In certain ments, the invention provides a inding agent (e. g., an antibody) that specifically binds VEGF, wherein the VEGF—binding agent comprises a heavy chain variable region having at least about 80% sequence identity to SEQ ID NO:11, and a light chain variable region having at least 80% sequence identity to SEQ ID NO:12. In certain embodiments, the VEGF-binding agent comprises a heavy chain variable region having at least about 85%, at least about 90%, at least about 95%, at least about 97%, or at least about 99% sequence identity to SEQ ID NO:11. In certain embodiments, the VEGF-binding agent comprises a light chain le region having at least about 85%, at least about 90%, at least about 95%, at least about 97%, or at least about 99% sequence ty to SEQ ID NO: 12.
In certain embodiments, the VEGF-binding agent comprises a heavy chain variable region having at least about 95% sequence identity to SEQ ID NO:1 l, and a light chain variable region having at least about 95% sequence identity to SEQ ID NO:12. In certain embodiments, the VEGF—binding agent comprises a heavy chain variable region comprising SEQ ID N02] 1, and a light chain variable region comprising SEQ ID NO:12. In certain ments, the inding agent comprises a heavy chain le region consisting essentially of SEQ ID NO:11, and a light chain variable region consisting essentially of SEQ ID NO:12. In some embodiments, the VEGF—binding agent ses a heavy chain comprising SEQ ID NO:49, and a light chain sing SEQ ID NO:8. In some embodiments, the VEGF-binding antibody or other agent comprises a heavy chain comprising SEQ IDNO:7, and a light chain comprising SEQ ID NO:8.
In some embodiments, the VEGF-binding agent binds VEGF with a KD of about 10nM or less. In some embodiments, the VEGF-binding agent binds VEGF with a KD of about 1nM or less. In some embodiments, the VEGF-binding agent binds VEGF with a KD of about 0.1nM or less. In some ments, the VEGF-binding agent binds VEGF with a KD of about 0.01nM or less. In some embodiments, at least one amino acid residue in at least one CDR of the VEGF-binding agent is substituted with a different amino acid so that the affinity of the VEGF-binding agent for VEGF is altered.
In some embodiments, the affinity of the VEGF—binding agent is increased. In some embodiments, the affinity of the VEGF-binding agent is decreased. In some embodiments, the inding agent binds human VEGF. In some embodiments, the VEGF—binding agent binds human VEGF and mouse VEGF.
In certain embodiments, the VEGF—binding agent comprises the heavy chain variable region and light chain variable region of the 219R45 dy. In certain embodiments, the VEGF-binding agent comprises the heavy chain and light chain of the 219R45 antibody (with or without the leader sequence).
In certain embodiments, a VEGF-binding agent is the 219R45 antibody.
In certain embodiments, a VEGF—binding agent comprises, consists essentially of, or consists of, the antibody 219R45.
In certain embodiments, a VEGF-binding agent (e.g., an antibody) binds the same epitope, or essentially the same epitope, on VEGF as an antibody of the invention. In another embodiment, a VEGF- binding agent is an antibody that binds an epitope on VEGF that overlaps with the e on VEGF bound by an antibody of the invention. In certain embodiments, a VEGF-binding agent (e.g., an antibody) binds the same e, or essentially the same epitope, on VEGF as antibody 219R45. In another embodiment, the VEGF-binding agent is an antibody that binds an epitope on VEGF that overlaps with the epitope on VEGF bound by antibody 219R45.
In some embodiments, the VEGF—binding agent inhibits binding ofVEGF to at least one VEGF receptor. In certain embodiments, the inding agent inhibits binding of human VEGF to VEGFR- l or VEGFR-Z. In some embodiments, the inding agent specifically binds VEGF and modulates angiogenesis. In some ments, the VEGF-binding agent specifically binds VEGF and inhibits angiogenesis. In some embodiments, the VEGF—binding agent specifically binds VEGF and inhibits tumor growth.
In certain embodiments, the invention provides a DLL4-binding agent (e.g., an antibody) that specifically binds human DLL4, wherein the DLL4-binding agent (e. g., an antibody) comprises one, two, three, four, five, and/or Six of the CDRs of antibody 21R79 (see Table 2). In some embodiments, the DLL4-binding agent ses one or more of the CDRs of 21R79, two or more of the CDRs of 21R79, three or more of the CDRs of 21R79, four or more of the CDRs of 21R79, five or more of the CDRs of 21R79, or all six of the CDRs of 21R79. In certain embodiments, the ion provides a DLL4-binding agent (e.g., an antibody) that specifically binds human DLL4, wherein the DLL4-binding agent (e.g., an antibody) comprises one, two, three, four, five, and/or six of the CDRs of antibody 21R75 (see Table 2).
In some embodiments, the DLL4-binding agent comprises one or more of the CDRs of 21R75, two or more ofthe CDRs of21R75, three or more ofthe CDRs of21R75, four or more ofthe CDRs of21R75, five or more of the CDRs of 21R75, or all six of the CDRs of 21R75. In certain embodiments, the invention provides a DLL4-binding agent (e.g., an antibody) that specifically binds human DLL4, wherein the DLL4-binding agent (e.g., an dy) comprises one, two, three, four, five, and/or six of the CDRs of antibody 21R83 (see Table 2). In some embodiments, the DLL4-binding agent ses one or more ofthe CDRs of21R83, two or more ofthe CDRs of21R83, three or more ofthe CDRs of21R83, four or more ofthe CDRs 3, five or more ofthe CDRs of21R83, or all six ofthe CDRs of21R83. In some embodiments, the DLL4-binding agent binds human DLL4 and mouse DLL4.
Table 2 _ 21R79 21R75 21R83 TAYYIH TAYYIH TAYYIH HC CDRl (SEQ ID NO: 13) (SEQ ID NO: l3) (SEQ ID NO: 13) YIANYNRATNYNQKFKG YIAGYKDATNYNQKFKG YISNYNRATNYNQKFKG HC CDR2 (SEQ ID NO: 14) (SEQ ID NO:59) (SEQ ID NO:65) VGMDY RDYDYDVGMDY RDYDYDVGMDY HC CDR3 (SEQ ID NO: 16) (SEQ ID NO: 16) (SEQ ID NO: 16) RASESVDNYGISFMK RASESVDNYGISFMK RASESVDNYGISFMK LC CDRI (SEQ ID NO:20) (SEQ ID NO:20) (SEQ ID NO:20) S AASNQGS AASNQGS LC CDR2 (SEQ ID NO:21) (SEQ ID NO:21) (SEQ ID NO:21) (SEQ ID NO:22) (SEQ ID NO:22) (SEQ ID NO:22) In certain ments, the heavy chain CDRl of the DLL4—binding dy is a minimal HC CDRl comprising AYYIH (SEQ ID .
In some embodiments, the binding agent is an antibody that binds human DLL4 and comprises a heavy chain CDRl comprising TAYYIH (SEQ ID NO:13) or AYYIH (SEQ ID NO:79), a heavy chain CDR2 comprising YX3X4ATNYNQKFKG (SEQ ID NO:80), wherein X1 is serine or alanine, X2 is serine, asparagine, or glycine, X3 is asparagine or lysine, and X4 is glysine, arginine,or aspartic acid and a heavy chain CDR3 comprising RDYDYDVGMDY (SEQ ID NO:16); and a light chain CDRI comprising RASESVDNYGISFMK (SEQ ID NO:20), a light chain CDR2 comprising AASNQGS (SEQ ID N021), and a light chain CDR3 comprising QQSKEVPWTFGG (SEQ ID NO:22).
In certain embodiments, the invention es a DLL4-binding agent (e.g., an antibody) that specifically binds human DLL4, n the DLL4—binding agent comprises a heavy chain CDRl comprising TAYYIH (SEQ ID NO:13), a heavy chain CDR2 comprising YIANYNRATNYNQKFKG (SEQ ID NO:14), and a heavy chain CDR3 comprising RDYDYDVGMDY (SEQ ID NO:16). In some embodiments, the DLL4-binding agent further comprises a light chain CDRl comprising DNYGISFMK (SEQ ID NO:20), a light chain CDR2 comprising AASNQGS (SEQ ID N021), and a light chain CDR3 comprising QQSKEVPWTFGG (SEQ ID NO:22). In n embodiments, the DLL4-binding agent comprises: (a) a heavy chain CDRl comprising TAYYIH (SEQ ID NO:13), a heavy chain CDR2 comprising YIANYNRATNYNQKFKG (SEQ ID NO:14), and a heavy chain CDR3 comprising RDYDYDVGMDY (SEQ ID NO:16), and (b) a light chain CDRl sing RASESVDNYGISFMK (SEQ ID NO:20), a light chain CDR2 comprising AASNQGS (SEQ ID N021), and a light chain CDR3 comprising QQSKEVPWTFGG (SEQ ID NO:22).
In n embodiments, the ion provides a DLL4—binding agent (e.g., an antibody) that specifically binds human DLL4, wherein the DLL4—binding agent comprises: (a) a heavy chain CDRl comprising TAYYIH (SEQ ID NO: 13), or a variant thereof comprising 1, 2, 3, or 4 amino acid substitutions; (b) a heavy chain CDR2 sing YIANYNRATNYNQKFKG (SEQ ID NO:14), or a variant thereof comprising 1, 2, 3, or 4 amino acid substitutions; (c) a heavy chain CDR3 comprising RDYDYDVGMDY (SEQ ID NO: 16), or a variant thereof comprising 1, 2, 3, or 4 amino acid substitutions; (d) a light chain CDRl comprising RASESVDNYGISFMK (SEQ ID NO:20), or a variant thereof comprising 1, 2, 3, or 4 amino acid substitutions; (e) a light chain CDR2 comprising AASNQGS (SEQ ID NO:21), or a variant thereof comprising 1, 2, 3, or 4 amino acid substitutions; and (f) a light chain CDR3 comprising QQSKEVPWTFGG (SEQ ID NO:22), or a variant thereof comprising 1, 2, 3, or 4 amino acid tutions. In n embodiments, the amino acid substitutions are conservative substitutions.
In certain embodiments, the invention provides a DLL4-binding agent (e.g., an antibody) that specifically binds DLL4, wherein the inding agent comprises a heavy chain variable region having at least about 80% sequence identity to SEQ ID NO:10, and a light chain variable region having at least 80% sequence identity to SEQ ID NO:12. In certain embodiments, the DLL4-binding agent comprises a heavy chain variable region having at least about 85%, at least about 90%, at least about 95%, at least about 97%, or at least about 99% sequence identity to SEQ ID NO: 10. In certain embodiments, the DLL4-binding agent comprises a light chain variable region having at least about 85%, at least about 90%, at least about 95%, at least about 97%, or at least about 99% ce identity to SEQ ID NO: 12.
In certain embodiments, the DLL4—binding agent comprises a heavy chain variable region having at least about 95% sequence identity to SEQ ID NO:10, and a light chain variable region having at least about 95% sequence ty to SEQ ID NO:12. In certain embodiments, the DLL4-binding agent comprises a heavy chain variable region comprising SEQ ID NO:10, and a light chain le region comprising SEQ ID NO:12. In certain embodiments, the DLL4—binding agent comprises a heavy chain variable region consisting essentially of SEQ ID NO: 10, and a light chain le region consisting essentially of SEQ ID NO:12. In some embodiments, the DLL4-binding agent comprises a heavy chain comprising SEQ ID NO:48, and a light chain comprising SEQ ID NO:8. In some embodiments, the DLL4-binding dy or other agent comprises a heavy chain comprising SEQ ID N026, and a light chain sing SEQ ID NO:8. In some embodiments, the antibody is a bispecific dy.
In n embodiments, the invention provides a DLL4-binding agent (e.g., an antibody) that specifically binds human DLL4, wherein the DLL4—binding agent comprises a heavy chain CDRl comprising TAYYIH (SEQ ID NO:13), a heavy chain CDR2 comprising YIAGYKDATNYNQKFKG (SEQ ID NO:59), and a heavy chain CDR3 comprising RDYDYDVGMDY (SEQ ID NO: 16). In some embodiments, the DLL4-binding agent further comprises a light chain CDRl comprising RASESVDNYGISFMK (SEQ ID NO:20), a light chain CDR2 sing AASNQGS (SEQ ID NO:21), and a light chain CDR3 comprising QQSKEVPWTFGG (SEQ ID NO:22). In certain ments, the DLL4-binding agent comprises: (a) a heavy chain CDRl comprising TAYYIH (SEQ ID NO: 13), a heavy chain CDR2 comprising YIAGYKDATNYNQKFKG (SEQ ID NO:59), and a heavy chain CDR3 comprising RDYDYDVGMDY (SEQ ID NO:16), and (b) a light chain CDRl comprising RASESVDNYGISFMK (SEQ ID NO:20), a light chain CDR2 comprising AASNQGS (SEQ ID NO:21), and a light chain CDR3 comprising PWTFGG (SEQ ID NO:22).
In certain embodiments, the invention provides a DLL4—binding agent (e.g., an antibody) that specifically binds human DLL4, wherein the DLL4—binding agent comprises: (a) a heavy chain CDRl comprising TAYYIH (SEQ ID NO:13), or a variant thereof comprising 1, 2, 3, or 4 amino acid substitutions; (b) a heavy chain CDR2 comprising YIAGYKDATNYNQKFKG (SEQ ID NO:59), or a variant thereof comprising 1, 2, 3, or 4 amino acid substitutions; (c) a heavy chain CDR3 comprising RDYDYDVGMDY (SEQ ID NO: 16), or a variant thereof comprising 1, 2, 3, or 4 amino acid substitutions; (d) a light chain CDRl comprising RASESVDNYGISFMK (SEQ ID , or a variant thereof comprising 1, 2, 3, or 4 amino acid substitutions; (e) a light chain CDR2 comprising AASNQGS (SEQ ID NO:21), or a variant thereof comprising 1, 2, 3, or 4 amino acid tutions; and (f) a light chain CDR3 comprising QQSKEVPWTFGG (SEQ ID , or a t thereof comprising 1, 2, 3, or 4 amino acid substitutions. In certain embodiments, the amino acid substitutions are conservative substitutions.
In certain embodiments, the invention provides a DLL4-binding agent (e.g., an antibody) that specifically binds DLL4, wherein the DLL4—binding agent comprises a heavy chain variable region having at least about 80% sequence identity to SEQ ID NO:58, and a light chain variable region having at least 80% sequence identity to SEQ ID NO:12. In certain embodiments, the DLL4-binding agent comprises a heavy chain le region having at least about 85%, at least about 90%, at least about 95%, at least about 97%, or at least about 99% sequence identity to SEQ ID NO:58. In certain embodiments, the DLL4-binding agent comprises a light chain variable region having at least about 85%, at least about 90%, at least about 95%, at least about 97%, or at least about 99% sequence identity to SEQ ID NO: 12.
In n embodiments, the DLL4-binding agent comprises a heavy chain variable region having at least about 95% ce identity to SEQ ID NO:58, and a light chain variable region having at least about 95% sequence identity to SEQ ID NO:12. In certain embodiments, the DLL4-binding agent comprises a heavy chain variable region comprising SEQ ID NO:58, and a light chain variable region comprising SEQ ID NO:12. In n embodiments, the inding agent comprises a heavy chain variable region consisting essentially of SEQ ID N025 8, and a light chain variable region consisting essentially of SEQ ID NO:12. In some embodiments, the DLL4—binding agent comprises a heavy chain comprising SEQ ID NO:56, and a light chain comprising SEQ ID N08 In n embodiments, the invention provides a DLL4—binding agent (e.g., an antibody) that specifically binds human DLL4, wherein the DLL4—binding agent comprises a heavy chain CDRl comprising TAYYIH (SEQ ID NO: 13), a heavy chain CDR2 comprising YISNYNRATNYNQKFKG (SEQ ID NO:65), and a heavy chain CDR3 comprising VGMDY (SEQ ID NO: 16). In some embodiments, the DLL4-binding agent r comprises a light chain CDRl comprising RASESVDNYGISFMK (SEQ ID NO:20), a light chain CDR2 comprising AASNQGS (SEQ ID NO:21), and a light chain CDR3 comprising QQSKEVPWTFGG (SEQ ID NO:22). In certain embodiments, the DLL4-binding agent comprises: (a) a heavy chain CDRl comprising TAYYIH (SEQ ID NO: 13), a heavy chain CDR2 comprising YISNYNRATNYNQKFKG (SEQ ID NO:65), and a heavy chain CDR3 comprising RDYDYDVGMDY (SEQ ID NO:16), and (b) a light chain CDRl comprising DNYGISFMK (SEQ ID NO:20), a light chain CDR2 comprising AASNQGS (SEQ ID N021), and a light chain CDR3 comprising QQSKEVPWTFGG (SEQ ID .
In certain embodiments, the invention provides a DLL4-binding agent (e.g., an antibody) that specifically binds human DLL4, wherein the DLL4—binding agent comprises: (a) a heavy chain CDRl comprising TAYYIH (SEQ ID NO: 13), or a variant thereof comprising 1, 2, 3, or 4 amino acid substitutions; (b) a heavy chain CDR2 comprising YISNYNRATNYNQKFKG (SEQ ID NO:65), or a variant thereof comprising 1, 2, 3, or 4 amino acid substitutions; (c) a heavy chain CDR3 comprising RDYDYDVGMDY (SEQ ID NO: 16), or a variant thereof comprising 1, 2, 3, or 4 amino acid substitutions; (d) a light chain CDRl comprising RASESVDNYGISFMK (SEQ ID NO:20), or a variant f comprising 1, 2, 3, or 4 amino acid substitutions; (e) a light chain CDR2 comprising AASNQGS (SEQ ID NO:21), or a variant f comprising 1, 2, 3, or 4 amino acid substitutions; and (f) a light chain CDR3 comprising PWTFGG (SEQ ID , or a variant thereof sing 1, 2, 3, or 4 amino acid substitutions. In certain embodiments, the amino acid substitutions are vative substitutions.
In certain embodiments, the invention provides a DLL4-binding agent (e.g., an antibody) that specifically binds DLL4, wherein the DLL4-binding agent comprises a heavy chain variable region having at least about 80% sequence identity to SEQ ID NO:64, and a light chain variable region having at least 80% sequence identity to SEQ ID NO:12. In n embodiments, the DLL4-binding agent comprises a heavy chain variable region having at least about 85%, at least about 90%, at least about 95%, at least about 97%, or at least about 99% sequence identity to SEQ ID NO:64. In certain embodiments, the DLL4-binding agent ses a light chain variable region having at least about 85%, at least about 90%, at least about 95%, at least about 97%, or at least about 99% sequence identity to SEQ ID NO:12.
In certain ments, the DLL4-binding agent comprises a heavy chain variable region having at least about 95% sequence identity to SEQ ID NO:64, and a light chain variable region having at least about 95% sequence identity to SEQ ID NO: 12. In certain embodiments, the DLL4-binding agent comprises a heavy chain variable region comprising SEQ ID NO:64, and a light chain variable region comprising SEQ ID NO: 12. In certain ments, the DLL4—binding agent comprises a heavy chain variable region consisting essentially of SEQ ID NO:64, and a light chain variable region consisting essentially of SEQ ID NO:12. In some embodiments, the DLL4-binding agent comprises a heavy chain comprising SEQ ID NO:62, and a light chain comprising SEQ ID NO:8.In some embodiments, the agent is a bispecific antibody.
In some embodiments, the DLL4—binding agent is an antibody that comprises a heavy chain comprising SEQ ID N025, and a light chain comprising SEQ ID NO:8. In some embodiments, the antibody is a bispecific antibody.
In some embodiments, the inding agent binds DLL4 with a KD of 25nM or less. In some embodiments, the DLL4-binding agent binds DLL4 with a KD of 10nM or less. In some embodiments, the DLL4-binding agent binds DLL4 with a KD of about lnM or less. In some embodiments, the DLL4- binding agent binds DLL4 with a KB of about 0.1nM or less. In some embodiments, the DLL4-binding agent binds DLL4 with a KD of about 0.01nM or less. In some embodiments, at least one amino acid residue in at least one CDR of the DLL4—binding agent is substituted with a different amino acid so that the affinity of the DLL4-binding agent for DLL4 is altered. In some embodiments, the affinity of the DLL4-binding agent is increased. In some embodiments, the affinity of the DLL4-binding agent is decreased.
In certain ments, the DLL4-binding agent comprises the heavy chain variable region and the light chain variable region of the 21R79 antibody. In certain embodiments, the inding agent comprises the heavy chain and light chain of the 21R79 antibody (with or without the leader sequence).
In certain embodiments, the inding agent is the 21R79 antibody.
In certain embodiments, a DLL4—binding agent comprises, consists essentially of, or consists of, the antibody 21R79.
In certain embodiments, the DLL4-binding agent comprises the heavy chain variable region and the light chain variable region of the 21R75 antibody. In certain embodiments, the DLL4-binding agent comprises the heavy chain and light chain of the 21R75 antibody (with or without the leader sequence).
In certain embodiments, the DLL4-binding agent is the 21R75 antibody.
In certain embodiments, a DLL4-binding agent comprises, consists essentially of, or ts of, the antibody 21R75.
In certain embodiments, the DLL4-binding agent comprises the heavy chain variable region and the light chain variable region of the 21R83 antibody. In certain ments, the DLL4-binding agent comprises the heavy chain and light chain of the 21R83 antibody (with or without the leader sequence).
In certain embodiments, the DLL4-binding agent is the 21R83 antibody.
In certain ments, a DLL4-binding agent comprises, ts essentially of, or consists of, the antibody 21R83.
In some embodiments, a DLL4—binding agent binds an N—terminal fragment of human DLL4 (amino acids 1-191 of SEQ ID NO:24). In some embodiments, the DLL4-binding agent binds an e comprising amino acids 40-47 of SEQ ID NO:25. In some embodiments, the inding agent binds an e comprising amino acids 113-120 of SEQ ID NO:25. In some embodiments, the DLL4-binding agent binds an epitope comprising amino acids 40—47 of SEQ ID NO:25 and amino acids 0 of SEQ ID NO:25.
In certain embodiments, a DLL4—binding agent (e.g., an antibody) binds the same epitope, or essentially the same epitope, on DLL4 as an antibody of the invention. In another embodiment, a DLL4- binding agent is an antibody that binds an epitope on DLL4 that overlaps with the epitope on DLL4 bound by an antibody of the invention. In certain embodiments, a DLL4-binding agent (e.g., an antibody) binds the same epitope, or essentially the same epitope, on DLL4 as antibody 21R79. In another embodiment, the DLL4-binding agent is an antibody that binds an e on DLL4 that overlaps with the epitope on DLL4 bound by dy 21R79. In certain ments, a DLL4—binding agent (e.g., an antibody) binds the same epitope, or essentially the same epitope, on DLL4 as antibody 21R75. In another embodiment, the DLL4-binding agent is an antibody that binds an epitope on DLL4 that overlaps with the epitope on DLL4 bound by antibody 21R75. In n embodiments, a DLL4-binding agent (e. g., an antibody) binds the same epitope, or essentially the same epitope, on DLL4 as antibody 21R83. In another embodiment, the DLL4-binding agent is an antibody that binds an epitope on DLL4 that overlaps with the epitope on DLL4 bound by antibody 21R83.
In some embodiments, the inding agent inhibits g of DLL4 to at least one Notch receptor. In certain embodiments, the Notch receptor is Notch] or Notch4. In some , Notch2, Notch3, embodiments, the DLL4-binding agent specifically binds DLL4 and inhibits DLL4 activity. In some embodiments, the inding agent specifically binds DLL4 and ts Notch signaling. In some embodiments, the DLL4-binding agent specifically binds DLL4 and modulates angiogenesis. In some embodiments, the DLL4-binding agent specifically binds DLL4 and ts tumor growth. In some embodiments, the DLL4-binding agent specifically binds DLL4 and inhibits tumorigenicity. In some embodiments, the DLL4-binding agent specifically binds DLL4 and reduces the number or frequency of CSCs in a tumor.
In certain embodiments, the invention es a VEGF/DLL4-binding agent that is a ific antibody. In some embodiments, the VEGF/DLL4 g agent is a bispecific antibody sing a first antigen-binding site that specifically binds human VEGF. In some embodiments, the VEGF/DLL4 binding agent is a bispecific antibody comprising a first antigen-binding site that specifically binds human VEGF and a second antigen-binding site that binds a tumor—associated target. In some embodiments, the LL4-binding agent is a ific antibody comprising: a first antigen-binding site that specifically binds human VEGF, wherein the first antigen—binding site comprises a heavy chain CDRl comprising NYWMH (SEQ ID NO:17), a heavy chain CDR2 comprising DINPSNGRTSYKEKFKR (SEQ ID NO:18), and a heavy chain CDR3 comprising HYDDKYYPLMDY (SEQ ID NO: 19). In some embodiments, the bispecific antibody further comprises: a light chain CDR] comprising RASESVDNYGISFMK (SEQ ID NO:20), a light chain CDR2 comprising AASNQGS (SEQ ID NO:21), and a light chain CDR3 comprising QQSKEVPWTFGG (SEQ ID NO:22). In some embodiments, the VEGF/DLL4-binding agent is a bispecific antibody comprising: a first antigen-binding site that specifically binds human VEGF, wherein the first antigen—binding site comprises (a) a heavy chain CDRl comprising NYWMH (SEQ ID NO: 17), a heavy chain CDR2 comprising DINPSNGRTSYKEKFKR (SEQ ID NO: 18), and a heavy chain CDR3 comprising HYDDKYYPLMDY (SEQ ID NO: 19), and (b) a light chain CDRl comprising RASESVDNYGISFMK (SEQ ID NO:20), a light chain CDR2 comprising AASNQGS (SEQ ID NO:21), and a light chain CDR3 comprising PWTFGG (SEQ ID NO:22).
In some embodiments, the VEGFKDLL4 g agent is a bispecific antibody comprising a first heavy chain variable region having at least about 80% sequence identity to SEQ ID NO:1 1. In some embodiments, the bispecific antibody r comprises a light chain variable region having at least 80% sequence identity to SEQ ID NO: 12. In n embodiments, the bispecific VEGF/DLL4-binding agent comprises a first heavy chain variable region having at least about 85%, at least about 90%, at least about 95%, at least about 97%, or at least about 99% sequence identity to SEQ ID NO:1 1, and a light chain variable region having at least about 85%, at least about 90%, at least about 95%, at least about 97%, or at least about 99% sequence identity to SEQ ID NO:12.
In certain embodiments, the invention es a VEGF/DLL4-binding agent that is a bispecific antibody. In some embodiments, the VEGF/DLL4 binding agent is a bispecific antibody comprising a first antigen-binding site that specifically binds human DLL4. In some embodiments, the VEGF/DLL4 binding agent is a bispecific antibody comprising a first antigen-binding site that specifically binds human DLL4 and a second antigen-binding site that binds a tumor—associated . In some ments, the VEGF/DLL4-binding agent is a bispecific antibody sing: a first antigen-binding site that specifically binds human DLL4, wherein the first antigen—binding site comprises a heavy chain CDRl comprising TAYYIH (SEQ ID NO: 13) or AYYIH (SEQ ID NO:79), a heavy chain CDR2 comprising YIXleYX3X4ATNYNQKFKG (SEQ ID NO:80), wherein X1 is serine or alanine, X2 is serine, asparagine, or glycine, X3 is asparagine or lysine, and X4 is glysine, arginine,or aspartic acid and a heavy chain CDR3 comprising RDYDYDVGMDY (SEQ ID NO:16); and a light chain CDRl comprising RASESVDNYGISFMK (SEQ ID NO:20), a light chain CDR2 comprising AASNQGS (SEQ ID N021), and a light chain CDR3 comprising QQSKEVPWTFGG (SEQ ID NO:22). In some embodiments, the LL4-binding agent is a bispecific antibody comprising: a first antigen-binding site that specifically binds human DLL4, wherein the first antigen—binding site comprises a heavy chain CDRl comprising TAYYIH (SEQ ID NO:13), a heavy chain CDR2 comprising YIANYNRATNYNQKFKG (SEQ ID , YISSYNGATNYNQKFKG (SEQ ID NO:15), YIAGYKDATNYNQKFKG (SEQ ID NO:59), or YISNYNRATNYNQKFKG (SEQ ID NO:65), and a heavy chain CDR3 comprising RDYDYDVGMDY (SEQ ID NO:16). In some embodiments, the bispecific dy comprises a first n-binding site comprising a heavy chain CDRl comprising TAYYIH (SEQ ID NO: 13), a heavy chain CDR2 comprising YIANYNRATNYNQKFKG (SEQ ID NO: 14), and a heavy chain CDR3 comprising RDYDYDVGMDY (SEQ ID NO:16). In some embodiments, the bispecific antibody comprises a first antigen-binding site comprising a heavy chain CDRl comprising TAYYIH (SEQ ID NO:13), a heavy chain CDR2 comprising YISSYNGATNYNQKFKG (SEQ ID NO: 15), and a heavy chain CDR3 comprising RDYDYDVGMDY (SEQ ID NO:16). In some embodiments, the bispecific antibody ses a first antigen-binding site comprising a heavy chain CDRl comprising TAYYIH (SEQ ID NO:13), a heavy chain CDR2 comprising YIAGYKDATNYNQKFKG (SEQ ID NO:59), and a heavy chain CDR3 comprising VGMDY (SEQ ID NO:16). In some embodiments, the ific antibody comprises a first antigen—binding site comprising a heavy chain CDRl comprising TAYYIH (SEQ ID NO:13), a heavy chain CDR2 sing YISNYNRATNYNQKFKG (SEQ ID NO:65), and a heavy chain CDR3 comprising RDYDYDVGMDY (SEQ ID NO: 16). In some embodiments, the bispecific antibody further comprises: a light chain CDRl comprising RASESVDNYGISFMK (SEQ ID NO:20), a light chain CDR2 comprising S (SEQ ID NO:21), and a light chain CDR3 comprising QQSKEVPWTFGG (SEQ ID . In some embodiments, the VEGF/DLL4-binding agent is a bispecific antibody comprising: a first antigen-binding site that specifically binds human DLL4, wherein the first antigen—binding site comprises (a) a heavy chain CDRl comprising TAYYIH (SEQ ID NO:13), a heavy chain CDR2 comprising YIANYNRATNYNQKFKG (SEQ ID NO: 14), YISSYNGATNYNQKFKG (SEQ ID NO: 15), YIAGYKDATNYNQKFKG (SEQ ID NO:59), or YISNYNRATNYNQKFKG (SEQ ID N0265), and a heavy chain CDR3 comprising VGMDY (SEQ ID NO:16), and (b) a light chain CDRl comprising RASESVDNYGISFMK (SEQ ID NO:20), a light chain CDR2 comprising AASNQGS (SEQ ID N021), and a light chain CDR3 sing QQSKEVPWTFGG (SEQ ID NO:22).
In some embodiments, the VEGF/DLL4 g agent is a bispecific antibody comprising a first heavy chain variable region having at least about 80% ce identity to SEQ ID NO:9, SEQ ID NO:10, SEQ ID NO:58, or SEQ ID NO:64. In some embodiments, the bispecific antibody further comprises a light chain variable region having at least 80% sequence identity to SEQ ID NO: 12. In certain embodiments, the bispecific LL4—binding agent comprises a first heavy chain le region having at least about 85%, at least about 90%, at least about 95%, at least about 97%, or at least about 99% sequence identity to SEQ ID NO:9, SEQ ID NO:10, SEQ ID NO:58, or SEQ ID NO:64; and/or a light chain variable region having at least about 85%, at least about 90%, at least about 95%, at least about 97%, or at least about 99% sequence identity to SEQ ID NO:12.
In certain embodiments, the invention provides a VEGF/DLL4—binding agent (e. g., a bispecific antibody) that specifically binds human VEGF and human DLL4. In some embodiments, the bispecific antibody comprises: a) a first antigen-binding site that specifically binds human VEGF, and b) a second antigen-binding Site that specifically binds human DLL4, wherein the first antigen-binding site comprises a heavy chain CDRl comprising NYWMH (SEQ ID NO:17), a heavy chain CDR2 comprising DINPSNGRTSYKEKFKR (SEQ ID NO:18), and a heavy chain CDR3 comprising HYDDKYYPLMDY (SEQ ID NO:19); wherein the second antigen—binding site ses a heavy chain CDRl comprising TAYYIH (SEQ ID NO:13) or AYYIH (SEQ ID N0279), a heavy chain CDR2 comprising YIXleYX3X4ATNYNQKFKG (SEQ ID NO:80), wherein X1 is serine or alanine, X2 is serine, asparagine, or glycine, X3 is asparagine or lysine, and X4 is glysine, arginine,or aspartic acid and a heavy chain CDR3 comprising RDYDYDVGMDY (SEQ ID NO: 16); and a light chain CDRl comprising RASESVDNYGISFMK (SEQ ID NO:20), a light chain CDR2 sing S (SEQ ID NO:21), and a light chain CDR3 comprising QQSKEVPWTFGG (SEQ ID NO:22). In some embodiments, a bispecific antibody comprises a first antigen—binding site that specifically binds human VEGF, and a second antigen-binding site that cally binds human DLL4, wherein the first antigen-binding site comprises a heavy chain CDRl comprising NYWMH (SEQ ID NO:17), a heavy chain CDR2 comprising DINPSNGRTSYKEKFKR (SEQ ID NO:18), and a heavy chain CDR3 sing HYDDKYYPLMDY (SEQ ID NO: 19), and the second antigen-binding site comprises a heavy chain CDRl comprising TAYYIH (SEQ ID NO: 13), a heavy chain CDR2 comprising YIANYNRATNYNQKFKG (SEQ ID NO: 14), GATNYNQKFKG (SEQ ID NO:15), YIAGYKDATNYNQKFKG (SEQ ID NO:59), or YISNYNRATNYNQKFKG (SEQ ID NO:65), and a heavy chain CDR3 comprising RDYDYDVGMDY (SEQ ID NO:16); and wherein both the first and second antigen—binding sites comprise a light chain CDRl comprising RASESVDNYGISFMK (SEQ ID NO:20), a light chain CDR2 sing AASNQGS (SEQ ID NO:21), and a light chain CDR3 comprising QQSKEVPWTFGG (SEQ ID NO:22).
In some embodiments, the bispecific dy comprises a first antigen-binding site that specifically binds human VEGF, and a second n—binding site that specifically binds human DLL4, wherein the first antigen-binding site comprises a heavy chain CDRl comprising NYWMH (SEQ ID NO: 17), a heavy chain CDR2 comprising DINPSNGRTSYKEKFKR (SEQ ID NO: 18), and a heavy chain CDR3 comprising HYDDKYYPLMDY (SEQ ID NO:19), and the second antigen-binding site comprises a heavy chain CDRl comprising TAYYIH (SEQ ID NO:13), a heavy chain CDR2 comprising YIANYNRATNYNQKFKG (SEQ ID NO:14), and a heavy chain CDR3 sing RDYDYDVGMDY (SEQ ID NO: 16); and wherein both the first and second antigen-binding sites comprise a light chain CDRl comprising RASESVDNYGISFMK (SEQ ID NO:20), a light chain CDR2 sing S (SEQ ID NO:21), and a light chain CDR3 comprising QQSKEVPWTFGG (SEQ ID NO:22). In some embodiments, the bispecific antibody is 219R45—MB—21R?9.
In some embodiments, the bispecific antibody comprises a first antigen—binding site that specifically binds human VEGF, and a second antigen-binding site that specifically binds human DLL4, wherein the first antigen-binding site comprises a heavy chain CDRl sing NYWMH (SEQ ID NO:17), a heavy chain CDR2 comprising DINPSNGRTSYKEKFKR (SEQ ID NO: 18), and a heavy chain CDR3 comprising HYDDKYYPLMDY (SEQ ID NO:19), and the second antigen-binding site ses a heavy chain CDRl comprising TAYYIH (SEQ ID N0213), a heavy chain CDR2 comprising YISSYNGATNYNQKFKG (SEQ ID NO:15), and a heavy chain CDR3 comprising RDYDYDVGMDY (SEQ ID NO: 16); and wherein both the first and second antigen—binding sites comprise a light chain CDRl comprising RASESVDNYGISFMK (SEQ ID NO:20), a light chain CDR2 comprising AASNQGS (SEQ ID N021), and a light chain CDR3 comprising QQSKEVPWTFGG (SEQ ID NO:22). In some ments, the bispecific antibody is 219R45—MB-21M18.
In some embodiments, the bispecific dy comprises a first antigen-binding site that specifically binds human VEGF, and a second antigen—binding site that specifically binds human DLL4, n the first antigen-binding site which comprises a heavy chain CDRl comprising NYWMH (SEQ ID NO:17), a heavy chain CDR2 comprising DINPSNGRTSYKEKFKR (SEQ ID NO: 18), and a heavy chain CDR3 comprising HYDDKYYPLMDY (SEQ ID N0219), and the second antigen-binding site comprises a heavy chain CDRl comprising TAYYIH (SEQ ID NO: 1 3), a heavy chain CDR2 comprising YIAGYKDATNYNQKFKG (SEQ ID NO:59), and a heavy chain CDR3 comprising RDYDYDVGMDY (SEQ ID NO:16); and wherein both the first and second antigen—binding sites comprise a light chain CDRl comprising RASESVDNYGISFMK (SEQ ID N0220), a light chain CDR2 comprising AASNQGS (SEQ ID N021), and a light chain CDR3 comprising QQSKEVPWTFGG (SEQ ID . In some embodiments, the bispecific antibody is —MB—21R75.
In some embodiments, the bispecific antibody comprises a first antigen-binding site that specifically binds human VEGF, and a second antigen—binding site that specifically binds human DLL4, n the first antigen-binding site comprises a heavy chain CDRl comprising NYWMH (SEQ ID NO: 17), a heavy chain CDR2 comprising DINPSNGRTSYKEKFKR (SEQ ID NO: 1 8), and a heavy chain CDR3 comprising HYDDKYYPLMDY (SEQ ID NO: 19), and the second n-binding site comprises a heavy chain CDRl sing TAYYIH (SEQ ID NO: 13), a heavy chain CDR2 comprising YISNYNRATNYNQKFKG (SEQ ID NO:65), and a heavy chain CDR3 sing RDYDYDVGMDY (SEQ ID NO: 16); and wherein both the first and second antigen-binding sites comprise a light chain CDRl comprising RASESVDNYGISFMK (SEQ ID NO:20), a light chain CDR2 comprising AASNQGS (SEQ ID N021), and a light chain CDR3 comprising QQSKEVPWTFGG (SEQ ID NO:22). In some embodiments, the bispecific antibody is 219R45—MB-21R83.
In some embodiments, the VEGF/DLL4 binding agent (e.g., a bispecific antibody) comprises a first heavy chain variable region having at least about 80% sequence identity to SEQ ID NO:11, a second heavy chain variable region having at least about 80% sequence identity to SEQ ID NO:9, SEQ ID NO: 10, SEQ ID N025 8, or SEQ ID NO:64, and a first and a second light chain le region having at least 80% sequence identity to SEQ ID NO:12. In certain embodiments, the bispecific VEGF/DLL4- binding agent comprises a first heavy chain variable region having at least about 85%, at least about 90%, at least about 95%, at least about 97%, or at least about 99% sequence identity to SEQ ID NO:11; a second heavy chain variable region having at least about 85%, at least about 90%, at least about 95%, at least about 97%, or at least about 99% sequence identity to SEQ ID NO:9, SEQ ID NO: 10, SEQ ID NOS8, or SEQ ID NO:64; and a first and a second light chain variable region having at least about 85%, at least about 90%, at least about 95%, at least about 97%, or at least about 99% sequence identity to SEQ ID NO: 12. In certain embodiments, the bispecific LL4-binding agent comprises a first heavy chain variable region having at least about 95% sequence identity to SEQ ID NO:11, a second heavy chain variable region having at least about 95% sequence identity to SEQ ID NO:9, and a first and a second light chain variable region having at least about 95% sequence identity to SEQ ID NO:12. In certain embodiments, the bispecific VEGFKDLL4—binding agent comprises a first heavy chain variable region having at least about 95% sequence identity to SEQ ID NO:1 1, a second heavy chain variable region having at least about 95% sequence identity to SEQ ID NO: 10, and a first and a second light chain le region having at least about 95% ce identity to SEQ ID NO:12. In certain embodiments, the bispecific VEGF/DLL4-binding agent comprises a first heavy chain variable region having at least about 95% sequence identity to SEQ ID NO:1 1, a second heavy chain variable region having at least about 95% sequence ty to SEQ ID NO:58, and a first and a second light chain variable region having at least about 95% sequence identity to SEQ ID NO:12. In certain embodiments, the bispecific VEGF/DLL4-binding agent ses a first heavy chain variable region having at least about 95% sequence identity to SEQ ID NO:11, a second heavy chain variable region having at least about 95% sequence identity to SEQ ID NO:64, and a first and a second light chain variable region having at least about 95% sequence ty to SEQ ID NO:12. In certain embodiments, the bispecific LL4- binding agent comprises a first heavy chain variable region comprising SEQ ID NO:11, a second heavy chain variable region comprising SEQ ID NO:9, and a first and a second light chain variable region comprising SEQ ID NO:12. In certain embodiments, the bispecific VEGF/DLL4-binding agent comprises a first heavy chain variable region comprising SEQ ID NO:11, a second heavy chain le region comprising SEQ ID NO:10, and a first and a second light chain variable region comprising SEQ ID NO:12. In certain embodiments, the bispecific LL4-binding agent comprises a first heavy chain variable region sing SEQ ID NO:11, a second heavy chain variable region sing SEQ ID NOS8, and a first and a second light chain le region comprising SEQ ID NO:12. In certain embodiments, the bispecific VEGF/DLL4—binding agent comprises a first heavy chain variable region comprising SEQ ID N021 1, a second heavy chain variable region comprising SEQ ID NO:64, and a first and a second light chain le region comprising SEQ ID NO:12. In certain embodiments, the bispecific VEGF/DLL4-binding agent comprises a first heavy chain variable region consisting essentially of SEQ ID NO:11, a second heavy chain variable region consisting essentially of SEQ ID NO:9, and a first and a second light chain variable region consisting essentially of SEQ ID NO:12. In certain embodiments, the bispecific VEGF/DLL4—binding agent comprises a first heavy chain variable region consisting essentially of SEQ ID N011 1, a second heavy chain variable region consisting essentially of SEQ ID NO: 10, and a first and a second light chain variable region consisting essentially of SEQ ID NO:12. In certain embodiments, the bispecific VEGF/DLL4—binding agent comprises a first heavy chain variable region consisting ially of SEQ ID NO:1 1, a second heavy chain variable region consisting essentially of SEQ ID N025 8, and a first and a second light chain variable region consisting essentially of SEQ ID NO: 12. In certain embodiments, the bispecific VEGF/DLL4-binding agent comprises a first heavy chain variable region consisting essentially of SEQ ID NO: 1 l, a second heavy chain variable region consisting essentially of SEQ ID NO:64, and a first and a second light chain variable region consisting essentially of SEQ ID NO:12.
In some ments, the VEGF/DLL4—binding agent is a bispecific antibody which comprises a heavy chain variable region from the anti-VEGF antibody 219R45. In some embodiments, the VEGF/DLL4-binding agent is a bispecific antibody which comprises a heavy chain variable region from the anti-DLL4 dy 21M18. In some embodiments, the VEGF/DLL4-binding agent is a bispecific antibody which comprises a heavy chain variable region from the anti-DLL4 antibody 21R79. In some embodiments, the VEGF/DLL4—binding agent is a bispecific antibody which comprises a heavy chain variable region from the anti-DLL4 antibody 21R75. In some embodiments, the VEGF/DLL4-binding agent is a bispecific antibody which comprises a heavy chain variable region from the anti-DLL4 antibody 21R83. In some ments, the VEGF/DLL4—binding agent is a bispecific antibody which comprises a heavy chain variable region from the anti-VEGF antibody 219R45, a heavy chain variable region from the anti-DLL4 antibody 21R79 and two identical light chain variable regions. In some embodiments, the VEGF/DLL4-binding agent is a bispecific antibody which comprises a heavy chain variable region from the anti-VEGF antibody 219R45, a heavy chain variable region from the LL4 antibody 21Ml8 and two identical light chain variable regions. In some embodiments, the VEGF/DLL4-binding agent is a bispecific dy which comprises a heavy chain variable region from the anti-VEGF antibody 219R45, a heavy chain variable region from the anti-DLL4 antibody 21R75 and two identical light chain variable regions. In some ments, the LL4—binding agent is a bispecific antibody which comprises a heavy chain variable region from the anti—VEGF antibody 219R45, a heavy chain variable region from the anti-DLL4 antibody 21R83 and two identical light chain variable regions.
In some embodiments, the VEGF/DLL4—binding agent is a bispecific antibody which comprises a first CH3 domain and a second CH3 domain, each of which is d to promote formation of heteromultimers. In some embodiments, the first and second CH3 domains are modified using a knobs- into-holes technique. In some ments, the first and second CH3 domains comprise changes in amino acids that result in altered electrostatic ctions. In some ments, the first and second CH3 domains comprise changes in amino acids that result in altered hydrophobic/hydrophilic ctions.
In some embodiments, the VEGF/DLL4—binding agent is a bispecific antibody which comprises heavy chain constant regions selected from the group consisting of: (a) a first human IgG1 nt region, wherein the amino acids at positions 253 and 292 are substituted with glutamate or aspartate, and a second human IgG1 constant region, wherein the amino acids at positions 240 and 282 are substituted with lysine; (b) a first human IgG2 constant region, wherein the amino acids at positions 249 and 288 are tuted with glutamate or aspartate, and a second human IgG2 constant region wherein the amino acids at ons 236 and 278 are substituted with lysine; (c) a first human IgG3 constant region, wherein the amino acids at positions 300 and 339 are substituted with glutamate or aspartate, and a second human IgG3 constant region wherein the amino acids at positions 287 and 329 are substituted with lysine; and (d) a first human IgG4 constant region, wherein the amino acids at positions 250 and 289 are substituted with glutamate or aspartate, and a second IgG4 constant region wherein the amino acids at positions 237 and 279 are substituted with lysine.
In some embodiments, the VEGF/DLL4-binding agent is a bispecific dy which ses a first human IgG1 constant region with amino acid tutions at positions 253 and 292, wherein the amino acids are glutamate or aspartate, and a second human IgG1 constant region with amino acid tutions at positions 240 and 282, wherein the amino acids are lysine. In some embodiments, the VEGF/DLL4-binding agent is a bispecific antibody which comprises a first human IgG2 constant region with amino acid substitutions at positions 249 and 288, n the amino acids are glutamate or aspartate, and a second human IgG2 constant region with amino acid substitutions at positions 236 and 278, wherein the amino acids are lysine. In some embodiments, the VEGF/DLL4-binding agent is a bispecific antibody which comprises a first human IgG3 constant region with amino acid substitutions at positions 300 and 339, wherein the amino acids are glutamate or ate, and a second human IgG2 constant region with amino acid substitutions at positions 287 and 329, n the amino acids are . In some embodiments, the VEGF/DLL4—binding agent is a bispecific antibody which comprises a first human IgG4 constant region with amino acid substitutions at positions 250 and 289, wherein the amino acids are glutamate or aspartate, and a second human IgG4 constant region with amino acid substitutions at ons 237 and 279, wherein the amino acids are lysine.
In some embodiments, the VEGF/DLL4—binding agent is a ific antibody which comprises a first human IgG2 constant region with amino acid substitutions at positions 249 and 288, wherein the amino acids are glutamate, and a second human IgG2 constant region with amino acid substitutions at positions 236 and 278, wherein the amino acids are lysine. In some embodiments, the VEGF/DLL4- binding agent is a bispecific antibody which ses a first human IgG2 constant region with amino acid substitutions at positions 249 and 288, wherein the amino acids are asparate, and a second human IgG2 constant region with amino acid substitutions at positions 236 and 278, wherein the amino acids are lysine.
In some embodiments, the VEGF/DLL4—binding agent is a bispecific antibody which comprises a heavy chain of SEQ ID N017. In some embodiments, the VEGF/DLL4-binding agent is a bispecific antibody which comprises a heavy chain of SEQ ID N025. In some embodiments, the VEGF/DLL4- binding agent is a bispecific antibody which comprises a heavy chain of SEQ ID NO:56. In some embodiments, the VEGF/DLL4-binding agent is a bispecific antibody which comprises a heavy chain of SEQ ID N0:62. In some embodiments, the bispecific antibody further ses a light chain of SEQ ID NO: 12. In some embodiments, the VEGF/DLL4—binding agent is a bispecific antibody which comprises a heavy chain of SEQ ID N027, a heavy chain of SEQ ID N0:5, and two light chains of SEQ ID N0:8. In some embodiments, the VEGF/DLL4-binding agent is a bispecific antibody which comprises a heavy chain of SEQ ID N027, a heavy chain of SEQ ID N0:6, and two light chains of SEQ ID N0:8. In some embodiments, the VEGF/DLL4-binding agent is a bispecific antibody which comprises a heavy chain of SEQ ID N027, a heavy chain of SEQ ID N0256, and two light chains of SEQ ID N0:8. In some embodiments, the VEGF/DLL4-binding agent is a bispecific antibody which comprises a heavy chain of SEQ ID N027, a heavy chain of SEQ ID N0262, and two light chains of SEQ ID N0:8.
In some embodiments, the VEGF/DLL4—binding agent is a bispecific antibody which binds VEGF with a KD of about SOnM or less, about 25nM or less, about 10nM or less, about 1nM or less, or about 0.1nM or less. In some embodiments, the VEGF/DLL4—binding agent is a bispecific antibody which binds DLL4 with a KB of about SOnM or less, about 25nM or less, about 10nM or less, about 1nM or less, or about 0.1nM or less. In some embodiments, the VEGF/DLL4-binding agent is a bispecific dy which binds VEGF with a KD of about SOnM or less and binds DLL4 with a KD of about SOnM or less. In some embodiments, the bispecific antibody binds VEGF with a KD of about 25nM or less and binds DLL4 with a KD of about 25nM or less. In some ments, the bispecific antibody binds VEGF with a KD of about 10nM or less and binds DLL4 with a KD of about 10nM or less. In some embodiments, the bispecific antibody binds VEGF with a KD of about 1nM or less and binds DLL4 with a KD of about 1nM or less.
In some embodiments, the LL4—binding agent is a bispecific antibody which comprises one antigen-binding site with a binding affinity that is weaker than the binding affinity of the second antigen-binding site. For example, in some ments, the bispecific antibody may bind VEGF with a KD ranging from about 0.1nM to 1nM and may bind DLL4 with a KD ranging from about 1nM to 10nM.
Or the ific antibody may bind VEGF with a KD ranging from about lnM to 10nM and may bind DLL4 with a KD ranging from about 0.1nM to 1nM. In some embodiments, the bispecific dy may bind DLL4 with a KD ranging from about 0.1nM to 1nM and may bind VEGF with a KD ranging from about 1nM to 10nM. Or the ific antibody may bind DLL4 with a KD ranging from about 1nM to 10nM and may bind VEGF with a KD ranging from about 0.1nM to lnMIn some embodiments, the ence in affinity between the two antigen-binding sites may be about 2-fold or more, about 3-fold or more, about 5-fold or more, about 8—fold or more, about 10—fold or more, about 15-fold or more, about 30- fold or more, about 50-fold or more, or about lOO—fold or more. In some embodiments, at least one amino acid residue in at least one CDR of the antigen—binding site for VEGF is substituted with a different amino acid so that the affinity of the VEGF-binding site is altered. In some embodiments, the affinity of the VEGF-binding site is increased. In some embodiments, the affinity of the VEGF-binding site is decreased. In some embodiments, at least one amino acid residue in at least one CDR of the antigen- binding site for DLL4 is substituted with a different amino acid so that the affinity of the DLL4-binding site is altered. In some ments, the affinity of the DLL4—binding site is increased. In some embodiments, the affinity of the DLL4-binding site is decreased. In some embodiments, the affinities of both the VEGF and DLL4 antigen-binding sites are altered.
The invention provides ptides, ing but not limited to antibodies, that cally bind VEGF and/or DLL4. In some embodiments, a polypeptide binds human VEGF. In some embodiments, a ptide binds human DLL4. In some embodiments, a polypeptide binds human VEGF and mouse VEGF. In some ments, a polypeptide binds human DLL4 and mouse DLL4.
In some embodiments, a VEGF—binding agent comprises a polypeptide comprising a sequence selected from the group consisting of: SEQ ID N023, SEQ ID NO:4, SEQ ID NO:7, SEQ ID NO:8, SEQ ID N0:11, SEQ ID NO: 12, SEQ ID NO:47, and SEQ ID NO:49.
In some embodiments, a DLL4—binding agent comprises a polypeptide comprising a ce selected from the group consisting of: SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:4, SEQ ID N0:5, SEQ N0 ID:6, SEQ ID NO:8, SEQ ID N0:9, SEQ ID N0:10, SEQ ID N0:12, SEQ ID NO:46, SEQ ID NO:48, SEQ ID N0:56, SEQ ID N0:57, SEQ ID N0258, SEQ ID NO:62, SEQ ID NO:63, and SEQ ID NO:64.
In some embodiments, a VEGF/DLL4-binding agent comprises a polypeptide comprising a sequence selected from the group consisting of: SEQ ID NO:1, SEQ ID NO:2, SEQ ID N0:3, SEQ ID NO:4, SEQ ID N0:5, SEQ ID NO:6, SEQ ID NO:7, SEQ ID NO:8, SEQ ID N0:9, SEQ ID N0:10, SEQ ID N0:11, SEQ ID NO: 12, SEQ ID NO:46, SEQ ID NO:47, SEQ ID N0:48, SEQ ID NO:49, SEQ ID N0:56, SEQ ID N0:57, SEQ ID N0258, SEQ ID NO:62, SEQ ID NO:63, and SEQ ID NO:64.
In some ments, a VEGF/DLL4—binding agent comprises a polypeptide comprising a sequence selected from the group ting of: SEQ ID NO:1, SEQ ID NO:2, SEQ ID N0:5, SEQ ID N026, SEQ ID N029, SEQ ID N0:10, SEQ ID NO:46, SEQ ID N0z48, SEQ ID N0256, SEQ ID N0:57, SEQ ID N0258, SEQ ID NO:62, SEQ ID NO:63, and SEQ ID NO:64. In some embodiments, the VEGF/DLL4 binding agent further ses a polypeptide comprising a sequence selected from the group consisting of: SEQ ID N023, SEQ ID NO:7, SEQ ID N0:1 1, SEQ ID NO:47, and SEQ ID NO:49.
In some embodiments, the VEGF/DLL4 g agent further comprises a polypeptide comprising a sequence selected from the group consisting of: SEQ ID NO:4, SEQ ID NO:8, and SEQ ID N0:12.
In some embodiments, a VEGF/DLL4—binding agent comprises a polypeptide comprising a sequence selected from the group consisting of: SEQ ID N0:3, SEQ ID NO:7, SEQ ID N0:11, SEQ ID NO:47, and SEQ ID NO:49. In some embodiments, the VEGF/’DLL4 binding agent further comprises a polypeptide comprising a sequence selected from the group consisting of: SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:5, SEQ ID NO:6, SEQ ID NO:9, SEQ ID NO:10, SEQ ID NO:46, SEQ ID NO:48, SEQ ID NO:56, SEQ ID NO:57, SEQ ID NO:58, SEQ ID NO:62, SEQ ID NO:63, and SEQ ID NO:64. In some embodiments, the VEGF/DLL4 binding agent fiirther comprises a polypeptide comprising a sequence selected from the group consisting of: SEQ ID NO:4, SEQ ID NO:8, and SEQ ID NO: 12.
In certain embodiments, a VEGFXDLL4—binding agent (e.g., antibody) competes for specific binding to VEGF with an antibody that comprises a heavy chain variable region comprising SEQ ID NO:11 and a light chain variable region comprising SEQ ID NO:12. In certain embodiments, a VEGF/DLL4-binding agent competes with antibody 219R45 for c binding to human VEGF. In some embodiments, a VEGF/DLL4-binding agent or antibody competes for c binding to VEGF in an in vitro competitive binding assay. In some embodiments, the VEGF is human VEGF. In some embodiments, the VEGF is mouse VEGF.
In certain embodiments, a VEGF—DLL4—binding agent (e.g., an antibody) binds the same epitope, or essentially the same epitope, on VEGF as an antibody of the invention. In another embodiment, a VEGF/DLL4-binding agent is an antibody that binds an epitope on VEGF that ps with the epitope on VEGF bound by an antibody of the ion. In certain embodiments, a VEGF/DLL4-binding agent (e.g., an antibody) binds the same epitope, or essentially the same epitope, on VEGF as antibody 219R45.
In r embodiment, the VEGF/DLL4-binding agent is an antibody that binds an e on VEGF that overlaps with the epitope on VEGF bound by antibody 219R45.
In n embodiments, the LL4—binding agent is an agent that es for specific binding to VEGF with the antibody 219R45 (e.g., in a competitive g assay).
In certain embodiments, a VEGF/DLL4—binding agent (e.g., antibody) competes for specific binding to DLL4 with an antibody that comprises a heavy chain variable region comprising SEQ ID NO:9 SEQ ID NO:10, SEQ ID N025 8, or SEQ ID NO:64 and a light chain variable region comprising SEQ ID NO:12. In certain embodiments, a VEGF/DLL4—binding agent competes with antibody 21R79 for specific binding to human DLL4. In n embodiments, a VEGF/DLL4-binding agent competes with antibody 21R75 for specific binding to human DLL4. In certain embodiments, a VEGF/DLL4-binding agent competes with dy 21R83 for specific binding to human DLL4. In some embodiments, a LL4-binding agent or antibody competes for specific binding to DLL4 in an in vitro competitive binding assay. In some embodiments, the DLL4 is human DLL4. In some embodiments, the DLL4 is mouse DLL4.
In certain embodiments, a VEGF/DLL4-binding agent (e.g., an antibody) binds the same epitope, or essentially the same epitope, on DLL4 as an antibody of the invention. In another embodiment, a VEGF/DLL4-binding agent is an dy that binds an epitope on DLL4 that overlaps with the epitope on DLL4 bound by an antibody of the invention. In certain embodiments, a VEGF/DLL4-binding agent binds the same epitope, or essentially the same epitope, on DLL4 as antibody 21R79. In certain embodiments, a LL4-binding agent binds the same epitope, or essentially the same epitope, on DLL4 as antibody 21R75. In certain embodiments, a LL4-binding agent binds the same epitope, or essentially the same epitope, on DLL4 as antibody 21R83. In another embodiment, the VEGF/DLL4- binding agent is an antibody that binds an epitope on DLL4 that overlaps with the epitope on DLL4 bound by antibody 21R79. In another embodiment, the VEGF/DLL4—binding agent is an antibody that binds an epitope on DLL4 that ps with the epitope on DLL4 bound by antibody 21R75. In another embodiment, the VEGF/DLL4-binding agent is an antibody that binds an epitope on DLL4 that overlaps with the epitope on DLL4 bound by dy 21R83.
In n embodiments, the VEGF/DLL4-binding agent is an agent that competes for specific binding to DLL4 with the antibody 21R79 (e.g., in a competitive binding assay). In certain embodiments, the VEGF/DLL4—binding agent is an agent that competes for specific g to DLL4 with the antibody 21R75 (e. g., in a competitive binding assay). In certain embodiments, the VEGF/DLL4-binding agent is an agent that es for specific binding to DLL4 with the antibody 21R83 (e. g., in a itive binding assay). In certain embodiments, the VEGF/DLL4—binding agent is an agent that competes for specific binding to DLL4 with the antibody 21M18 (e.g., in a competitive binding assay).
In certain embodiments, the VEGF/DLL4—binding agent is an agent that es for c binding to VEGF and/or DLL4 with the ific antibody 219R45-MB-21M18 (e.g., in a competitive binding . In certain embodiments, the VEGF/DLL4—binding agent is an agent that competes for specific binding to VEGF and/or DLL4 with the bispecific antibody 219R45-MB-21M79 (e.g., in a competitive binding . In certain embodiments, the VEGF/DLL4-binding agent is an agent that competes for specific binding to VEGF and/or DLL4 with the bispecific antibody 219R45-MB-21M75 (e.g., in a competitive binding assay). In certain embodiments, the VEGF/DLL4-binding agent is an agent that competes for c binding to VEGF and/or DLL4 with the bispecific antibody 219R45-MB- 21M83 (e. g., in a itive binding assay).
In certain ments, the VEGFXDLL4-binding agent (e.g., an antibody) described herein binds VEGF and modulates VEGF activity. In some embodiments, the VEGF/DLL4-binding agent is a VEGF antagonist and inhibits VEGF activity. In some embodiments, the VEGF/DLL4-binding agent is a VEGF antagonist and modulates enesis. In some embodiments, the VEGF/DLL4—binding agent is a VEGF antagonist and inhibits angiogenesis. In some embodiments, the VEGF/DLL4-binding agent is a VEGF antagonist and inhibits tumor growth.
In certain embodiments, a VEGF/DLL4-binding agent (e.g., an antibody) described herein binds human DLL4 and modulates DLL4 activity. In some embodiments, a VEGF/DLL4-binding agent is a DLL4 antagonist and inhibits DLL4 activity. In some embodiments, a VEGF/DLL4-binding agent is a DLL4 antagonist and inhibits Notch activity. In some embodiments, a VEGF/DLL4-binding agent is a DLL4 antagonist and inhibits Notch signaling. In some embodiments, a VEGF/DLL4-binding agent is a DLL4 nist and modulates angiogenesis. In some embodiments, a VEGF/DLL4-binding agent is a DLL4 antagonist and es aberrant angiogenesis. In some ments, a LL4-binding agent is a DLL4 antagonist and inhibits tumor growth.
In n embodiments, a VEGFXDLL4—binding agent (e.g., an antibody) described herein is a bispecific antibody that binds human VEGF and modulates VEGF activity. In certain embodiments, a VEGF/DLL4-binding agent (e.g., an antibody) described herein is a bispecific antibody that binds human DLL4 and tes DLL4 activity. In certain embodiments, a VEGF/DLL4—binding agent (e.g., an antibody) described herein is a bispecific antibody that binds human VEGF and human DLL4 and modulates both VEGF and DLL4 activity. In some embodiments, the bispecific dy is a VEGF antagonist and a DLL4 antagonist and inhibits both VEGF activity and DLL4 activity. In some embodiments, the bispecific dy is a VEGF antagonist and a DLL4 antagonist and inhibits VEGF activity and Notch activity. In some embodiments, the bispecific antibody is a VEGF nist and a DLL4 antagonist and inhibits VEGF activity and Notch signaling. In some embodiments, the bispecific antibody is a VEGF antagonist and a DLL4 antagonist and modulates angiogenesis. In some embodiments, the bispecific antibody is a VEGF antagonist and a DLL4 antagonist and promotes aberrant angiogenesis. In some embodiments, the bispecific antibody is a VEGF antagonist and a DLL4 antagonist and inhibits angiogenesis. In some embodiments, the bispecific antibody is a VEGF antagonist and a DLL4 antagonist and inhibits tumor growth.
In certain embodiments, the VEGF/DLL4—binding agent (e.g., an antibody or a bispecific dy) is an nist ofVEGF. In some embodiments, the VEGF/DLL4-binding agent is an antagonist ofVEGF and ts VEGF activity. In certain embodiments, the VEGF/DLL4-binding agent inhibits VEGF activity by at least about 10%, at least about 20%, at least about 30%, at least about 50%, at least about 75%, at least about 90%, or about 100%. In certain embodiments, a LL4-binding agent that inhibits human VEGF ty is antibody 219R45. In certain embodiments, a VEGF/DLL4- binding agent that inhibits human VEGF activity is a bispecific antibody comprising the antigen-binding site of 219R45. In certain embodiments, a VEGF/DLL4—binding agent that inhibits human VEGF activity is the bispecific antibody 219R45-MB-21M18. In certain embodiments, a VEGF/DLL4-binding agent that inhibits human VEGF activity is the bispecific antibody 219R45—MB—21R79. In certain embodiments, a VEGF/DLL4-binding agent that inhibits human VEGF activity is the bispecific antibody 219R45-MB-21R75. In certain embodiments, a VEGF/DLL4-binding agent that inhibits human VEGF activity is the bispecific antibody 219R45-MB-21R83.
In certain embodiments, the VEGF/DLL4—binding agent (e.g., an antibody) is an antagonist of DLL4. In some embodiments, the VEGF/DLL4—binding agent is an antagonist of DLL4 and inhibits DLL4 activity. In n embodiments, the VEGF/DLL4—binding agent inhibits DLL4 activity by at least about 10%, at least about 20%, at least about 30%, at least about 50%, at least about 75%, at least about 90%, or about 100%. In certain embodiments, a VEGF/DLL4-binding agent that inhibits human DLL4 ty is antibody 21R79. In certain embodiments, a VEGF/DLL4-binding agent that inhibits human DLL4 activity is antibody 21R75. In certain embodiments, a VEGF/DLL4-binding agent that inhibits human DLL4 activity is antibody 21R83. In certain embodiments, a VEGF/DLL4-binding agent that inhibits human DLL4 activity is a bispecific antibody comprising the antigen-binding site of 21R79. In certain embodiments, a VEGF/DLL4-binding agent that inhibits human DLL4 activity is a bispecific antibody comprising the antigen-binding site of 21R75. In certain embodiments, a VEGF/DLL4-binding agent that inhibits human DLL4 activity is a bispecific dy comprising the antigen-binding site of 21R83. In certain embodiments, a VEGF/DLL4-binding agent that inhibits human DLL4 ty is the bispecific antibody 219R45-MB-21M18. In certain embodiments, a VEGF/DLL4-binding agent that inhibits human DLL4 activity is the bispecific antibody —MB-21R79. In certain embodiments, a VEGF/DLL4-binding agent that inhibits human DLL4 activity is the bispecific antibody 219R45-MB- 21R75. In n embodiments, a VEGF/DLL4—binding agent that inhibits human DLL4 activity is the bispecific antibody 2 1 9R45-MB-2 1 R83.
In certain embodiments, the VEGF/DLL4—binding agent (e.g., antibody) is an antagonist h signaling. In certain embodiments, the VEGF/DLL4—binding agent inhibits Notch signaling by at least about 10%, at least about 20%, at least about 30%, at least about 50%, at least about 75%, at least about 90%, or about 100%. In certain embodiments, a VEGF/DLL4-binding agent that inhibits Notch ing is antibody 21R79. In certain embodiments, a LL4-binding agent that ts Notch signaling is antibody 21R75. In certain embodiments, a VEGF/DLL4-binding agent that inhibits Notch signaling is antibody 21R83. In n embodiments, a VEGF/DLL4-binding agent that inhibits Notch ing is a bispecific antibody comprising the antigen-binding site of 21R79. In certain ments, a VEGF/DLL4-binding agent that ts Notch ing is a bispecific antibody comprising the antigen- binding site of 21R75. In certain embodiments, a VEGF/DLL4-binding agent that inhibits Notch signaling is a ific antibody comprising the antigen—binding site of 21R83. In certain embodiments, a VEGF/DLL4-binding agent that inhibits Notch signaling is the bispecific antibody 219R45-MB-21M18.
In certain embodiments, a VEGF/DLL4—binding agent that inhibits Notch signaling is the bispecific antibody -MB-21R79. In certain embodiments, a VEGF/DLL4—binding agent that inhibits Notch signaling is the bispecific antibody 219R45-MB-21R75. In certain embodiments, a VEGF/DLL4-binding agent that inhibits Notch signaling is the bispecific antibody 219R45-MB-21R83.
In certain embodiments, the VEGF/DLL4-binding agent (e.g., antibody) inhibits binding of VEGF to at least one receptor. In some ments, the VEGF/DLL4-binding agent inhibits binding of VEGF to VEGFR-l or VEGFR—2. In certain embodiments, the VEGF/DLL4-binding agent inhibits binding ofVEGF to at least one VEGF receptor by at least about 10%, at least about 25%, at least about 50%, at least about 75%, at least about 90%, or at least about 95%. In certain embodiments, a VEGF/DLL4-binding agent that inhibits binding of human VEGF to at least one VEGF receptor is antibody 219R45. In certain embodiments, a VEGF/DLL4-binding agent that inhibits binding of human VEGF to at least one VEGF receptor is a bispeciflc antibody comprising the n-binding site of 2 l 9R45. In certain embodiments, a VEGFXDLL4—binding agent that inhibits binding of human VEGF to at least one VEGF receptor is the bispecific antibody 219R45—MB—21M18. In certain embodiments, a VEGF/DLL4-binding agent that inhibits binding of human VEGF to at least one VEGF or is the bispecific antibody 219R45-MB-21R79. In certain ments, a VEGF/DLL4—binding agent that ts g of human VEGF to at least one VEGF receptor is the bispecific antibody 219R45-MB- 21R75. In certain embodiments, a VEGF/DLL4-binding agent that inhibits binding of human VEGF to at least one VEGF or is the bispecific dy 219R45-MB-21R83.
In certain embodiments, the VEGF/DLL4—binding agent (e.g., antibody) inhibits binding of DLL4 protein to at least one Notch receptor. In some embodiments, the LL4-binding agent inhibits g of DLL4 to , Notch2, Notch3, and/or Notch4. In certain embodiments, the VEGF/DLL4- binding agent inhibits binding of DLL4 to at least one Notch receptor by at least about 10%, at least about %, at least about 50%, at least about 75%, at least about 90%, or at least about 95%. In certain embodiments, a VEGF/DLL4-binding agent that inhibits binding of human DLL4 to at least one Notch receptor is antibody 21R79. In certain embodiments, a VEGF/DLL4-binding agent that inhibits binding of human DLL4 to at least one Notch or is antibody 21R75. In certain embodiments, a VEGF/DLL4-binding agent that inhibits binding of human DLL4 to at least one Notch receptor is antibody 21R83. In certain embodiments, a VEGF/DLL4-binding agent that inhibits binding of human DLL4 to at least one Notch receptor is a bispecific antibody comprising the antigen-binding site of 21R79.
In certain embodiments, a VEGF/DLL4-binding agent that inhibits binding of human DLL4 to at least one Notch receptor is a bispecific antibody comprising the antigen-binding site of 21R75. In certain embodiments, a VEGF/DLL4-binding agent that inhibits binding of human DLL4 to at least one Notch receptor is a bispecific antibody comprising the antigen—binding site of 21R83. In certain embodiments, a VEGF/DLL4-binding agent that inhibits binding of human DLL4 to at least one Notch receptor is the bispecific antibody 219R45-MB-21M18. In certain embodiments, a VEGF/DLL4-binding agent that inhibits binding of human DLL4 to at least one Notch receptor is the bispecific antibody 219R45-MB- 21R79. In certain embodiments, a LL4-binding agent that ts binding of human DLL4 to at least one Notch or is the bispeciflc antibody 219R45-MB-21R75. In certain embodiments, a VEGF/DLL4-binding agent that inhibits binding of human DLL4 to at least one Notch receptor is the bispecific dy —MB—21R83.
In vivo and in vitro assays for determining whether a VEGF/DLL4-binding agent (or candidate VEGF/DLL4-binding agent) inhibits VEGF or affects angiogenesis are known in the art. In vitro assays of enesis include but are not limited to, HUVEC proliferation assays, endothelial cell tube formation assays, sprouting (or sprout ion) assays, HUVEC cell migration assays, and invasion assays. In some embodiments, cells in the presence ofVEGF and the presence of a VEGF/DLL4-binding agent are compared to cells in the presence ofVEGF without the LL4-binding agent present, and ted for effects on angiogenesis (or biological effects associated with angiogenesis). In vivo assays of angiogenesis include, but are not limited to, matrigel plug assays, corneal micropocket assays, and n allantoic membrane (CAM) assays.
In vivo and in vitro assays for determining whether a VEGF/DLL4—binding agent (or candidate VEGF/DLL4-binding agent) inhibits Notch activation or signaling are known in the art. For example, ased, rase reporter assays utilizing a c reporter vector containing multiple copies of the TCF-binding domain upstream of a firefly luciferase reporter gene may be used to measure Notch signaling levels in vitro (Gazit et al., 1999, Oncogene, 18; 5959—66; TOPflash, Millipore, Billerica MA).
In some embodiments, a ased, luciferase reporter assay ing a CBF/Luc reporter vector containing multiple copies of the CBF—binding domain upstream of a firefly luciferase report genes may be used. The level ofNotch signaling in the presence of one or more Notch ligands (e.g., DLL4 expressed on the surface of transfected cells or soluble DLL4—F0 fusion protein) and in the presence of a VEGF/DLL4-binding agent is compared to the level ofNotch signaling without the VEGF/DLL4-binding agent present.
In certain embodiments, the VEGF/DLL4-binding agents have one or more of the following effects: inhibit proliferation of tumor cells, t tumor growth, reduce the tumorigenicity of a tumor, reduce the frequency of cancer stem cells in a tumor, trigger cell death of tumor cells, prevent metastasis of tumor cells, decrease survival of tumor cells, modulate angiogenesis, inhibit angiogenesis, inhibit productive angiogenesis, or promote aberrant angiogenesis.
In certain embodiments, the VEGF/DLL4—binding agents are capable of inhibiting tumor growth.
In certain embodiments, the VEGF/DLL4—binding agents are capable of inhibiting tumor growth in vivo (e.g., in a aft mouse model, and/or in a human having cancer). In certain embodiments, tumor growth is inhibited at least about two-fold, about three—fold, about five—fold, about ten-fold, about 50-fold, about 100-fold, or about IOOO-fold as compared to an untreated tumor.
In certain embodiments, the VEGF/DLL4—binding agents are capable of reducing the tumorigenicity of a tumor. In n embodiments, the VEGF/DLL4-binding agent or antibody is capable of reducing the tumorigenicity of a tumor comprising cancer stem cells in an animal model, such as a mouse xenograft model. In n ments, the VEGF/DLL4-binding agent or antibody is capable of reducing the tumorigenicity of a tumor by decreasing the number or ncy of cancer stem cells in the tumor. In certain embodiments, the number or frequency of cancer stem cells in a tumor is reduced by at least about two-fold, about three—fold, about five—fold, about ld, about 50-fold, about 100-fold, or about 1000-fold. In certain embodiments, the reduction in the number or frequency of cancer stem cells is determined by limiting dilution assay using an animal model. Additional examples and guidance regarding the use of limiting dilution assays to determine a reduction in the number or frequency of cancer stem cells in a tumor can be found, e.g., in International Publication Number ; US.
Patent Publication No. 2008/0064049; and US. Patent Publication No. 2008/0178305.
In certain embodiments, the LL4—binding agents are capable of modulating angiogenesis.
In certain embodiments, the LL4-binding agents are capable of modulating angiogenesis in vivo (e.g., in a xenograft mouse model, and/or in a human having cancer). In certain ments, VEGF/DLL4-binding agents are capable of inhibiting angiogenesis. In certain ments, LL4-binding agents are capable ofpromoting aberrant angiogenesis. In certain embodiments, LL4-binding agents are e of inhibiting angiogenesis and/or promoting nt enesis, leading to unproductive vascularization.
In certain embodiments, the VEGF/DLL4—binding agents described herein have a circulating half- life in mice, cynomolgus monkeys, or humans of at least about 2 hours, at least about 5 hours, at least about 10 hours, at least about 24 hours, at least about 3 days, at least about 1 week, or at least about 2 weeks. In certain embodiments, the VEGF/DLL4—binding agent is an IgG (e.g., IgGl or IgG2) antibody that has a circulating half-life in mice, cynomolgus monkeys, or humans of at least about 2 hours, at least about 5 hours, at least about 10 hours, at least about 24 hours, at least about 3 days, at least about 1 week, or at least about 2 weeks. Methods of increasing (or decreasing) the half-life of agents such as polypeptides and antibodies are known in the art. For example, known methods of increasing the circulating half-life of IgG dies include the introduction of mutations in the Fc region which increase the pH-dependent binding of the antibody to the neonatal Fc receptor (FcRn) at pH 6.0 (see, e. g., US. Patent Publication Nos. 2005/0276799, 2007/0148164, and 2007/0122403). Known methods of increasing the circulating half-life of antibody fragments lacking the Fc region include such techniques as PEGylation.
In some embodiments, the VEGFXDLL4—binding agents are antibodies. Polyclonal dies can be ed by any known method. In some embodiments, polyclonal antibodies are produced by immunizing an animal (e.g, a rabbit, rat, mouse, goat, donkey) with an antigen of interest (e.g., a purified peptide fragment, full-length recombinant protein, or fiision protein) by multiple subcutaneous or intraperitoneal injections. The antigen can be optionally conjugated to a r such as keyhole limpet hemocyanin (KLH) or serum albumin. The antigen (with or without a carrier protein) is diluted in sterile saline and usually ed with an adjuvant (e.g., Complete or Incomplete Freund’s Adjuvant) to form a stable emulsion. After a sufficient period of time, polyclonal antibodies are recovered from the immunized animal, y from blood or ascites. The polyclonal dies can be purified from serum or ascites according to standard s in the art including, but not limited to, affinity chromatography, ion-exchange chromatography, gel electrophoresis, and dialysis.
In some embodiments, the VEGF/DLL4—binding agents are monoclonal antibodies. Monoclonal antibodies can be prepared using hybridoma s known to one of skill in the art (see e. g., Kohler and Milstein, 1975, Nature, 5-497). In some embodiments, using the hybridoma method, a mouse, hamster, or other appropriate host animal, is immunized as described above to elicit from lymphocytes the production of antibodies that cally bind the immunizing antigen. In some embodiments, lymphocytes can be immunized in vitro. In some embodiments, the immunizing antigen can be a human protein or a portion thereof. In some embodiments, the immunizing antigen can be a mouse protein or a portion thereof.
Following zation, lymphocytes are isolated and fused with a suitable myeloma cell line using, for example, polyethylene . The hybridoma cells are selected using specialized media as known in the art and unfused lymphocytes and myeloma cells do not survive the selection process.
Hybridomas that produce monoclonal antibodies directed specifically against a chosen antigen may be identified by a y of methods including, but not limited to, immunoprecipitation, immunoblotting, and in vitro binding assays (e.g., flow cytometry, FACS, ELISA, and radioimmunoassay). The hybridomas can be propagated either in in vitro culture using rd methods (J.W. , 1996, Monoclonal Antibodies: Principles and Practice, 3rd Edition, ic Press, San Diego, CA) or in vivo as ascites tumors in an animal. The monoclonal antibodies can be purified from the culture medium or ascites fluid according to standard methods in the art including, but not limited to, affinity chromatography, ion-exchange chromatography, gel electrophoresis, and dialysis.
In certain embodiments, monoclonal antibodies can be made using recombinant DNA techniques as known to one skilled in the art. The polynucleotides encoding a monoclonal antibody are isolated from mature B-cells or hybridoma cells, such as by RT—PCR using oligonucleotide primers that specifically y the genes encoding the heavy and light chains of the antibody, and their sequence is determined using standard techniques. The isolated polynucleotides encoding the heavy and light chains are then cloned into suitable expression vectors which produce the monoclonal antibodies when transfected into host cells such as E. coli, simian COS cells, Chinese r ovary (CHO) cells, or a cells that do not otherwise produce immunoglobulin proteins.
In certain other ments, recombinant monoclonal antibodies, or fragments thereof, can be isolated from phage display libraries expressing variable domains or CDRs of a d s (see e.g., McCafferty et al., 1990, Nature, 2-554; Clackson et al., 1991, , 352:624-628; and Marks et al., 1991, J. Mol. Biol, 222:581-597).
The polynucleotide(s) encoding a monoclonal antibody can be modified, for example, by using recombinant DNA technology to generate alternative antibodies. In some embodiments, the constant domains of the light and heavy chains of, for example, a mouse monoclonal antibody can be substituted for those regions of, for example, a human antibody to generate a chimeric antibody, or for a non- globulin polypeptide to generate a fusion antibody. In some embodiments, the constant regions are truncated or removed to generate the desired antibody fragment of a monoclonal antibody. Site- directed or high-density mutagenesis of the variable region can be used to optimize specificity, affinity, etc. of a monoclonal antibody.
In some embodiments, a monoclonal antibody against VEGF and/or DLL4 is a zed antibody. Typically, humanized antibodies are human immunoglobulins in which residues from the CDRs are replaced by residues from a CDR of a non—human s (e.g., mouse, rat, rabbit, hamster, etc.) that have the desired specificity, affinity, and/or binding capability using methods known to one skilled in the art. In some embodiments, the Fv ork region residues of a human immunoglobulin are replaced with the corresponding residues in an antibody from a non-human species that has the desired specificity, affinity, and/or binding capability. In some embodiments, a humanized antibody can be further modified by the tution of additional residues either in the FV framework region and/or within the replaced non-human residues to refine and optimize antibody specificity, y, and/or capability.
In general, a humanized antibody will comprise substantially all of at least one, and typically two or three, variable domain regions containing all, or substantially all, of the CDRs that correspond to the non-human immunoglobulin whereas all, or substantially all, of the framework regions are those of a human immunoglobulin consensus sequence. In some embodiments, a humanized antibody can also comprise at least a portion of an globulin nt region or domain (Fc), typically that of a human immunoglobulin. In certain embodiments, such humanized antibodies are used eutically because they may reduce antigenicity and HAMA (human anti—mouse antibody) responses when administered to a human subject. One skilled in the art would be able to obtain a functional humanized antibody with d immunogenicity following known ques (see e.g., US. Patent Nos. 5,225,539; 089; ,693,761; and 5,693,762).
In certain embodiments, the VEGF/DLL4—binding agent is a human dy. Human antibodies can be directly prepared using various techniques known in the art. In some embodiments, human antibodies may be generated from immortalized human B lymphocytes zed in vitro or from lymphocytes isolated from an immunized individual. In either case, cells that produce an antibody directed t a target antigen can be ted and isolated (see, e.g., Cole et al., 1985, onal Antibodies and Cancer Therapy, Alan R. Liss, p. 77; Boemer et al., 1991, J. Immunol, -95; and US. Patent Nos. 5,750,373; 5,567,610; and 5,229,275). In some embodiments, the human antibody can be selected from a phage library, where that phage library expresses human antibodies (Vaughan et al., 1996, Nature Biotechnology, 14:309—3 14; Sheets et al., 1998, PNAS, 95:6157-6162; Hoogenboom and Winter, 1991, J. Mol. Biol, 2272381; Marks et al., 1991, J. Mol. Biol, 222:581). Alternatively, phage display technology can be used to produce human antibodies and antibody fragments in vitro, from immunoglobulin variable domain gene repertoires from unimmunized donors. Techniques for the generation and use of antibody phage libraries are also described in U.S. Patent Nos. 5,969,108; 6,172,197; 5,885,793; 6,521,404; 6,544,731; 6,555,313; 6,582,915; 6,593,081; 6,300,064; 6,653,068; 6,706,484; and 7,264,963; and Rothe et al., 2008, J. Mol. Bio., 376:1182-1200. Once antibodies are identified, affinity maturation strategies known in the art, including but not limited to, chain shuffling (Marks et al., 1992, Bio/Technology, 102779—783) and site—directed mutagenesis, may be employed to generate high affinity human antibodies.
In some embodiments, human dies can be made in transgenic mice that contain human immunoglobulin loci. Upon immunization these mice are e of producing the full repertoire of human antibodies in the absence of endogenous immunoglobulin production. This approach is described in U.S. Patent Nos. 5,545,807; 5,545,806; 825; 5,625,126; 5,633,425; and 5,661,016.
This invention also encompasses bispecific antibodies. Bispecific antibodies are capable of specifically izing and binding at least two different antigens or epitopes. The different epitopes can either be within the same molecule (e. g., two epitopes on a single protein) or on ent molecules (e. g., one epitope on a protein and one epitope on a second protein). In some embodiments, a bispecific antibody has enhanced potency as compared to an individual antibody or to a combination ofmore than one antibody. In some ments, a ific antibody has reduced toxicity as compared to an individual antibody or to a combination ofmore than one antibody. It is known to those of skill in the art that any g agent (e.g., antibody) may have unique pharmacokinetics (PK) (e. g., circulating half- life). In some embodiments, a bispecific antibody has the ability to synchronize the PK of two active binding agents wherein the two individual binding agents have different PK profiles. In some embodiments, a ific dy has the ability to concentrate the actions of two binding agents (e.g., antibodies) in a common area (e. g., a tumor and/or tumor environment). In some embodiments, a bispecific antibody has the ability to concentrate the actions of two binding agents (e. g., antibodies) to a common target (e. g., a tumor or a tumor cell). In some embodiments, a bispecific antibody has the ability to target the actions of two binding agents (e.g., antibodies) to more than one biological pathway or function.
In certain embodiments, the bispecific antibody cally binds VEGF and a second target. In certain embodiments, the bispecific dy specifically binds DLL4 and a second target. In certain embodiments, the bispecific antibody specifically binds VEGF and DLL4. In some embodiments, the bispecific antibody cally binds human VEGF and human DLL4. In some embodiments, the bispecific dy is a monoclonal human or a humanized antibody. In some embodiments, the bispecific antibody inhibits angiogenesis and reduces cancer stem cell number or frequency. In some embodiments, the bispecific antibody inhibits blood vessel growth and inhibits blood vessel maturation.
In some embodiments, the ific antibody prevents endothelial roliferation. In some embodiments, the bispecific antibody has decreased toxicity and/or side effects. In some embodiments, the ific antibody has decreased toxicity and/or side effects as compared to a mixture of the two individual antibodies or the antibodies as single agents. In some ments, the bispeciflc antibody has an increased eutic index. In some embodiments, the bispecific antibody has an increased therapeutic index as compared to a mixture of the two individual antibodies or the antibodies as single agents.
In some embodiments, the bispecific antibody can specifically recognize and bind a first antigen target, (e.g., DLL4) as well as a second antigen target, such as an effector molecule on a leukocyte (e.g., CD2, CD3, CD28, or B7) or a PC receptor (e.g., CD64, CD32, or CD16) so as to focus cellular defense mechanisms to the cell expressing the first antigen target. In some embodiments, the bispecific dies can be used to direct cytotoxic agents to cells which express a particular target antigen. These antibodies possess an antigen—binding site (e. g., to human DLL4) and a second site which binds a cytotoxic agent or a radionuclide or, such as EOTUBE, DPTA, DOTA, or TETA.
Techniques for making bispecific antibodies are known by those skilled in the art, see for example, Millstein et al., 1983, Nature, 305:537—539; Brennan et al., 1985, Science, 229:81; Suresh et al., 1986, Methods in Enzymol., 121 : 120; Traunecker et al., 1991, EMBO J., 10:3655-3659; Shalaby et al., 1992, J. Exp. Med, 175:217-225; Kostelny et al., 1992, J. Immunol., 148:1547-1553; Gruber et al., 1994, J. Immunol., 152:5368; U.S. Patent No. 5,731,168; International Publication No. ; and US. Patent ation No. 2011/0123532. In some embodiments, the bispecific antibodies comprise heavy chain constant regions with modifications in the amino acids which are part of the interface between the two heavy chains. In some embodiments, the bispecific antibodies can be generated using a “knobs-into-holes” strategy (see. e.g., US. Patent No. 5,731,168; Ridgway et. al., 1996, Prat. Engin, 9:617-621). At times the “knobs” and “holes” terminology is replaced with the terms “protuberances” and “cavities”. In some embodiments, the bispecific antibodies may comprise variant hinge regions incapable of g de linkages between the heavy chains (see, e.g., ). In some embodiments, the modifications may comprise changes in amino acids that result in altered ostatic interactions. In some embodiments, the modifications may comprise changes in amino acids that result in altered hobic/hydrophilic interactions.
Bispecific antibodies can be intact antibodies or antibody fragments sing antigen-binding sites. Antibodies with more than two valencies are also contemplated. For example, trispecific antibodies can be prepared (Tutt et al., 1991, J. l, 147:60). Thus, in certain embodiments the antibodies to VEGF and/or DLL4 are multispecific.
In certain ments, the antibodies (or other ptides) described herein may be monospecific. In certain embodiments, each of the one or more n-binding sites that an antibody contains is capable of binding (or binds) a homologous epitope on different proteins.
In certain embodiments, the VEGF/DLL4—binding agent is an antibody fragment. Antibody fragments may have ent functions or capabilities than intact antibodies; for example, antibody nts can have increased tumor penetration. Various techniques are known for the production of antibody fragments including, but not limited to, proteolytic digestion of intact antibodies. In some embodiments, antibody fragments include a F(ab')2 nt produced by pepsin digestion of an antibody molecule. In some embodiments, dy fragments include a Fab fragment generated by reducing the disulfide bridges of an F(ab')2 fragment. In other embodiments, antibody fragments include a Fab fragment generated by the treatment of the antibody molecule with papain and a reducing agent. In certain embodiments, antibody fragments are produced recombinantly. In some embodiments, antibody fragments e Fv or single chain Fv (scFv) fragments. Fab, Fv, and scFv antibody fragments can be expressed in and secreted from E. coli or other host cells, ng for the production of large amounts of these fragments. In some embodiments, antibody fragments are isolated from dy phage libraries as discussed herein. For example, methods can be used for the construction of Fab sion libraries (Huse et al., 1989, Science, 246: 1275—1281) to allow rapid and effective identification of monoclonal Fab fragments with the desired specificity for VEGF and/or DLL4 or derivatives, fragments, analogs or homologs thereof. In some embodiments, antibody fragments are linear antibody fragments. In certain embodiments, antibody fragments are monospecific or bispecific. In certain embodiments, the LL4-binding agent is a scFv. Various techniques can be used for the production of single-chain dies specific to VEGF or DLL4 (see, e.g., U.S. Patent No. 4,946,778).
It can further be ble, especially in the case of dy fragments, to modify an antibody in order to alter (e.g., increase or decrease) its serum half-life. This can be achieved, for example, by incorporation of a salvage receptor g epitope into the antibody fragment by mutation of the riate region in the antibody fragment or by incorporating the epitope into a peptide tag that is then fused to the dy nt at either end or in the middle (e.g., by DNA or peptide synthesis).
Heteroconjugate dies are also within the scope of the present invention. Heteroconjugate antibodies are composed of two ntly joined antibodies. Such antibodies have, for example, been proposed to target immune cells to unwanted cells (see, e.g., U.S. Patent No. 4,676,980). It is also contemplated that the heteroconjugate antibodies can be ed in vitro using known methods in synthetic protein chemistry, including those involving crosslinking agents. For example, immunotoxins can be constructed using a disulfide exchange reaction or by forming a thioether bond. Examples of suitable reagents for this purpose include iminothiolate and methylmercaptobutyrimidate.
For the purposes of the present invention, it should be appreciated that modified dies can se any type of variable region that provides for the association of the antibody with the target (i.e., human VEGF or human DLL4). In this regard, the variable region may comprise or be derived from any type of mammal that can be induced to mount a humoral response and generate immunoglobulins against the desired antigen. As such, the variable region of the modified antibodies can be, for example, of human, murine, non-human primate (e.g. cynomolgus monkeys, macaques, etc.) or rabbit origin. In some embodiments, both the variable and constant regions of the modified globulins are human. In other embodiments, the variable regions of compatible antibodies (usually derived from a non-human source) can be engineered or specifically tailored to improve the binding properties or reduce the immunogenicity of the molecule. In this respect, variable s usefiil in the present invention can be humanized or otherwise altered through the inclusion of imported amino acid sequences.
In certain embodiments, the le domains in both the heavy and light chains are d by at least partial replacement of one or more CDRs and, if necessary, by partial framework region replacement and sequence modification and/or tion. Although the CDRs may be d from an antibody of the same class or even subclass as the antibody from which the framework regions are derived, it is envisaged that the CDRs may be derived from an antibody of different class and often from an antibody from a different species. It may not be necessary to replace all of the CDRs with all of the CDRs from the donor variable region to transfer the antigen binding capacity of one variable domain to another. Rather, it may only be necessary to transfer those residues that are required to maintain the activity of the antigen- binding site.
Alterations to the le region notwithstanding, those skilled in the art will iate that the modified antibodies of this invention will comprise antibodies (e. g., full-length antibodies or immunoreactive fragments thereof) in which at least a fraction of one or more of the nt region domains has been deleted or otherwise altered so as to provide desired biochemical characteristics such as increased tumor localization or increased serum half—life when compared with an antibody of approximately the same genicity comprising a native or unaltered constant region. In some embodiments, the constant region of the modified antibodies will comprise a human constant region.
Modifications to the constant region compatible with this ion comprise additions, deletions or substitutions of one or more amino acids in one or more domains. The d antibodies disclosed herein may comprise alterations or modifications to one or more of the three heavy chain constant domains (CH1, CH2 or CH3) and/or to the light chain constant domain (CL). In some embodiments, one or more domains are lly or entirely deleted from the constant s of the modified antibodies. In some ments, the modified antibodies will comprise domain deleted constructs or variants wherein the entire CH2 domain has been removed (ACH2 ucts). In some embodiments, the omitted constant region domain is replaced by a short amino acid spacer (e.g., 10 amino acid residues) that es some of the molecular flexibility typically imparted by the absent constant region.
In some embodiments, the modified antibodies are engineered to fuse the CH3 domain directly to the hinge region of the antibody. In other embodiments, a e spacer is inserted between the hinge region and the modified CH2 and/or CH3 s. For example, constructs may be expressed wherein the CH2 domain has been deleted and the ing CH3 domain (modified or unmodified) is joined to the hinge region with a 5-20 amino acid spacer. Such a spacer may be added to ensure that the regulatory elements of the constant domain remain free and accessible or that the hinge region remains e.
However, it should be noted that amino acid spacers may, in some cases, prove to be immunogenic and elicit an unwanted immune response against the construct. Accordingly, in certain embodiments, any spacer added to the construct will be relatively non—immunogenic so as to maintain the d biological qualities of the modified antibodies.
In some embodiments, the modified antibodies may have only a partial deletion of a constant domain or substitution of a few or even a single amino acid. For example, the mutation of a single amino acid in selected areas of the CH2 domain may be enough to substantially reduce Fc binding and thereby se cancer cell localization and/or tumor penetration. rly, it may be desirable to simply delete the part of one or more constant region domains that l a specific effector function (e.g. complement Clq binding) to be modulated. Such partial deletions of the constant regions may improve selected characteristics of the antibody (serum half—life) while leaving other desirable functions associated with the subject constant region domain intact. Moreover, as alluded to above, the constant regions of the disclosed dies may be modified through the mutation or substitution of one or more amino acids that enhances the profile of the resulting construct. In this respect it may be possible to disrupt the activity provided by a conserved binding site (e.g., Fc binding) while substantially maintaining the configuration and immunogenic profile of the modified antibody. In certain embodiments, the modified dies comprise the addition of one or more amino acids to the constant region to enhance desirable characteristics such as decreasing or increasing or fimction or provide for more cytotoxin or carbohydrate attachment sites.
It is known in the art that the constant region mediates l effector functions. For example, binding of the Cl component of complement to the Fc region of IgG or IgM antibodies (bound to antigen) activates the complement system. Activation of complement is important in the opsonization and lysis of cell pathogens. The activation of complement also stimulates the inflammatory response and can also be involved in autoimmune hypersensitivity. In addition, the Fc region of an antibody can bind a cell expressing a PC receptor (FcR). There are a number of Fc receptors which are specific for different s of antibody, including IgG (gamma receptors), IgE on receptors), IgA (alpha receptors) and IgM (mu receptors). Binding of antibody to Fc receptors on cell es triggers a number of important and diverse biological responses ing ment and destruction of antibody-coated particles, clearance of immune complexes, lysis of antibody—coated target cells by killer cells (called antibody- dependent cell cytotoxicity or ADCC), release of inflammatory mediators, placental transfer, and l of immunoglobulin production.
In certain embodiments, the modified antibodies e for altered effector functions that, in turn, affect the biological profile of the administered antibody. For example, in some ments, the deletion or inactivation (through point mutations or other means) of a constant region domain may reduce Fc receptor binding of the circulating modified antibody thereby increasing cancer cell localization and/or tumor penetration. In other embodiments, the constant region modifications se the serum ife of the antibody. In other embodiments, the constant region modifications reduce the serum half-life of the dy. In some embodiments, the constant region is modified to eliminate disulfide linkages or oligosaccharide moieties. Modifications to the constant region in accordance with this invention may easily be made using well known biochemical or molecular engineering techniques known to those of skill in the art.
In certain embodiments, a VEGF/DLL4—binding agent that is an antibody does not have one or more effector ons. For ce, in some embodiments, the antibody has no ADCC activity, and/or no complement-dependent cytotoxicity (CDC) ty. In certain embodiments, the antibody does not bind an Fc receptor, and/or ment factors. In certain embodiments, the antibody has no effector function.
The present invention further embraces variants and equivalents which are substantially gous to the chimeric, humanized, and human antibodies, or antibody fragments thereof, set forth herein. These can contain, for example, conservative substitution mutations, i.e. the substitution of one or more amino acids by similar amino acids. For example, conservative substitution refers to the substitution of an amino acid with another amino acid within the same general class such as, for example, one acidic amino acid with another acidic amino acid, one basic amino acid with another basic amino acid or one neutral amino acid by another neutral amino acid. What is intended by a conservative amino acid substitution is well known in the art and described herein.
Thus, the present invention provides methods for producing an antibody that binds VEGF and/or DLL4, including ific antibodies that specifically bind both VEGF and DLL4. In some embodiments, the method for producing an antibody that binds VEGF and/or DLL4 comprises using hybridoma techniques. In some ments, the method of generating an antibody that binds VEGF or DLL4 or a bispecific antibody that binds VEGF and DLL4 comprises screening a human phage library.
The t invention further provides methods of identifying an antibody that binds VEGF and/or DLL4.
In some embodiments, the antibody is identified by FACS screening for binding to VEGF or a portion thereof. In some embodiments, the antibody is identified by FACS screening for binding to DLL4 or a n thereof. In some ments, the antibody is identified by FACS screening for binding to both VEGF and DLL4 or a portion thereof. In some embodiments, the antibody is fied by screening using ELISA for g to VEGF. In some embodiments, the dy is identified by screening using ELISA for binding to DLL4. In some embodiments, the antibody is identified by screening using ELISA for binding to VEGF and DLL4. In some embodiments, the antibody is identified by FACS screening for blocking of binding of human VEGF to a human VEGF receptor. In some ments, the antibody is identified by FACS screening for blocking of binding of human DLL4 to a human Notch receptor. In some embodiments, the antibody is identified by screening for inhibition or blocking ofNotch signaling.
In some embodiments, the antibody is identified by screening for inhibition or ng ofVEGF activity (e.g., induction of HUVEC proliferation). In some embodiments, the antibody is identified by ing for modulation of angiogenesis.
In some embodiments, a method of generating an antibody to human VEGF comprises immunizing a mammal with a polypeptide comprising amino acids 27-232 of human VEGF. In some embodiments, a method of ting an antibody to human VEGF comprises immunizing a mammal with a polypeptide comprising at least a portion of amino acids 27-232 of human VEGF. In some embodiments, the method further comprises isolating antibodies or antibody-producing cells from the mammal. In some ments, a method of generating a monoclonal antibody which binds VEGF comprises: immunizing a mammal with a ptide comprising at least a n of amino acids 27-232 of human VEGF, and isolating antibody-producing cells from the immunized mammal. In some embodiments, the method r comprises fusing the antibody-producing cells with cells of a a cell line to form hybridoma cells. In some embodiments, the method further comprises selecting a oma cell expressing an dy that binds VEGF. In certain embodiments, the mammal is a mouse. In some embodiments, the antibody is selected using a polypeptide comprising at least a portion of amino acids 27-232 of human VEGF.
In some embodiments, a method of generating an antibody to human DLL4 comprises immunizing a mammal with a polypeptide comprising amino acids 27-529 of human DLL4. In some embodiments, a method of generating an antibody to human DLL4 comprises immunizing a mammal with a polypeptide comprising at least a portion of amino acids 2?—529 of human DLL4. In some embodiments, a method of generating a monoclonal antibody which binds DLL4 comprises: immunizing a mammal with a polypeptide comprising at least a portion of amino acids 27-529 of human DLL4, and isolating antibody producing cells from the immunized mammal. In some ments, the method further comprises fusing the antibody-producing cells with cells of a myeloma cell line to form hybridoma cells. In some embodiments, the method further comprises selecting a hybridoma cell expressing an antibody that binds DLL4. In certain embodiments, the mammal is a mouse. In some embodiments, the antibody is selected using a polypeptide comprising at least a portion of amino acids 27-529 of human DLL4.
In some embodiments, a method of generating an dy to human VEGF comprises screening an antibody-expressing library for antibodies that bind human VEGF. In some embodiments, a method of generating an antibody human DLL4 ses screening an antibody-expressing library for antibodies that bind human DLL4. In some embodiments, a method of generating an antibody to human VEGF and/or human DLL4 comprises ing an antibody—expressing library for bispecific antibodies that bind human VEGF and human DLL4. In some embodiments, the antibody-expressing y is a phage library. In some embodiments, the screening comprises panning. In some embodiments, the antibody- expressing library (e.g., a phage library) is screened using at least a portion of amino acids 27-232 of human VEGF. In some embodiments, antibodies identified in the first screening, are screened again using at least a portion of amino acids 27-529 of human DLL4 to identify a bispecific antibody that binds VEGF and DLL4. In some ments, the antibody-expressing library (e. g., a phage library) is ed using at least a portion of amino acids 27-529 of human DLL4. In some embodiments, antibodies identified in the first screening, are screened again using at least a portion of amino acids 27-232 of human VEGF to identify a bispecific antibody that binds VEGF and DLL4. In some embodiments, the antibody identified in the screening is a VEGF antagonist. In some embodiments, the antibody identified in the screening ts biological activities induced by VEGF. In some embodiments, the antibody identified in the screening is a DLL4 antagonist. In some embodiments, the antibody identified in the screening inhibits Notch ing d by DLL4. In some embodiments, the antibody fied in the screening binds both human VEGF and mouse VEGF. In some embodiments, the antibody identified in the screening binds both human DLL4 and mouse DLL4.
In certain ments, the antibodies described herein are ed. In certain embodiments, the antibodies described herein are substantially pure.
In some embodiments of the present invention, the VEGF/DLL4-binding agents are polypeptides.
The polypeptides can be recombinant polypeptides, natural polypeptides, or synthetic polypeptides comprising an antibody, or fragment thereof, that bind VEGF and/or DLL4. It will be recognized in the art that some amino acid sequences of the binding agents described herein can be varied without significant effect on the structure or function of the protein. Thus, the invention further includes variations of the polypeptides which show substantial activity or which include regions of an antibody, or fragment thereof, against human VEGF andfor DLL4. In some embodiments, amino acid sequence variations of VEGF/DLL4-binding polypeptides include deletions, insertions, inversions, repeats, and/or other types of substitutions.
In some embodiments, the polypeptides described herein are isolated. In some embodiments, the ptides described herein are ntially pure.
The polypeptides, analogs and variants thereof, can be r modified to contain additional chemical moieties not normally part of the polypeptide. The derivatized moieties can improve or ise modulate the solubility, the biological half—life, and/or absorption of the polypeptide. The es can also reduce or eliminate undesirable side effects of the polypeptides and variants. An overview for chemical moieties can be found in Remington: The Science and Practice ofPharmacy, 21” Edition, 2005, University of the Sciences, Philadelphia, PA.
The polypeptides described herein can be produced by any suitable method known in the art.
Such methods range from direct protein synthesis methods to ucting a DNA sequence encoding polypeptide sequences and sing those sequences in a suitable host. In some embodiments, a DNA sequence is constructed using recombinant technology by isolating or synthesizing a DNA sequence encoding a wild-type protein of st. Optionally, the sequence can be mutagenized by site-specific mutagenesis to provide functional analogs thereof. See, e.g., Zoeller et al., 1984, PNAS, 81:5662-5066 and US Patent No. 4,588,585.
In some embodiments, a DNA sequence ng a polypeptide of interest may be constructed by chemical synthesis using an ucleotide synthesizer. Oligonucleotides can be designed based on the amino acid sequence of the desired polypeptide and selecting those codons that are d in the host cell in which the recombinant polypeptide of st will be produced. Standard methods can be applied to synthesize a polynucleotide sequence encoding an isolated polypeptide of st. For example, a complete amino acid sequence can be used to construct a back—translated gene. Further, a DNA oligomer containing a nucleotide sequence coding for the particular isolated polypeptide can be synthesized. For example, l small Oligonucleotides coding for portions of the d polypeptide can be synthesized and then ligated. The individual Oligonucleotides typically n 5' or 3' overhangs for complementary assembly.
Once assembled (by synthesis, site-directed mutagenesis, or another method), the polynucleotide sequences encoding a particular polypeptide of interest can be ed into an expression vector and operatively linked to an expression control sequence appropriate for expression of the protein in a desired host. Proper assembly can be confirmed by nucleotide sequencing, restriction enzyme mapping, and/or expression of a biologically active polypeptide in a suitable host. As is well-known in the art, in order to obtain high expression levels of a transfected gene in a host, the gene must be operatively linked to riptional and translational expression control sequences that are functional in the chosen expression host.
In certain embodiments, inant expression vectors are used to amplify and express DNA encoding antibodies, or nts thereof, against human VEGF and/or DLL4. For example, recombinant sion vectors can be replicable DNA constructs which have synthetic or cDNA-derived DNA fragments encoding a polypeptide chain of a VEGF/DLL4-binding agent, such as an anti-VEGF antibody or an anti-DLL4 antibody, or fragment thereof, ively linked to suitable transcriptional and/or translational tory elements derived from mammalian, microbial, viral, or insect genes. A transcriptional unit generally comprises an assembly of (1) a genetic element or elements having a regulatory role in gene expression, for example, transcriptional promoters or ers, (2) a structural or coding sequence which is transcribed into mRNA and translated into protein, and (3) appropriate transcription and translation initiation and termination sequences. Regulatory elements can include an operator sequence to control transcription. The ability to replicate in a host, usually red by an origin of replication, and a selection gene to facilitate recognition of transformants can additionally be incorporated. DNA regions are “operatively linked” when they are functionally related to each other. For example, DNA for a signal peptide (secretory leader) is operatively linked to DNA for a polypeptide if it is expressed as a precursor which participates in the secretion of the polypeptide; a promoter is operatively linked to a coding sequence if it controls the transcription of the sequence; or a ribosome binding site is operatively linked to a coding sequence if it is positioned so as to permit ation. In some embodiments, structural elements intended for use in yeast expression systems include a leader sequence enabling extracellular secretion of translated protein by a host cell. In other embodiments, in situations where recombinant protein is sed without a leader or transport sequence, it can include an N- terminal methionine residue. This residue can optionally be subsequently cleaved from the expressed recombinant protein to provide a final product.
The choice of an expression control sequence and an expression vector depends upon the choice of host. A wide y of expression host/vector combinations can be employed. Useful sion vectors for eukaryotic hosts include, for example, vectors sing expression control sequences from SV40, bovine papilloma virus, adenovirus, and cytomegalovirus. Useful expression vectors for bacterial hosts include known bacterial plasmids, such as plasmids from E. coli, including pCRl, pBR322, pMB9, and their tives, and wider host range ds, such as M13 and other filamentous single-stranded DNA phages.
The VEGF/DLL4-binding agents (e.g., polypeptides) of the present invention can be sed from one or more vectors. For example, in some embodiments, one heavy chain polypeptide is expressed by one vector, a second heavy chain polypeptide is expressed by a second vector and a light chain polypeptide is expressed by a third vector. In some embodiments, a first heavy chain polypeptide and a light chain polypeptide is expressed by one vector and a second heavy chain polypeptide is sed by a second vector. In some embodiments, two heavy chain ptides are expressed by one vector and a light chain ptide is expressed by a second vector. In some embodiments, three polypeptides are sed from one vector. Thus, in some embodiments, a first heavy chain polypeptide, a second heavy chain polypeptide, and a light chain polypeptide are expressed by a single vector.
Suitable host cells for sion of a VEGF/DLL4-binding polypeptide or antibody (or a VEGF or DLL4 protein to use as an antigen) include prokaryotes, yeast cells, insect cells, or higher eukaryotic cells under the l of appropriate promoters. Prokaryotes e gram-negative or ositive organisms, for example E. coli or Bacillus. Higher eukaryotic cells include established cell lines of mammalian origin as described below. Cell—free translation systems may also be ed. Appropriate cloning and expression vectors for use with bacterial, fungal, yeast, and mammalian cellular hosts are described in Pouwels et al., 1985, Cloning Vectors: A Laboratory Manual, ElseVier, New York, NY. onal information regarding methods of protein production, ing antibody production, can be found, e.g., in US. Patent Publication No. 2008/0187954; US. Patent Nos. 6,413,746; 6,660,501; and International Patent Publication No. WO 04f009823.
Various mammalian or insect cell e systems may be used to express recombinant polypeptides. Expression of recombinant proteins in mammalian cells may be desirable because these proteins are generally correctly folded, appropriately d, and biologically onal. Examples of le mammalian host cell lines include, but are not limited to, COS-7 (monkey kidney-derived), L-929 (murine fibroblast-derived), C127 (murine mammary tumor-derived), 3T3 (murine fibroblast-derived), CHO (Chinese hamster ovary—derived), HeLa (human cervical cancer-derived), BHK (hamster kidney fibroblast-derived), HEK-293 (human embryonic kidney—derived) cell lines and variants of these cell lines. Mammalian expression vectors can comprise non—transcribed elements such as an origin of replication, a suitable promoter and enhancer linked to the gene to be expressed, and other 5' or 3' flanking non-transcribed sequences, and 5' or 3' non—translated sequences, such as necessary ribosome binding sites, a polyadenylation site, splice donor and acceptor sites, and transcriptional termination sequences.
Expression of recombinant proteins in baculovirus also offers a robust method for producing correctly folded and biologically functional proteins. Baculovirus s for production of logous proteins in insect cells are well-known to those of skill in the art (see, e.g., Luckow and Summers, 1988, Bio/Technology, 6:47).
Thus, the present invention provides cells comprising the VEGF/DLL4-binding agents described herein. In some embodiments, the cells produce the VEGF/DLL4-binding agents described herein. In certain embodiments, the cells produce an antibody. In some embodiments, the cells produce a VEGF- binding agent, such as an anti-VEGF antibody. In some embodiments, the cells produce a bispeciflc antibody that binds VEGF. In some embodiments, the cells produce a DLL4-binding agent, such as an anti-DLL4 antibody. In some embodiments, the cells produce a ific antibody that binds DLL4. In certain embodiments, the cells produce a bispecific VEGF/DLL4—binding agent, such as a bispecific dy that binds VEGF and DLL4. In certain embodiments, the cells produce antibody 219R45. In certain embodiments, the cells produce dy 21R79. In certain ments, the cells produce dy 21R75. In certain embodiments, the cells produce antibody 21R83. In certain embodiments, the cells produce a bispeciflc antibody which comprises an n-binding site from dy . In certain ments, the cells produce a bispecific antibody which comprises an antigen-binding site from antibody 21R79. In certain embodiments, the cells produce a bispecific antibody which ses an antigen-binding site from antibody 21R75. In certain embodiments, the cells produce a bispecific antibody which ses an antigen-binding site from antibody 21R83. In certain embodiments, the cells produce a bispecific antibody which comprises an antigen-binding site from antibody 219R45 and an antigen-binding site from antibody 21R79. In certain embodiments, the cells produce a bispecific dy which comprises an antigen-binding site from antibody 219R45 and an antigen-binding site from dy 21M18. In certain embodiments, the cells produce a ific antibody which comprises an antigen-binding site from antibody 219R45 and an antigen—binding site from dy 21R75. In certain embodiments, the cells produce a bispecific antibody which comprises an n-binding site from antibody 219R45 and an antigen-binding site from antibody 21R83. In certain embodiments, the cells e the ific antibody 219R45-MB-21M18. In certain embodiments, the cells produce the bispecific antibody 219R45-MB-21R79. In certain embodiments, the cells produce the bispecific antibody 219R45-MB-21R75. In n embodiments, the cells produce the bispecific antibody 219R45- The proteins produced by a transformed host can be purified according to any suitable method.
Standard methods include chromatography (e.g., ion exchange, affinity, and sizing column chromatography), centrifugation, differential solubility, or by any other standard technique for protein purification. Affinity tags such as hexa—histidine, maltose g domain, influenza coat sequence, and glutathione-S-transferase can be attached to the protein to allow easy purification by passage over an appropriate affinity column. y chromatography used for purifying immunoglobulins can include n A, n G, and Protein L chromatography. Isolated proteins can be physically characterized using such techniques as proteolysis, size exclusion chromatography (SEC), mass spectrometry (MS), nuclear magnetic resonance (NMR), isoelectric focusing (IEF), high mance liquid chromatography (HPLC), and x-ray crystallography. The purity of ed proteins can be determined using techniques known to those of skill in the art, including but not limited to, SDS-PAGE, SEC, capillary gel electrophoresis, IEF, and capillary isoelectric focusing (cIEF).
In some embodiments, supernatants from expression systems which secrete recombinant protein into culture media can be first concentrated using a commercially available protein concentration filter, for example, an Amicon or Millipore Pellicon ltration unit. Following the concentration step, the concentrate can be d to a suitable purification matrix. In some embodiments, an anion exchange resin can be employed, for example, a matrix or substrate having pendant diethylaminoethyl (DEAE) groups. The matrices can be acrylamide, agarose, dextran, cellulose, or other types commonly employed in protein ation. In some embodiments, a cation exchange step can be employed. le cation exchangers include various insoluble matrices comprising sulfopropyl or carboxymethyl groups. In some embodiments, a hydroxyapatite media can be employed, including but not limited to, ceramic hydroxyapatite (CHT). In certain embodiments, one or more reverse-phase HPLC steps ing hydrophobic RP-HPLC media, e.g., silica gel having pendant methyl or other aliphatic groups, can be employed to further purify a recombinant protein (e.g., a VEGF/DLL4-binding agent). Some or all of the foregoing purification steps, in various combinations, can be employed to provide a homogeneous inant protein.
In some ments, heterodimeric proteins such as bispecific antibodies are purified according the any of the methods described herein. In some embodiments, anti-VEGF/anti-DLL4 bispecific antibodies are isolated and/or purified using at least one chromatography step. In some embodiments, the at least one chromatography step comprises affinity chromatography. In some embodiments, the at least one chromatography step further comprises anion exchange chromatography. In some ments, the isolated and/or purified antibody product comprises at least 90% dimeric antibody. In some embodiments, the isolated and/or purified antibody t comprises at least 95%, 96%, 97%, 98% or 99% heterodimeric antibody. In some embodiments, the isolated and/or purified antibody product comprises about 100% heterodimeric antibody.
In some embodiments, recombinant protein produced in bacterial culture can be isolated, for example, by initial extraction from cell pellets, followed by one or more concentration, salting-out, aqueous ion exchange, or size exclusion chromatography steps. HPLC can be employed for final purification steps. Microbial cells employed in expression of a recombinant protein can be disrupted by any convenient method, including -thaw cycling, sonication, mechanical disruption, or use of cell lysing agents.
Methods known in the art for purifying antibodies and other proteins also e, for example, those described in U.S. Patent ation Nos. 2008/0312425; 2008/0177048; and 2009/0187005.
In certain embodiments, the VEGF/DLL4—binding agent is a ptide that is not an antibody.
A variety ofmethods for fying and ing non-antibody polypeptides that bind with high affinity to a protein target are known in the art. See, e.g., Skerra, 2007, Curr. Opin. Biotechnol, 18:295-304; Hosse et al., 2006, Protein Science, 15:14-27; Gill et al., 2006, Curr. Opin. Biotechnol., 17:653-658; Nygren, 2008, FEBSJ, 275:2668-76; and Skerra, 2008, FEBSJ, 275:2677-83. In n embodiments, phage or mammalian cell display technology may be used to produce and/or identify a LL4- binding polypeptide that is not an antibody. In certain embodiments, the ptide comprises a protein scaffold of a type ed from the group consisting of n A, protein G, a lipocalin, a fibronectin domain, an ankyrin consensus repeat , and thioredoxin.
In certain embodiments, the VEGF/DLL4-binding agents or antibodies can be used in any one of a number of ated (i.e. an immunoconjugate or radioconjugate) or non-conjugated forms. In certain embodiments, the antibodies can be used in a non-conjugated form to harness the subject’s natural defense mechanisms including ment—dependent cytotoxicity and antibody-dependent cellular toxicity to eliminate malignant or cancer cells.
In some embodiments, the VEGF/DLL4—binding agent (e.g., an antibody or polypeptide) is conjugated to a cytotoxic agent. In some embodiments, the cytotoxic agent is a chemotherapeutic agent including, but not limited to, methotrexate, adriamicin, doxorubicin, melphalan, mitomycin C, chlorambucil, ubicin or other intercalating agents. In some ments, the cytotoxic agent is an enzymatically active toxin of bacterial, fungal, plant, or animal origin, or fragments thereof, including, but not limited to, diphtheria A chain, non-binding active fragments of diphtheria toxin, exotoxin A chain, ricin A chain, abrin A chain, modeccin A chain, alpha—sarcin, Aleurites fordii proteins, dianthin ns, Phytolaca americana proteins (PAPI, PAPII, and PAP—S), ica charantia inhibitor, curcin, crotin, Sapaonaria officinalis inhibitor, gelonin, mitogellin, restrictocin, phenomycin, enomycin, and the tricothecenes. In some embodiments, the cytotoxic agent is a radioisotope to produce a radioconjugate or a radioconjugated antibody. A variety of radionuclides are available for the production of radioconjugated antibodies including, but not d to, 90Y, 125I, 131I, 123I, 111In, 131In, 105Rh, 1538m, 67Cu, 67Ga, 166Ho, 177Lu, 186Re, 188Re and 212Bi. Conjugates of an antibody and one or more small le , such as calicheamicins, maytansinoids, trichothecenes, and CC1065, and the tives of these toxins that have toxin activity, can also be used. Conjugates of an dy and cytotoxic agent can be made using a variety of bifunctional protein—coupling agents including, but not limited to, N- succinimidyl(2-pyridyidithiol) propionate (SPDP), iminothiolane (IT), bifunctional tives of imidoesters (such as dimethyl adipimidate HCl), active esters (such as disuccinimidyl suberate), aldehydes (such as eldehyde), ido compounds (such as bis(p-azidobenzoyl) hexanediamine), bis- diazonium derivatives (such as bis-(p-diazoniumbenzoyl)—ethylenediamine), diisocyanates (such as toluene 2,6-diisocyanate), and bis-active fluorine compounds (such as 1,5-difluoro-2,4-dinitrobenzene).
III. cleotides In certain embodiments, the invention encompasses polynucleotides comprising polynucleotides that encode a polypeptide (or a fragment of a polypeptide) that specifically binds VEGF, DLL4, both VEGF and DLL4. The term “polynucleotides that encode a polypeptide” encompasses a polynucleotide which includes only coding sequences for the polypeptide, as well as a polynucleotide which includes additional coding and/or non-coding sequences. For example, in some embodiments, the invention provides a polynucleotide comprising a polynucleotide sequence that encodes an antibody to human VEGF or encodes a fragment of such an antibody (e.g., a nt comprising the n-binding site).
In some embodiments, the invention provides a polynucleotide comprising a polynucleotide sequence that encodes an dy to human DLL4 or encodes a fragment of such an antibody (e. g., a fragment comprising the antigen-binding site). The polynucleotides of the invention can be in the form ofRNA or in the form of DNA. DNA includes cDNA, c DNA, and synthetic DNA; and can be double- stranded or single-stranded, and if single—stranded can be the coding strand or non-coding (anti-sense) strand.
In certain embodiments, the polynucleotide comprises a cleotide encoding a polypeptide comprising a sequence selected from the group ting of SEQ ID N021, SEQ ID N022, SEQ ID N0:3, SEQ ID N024, SEQ ID N025, SEQ ID N0:6, SEQ ID N027, SEQ ID N028, SEQ ID N0:9, SEQ ID N0:10, SEQ ID N0:11, SEQ ID N0:12, SEQ ID N0:46, SEQ ID N0:47, SEQ ID N0:48, SEQ ID N0:49, SEQ ID N0:56, SEQ ID N0:57, SEQ ID N0:58, SEQ ID N0:62, SEQ ID N0:63, and SEQ ID N0:64. In certain embodiments, the polynucleotide comprises a polynucleotide encoding a polypeptide comprising a sequence selected from the group consisting of SEQ ID N0z5, SEQ ID N026, SEQ ID N027, SEQ ID N028, SEQ ID N029, SEQ ID N0:10, SEQ ID N0:11, SEQ ID N0:12, SEQ ID N0:48, SEQ ID N0249, SEQ ID N0256, SEQ ID NO:58, SEQ ID N0:62, and SEQ ID NO: 64. In some embodiments, the polynucleotide comprises a polynucleotide sequence selected from the group ting of SEQ ID N0229, SEQ ID N0:30, SEQ ID N0:3], SEQ ID N0:32, SEQ ID N0:33, SEQ ID N0:34, SEQ ID N0:35, SEQ ID N0:36, SEQ ID N0:37, SEQ ID N0:38, SEQ ID N0:39, SEQ ID N0:40, SEQ ID NO:50, SEQ ID N0:51, SEQ ID N0:52, SEQ ID NO:53, SEQ ID N0:54, SEQ ID NO:55, SEQ ID N0:60, SEQ ID N0:61, SEQ ID N0:66, SEQ ID N0:67, SEQ ID N0:68, SEQ ID N0:69, SEQ ID N0:70, SEQ ID N0:71, SEQ ID N0:72, SEQ ID N0:73, SEQ ID N0:74, SEQ ID N0:75, SEQ ID N0:76, SEQ ID N077, and SEQ ID N0:78.
In certain embodiments, the polynucleotide comprises a polynucleotide haVing a nucleotide sequence at least about 80% identical, at least about 85% identical, at least about 90% identical, at least about 95% identical, and in some embodiments, at least about 96%, 97%, 98% or 99% identical to a cleotide comprising a sequence selected from the group consisting of SEQ ID N0229, SEQ ID N0:30, SEQ ID N0:31, SEQ ID N0:32, SEQ ID N0:33, SEQ ID N0:34, SEQ ID N0:52, SEQ ID N0:53, SEQ ID N0:55, SEQ ID N0:60, SEQ ID N0:61, SEQ ID N0:66, SEQ ID N0:67, SEQ ID N0:68, SEQ ID N0:69, SEQ ID N0:70, SEQ ID N0:71, SEQ ID N0:72, SEQ ID N0:73, SEQ ID N0:74, SEQ ID N0:75, SEQ ID N0276, SEQ ID N0:77, and SEQ ID N0:78. In certain embodiments, the polynucleotide comprises a cleotide having a nucleotide sequence at least about 80% cal, at least about 85% identical, at least about 90% identical, at least about 95% identical, and in some embodiments, at least about 96%, 97%, 98% or 99% identical to a polynucleotide comprising a sequence selected from the group consisting of SEQ ID N0:35, SEQ ID N0:36, SEQ ID N0:37, SEQ ID N0:38, SEQ ID N0239, SEQ ID N0240, SEQ ID NO:50, SEQ ID N0:51, SEQ ID N0:54, SEQ ID N0:68, SEQ ID NO:69, SEQ ID NO:70, SEQ ID N0:71, SEQ ID N0:72, SEQ ID N0:73, SEQ ID N0:74, SEQ ID N0:75, SEQ ID N0276, SEQ ID N0277, and SEQ ID N0:78. Also ed is a polynucleotide that comprises a polynucleotide that hybridizes to SEQ ID N0:29, SEQ ID N0:30, SEQ ID N0:31, SEQ ID N0:32, SEQ ID N0:33, SEQ ID N0:34, SEQ ID N0:35, SEQ ID N0:36, SEQ ID N0:37, SEQ ID N0:38, SEQ ID N0:39, SEQ ID N0240, SEQ ID NO:50, SEQ ID N0:51, SEQ ID N0:52, SEQ ID N0:53, SEQ ID N0:54, SEQ ID NO:55, SEQ ID N0:60, SEQ ID N0:61, SEQ ID N0:66, SEQ ID NO:67, SEQ ID NO:68, SEQ ID NO:69, SEQ ID NO:70, SEQ ID NO:71, SEQ ID NO:72, SEQ ID NO:73, SEQ ID NO:74, SEQ ID NO:75, SEQ ID NO:76, SEQ ID NO:77, and SEQ ID NO:78. In certain embodiments, the hybridization is under ions of high stringency.
In certain embodiments, the polynucleotides comprise the coding sequence for the mature polypeptide fused in the same reading frame to a polynucleotide which aids, for example, in sion and secretion of a polypeptide from a host cell (e.g., a leader sequence which fimctions as a secretory sequence for controlling transport of a polypeptide from the cell). The polypeptide having a leader sequence is a preprotein and can have the leader sequence cleaved by the host cell to form the mature form of the ptide. The polynucleotides can also encode for a proprotein which is the mature protein plus additional 5 ' amino acid residues. A mature protein having a prosequence is a proprotein and is an inactive form of the protein. Once the prosequence is d an active mature protein remains.
In certain embodiments, the polynucleotides comprise the coding sequence for the mature polypeptide fused in the same reading frame to a marker sequence that allows, for example, for ation of the encoded polypeptide. For example, the marker sequence can be a hexa-histidine tag supplied by a pQE-9 vector to e for ation of the mature polypeptide fused to the marker in the case of a bacterial host, or the marker ce can be a hemagglutinin (HA) tag derived from the influenza hemagglutinin protein when a mammalian host (e.g., COS-7 cells) is used. In some embodiments, the marker sequence is a FLAG-tag, a peptide of sequence DYKDDDDK (SEQ ID NO:45) which can be used in conjunction with other affinity tags.
The present invention further relates to variants of the hereinabove described cleotides encoding, for example, fragments, analogs, and/or derivatives.
In certain embodiments, the present invention provides polynucleotides comprising polynucleotides having a nucleotide sequence at least about 80% identical, at least about 85% identical, at least about 90% identical, at least about 95% identical, and in some embodiments, at least about 96%, 97%, 98% or 99% identical to a polynucleotide encoding a polypeptide comprising a VEGF/DLL4- binding agent (e.g., an antibody), or fragment f, described herein.
As used herein, the phrase a polynucleotide having a nucleotide sequence at least, for example, 95% “identical” to a reference nucleotide ce is ed to mean that the nucleotide sequence of the polynucleotide is identical to the reference sequence except that the polynucleotide sequence can include up to five point mutations per each 100 nucleotides of the reference nucleotide sequence. In other words, to obtain a polynucleotide having a nucleotide ce at least 95% identical to a reference nucleotide sequence, up to 5% of the nucleotides in the reference sequence can be deleted or tuted with another nucleotide, or a number of nucleotides up to 5% of the total nucleotides in the reference sequence can be inserted into the nce sequence. These mutations of the reference sequence can occur at the 5' or 3' terminal positions of the nce nucleotide sequence or anywhere n those terminal positions, interspersed either individually among nucleotides in the nce ce or in one or more contiguous groups within the reference sequence.
The polynucleotide variants can contain tions in the coding regions, non-coding regions, or both. In some embodiments, a polynucleotide variant contains tions which produce silent substitutions, additions, or deletions, but does not alter the properties or activities of the encoded polypeptide. In some embodiments, a polynucleotide variant comprises silent tutions that results in no change to the amino acid sequence of the polypeptide (due to the degeneracy of the genetic code).
Polynucleotide variants can be produced for a variety of reasons, for example, to optimize codon expression for a particular host (i.e., change codons in the human mRNA to those preferred by a bacterial host such as E. coli). In some embodiments, a polynucleotide variant comprises at least one silent mutation in a non-coding or a coding region of the sequence.
In some embodiments, a polynucleotide variant is produced to modulate or alter expression (or expression levels) of the encoded polypeptide. In some embodiments, a polynucleotide variant is produced to increase expression of the encoded ptide. In some embodiments, a polynucleotide variant is produced to decrease expression of the encoded polypeptide. In some embodiments, a polynucleotide t has increased expression of the d polypeptide as compared to a parental polynucleotide sequence. In some ments, a polynucleotide variant has sed expression of the encoded polypeptide as compared to a parental polynucleotide sequence.
In some embodiments, at least one cleotide variant is produced (without changing the amino acid sequence of the encoded polypeptide) to increase production of a heteromultimeric molecule.
In some embodiments, at least one polynucleotide variant is produced (without changing the amino acid sequence of the encoded polypeptide) to increase production of a bispeciflc antibody.
In certain embodiments, the polynucleotides are ed. In n embodiments, the polynucleotides are substantially pure.
Vectors and cells comprising the polynucleotides described herein are also provided. In some embodiments, an expression vector ses a polynucleotide molecule. In some embodiments, a host cell comprises an expression vector sing the polynucleotide molecule. In some ments, a host cell ses a polynucleotide molecule.
IV. Methods of use and pharmaceutical compositions The -binding agents (including polypeptides and antibodies) of the invention that bind (e.g., specifically bind) VEGF and/or DLL4 are useful in a variety of applications including, but not d to, therapeutic treatment methods, such as the treatment of cancer. In certain embodiments, the agents are useful for inhibiting VEGF activity, ting DLL4—induced Notch signaling, inhibiting tumor growth, reducing tumor volume, reducing the frequency of cancer stem cells in a tumor, reducing the tumorigenicity of a tumor, modulating angiogenesis, and/or inhibiting enesis. The methods of use may be in vitro, ex vivo, or in vivo. In certain embodiments, a VEGF/DLL4-binding agent is an antagonist of human VEGF. In certain embodiments, a VEGF/DLL4—binding agent is an antagonist of human DLL4.
In certain embodiments, a VEGF/DLL4-binding agent is an antagonist of both VEGF and DLL4.
In certain embodiments, the VEGFXDLL4—binding agents are used in the treatment of a disease associated with angiogenesis, i.e. sed angiogenesis and/or aberrant angiogenesis. In n embodiments, the disease is a disease dependent upon angiogenesis. In certain embodiments, the VEGF/DLL4-binding agents are used in the treatment of disorders characterized by increased levels of stem cells and/or progenitor cells.
The present invention provides methods for inhibiting growth of a tumor using the VEGF/DLL4- binding agents or antibodies described herein. In certain embodiments, the method of inhibiting growth of a tumor comprises ting a tumor cell with a VEGF/DLL4-binding agent (e.g., antibody) in vilro.
For example, an immortalized cell line or a cancer cell line is cultured in medium to which is added an EGF antibody, an anti-DLL4 antibody, or an anti—VEGF/anti-DLL4 bispeciflc antibody to t tumor cell growth. In some embodiments, tumor cells are ed from a patient sample such as, for example, a tissue biopsy, pleural effusion, or blood sample and cultured in medium to which is added a VEGF/DLL4-binding agent to inhibit tumor cell growth.
In some ments, the method of inhibiting growth of a tumor comprises ting a tumor or tumor cells with a VEGF/DLL4-binding agent (e.g., dy) in vivo. In certain embodiments, contacting a tumor or tumor cell with a VEGF/DLL4—binding agent is undertaken in an animal model.
For e, an anti-VEGF antibody, an anti—DLL4 antibody, or an EGF/anti-DLL4 bispeciflc antibody may be administered to an immunocompromised host animal (e.g., NOD/SCID mice) which has a tumor xenograft. In some embodiments, tumor cells and/or cancer stem cells are isolated from a patient sample such as, for example, a tissue biopsy, pleural effusion, or blood sample and injected into an immunocompromised host animal (e.g., NOD/SCID mice) that is then administered a VEGF/DLL4- binding agent to inhibit tumor cell growth. In some embodiments, the VEGF/DLL4-binding agent is administered at the same time or shortly after introduction of tumorigenic cells into the animal to t tumor growth (“preventative ). In some embodiments, the VEGF/DLL4—binding agent is administered as a therapeutic after tumors have grown to a specified size (“therapeutic model”). In certain embodiments, the VEGF/DLL4-binding agent is a bispecific antibody that specifically binds human VEGF and human DLL4.
In certain embodiments, the method of inhibiting growth of a tumor comprises administering to a subject a therapeutically effective amount of a VEGF/DLL4—binding agent. In certain embodiments, the subject is a human. In certain ments, the t has a tumor or has had a tumor which was removed. In certain embodiments, the tumor comprises cancer stem cells. In certain embodiments, the frequency of cancer stem cells in the tumor is reduced by stration of the VEGF/DLL4-binding agent. The invention also provides a method of reducing the frequency of cancer stem cells in a tumor, comprising contacting the tumor with an effective amount of a VEGF/DLL4-binding agent (e. g., an anti- VEGF/anti-DLL4 bispecific antibody). In some embodiments, a method of reducing the frequency of cancer stem cells in a tumor in a subject, comprises administering to the subject a therapeutically effective amount of a LL4-binding agent.
In some embodiments, the tumor is a solid tumor. In certain embodiments, the tumor is a tumor selected from the group ting of colorectal tumor, colon tumor, pancreatic tumor, lung tumor, ovarian tumor, liver tumor, breast tumor, kidney tumor, prostate tumor, gastrointestinal tumor, melanoma, cervical tumor, bladder tumor, glioblastoma, and head and neck tumor. In certain ments, the tumor is a colorectal tumor or a colon tumor. In certain embodiments, the tumor is an ovarian tumor. In some ments, the tumor is a lung tumor. In certain embodiments, the tumor is a pancreatic tumor.
In certain embodiments, the tumor is a breast tumor.
The present invention further provides methods for treating cancer comprising administering a therapeutically effective amount of a VEGF/DLL4—binding agent to a subject. In some embodiments, the VEGF/DLL4-binding agent binds VEGF, and inhibits or s growth of the cancer. In some embodiments, the VEGF/DLL4-binding agent binds DLL4, and inhibits or s growth of the cancer.
In some embodiments, the VEGF/DLL4-binding agent is a bispecific antibody that binds VEGF and DLL4, and inhibits or s grth of the cancer. In some embodiments, the VEGF/DLL4-binding agent binds VEGF, interferes with VEGF/VEGF receptor interactions, and inhibits or reduces growth of the cancer. In some embodiments, the VEGF/DLL4—binding agent binds DLL4, interferes with DLL4/Notch interactions, and inhibits or reduces growth of the cancer. In some embodiments, the VEGF/DLL4-binding agent binds both VEGF and DLL4, eres with VEGF/VEGF receptor interactions and with otch interactions, and inhibits or reduces growth of the cancer. In some embodiments, the VEGF/DLL4-binding agent binds DLL4, and reduces the frequency of cancer stem cells in the cancer.
The t invention provides s of treating cancer comprising administering a therapeutically effective amount of a VEGF/DLL4—binding agent to a subject (e.g., a subject in need of treatment). In certain embodiments, the t is a human. In certain embodiments, the subject has a cancerous tumor. In certain ments, the subject has had a tumor removed.
The subject’s cancer/tumor, may, in some embodiments, be refractory to certain treatment(s). As a non-limiting example, the subj ect’s cancer (or tumor) may be efractory. In certain embodiments, the subj ect’s cancer may be resistant to anti—VEGF therapy or anti-DLL4 therapy, or both.
In certain embodiments, the cancer is a cancer selected from the group consisting of colorectal cancer, pancreatic cancer, lung cancer, ovarian cancer, liver cancer, breast cancer, kidney cancer, prostate cancer, gastrointestinal cancer, ma, cervical cancer, bladder cancer, glioblastoma, and head and neck . In certain embodiments, the cancer is ovarian cancer. In certain embodiments, the cancer is colorectal cancer or colon cancer. In n embodiments, the cancer is atic cancer. In n embodiments, the cancer is breast cancer. In certain embodiments, the cancer is prostate cancer. In certain embodiments, the cancer is lung cancer. In some embodiments, the cancer is a hematologic cancer such as leukemia or ma. In some embodiments, the leukemia or lymphoma is a B-cell leukemia or lymphoma. In some embodiments, the leukemia or lymphoma is a T—cell leukemia or lymphoma. In some embodiments the hematologic cancer is acute myelogenous leukemia, Hodgkin lymphoma, non- ns’s lymphoma, acute lymphocytic leukemia, hairy cell leukemia, chronic lymphocytic leukemia, multiple myeloma, ous T-cell lymphoma, or T-cell acute lymphoblastic ia.
The invention also provides methods of treating a disease or disorder in a subject, wherein the disease or disorder is associated with angiogenesis. In some embodiments, the e or disorder is associated with aberrant angiogenesis. In some embodiments, the disease or disorder is associated with increased angiogenesis. Thus, the present invention es methods for modulating angiogenesis in a subject, sing administering to the subject a therapeutically effective amount of any of the VEGF/DLL4-binding agents described herein. In some embodiments, the VEGF/DLL4-binding agent is an antibody that binds human VEGF. In some ments, the VEGF/DLL4-binding agent is an antibody that binds human DLL4. In some embodiments, the VEGF/DLL4-binding agent is a bispecific antibody that binds human VEGF. In some embodiments, the VEGF/DLL4-binding agent is a bispecific antibody that binds human DLL4. In some embodiments, the VEGF/DLL4-binding agent is a bispecific antibody that binds human VEGF and human DLL4.
Methods of treating a disease or disorder in a subject, wherein the disease or disorder is terized by an increased level of stem cells and/or progenitor cells are further provided. In some embodiments, the treatment methods comprise administering a therapeutically effective amount of a VEGF/DLL4-binding agent, polypeptide, or antibody to the t.
In certain ments of any of the methods described herein, the VEGF/DLL4-binding agent is a bispecific antibody that specifically binds human VEGF and human DLL4. In some embodiments, the ific antibody comprises a first antigen—binding site that specifically binds human VEGF and a second antigen-binding site that specifically binds human DLL4, wherein the first antigen-binding site comprises a heavy chain CDRl comprising NYWMH (SEQ ID NO:17), a heavy chain CDR2 comprising DINPSNGRTSYKEKFKR (SEQ ID NO:18), and a heavy chain CDR3 comprising HYDDKYYPLMDY (SEQ ID NO: 19), and the second antigen—binding site comprises a heavy chain CDRl comprising TAYYIH (SEQ ID NO: 13), a heavy chain CDR2 comprising YIANYNRATNYNQKFKG (SEQ ID NO: 14), YISSYNGATNYNQKFKG (SEQ ID NO: 15), YIAGYKDATNYNQKFKG (SEQ ID NO:59), or YISNYNRATNYNQKFKG (SEQ ID NO:65), and a heavy chain CDR3 comprising RDYDYDVGMDY (SEQ ID NO: 16); and wherein both the first and second antigen-binding sites comprise a light chain CDRl comprising RASESVDNYGISFMK (SEQ ID NO:20), a light chain CDR2 sing AASNQGS (SEQ ID N021), and a light chain CDR3 comprising QQSKEVPWTFGG (SEQ ID NO:22). In some embodiments, the ific antibody ses a first antigen—binding site that specifically binds human VEGF and a second antigen-binding site that specifically binds human DLL4, wherein the first antigen- binding site ses a heavy chain CDRl comprising NYWMH (SEQ ID NO:17), a heavy chain CDR2 comprising DINPSNGRTSYKEKFKR (SEQ ID NO: 18), and a heavy chain CDR3 comprising HYDDKYYPLMDY (SEQ ID NO:19), and the second antigen-binding site comprises a heavy chain CDRl comprising TAYYIH (SEQ ID NO:13), a heavy chain CDR2 comprising YIANYNRATNYNQKFKG (SEQ ID , and a heavy chain CDR3 comprising VGMDY (SEQ ID NO: 16); and wherein both the first and second antigen—binding sites comprise a light chain CDRl comprising RASESVDNYGISFMK (SEQ ID NO:20), a light chain CDR2 comprising AASNQGS (SEQ ID N021), and a light chain CDR3 comprising QQSKEVPWTFGG (SEQ ID NO:22). In some ments, the bispecific antibody comprises a first antigen—binding site that specifically binds human VEGF and a second antigen-binding site that specifically binds human DLL4, wherein the first antigen- binding site comprises a heavy chain CDRl comprising NYWMH (SEQ ID NO: 17), a heavy chain CDR2 comprising DINPSNGRTSYKEKFKR (SEQ ID NO:18), and a heavy chain CDR3 sing HYDDKYYPLMDY (SEQ ID NO: 19), and the second antigen-binding site comprises a heavy chain CDRl comprising TAYYIH (SEQ ID NO:13), a heavy chain CDR2 comprising YISSYNGATNYNQKFKG (SEQ ID , and a heavy chain CDR3 comprising RDYDYDVGMDY (SEQ ID NO: 16); and wherein both the first and second antigen-binding sites comprise a light chain CDRl comprising RASESVDNYGISFMK (SEQ ID NO:20), a light chain CDR2 comprising AASNQGS (SEQ ID N021), and a light chain CDR3 comprising QQSKEVPWTFGG (SEQ ID NO:22). In some ments, the bispecific antibody comprises a first antigen-binding site that specifically binds human VEGF and a second antigen-binding site that specifically binds human DLL4, wherein the first antigen- binding site comprises a heavy chain CDRl comprising NYWMH (SEQ ID NO:17), a heavy chain CDR2 comprising DINPSNGRTSYKEKFKR (SEQ ID NO: 18), and a heavy chain CDR3 comprising HYDDKYYPLMDY (SEQ ID NO:19), and second antigen—binding site which comprises a heavy chain CDRl comprising TAYYIH (SEQ ID NO: 13), a heavy chain CDR2 comprising YIAGYKDATNYNQKFKG (SEQ ID NO:59), and a heavy chain CDR3 comprising RDYDYDVGMDY (SEQ ID NO: 16); and wherein both the first and second antigen-binding sites comprise a light chain CDRl comprising RASESVDNYGISFMK (SEQ ID NO:20), a light chain CDR2 comprising AASNQGS (SEQ ID N021), and a light chain CDR3 comprising QQSKEVPWTFGG (SEQ ID NO:22). In some embodiments, the bispecific dy comprises a first antigen—binding site that specifically binds human VEGF and a second antigen-binding site that cally binds human DLL4, wherein the first antigen- binding site comprises a heavy chain CDRl sing NYWMH (SEQ ID NO: 17), a heavy chain CDR2 comprising DINPSNGRTSYKEKFKR (SEQ ID NO: 18), and a heavy chain CDR3 comprising HYDDKYYPLMDY (SEQ ID NO: 19), and the second antigen-binding site comprises a heavy chain CDRl comprising TAYYIH (SEQ ID NO:13), a heavy chain CDR2 comprising YISNYNRATNYNQKFKG (SEQ ID NO:65), and a heavy chain CDR3 comprising RDYDYDVGMDY (SEQ ID NO: 16); and wherein both the first and second n—binding sites comprise a light chain CDRl comprising RASESVDNYGISFMK (SEQ ID NO:20), a light chain CDR2 comprising AASNQGS (SEQ ID NO:21), and a light chain CDR3 comprising QQSKEVPWTFGG (SEQ ID .
In certain embodiments of any of the methods described herein, the VEGF/DLL4 bispecific antibody comprises a first heavy chain variable region having at least about 80% sequence identity to SEQ ID NO:11, a second heavy chain variable region having at least about 80% sequence identity to SEQ ID N019, SEQ ID NO:10, SEQ ID NO:58, or SEQ ID N0264, and a first and a light chain variable region having at least 80% sequence identity to SEQ ID NO: 12. In some embodiments, the VEGF/DLL4 bispecific antibody comprises a first heavy chain variable region having at least about 80% sequence identity to SEQ ID NO:11, a second heavy chain variable region having at least about 80% sequence identity to SEQ ID N019, and a first and a second light chain variable region having at least 80% sequence identity to SEQ ID NO:12. In some embodiments, the VEGF/DLL4 bispecific antibody comprises a first heavy chain variable region having at least about 80% sequence identity to SEQ ID NO:11, a second heavy chain variable region having at least about 80% sequence identity to SEQ ID NO: 10, and a first and a second light chain variable region having at least 80% sequence identity to SEQ ID NO: 12. In some embodiments, the VEGF/DLL4 bispecific antibody comprises a first heavy chain le region having at least about 80% sequence ty to SEQ ID NO:11, a second heavy chain le region having at least about 80% sequence ty to SEQ ID NO:58, and a first and a second light chain variable region having at least 80% sequence identity to SEQ ID NO:12. In some embodiments, the VEGF/DLL4 bispecific dy comprises a first heavy chain variable region having at least about 80% sequence identity to SEQ ID NO:11, a second heavy chain variable region having at least about 80% sequence identity to SEQ ID NO:64, and a first and a second light chain le region having at least 80% sequence ty to SEQ ID NO:12.
In some embodiments of any of the methods bed herein, the VEGF/DLL4-binding agent is an antibody. In some embodiments, the LL4-binding agent is an anti-VEGF antibody. In some embodiments, the anti-VEGF antibody is antibody . In some embodiments, the VEGF/DLL4- binding agent is an anti—DLL4 antibody. In some embodiments, the anti-DLL4 antibody is antibody 21R79. In some embodiments, the LL4 antibody is antibody 21R75. In some embodiments, the anti-DLL4 antibody is antibody 21R83. In some embodiments, the VEGF/DLL4-binding agent is a bispecific antibody comprising an antigen—binding site from antibody 219R45. In some embodiments, the VEGF/DLL4-binding agent is a bispecific antibody comprising an antigen-binding site from dy 21R79. In some embodiments, the VEGF/DLL4—binding agent is a bispecific dy comprising an antigen-binding site from antibody 21R75. In some embodiments, the VEGF/DLL4-binding agent is a bispecific antibody comprising an n—binding site from antibody 21R83. In some embodiments, the VEGF/DLL4-binding agent is a ific antibody comprising a first antigen-binding site from antibody 219R45 and a second antigen-binding site from antibody 21R?9. In some embodiments, the VEGF/DLL4-binding agent is a bispecific antibody comprising a first antigen—binding site from antibody 219R45 and a second antigen-binding site from dy 21M18. In some embodiments, the LL4-binding agent is a bispecific antibody comprising a first antigen-binding site from antibody 219R45 and a second antigen-binding site from antibody 21R75. In some ments, the LL4-binding agent is a bispecific antibody comprising a first antigen-binding site from antibody 219R45 and a second antigen—binding site from antibody 21R83. In some embodiments, the VEGF/DLL4-binding agent is the bispecific antibody 219R45—MB-21M18. In some embodiments, the VEGF/DLL4-binding agent is the bispecific antibody 219R45—MB-21R79. In some ments, the VEGF/DLL4-binding agent is the bispecific antibody 219R45—MB-21R75. In some embodiments, the VEGF/DLL4-binding agent is the bispecific antibody 219R45-MB-21R83.
The present invention further provides pharmaceutical compositions comprising the binding agents described herein. In certain embodiments, the pharmaceutical compositions further comprise a pharmaceutically acceptable vehicle. These pharmaceutical compositions find use in inhibiting tumor growth and/or treating cancer in a subject (e.g., a human patient).
In certain embodiments, the invention provides pharmaceutical compositions comprising bispecific antibodies, wherein at least about 90%, at least about 95%, at least about 98%, at least about 99% of the antibodies in the ition are bispecific antibodies or heterodimeric antibodies. In certain embodiments, the bispecific antibodies are IgG (e.g., IgG2 or IgG1) dies. In certain ments, less than about 10%, less than about 5%, less than about 2% or less than about 1% of the total antibodies in the compositions are monospecific antibodies or homodimeric antibodies. In certain embodiments, the antibodies in the composition are at least about 98% heterodimeric.
In n embodiments, formulations are prepared for storage and use by combining a purified dy or agent of the present invention with a pharmaceutically acceptable vehicle (e.g., a carrier or excipient). Suitable pharmaceutically acceptable vehicles include, but are not limited to, non-toxic buffers such as phosphate, citrate, and other organic acids; salts such as sodium chloride; antioxidants ing ic acid and methionine; preservatives such as octadecyldimethylbenzyl ammonium chloride, hexamethonium de, benzalkonium chloride, benzethonium chloride, , butyl or benzyl alcohol, alkyl parabens, such as methyl or propyl paraben, catechol, resorcinol, cyclohexanol, 3-pentanol, and m- cresol; low molecular weight polypeptides (e.g., less than about 10 amino acid residues); proteins such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, ine, asparagine, histidine, arginine, or lysine; carbohydrates such as monosaccharides, disaccharides, glucose, mannose, or dextrins; chelating agents such as EDTA; sugars such as sucrose, mannitol, trehalose or sorbitol; salt-forming counter-ions such as sodium; metal complexes such as Zn-protein complexes; and non-ionic surfactants such as TWEEN or polyethylene glycol (PEG). (Remington: The Science and Practice ofPharmacy, 21st n, 2005, sity of the Sciences, Philadelphia, PA).
The pharmaceutical compositions of the present invention can be administered in any number of ways for either local or systemic ent. Administration can be topical by epidermal or transdermal patches, nts, s, creams, gels, drops, suppositories, sprays, liquids, and powders; pulmonary by inhalation or insufflation of s or aerosols, ing by nebulizer, intratracheal, and intranasal; oral; or parenteral including intravenous, intraarterial, intratumoral, subcutaneous, intraperitoneal, intramuscular (e. g., injection or on), or intracranial (e.g., intrathecal or intraventricular).
The eutic formulation can be in unit dosage form. Such formulations include s, pills, capsules, powders, granules, solutions or suspensions in water or non-aqueous media, or suppositories. In solid compositions such as tablets the principal active ingredient is mixed with a pharmaceutical carrier.
Conventional tableting ingredients include corn starch, lactose, sucrose, sorbitol, talc, stearic acid, magnesium stearate, ium phosphate or gums, and diluents (e.g., water). These can be used to form a solid preformulation composition containing a neous mixture of a compound of the present invention, or a non-toxic pharmaceutically acceptable salt thereof. The solid preformulation composition is then subdivided into unit dosage forms of a type described above. The tablets, pills, etc. of the ation or composition can be coated or otherwise compounded to provide a dosage form affording the advantage of prolonged action. For example, the tablet or pill can comprise an inner composition covered by an outer component. Furthermore, the two components can be separated by an enteric layer that serves to resist disintegration and permits the inner component to pass intact through the h or to be delayed in release. A variety of materials can be used for such enteric layers or gs, such materials include a number of polymeric acids and mixtures of polymeric acids with such materials as shellac, cetyl alcohol and cellulose acetate.
The VEGF/DLL4-binding agents or antibodies described herein can also be entrapped in microcapsules. Such apsules are prepared, for example, by coacervation techniques or by interfacial polymerization, for example, hydroxymethylcellulose or gelatin-microcapsules and poly- (methylmethacylate) microcapsules, respectively, in colloidal drug delivery systems (for e, liposomes, n microspheres, microemulsions, rticles and nanocapsules) or in macroemulsions as described in Remington: The Science and Practice ofPharmacy, 21st Edition, 2005, University of the Sciences in Philadelphia, PA.
In certain embodiments, pharmaceutical formulations include a VEGF/DLL4-binding agent (e.g., an antibody) of the present invention complexed with liposomes. Methods to produce liposomes are known to those of skill in the art. For example, some liposomes can be generated by reverse phase evaporation with a lipid composition comprising phosphatidylcholine, cholesterol, and PEG-derivatized phosphatidylethanolamine (PEG-PE). Liposomes can be extruded through filters of defined pore size to yield liposomes with the d diameter.
In certain embodiments, sustained—release preparations can be ed. Suitable examples of sustained-release ations e semi-permeable matrices of solid hydrophobic polymers containing a LL4-binding agent (e.g., an antibody), where the es are in the form of shaped articles (e.g., films or microcapsules). Additional examples of sustained-release matrices include polyesters, hydrogels such as poly(2—hydroxyethyl—methacrylate) or poly(vinyl alcohol), polylactides, copolymers of L-glutamic acid and 7 ethyl—L—glutamate, non—degradable ethylene-vinyl acetate, degradable lactic acid- ic acid copolymers such as the LUPRON DEPOTTM (injectable microspheres composed of lactic lycolic acid copolymer and leuprolide e), sucrose e isobutyrate, and poly-D-(-) hydroxybutyric acid.
In certain embodiments, in addition to administering a VEGF/DLL4-binding agent (e.g., an antibody), the method or treatment further comprises administering at least one additional therapeutic agent. An additional therapeutic agent can be stered prior to, concurrently with, and/or subsequently to, administration of the VEGF/DLL4—binding agent. Pharmaceutical compositions comprising a VEGF/DLL4-binding agent and the additional therapeutic agent(s) are also ed. In some embodiments, the at least one additional therapeutic agent comprises 1, 2, 3, or more additional therapeutic agents.
Combination therapy with at least two therapeutic agents often uses agents that work by different mechanisms of action, although this is not required. Combination therapy using agents with different mechanisms of action may result in additive or synergetic effects. Combination therapy may allow for a lower dose of each agent than is used in erapy, thereby reducing toxic side effects and/or increasing the therapeutic index of at least one of the agents. Combination therapy may decrease the likelihood that resistant cancer cells will develop. In some embodiments, combination therapy comprises a therapeutic agent that primarily affects (e.g., inhibits or kills) morigenic cells and a therapeutic agent that primarily affects (e.g., inhibits or kills) tumorigenic CSCs.
Useful classes of therapeutic agents include, for e, bulin agents, atins, DNA minor groove binders, DNA replication inhibitors, alkylating agents (e.g., platinum complexes such as cisplatin, mono(platinum), bis(platinum) and tri—nuclear platinum xes and carboplatin), cyclines, antibiotics, antifolates, antimetabolites, chemotherapy sensitizers, duocarmycins, etoposides, fluorinated pyrimidines, ionophores, lexitropsins, nitrosoureas, platinols, purine antimetabolites, puromycins, ion sensitizers, steroids, taxanes, topoisomerase inhibitors, vinca alkaloids, or the like. In certain embodiments, the second therapeutic agent is an alkylating agent, an antimetabolite, an totic, a topoisomerase inhibitor, or an angiogenesis inhibitor. In some embodiments, the second eutic agent is a platinum x such as carboplatin or cisplatin. In some embodiments, the additional therapeutic agent is a platinum complex in combination with a .
Therapeutic agents that may be administered in combination with the VEGF/DLL4-binding agents include chemotherapeutic agents. Thus, in some embodiments, the method or treatment involves the administration of an anti-VEGF-binding agent or antibody of the present invention in ation with a chemotherapeutic agent or cocktail of multiple different herapeutic agents. In some embodiments, the method or treatment involves the administration of an anti-DLL4-binding agent or antibody of the t invention in ation with a herapeutic agent or cocktail of multiple different chemotherapeutic agents. In some embodiments, the method or treatment involves the administration of a bispecific antibody of the present invention that binds VEGF and DLL4 in combination with a chemotherapeutic agent or cocktail of multiple different chemotherapeutic .
Chemotherapeutic agents useful in the instant invention include, but are not limited to, alkylating agents such as thiotepa and cyclophosphamide (CYTOXAN); alkyl sulfonates such as an, improsulfan and piposulfan; aziridines such as benzodopa, carboquone, meturedopa, and uredopa; ethylenimines and methylamelamines including altretamine, triethylenemelamine, trietylenephosphoramide, triethylenethiophosphaoramide and trimethylolomelamime; en mustards such as chlorambucil, chlomaphazine, cholophosphamide, estramustine, ifosfamide, mechlorethamine, mechlorethamine oxide hydrochloride, melphalan, novembichin, phenesterine, prednimustine, trofosfamide, uracil mustard; ureas such as carmustine, chlorozotocin, fotemustine, lomustine, nimustine, ranimustine; antibiotics such as aclacinomysins, actinomycin, authramycin, ine, bleomycins, cactinomycin, calicheamicin, carabicin, caminomycin, carzinophilin, chromomycins, dactinomycin, daunorubicin, bicin, 6—diazo—5—oxo—L—norleucine, doxorubicin, epirubicin, esorubicin, idarubicin, marcellomycin, mitomycins, enolic acid, nogalamycin, olivomycins, ycin, potfiromycin, puromycin, quelamycin, rodorubicin, streptonigrin, streptozocin, tubercidin, ubenimex, atin, zorubicin; etabolites such as methotrexate and 5—fluorouracil (S-FU); folic acid analogues such as denopterin, methotrexate, pteropterin, trimetrexate; purine analogs such as fludarabine, 6-mercaptopurine, thiamiprine, thioguanine; pyrimidine analogs such as bine, azacitidine, 6-azauridine, carrnofur, cytosine arabinoside, dideoxyuridine, doxifluridine, enocitabine, floxuridine, S-FU; androgens such as calusterone, dromostanolone propionate, epitiostanol, mepitiostane, testolactone; anti-adrenals such as aminoglutethimide, mitotane, trilostane; folic acid replenishers such as folinic acid; aceglatone; aldophosphamide glycoside; aminolevulinic acid; amsacrine; bestrabucil; bisantrene; xate; defofamine; demecolcine; diaziquone; elformithine; elliptinium acetate; etoglucid; gallium nitrate; hydroxyurea; lentinan; mine; mitoguazone; mitoxantrone; mopidamol; nitracrine; pentostatin; phenamet; pirarubicin; podophyllinic acid; 2-ethylhydrazide; procarbazine; PSK; razoxane; ran; spirogermanium; tenuazonic acid; triaziquone; 2,2',2"-trichlorotriethylamine; urethan; vindesine; dacarbazine; mannomustine; mitobronitol; mitolactol; pipobroman; gacytosine; arabinoside (Ara-C); taxoids, e. g. paclitaxel (TAXOL) and docetaxel (TAXOTERE); chlorambucil; gemcitabine; 6- thioguanine; mercaptopurine; platinum analogs such as cisplatin and carboplatin; vinblastine; platinum; ide ); ifosfamide; mitomycin C; mitoxantrone; vincristine; vinorelbine; navelbine; novantrone; teniposide; daunomycin; aminopterin; ibandronate; CPTl l; topoisomerase inhibitor RFS 2000; difluoromethylornithine (DMFO); retinoic acid; esperamicins; capecitabine (XELODA); and pharmaceutically acceptable salts, acids or derivatives of any of the above. Chemotherapeutic agents also include anti-hormonal agents that act to regulate or t hormone action on tumors such as anti- estrogens including, for example, tamoxifen, raloxifene, aromatase inhibiting 4(5)-imidazoles, 4- hydroxytamoxifen, trioxifene, keoxifene, LYl 17018, onapristone, and toremifene (FARESTON); and anti-androgens such as flutamide, mide, bicalutamide, leuprolide, and goserelin; and pharmaceutically acceptable salts, acids or derivatives of any of the above. In certain embodiments, the second therapeutic agent is tin. In certain embodiments, the second therapeutic agent is carboplatin.
In n embodiments, the second therapeutic agent is paclitaxel.
In certain embodiments, the chemotherapeutic agent is a topoisomerase inhibitor. omerase inhibitors are chemotherapeutic agents that ere with the action of a topoisomerase enzyme (e.g., topoisomerase I or II). Topoisomerase inhibitors include, but are not limited to, bicin HCl, daunorubicin citrate, mitoxantrone HCl, actinomycin D, etoposide, can HCl, teniposide (VM-26), and irinotecan, as well as pharmaceutically acceptable salts, acids, or derivatives of any of these. In n embodiments, the second eutic agent is irinotecan.
In certain embodiments, the chemotherapeutic agent is an anti-metabolite. An anti-metabolite is a chemical with a structure that is similar to a metabolite required for normal biochemical reactions, yet different enough to interfere with one or more normal fimctions of cells, such as cell division. Anti- metabolites include, but are not limited to, gemcitabine, fluorouracil, tabine, methotrexate sodium, ralitrexed, pemetrexed, tegafur, cytosine arabinoside, thioguanine, S—azacytidine, aptopu1ine, azathioprine, 6-thioguanine, tatin, fludarabine phosphate, and cladribine, as well as pharmaceutically acceptable salts, acids, or derivatives of any of these. In certain embodiments, the second eutic agent is gemcitabine.
In certain embodiments, the chemotherapeutic agent is an antimitotic agent, including, but not d to, agents that bind tubulin. In some embodiments, the agent is a . In certain embodiments, the agent is paclitaxel or docetaxel, or a pharmaceutically acceptable salt, acid, or derivative of paclitaxel or docetaxel. In certain embodiments, the agent is paclitaxel (TAXOL), docetaxel (TAXOTERE), albumin-bound paclitaxel (ABRAXANE), DHA—paclitaxel, or PG-paclitaxel. In certain alternative embodiments, the antimitotic agent comprises a vinca alkaloid, such as stine, binblastine, vinorelbine, or vindesine, or pharmaceutically acceptable salts, acids, or derivatives f. In some embodiments, the antimitotic agent is an inhibitor of kinesin EgS or an inhibitor of a mitotic kinase such as Aurora A or Plkl. In certain embodiments, where the chemotherapeutic agent administered in combination with a VEGF/DLL4-binding agent is an anti—mitotic agent, the cancer or tumor being treated is breast cancer or a breast tumor.
In some embodiments, a second therapeutic agent comprises an agent such as a small le.
For example, treatment can involve the combined stration of a VEGF/DLL4-binding agent (e.g. an antibody) of the t ion with a small molecule that acts as an inhibitor against additional tumor- associated ns including, but not limited to, EGFR, ErbB2, HERZ, and/or VEGF. In certain embodiments, the second therapeutic agent is a small molecule that inhibits a cancer stem cell pathway.
In some embodiments, the second eutic agent is a small molecule inhibitor of the Notch pathway.
In some embodiments, the second therapeutic agent is a small molecule inhibitor of the Wnt pathway. In some embodiments, the second therapeutic agent is a small molecule inhibitor of the BMP y. In some embodiments, the second eutic agent is a small molecule that inhibits B-catenin signaling.
In some embodiments, a second therapeutic agent comprises a biological molecule, such as an antibody. For example, treatment can involve the combined administration of a VEGF/DLL4-binding agent (e. g. an antibody) of the present invention with other antibodies against additional tumor-associated proteins including, but not limited to, antibodies that bind EGFR, ErbB2, HERZ, and/or VEGF. In certain embodiments, the second therapeutic agent is an antibody that is an anti-cancer stem cell marker antibody.
In some embodiments, the second therapeutic agent is an dy that binds a component of the Notch pathway. In some ments, the second therapeutic agent is an antibody that binds a ent of the Wnt pathway. In certain embodiments, the second therapeutic agent is an antibody that ts a cancer stem cell pathway. In some embodiments, the second therapeutic agent is an antibody tor of the Notch pathway. In some embodiments, the second therapeutic agent is an antibody inhibitor of the Wnt pathway. In some embodiments, the second therapeutic agent is an antibody inhibitor of the BMP pathway. In some embodiments, the second therapeutic agent is an antibody that inhibits B-catenin signaling. In certain embodiments, the second eutic agent is an antibody that is an angiogenesis inhibitor or modulator (e.g., an anti-VEGF or VEGF receptor antibody). In certain ments, the second therapeutic agent is bevacizumab (AVASTIN), trastuzumab (HERCEPTIN), panitumumab (VECTIBIX), or cetuximab (ERBITUX). Combined administration can include co-administration, either in a single pharmaceutical formulation or using separate formulations, or consecutive stration in either order but generally within a time period such that all active agents can exert their biological activities simultaneously.
Furthermore, treatment with a VEGF/DLL4—binding agent described herein can include combination ent with other biologic molecules, such as one or more cytokines (e.g., lymphokines, eukins, tumor necrosis factors, and/0r growth factors) or can be accompanied by al removal of tumors, cancer cells, or any other therapy deemed ary by a ng physician.
In certain embodiments, the treatment involves the administration of a VEGF/DLL4-binding agent (e. g. an antibody) of the present invention in combination with ion therapy. Treatment with a VEGF/DLL4-binding agent can occur prior to, rently with, or subsequent to administration of radiation therapy. Dosing schedules for such radiation therapy can be determined by the d medical practitioner.
It will be appreciated that the combination of a VEGF/DLL4-binding agent and an additional therapeutic agent may be administered in any order or rently. Treatment with a VEGF/DLL4- binding agent (e.g., an antibody) can occur prior to, concurrently with, or subsequent to administration of chemotherapies. ed administration can include co—administration, either in a single pharmaceutical formulation or using separate formulations, or consecutive administration in either order but lly within a time period such that all active agents can exert their biological activities simultaneously. Preparation and dosing schedules for such chemotherapeutic agents can be used according to manufacturers' instructions or as determined empirically by the skilled practitioner.
Preparation and dosing schedules for such chemotherapy are also bed in The Chemotherapy Source Book, 4’17 Edition, 2008, M. C. Perry, Editor, Lippincott, Williams & Wilkins, Philadelphia, PA.
In some embodiments, the VEGF/DLL4—binding agent will be administered to patients that have previously undergone treatment with a second therapeutic agent. In certain other embodiments, the VEGF/DLL4-binding agent and a second therapeutic agent will be administered substantially simultaneously or concurrently. For example, a subject may be given a VEGF/DLL4-binding agent (e.g., an antibody) while undergoing a course oftreatment with a second therapeutic agent (e.g., chemotherapy).
In certain embodiments, a VEGF/DLL4—binding agent will be administered within 1 year of the treatment with a second therapeutic agent. In certain alternative embodiments, a LL4-binding agent will be administered within 10, 8, 6, 4, or 2 months of any treatment with a second therapeutic agent. In certain other embodiments, a VEGF/DLL4-binding agent will be administered within 4, 3, 2, or 1 weeks of any treatment with a second therapeutic agent. In some embodiments, a VEGF/DLL4-binding agent will be administered within 5, 4, 3, 2, or 1 days of any ent with a second therapeutic agent. It will further be appreciated that the two (or more) agents or treatments may be stered to the subject within a matter of hours or minutes (i.e., substantially simultaneously).
For the treatment of a disease, the appropriate dosage of an VEGF/DLL4-binding agent (e.g., an antibody) of the present invention depends on the type of disease to be treated, the severity and course of the disease, the responsiveness of the disease, whether the LL4-binding agent or antibody is administered for eutic or preventative purposes, previous therapy, the patient’s clinical history, and so on, all at the discretion of the treating physician. The VEGF/DLL4-binding agent or antibody can be administered one time or as a series of treatments spread over several days to several months, or until a cure is effected or a diminution of the e state is ed (e.g., reduction in tumor size). Optimal dosing schedules can be calculated from measurements of drug accumulation in the body of the patient and will vary depending on the relative potency of an individual antibody or agent. The stering physician can determine optimum dosages, dosing methodologies, and repetition rates. In certain ments, dosage of a VEGF/DLL4-binding agent or antibody is from about 0.01ug to about 100mg/kg of body weight, from about 0.1ug to about 100mg/kg of body weight, from about lug to about 100mg/kg of body weight, from about 1mg to about 100mg/kg of body weight, about 1mg to about 80mg/kg of body weight from about 10mg to about lOOmg/kg of body weight, from about 10mg to about 75mg/kg of body weight, or from about 10mg to about 50mg/kg ofbody . In n embodiments, the dosage of the antibody or other VEGF/DLL4—binding agent is from about 0.1mg to about 20mg/kg of body weight. In certain embodiments, dosage can be given once or more daily, weekly, monthly, or yearly. In certain embodiments, the antibody or other VEGF/DLL4-binding agent is given once every week, once every two weeks, once every three weeks, or once every month.
In some embodiments, a LL4-binding agent (e.g., an antibody) may be administered at an initial higher “loading” dose, ed by one or more lower doses. In some embodiments, the frequency of administration may also change. In some embodiments, a dosing regimen may comprise administering an initial dose, followed by additional doses (or enance” doses) once a week, once every two weeks, once every three weeks, or once every month. For example, a dosing regimen may comprise administering an initial loading dose, followed by a weekly maintenance dose of, for example, one-half of the l dose. Or a dosing regimen may comprise administering an initial loading dose, followed by maintenance doses of, for e one-half of the l dose every other week. Or a dosing regimen may comprise administering three initial doses for 3 weeks, followed by maintenance doses of, for e, the same amount every other week. Or a dosing regimen may comprise administering an initial dose followed by additional doses every 3 weeks or once a month. The treating physician can estimate repetition rates for dosing based on measured nce times and concentrations of the drug in bodily fluids or tissues. The progress of therapy can be monitored by conventional techniques and assays.
As is known to those of skill in the art, administration of any therapeutic agent may lead to side effects and/or toxicities. In some cases, the side effects and/or toxicities are so severe as to preclude stration of the particular agent at a therapeutically effective dose. In some cases, drug therapy must be tinued, and other agents may be tried. However, many agents in the same therapeutic class often display similar side effects and/or toxicities, meaning that the patient either has to stop therapy, or if possible, suffer from the unpleasant side effects associated with the therapeutic agent.
Side effects from therapeutic agents may include, but are not limited to, hives, skin rashes, g, nausea, vomiting, decreased appetite, diarrhea, chills, fever, fatigue, muscle aches and pain, headaches, low blood pressure, high blood pressure, hypokalemia, low blood counts, bleeding, and cardiac problems.
Thus, one aspect of the present invention is directed to methods of treating cancer in a patient comprising administering an anti-VEGFfanti—DLL4 bispecific antibody using an intermittent dosing regimen, which may reduce side effects andfor toxicities associated with administration of the anti- VEGF/anti-DLL4 bispecific antibody. As used herein, mittent dosing” refers to a dosing regimen using a dosing interval of more than once a week, e.g., dosing once every 2 weeks, once every 3 weeks, once every 4 weeks, etc. In some ments, a method for treating cancer in a human patient comprises stering to the patient an effective dose of an anti-VEGF/anti-DLL4 bispecific antibody according to an intermittent dosing regimen. In some embodiments, a method for treating cancer in a human patient comprises administering to the patient an ive dose of an anti-VEGF/anti-DLL4 bispecific antibody according to an intermittent dosing regimen, and increasing the therapeutic index of the anti-VEGF/anti-DLL4 bispecific antibody. In some embodiments, the intermittent dosing regimen comprises stering an initial dose of an anti—VEGF/anti—DLL4 ific antibody to the patient, and administering subsequent doses of the anti-VEGF/anti—DLL4 bispecific antibody about once every 2 weeks. In some embodiments, the intermittent dosing regimen comprises administering an initial dose of an anti-VEGF/anti-DLL4 bispecific antibody to the t, and stering subsequent doses of the anti-VEGF/anti-DLL4 bispecific antibody about once every 3 weeks. In some embodiments, the ittent dosing regimen comprises administering an initial dose of an anti-VEGF/anti-DLL4 if1c antibody to the patient, and administering subsequent doses of the EGF/anti-DLL4 bispeciflc antibody about once every 4 weeks.
In some embodiments, the subsequent doses in an intermittent dosing regimen are about the same amount or less than the initial dose. In other embodiments, the subsequent doses are a greater amount than the initial dose. As is known by those of skill in the art, doses used will vary depending on the clinical goals to be achieved. In some embodiments, the initial dose is about 0.25mg/kg to about g. In some embodiments, the initial dose is about 0.5, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20mg/kg. In certain embodiments, the initial dose is about 0.5mg/kg. In certain embodiments, the initial dose is about 1mg/kg. In certain embodiments, the initial dose is about 2.5mg/kg. In certain embodiments, the initial dose is about 5mg/kg. In n embodiments, the initial dose is about 7.5mg/kg. In certain embodiments, the initial dose is about g. In certain embodiments, the l dose is about 12.5mg/kg. In certain embodiments, the initial dose is about 15mg/kg. In certain embodiments, the l dose is about 20mg/kg. In some embodiments, the subsequent doses are about 0.25mg/kg to about 15mg/kg. In certain embodiments, the subsequent doses are about 0.5, l, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14 or 15mg/kg. In certain embodiments, the subsequent doses are about 0.5mg/kg. In certain embodiments, the uent doses are about 1mg/kg.
In certain embodiments, the subsequent doses are about kg. In certain embodiments, the subsequent doses are about 5mg/kg. In some embodiments, the subsequent doses are about 7.5mg/kg. In some embodiments, the subsequent doses are about lOmg/kg. In some embodiments, the subsequent doses are about 12.5mg/kg.
In some embodiments, the intermittent dosing regimen comprises: (a) administering to the patient an initial dose of an anti-VEGF/anti-DLL4 bispecific antibody of about 2.5mg/kg and (b) stering subsequent doses of about 2.5 mg/kg once every 2 weeks. In some ments, the intermittent dosing regimen comprises: (a) administering to the patient an l dose of an anti-VEGF/anti-DLL4 bispecific antibody of about 5mg/kg and (b) administering subsequent doses of about 5 mg/kg once every 2 weeks.
In some embodiments, the intermittent dosing regimen comprises: (a) administering to the patient an initial dose of an anti-VEGF/anti—DLL4 ific dy of about 2.5mg/kg and (b) administering subsequent doses of about 2.5 mg/kg once every 3 weeks. In some embodiments, the intermittent dosing regimen comprises: (a) administering to the t an initial dose of an EGF/anti-DLL4 bispecifrc antibody of about 5mg/kg and (b) administering subsequent doses of about 5 mg/kg once every 3 weeks.
In some embodiments, the intermittent dosing regimen comprises: (a) administering to the patient an l dose of an anti-VEGF/anti-DLL4 bispecific antibody of about 2.5mg/kg and (b) administering subsequent doses of about 2.5 mg/kg once every 4 weeks. In some embodiments, the intermittent dosing regimen comprises: (a) stering to the patient an initial dose of an anti-VEGF/anti-DLL4 bispecifrc antibody of about 5mg/kg and (b) administering subsequent doses of about 5 mg/kg once every 4 weeks.
In certain embodiments, the initial dose and the maintenance doses are different, for example, the initial dose is about 5mg/kg and the subsequent doses are about 2.5mg/kg. In certain embodiments, an intermittent dosing regimen may comprise a g dose, for example, the initial dose is about 20mg/kg and the subsequent doses are about 2.5mgfkg or about 5mg/kg administered once every 2 weeks, once every 3 weeks, or once every 4 weeks.
Another aspect of the present invention is directed to methods for reducing toxicity of an anti- VEGF/anti-DLL4 bispecific antibody in a human patient comprises administering to the t the anti- nti-DLL4 bispecifrc antibody using an intermittent dosing regimen. Another aspect of the present invention is directed to methods for reducing side effects of an anti-VEGF/anti-DLL4 bispecific antibody in a human patient comprises administering to the patient the anti-VEGF/anti-DLL4 bispecific antibody using an intermittent dosing regimen. Another aspect of the t invention is directed to methods for increasing the therapeutic index of an EGF/anti—DLL4 bispecifrc antibody in a human patient comprises administering to the patient the anti—VEGF/anti—DLL4 ific antibody using an intermittent dosing regimen.
The choice of delivery method for the initial and subsequent doses is made ing to the ability of the animal or human patient to tolerate introduction of the anti-VEGF/anti-DLL4 bispecific antibody into the body. Thus, in any ofthe s and/or embodiments described herein, the administration of the anti-VEGF/anti-DLL4 bispecific antibody may be by intravenous injection or enously. In some embodiments, the administration is by intravenous infusion. In any of the aspects and/or embodiments described herein, the administration of the anti—VEGF/anti-DLL4 bispecific antibody may be by a non-intravenous route.
V. Kits comprising VEGF/DLL4-binding agents The present invention provides kits that comprise the VEGF/DLL4-binding agents (e.g., antibodies) described herein and that can be used to perform the methods described herein. In certain embodiments, a kit ses at least one purified antibody against VEGF and/or DLL4 in one or more containers. In some embodiments, the kits contain all of the components necessary and/or sufficient to perform a detection assay, ing all controls, ions for performing assays, and any necessary software for analysis and tation of results. One skilled in the art will readily recognize that the disclosed VEGF/DLL4-binding agents of the present invention can be readily incorporated into one of the established kit formats which are well known in the art.
Further provided are kits comprising a VEGF/DLL4-binding agent (e.g., an anti-VEGF/anti- DLL4 bispecific antibody), as well as at least one onal therapeutic agent. In certain embodiments, the second (or more) therapeutic agent is a chemotherapeutic agent. In certain embodiments, the second (or more) therapeutic agent is an angiogenesis inhibitor.
Embodiments of the present disclosure can be further defined by reference to the following non- limiting examples, which describe in detail preparation of certain antibodies of the t sure and s for using antibodies of the present disclosure. It will be apparent to those skilled in the art that many modifications, both to materials and methods, may be practiced without departing from the scope of the present disclosure.
EXAMPLES Example 1 g affinities of anti-VEGF/anti-DLL4 antibodies The KDs of parental antibodies anti—VEGF 219R45 (IgG format), anti-DLL4 21R79 (IgG format), LL4 21M18 (IgG format) and bispecific antibodies 219R45-MB-21M18 and 219R45-MB-21R79 were determined using a Biacore 2000 system from e LifeSciences (GE Healthcare). Recombinant human DLL4-Fc or mouse DLL4-Fc proteins were immobilized on CMS carboxyl chips using standard amine-based try (NHS/EDC) and blocked with ethanolamine. Recombinant human VEGF165 or mouse VEGF165 were biotinylated and immobilized on streptavidin chips. The antibodies were serially diluted 2-fold from 100nM to 0.78nM in HBS—P (0.01M HEPES pH7.4, 0.15M NaCl, 0.005% v/v Polysorbate 20). For each antibody, all 8 dilutions were sequentially injected over a c chip.
Kinetic data were collected over time and were fit using the simultaneous global fit equation to yield affinity constants (KD values) for each bispecific antibody.
Table 3 219R45-MB-21M18 0.36 . 16 219R45-MB-21R79 0.68 . .
As shown in Table 3, ific antibody 219R45—MB-21M18 had an affinity constant (KD) for human VEGF of 0.36nM and a KD for human DLL4 of 16nM. Bispecific dy 219R45-MB-21R79 had a KD for human VEGF of 0.68nM and a KD for human DLL4 of 0.53nM. Both bispecific antibodies demonstrated weaker binding to mouse VEGF as compared to human VEGF and r antibody bound mouse DLL4. Thus, both bispecific antibodies trated similar binding affinity to human VEGF and 219R45-MB-21R79 demonstrated approximately 30—fold stronger binding to human DLL4 than 219R45- MB-21M18. Furthermore, bispecific antibody 219R45-MB—21R79 had a similar g affinity to human VEGF despite the fact the bispecific antibody is monovalent for VEGF as compared to the bivalent parental antibody.
Several additional anti-DLL4 antibodies were identified that had g affinities intermediate to the KDs of 21M] 8 and 21R79. Two of these anti—DLL4 antibodies were used to produce anti-VEGF/anti- DLL4 ific antibodies 219R45-MB—21R75 and 219R45—MB—21R83. Using the e 2000 system as described above, the KDs of the bispecific antibodies 219R45—MB—21R75 and 219R45-MB-21R83 to human DLL4 were determined. A comparison of the binding affinity to human DLL4 of these four anti- VEGF/anti-DLL4 bispecific antibodies is shown in Table 4.
The CDRs for anti-VEGF/anti-DLL4 bispecific antibodies 219R45-MB-21M18, 219R45-MB- 21R79, 219R45-MB—21R75, and 219R45—MB—21R83 are shown in Figure 1A. The heavy chain and light chain variable region SEQ ID NOs are shown in Figure 1B and the heavy chain and light chain SEQ ID NOs (with and t signal sequence) are shown in Figure 1C.
EGF/anti-DLL4 bispecific antibody 219R45-MB-21M18 comprises a (a) heavy chain encoded by the DNA comprising SEQ ID NO:75 deposited with American Type Culture Collection (ATCC), 10801 University Boulevard, Manassas, VA, USA, under the conditions of the Budapest Treaty on September 21, 2012 and assigned designation number PTA— a heavy chain encoded by the , (b) DNA comprising SEQ ID NO:33 deposited with ATCC under the conditions of the Budapest Treaty on September 21, 2012 and ed designation number PTA— and (c) a light chain encoded by the DNA comprising SEQ ID NO:34 deposited with ATCC under the conditions of the st Treaty on September 21, 2012 and assigned designation number PTA-_ .
Anti-VEGF/anti-DLL4 bispecific antibody 219R45-MB-21R79 comprises a (a) heavy chain encoded by the DNA comprising SEQ ID NO:31 deposited with ATCC under the conditions of the Budapest Treaty on September 21, 2012 and assigned designation number PTA- a heavy chain , (b) encoded by the DNA comprising SEQ ID NO:33 ted with ATCC under the conditions of the Budapest Treaty on September 21, 2012 and assigned ation number PTA- and (c) a light chain encoded by the DNA comprising SEQ ID NO:34 deposited with ATCC under the conditions of the Budapest Treaty on September 21, 2012 and assigned designation number PTA-— Anti-VEGF/anti-DLL4 bispecific antibody —MB-21R83 comprises (a) a heavy chain encoded by the DNA comprising SEQ ID NO:72 ted with ATCC under the ions of the Budapest Treaty on and assigned designation number PTA- a heavy chain encoded by , (b) the DNA comprising SEQ ID NO:33 deposited with ATCC under the conditions of the Budapest Treaty on September 21, 2012 and assigned designation number PTA- and (c) a light chain encoded by the DNA comprising SEQ ID NO:34 deposited with ATCC under the conditions of the Budapest Treaty on September 21, 2012 and assigned designation number PTA-_ .
Anti-VEGF/anti-DLL4 bispecific antibody 219R45-MB-21R75 comprises (a) a heavy chain encoded by the DNA sing SEQ ID NO:74 deposited with ATCC under the conditions of the Budapest Treaty on September 21, 2012 and assigned ation number PTA- a heavy chain , (b) encoded by the DNA comprising SEQ ID NO:33 deposited with ATCC under the conditions of the Budapest Treaty on September 21, 2012 and assigned designation number PTA- and (c) a light chain encoded by the DNA comprising SEQ ID NO:34 deposited with ATCC under the conditions of the Budapest Treaty on September 21, 2012 and assigned designation number PTA- Table 4 Antibody Heavy chain CDR2 hDLL4 (nM) Y I S S YNGATNYNQKFKG 219R45-MB-21M18 16.00 SEQ ID NO:15 YIANYNRATNYNQKFKG 219R45-MB-21R79 SEQ ID NO:14 YIAGYKDATNYNQKFKG YISNYNRATNYNQKFKG Example 2 HTRF Assay for simultaneous binding of bispecific dies to human VEGF and human DLL4 To characterize the binding capabilities of certain antibodies and/or antibody mixtures to both VEGF and DLL4, homogeneous time resolved fluorescence (HTRF) assays were performed. Antibodies tested were anti-VEGF/anti-DLL4 bispecific antibodies -MB-21M18 and -MB-21R79, parental antibodies 219R45 (anti-VEGF), 21M18 (anti-DLL4), 21R79 (anti-DLL4), a combination of 219R45 and 21M18, or a ation of 219R45 and 21R79. The antibodies or dy mixtures were serially diluted 2-fold from 3000nM to 2.9nM in binding buffer (1X PBS, 0.1% gelatin, 0.1% Polysorbate , 400mM potassium fluoride) and placed in a white 96—well plate. An equal volume of on containing 4ug/ml of d2-labeled hDLL4—F0 and 21.4ng/ml Europium crypate-labeled hVEGF165 was added to each well for a final volume of 100p] (final concentrations of acceptor and donor fluorophores were 2ug/ml and 10.7ng/ml, respectively). The assay plates were incubated for 2 hours to overnight and read on a SpectraMax M5e Microplate reader (Molecular Devices, Sunnyvale CA) at an excitation wavelength of 314nm.
As shown in Figure 2, anti-VEGF/anti-DLL4 bispeciflc antibodies 219R45-MB-21M18 and 219R45-MB-21R79, were able to bind both hVEGF and hDLL4 simultaneously. antly, neither of the combinations of the parental dies (i.e., 219R45 and 21M18 or 219R45 and 21R79) was able to bind VEGF and DLL4 simultaneously. These results clearly demonstrate that the anti-VEGF/anti-DLL4 bispeciflc antibodies 219R45-MB-21M18 and 219R45—MB-21R79 are capable of oning differently than just a mixture of the two individual antibodies.
Example 3 Inhibition ofHUVEC proliferation by anti—VEGF/anti—DLL4 bispecific antibodies HUVEC cells were obtained from Lonza (Walkersville MD) and cultured in growth media (M199, 10% heat-inactivated FBS (HI-PBS), SOug/ml EGS, 1X heparin, 1mM L-glutamine). For the HUVEC proliferation assay, a 96-well plate was ated with 50p] of l rat tail collagen type I solution (collagen 1 in 0.02N acetic acid) and incubated at 4°C overnight. After incubation, the plate was thoroughly aspirated to remove unbound collagen I solution and washed once with 200ul DPBS. The HUVEC cells were removed from the surface of the growth flasks using an endothelial cell subclone reagent and centrifuged at 1200 rpm for 5 minutes at 4°C. The cells were resuspended in starvation/assay medium (M199 and 2% HI-FBS, 1X heparin, 5U/ml n—glutamine) at a density of 105 cells/ml. The cells were seeded into the collagen-coated assay plate at 5000 cells/well, 50ul/well. The cells were incubated for 3 hours at 37°C, washed one time, refed with 100ul assay media, and incubated overnight at 37°C. The next day, bispecific antibodies 219R45—MB—21Ml8, 219R45-MB-21R79, parental antibody 219R45, or control antibody LZl were prepared in a mixture with human VEGF (R&D Biosystems, Minneapolis MN). The antibodies were ly diluted 5—fold from 20”M to 0.25nM in assay buffer in combination with hVEGF (final concentration Sng/ml). The mixture was cubated at 37°C for 2 hours. The medium was removed from the assay plate, and 100p] of the antibody/hVEGF mixture was added to each well. After 3-4 days incubation, medium was removed and a fresh aliquot of the dy/hVEGF mixture was added to each well and allowed to te for another 4 days. On day 7, 20ul of Alamar Blue t (Invitrogen, Carlsbad, CA) was added to each well and incubated at 37°C for -6 hours. The plate was read with a SpectraMax MSe Microplate reader (Molecular Devices, Sunnyvale CA) using a excitation wavelength of 539nm and an emission wavelength of 590nm.
As shown in Figure 3, anti—VEGF/anti—DLL4 bispecific antibodies 219R45-MB-21M18 and 219R45-MB-21R79, as well as parental anti—VEGF antibody 219R45 inhibited HUVEC proliferation.
These results demonstrated that the bispecific antibodies were capable of inhibiting VEGF-induced proliferation of HUVEC cells.
Example 4 Inhibition of nduced Notch signalling by bispecific antibodies Human PC3 cells were transfected with an expression vector encoding a full-length human Notch2 receptor and a firefly luciferase reporter vector (8xCBF-luciferase reporter) that is responsive to Notch signaling. The cells were also ected with a Renilla luciferase reporter (Promega, n WI) as an internal control for transfection efficiency. Purified human DLL4 protein was coated onto 96- well plates at lOOng/well and Notch2-expressing PC3—luc cells were added to the wells. Anti-VEGF/anti- DLL4 bispecific antibodies 219R45-MB-21M18, 2 l 9R45—MB—2 1 R79, parental anti-DLL4 antibodies 21M18, 21R79 or a control antibody LZl were serially diluted 55—fold from 20ug/ml to 0.064ug/ml, added to the appropriate wells, and ted overnight. Luciferase ty was determined using a dual luciferase assay kit (Promega, Madison, WI) with firefly luciferase ty normalized to Renilla luciferase activity.
As shown in Figure 4, anti-VEGF/anti-DLL4 bispecific antibody 219R45-MB-21R79 and parental LL4 antibodies 21M18 and 21R79 inhibited DLL4-induced Notch signaling. Bispecific antibody 219R45-MB-21M18 inhibited nduced Notch signaling only at high antibody concentrations. These results trated that bispecific antibody -MB-21R79, and to a lesser extent bispecific antibody 219R45—MB—21M18, were capable of inhibiting DLL4-induced Notch signaling. Thus, in combination with the results presented in Example 3, the anti-VEGF/anti-DLL4 bispecific antibodies 219R45-MB-21R79 and 219R45-MB-2 1M1 8 have demonstrated the ability to inhibit both VEGF-induced and DLL4-induced signaling and/or proliferation functions.
Example 5 Inhibition of tumor growth in vivo by a bispecific antibody in a human skin graft model A human skin graft model has been reported which comprises a human skin graft and human tumor cells. A human skin graft is established and then human tumor cells are implanted into the skin graft, allowing the tumor cells to grow in an nment with human stroma and vasculature (Tahtis et al., 2003, Mol. Cancer Ther. 22229-737). Human skin samples were obtained from neonatal foreskin tissue and grafted onto the lateral flank ofNOD-SCID mice. After establishment of the skin graft, luciferase-labeled OMP—C8 colon tumor cells (20,000 cells) were injected intradermally into the human skin. Tumor growth was monitored by bioluminescence imaging using an IVIS imaging system (Caliper Life Sciences, in View, CA). Tumors were d to grow until they reached 1.2 x 106 photons per second. Tumor-bearing mice (n = 6 mice/group) were randomized and treated with control Ab, anti- hDLL4 antibody 21M18, anti-VEGF antibody bevacizumab, or anti-VEGF/anti-DLL4 bispecific dy 219R45-MB-21M18. Animals were treated once a week and antibodies were administered eritoneally at a dose of 25mg/kg. Tumor growth was monitored by bioluminescence imaging on the indicated days.
As shown in Figure 5, both anti-hDLL4 antibody 21M18 and anti-VEGF antibody bevacizumab ted tumor grth in this human skin graft/human tumor model. Furthermore, bispecif1c anti-VEGF/ LL4 ific antibody 219R45-MB-21M18 was more effective than either the anti-DLL4 antibody or the anti-VEGF antibody alone. These data demonstrate the utility of simultaneously targeting DLL4 and VEGF with a bispecific antibody.
Example 6 Tumorigenicity of 8 atic tumor cells after treatment with anti-VEGF/anti-DLL4 bispeciflc antibodies Mice bearing OMP-PN8 pancreatic tumors were treated with control antibody (15 mg/kg), antihDLL4 antibody 21M18 (15 mg/kg), anti-VEGF antibody bevacizumab (15 mg/kg), or anti-VEGF/anti- DLL4 bispeciflc antibodies 219R45-MB-21M18 or 219R45-MB-21R79 (30 mg/kg) with or without gemcitabine (70 mg/kg). Following four weeks of treatment, tumors were harvested, processed to single cell suspensions and the human tumor cells were purified by immunomagnetic depletion of murine cells. 90 human tumor cells from each treatment group were erred to a new cohort ofmice (n = 10 mice/group). Tumors were allowed to grow for 55 days without any treatment and tumor volumes were measured with electronic calipers.
Figure 6 shows the tumor volume from the individual mice in each group. Cells ed from mice treated with anti-hDLL4 antibody 21M18 had greatly decreased tumorigenicity, 5 out of 10 mice had tumors, as compared to cells isolated from mice treated with control antibody where 9 out of 10 mice had tumors. The reduction in tumor growth frequency indicates a reduction in cancer stem cell frequency. In st, bevacizumab treatment resulted in no reduction of tumor growth ncy, 10 out of 10 mice had tumors. Similar to bevacizumab, treatment with gemcitabine as a single agent had no effect on tumor growth frequency as 10 out of 10 mice had tumors. The anti—VEGF/anti—DLL4 ific antibodies 219R45-MB-21M18 and 219R45-MB-21R79 both reduced tumor growth frequency (5 out of 10 mice had tumors and 4 out of 10 mice had tumors, respectively). ation treatment with gemcitabine ed to have no effect on tumor growth frequency. These data indicate that targeting DLL4 reduces cancer stem cell frequency while targeting VEGF alone does not. Importantly, these data indicate that the anti-CSC activity of the anti—DLL4 antibody is retained in a ific antibody.
Example 7 Bispeciflc Antibody ELISA VEGF (ATGEN, South Korea) was coated onto Nunc maxisorb plates at 2ug/ml (100ul/well) and incubated overnight at 2-8°C. Bispeciflc antibodies 219R45—MB-21M18, 2 l 9R45-MB-2 1 R79, 219R45- 75, and 219R45-MB-21R83 were diluted in blocking buffer (1x PBS, 0. 1% gelatin, 0.1% Polysorbate-20, pH 7.4) containing 2ug/ml biotin—DLL4—hFc. The antibodies were serially diluted 3-fold from 500ng/ml to g/ml. The antibody samples were incubated for 2 hours in blocking buffer containing the -DLL4-hFc. After incubation, the antibody samples were transferred to the VEGF- coated assay plate (100 ul/well) and incubated for 2 hours. Streptavidin—HRP (Jackson lmmunoResearch, West Grove, PA) was added to each well and incubated for 1 hr. TMB substrate was added to the wells with a 10 minute color development and the reaction was stopped with 2M sulfuric acid. Absorbance was read at 450—65Onm and the data analyzed using the 4—parameter fit within the Softrnax Pro analysis program (Molecular Devices, Sunnyvale, CA).
Figure 7 shows the titration curves of bispecific antibodies 219R45—MB-21M18 (open circles), 219R45-MB-21R79 (open squares), 219R45—MB—21R75 (open triangles), and 219R45-MB-21R83 (open ds) in comparison to a reference anti-VEGF/anti-DLL4 bispecific antibody (solid circles). ve potencies for the bispecific antibodies as compared to the reference bispecific antibody are shown in Table 5.
Table 5 Antibody Relatlvgfl)Potency —MB—21M18 219R45—MB-21R79 501 -MB—21R75 422 -MB—21R83 222 Bispecific dy 219R45-MB—21R79 was the most potent, about 7-fold more potent than 219R45-MB-21M18, which reflected the higher affinity of the 21R79 antigen-binding site.
Example 8 Bispecific Antibody Production Bispecific antibodies were produced using a GS-CHO cell line. CHOK] SV cells (Lonza Biologics) were transfected via electroporation with the gene(s) of interest coupled with glutamine synthetase (GS) as the able marker. Transfectants and subclones were screened for antibody productivity and the high ers were selected for scaled—up production. Cells were grown using a fed-batch process and fed-batch bioreactors. Accumulated antibody in harvested cell culture fluid (HCCF) was isolated and purified using chromatography techniques.
Bispecific antibody cell lines 219R45—MB—21M18.010.017 and 219R45-MB-21R79.017.003 were cultured in SL stirred tank ctors for 14 days. Cell line -MB-21M18.010.017 produced a final antibody titer of 3.0g/L and cell line 219R45—MB—21R79.017.003 ed a final antibody titer of 0.8g/L. Cell lines -MB-21R75.101 and 219R45—MB—21R83.1 13 were cultured in 25L WAVE bioreactor systems (GE Healthcare) using a fed-batch process that achieved final antibody titers of .
Bispecific antibody cell lines 219R45-MB-21M18AG.138.007, 219R45-MB-21M18AG.038.009, 219R45-MB-21M18AG.142.002, 219R45-MB—21R79AG.072.014 and -MB-21R83AG.129.003 were cultured in 5 L stirred tank bioreactors for 14 — 15 days. Cell line 219R45-MB-21M18AG. 138.007 produced a final antibody titer of 1.0 g/L after 14 days. Cell line 219R45-MB-21M18AG.038.009 produced a final antibody titer of 1.6 g/L after 14 days. Cell line 219R45-MB-21M18AG. 142.002 produced a final antibody titer of2.6 g/L after 14 days. Cell line 219R45-MB-21R79AG.072.014 produced a final antibody titer of2.1 g/L after 15 days. Cell line 219R45-MB-21Ml8AG.038.009 produced a final antibody titer of 2.4 g/L after 15 days. Culture fluid was harvested by filtration from each of these four cell lines and subjected to Protein A affinity chromatography. The Protein A column was washed with a series of buffers and the antibodies were eluted using a low pH elution buffer. l characterization of the purity of the bispecific antibodies was performed using size exclusion chromatography (SEC-HPLC) and isoelectric focusing (IEF).
Size exclusion chromatography (SEC) was used to determine the purity of the antibody product.
SEC is a well known chromatographic method in which molecules (e. g., antibodies) in solution are separated by their size. SEC may be used to distinguish an antibody product from aggregate and/or impurities, and to determine the percentage of the antibody product as ed to the total mixture. As used herein, SEC does not distinguish between a homomeric antibody and a heterodimeric bispecif1c antibody. lmaged capillary isoelectric ng (icIEF) was used to determine identity and purity of the bispecific antibody heterodimers. Using iclEF, the charge isoforms of an antibody are separated according to their pl and the result is a print” of the antibody’s charge distribution. The iclEF method can also serve as a determination of purity by separating the bispecific antibody heterodimers by their distinct p1 from any homodimer products or impurities.
Bispecific antibody samples were analyzed by icIEF on a nSimple ICE280 instrument (ProteinSimple, Santa Clara, CA). For this analysis, a protein mixture is introduced into a capillary, high voltage is applied across the capillary and ampholytes establish a linear pH gradient along the length of the capillary. Under the influence of the electric field, the p] markers and the protein e both migrate the length of the capillary until a pH value is reached where the net charge is zero. Once focused, the ICE280 instrument uses whole—column imaging detection with a 280-nm UV camera to monitor the n of n isoforms within the capillary. The resulting opherogram is calibrated using internal pl markers and integrated to establish the respective percentage areas of the different charged isoforms of the protein mixture. The charge profiles from several anti-VEGF/anti-DLL4 bispecific antibodies are shown in Figure 8. For this experiment, Protein A s were diluted with MilliQ water to a concentration of ml. A total of 18p] ofthe sample was mixed with 100uL of 8M urea, 70ul of 0.5% cellulose, 8”L of 3-10 Pharmalyte, 2”] of high pl marker and 2ul of low pl marker to a final volume of 200ul. Table 6 shows the percentage of antibody product from cell lines 219R45-MB- 21M18.010.017, 219R45-MB-21R79.017.002, 219R45-MB-21R75.101, 219R45-MB-21R83.113, 219R45-MB-21M18. 138.007, 219R45-MB—21M18AG.038.009, 219R45-MB-21M18AG. 142.002, 219R45-MB-21R79AG.072.014, and 219R45—MB—2 . 129.003 after Protein A affinity chromatography as determined by SEC-HPLC. Table 6 also shows the percentage of heterodimeric antibodies from cell lines 219R45-MB-21M18.010.017, 219R45—MB-21R79.017.002, 219R45-MB- 21R75.101, 219R45-MB-21R83.113, —MB—21M18.138.007, 219R45-MB-21Ml8AG.038.009, 219R45-MB-21M18AG.142.002, 219R45—MB—21R79AG.072.014, and 219R45-MB-21R83AG.129.003 after Protein A affinity chromatography as analyzed by iclEF.
Table 6 . dy Purity by SEC Purity by IEF CC” Lme Titer (g/L) (% heterodimer) The purity of the bispecific antibody product can be increased fiarther by additional chromatography steps. After Protein A affinity chromatography, the eluate fraction was held at a low pH for no less than 60 minutes at room temperature for viral inactivation. The antibody solution (Protein A column , pH adjusted) was loaded onto a strong anion-exchange . Product— and process- related impurities bound to the anion exchange chromatography resin and the flow-through fraction (antibody product) was collected. In some cases, purity was further ed by use of a multi-modal chromatography resin such as ceramic hydroxyapatite. In some cases, buffer exchange of the antibody product was undertaken using ultrafiltration and ration techniques, after which excipients were added. The formulated antibody was sterile filtered into sterile containers and stored refrigerated or . Purity of the bispecific antibodies was re—assessed using SEC-HPLC and IEF.
Table 7 Purity by SEC Purity by IEF Cell Line (%) (% heterodimer) 2l9R45-\/lB-21Ml8.010.017 98.9 98.5 2l9R45-\/lB-21R79.0l7.002 95.1 99.3 2l9R45-\/lB-21R75.101 97.2 98.2 2l9R45-\/lB-21R83.ll3 95.3 91.4 2l9R45-\/lB-21Ml8.l38.007 98.1 100 2l9R45-VlB-21M18AG. 142.002 100 2l9R45-VlB-21R79AG.072.014 98.2 100 219R45-VIB-21R83AG. 129.003 998.6 100 As shown in Table 7, the purification of the anti-VEGF/anti-DLL4 bispecific antibodieswith additional chromatography steps after Protein A resulted in isolation of antibody ts that were 95% to about 99% pure as analyzed by SEC. Analysis by IEF ined that purified EGF/anti-DLL4 bispecific antibody from cell line 219R45—MB—21M18.010.0l7 was 98.5% heterodimeric, anti- VEGF/anti-DLL4 bispecific antibody from cell line 219R45—MB-21R79.017.002 was 99.3% heterodimeric, anti-VEGF/anti-DLL4 ific antibody from cell line 219R45-MB-21R75.101 was 98.2% heterodimeric, anti-VEGF/anti-DLL4 bispecific antibody from cell line 219R45-MB-21R83. 1 l3 was 91.4% heterodimeric, anti-VEGF/anti-DLL4 ific antibody from cell line -MB- 21M18. 7 was 100% heterodimeric, anti—VEGF/anti-DLL4 bispecific antibody from cell line 219R45-MB-21M18AG. 142.002 was 100% heterodimeric, anti-VEGF/anti-DLL4 bispecific antibody from cell line 219R45-MB-21R79AG.072.014 was 100% heterodimeric, and EGF/anti-DLL4 bispecific antibody from cell line -MB-21R83AG. 129.003 was 100% heterodimeric. These results demonstrated that the anion-exchange chromatography step greatly increased the percentage of heterodimeric antibodies as ed to purification with Protein A chromatography alone. The addition of a multi-modal chromatography step such as c hydroxyapatite can also improve monomeric purity (as determined by SEP-HPLC).
Example 9 Inhibition of OMP-C8 colon tumor growth in viva tumor recurrence model Single cell suspensions of OMP—C8 colon tumor xenografts (20,000 cells) were injected subcutaneously into the flanks of 6-8 week old ID mice. Tumors were allowed to grow for 33 days until they reached an average volume of 240mm}. The mice were randomized (n = 10 per group) and treated with anti-hDLL4 antibody 21M18, anti—VEGF antibody bevacizumab, a combination of antibodies 21M18 and bevacizumab, anti-VEGF/anti—DLL4 bispecific antibody 219R45-MB-21M18, anti-VEGF/anti-DLL4 bispecific dy 219R45-MB—21R79, or control dy, all in combination with irinotecan. Antibodies and irinotecan were dosed weekly by injection into the intraperitoneal cavity.
Antibodies 21M18 and bevacizumab were dosed at 7.5mg/kg, bispecific antibodies 219R45-MB-21M18 and 219R45-MB-21R79 were dosed at 15mg/kg, and irinotecan was dosed at 45mg/kg. Irinotecan was dosed for four weeks, at which time, it was discontinued and the administration of the dies continued. Tumor growth was monitored and tumor volumes were measured with electronic calipers at the indicated time points. Data are sed as mean i S.E.M.
As shown in Figure 9, anti-hDLL4 antibody 21M18 continued to inhibit tumor growth after treatment with irinotecan was stopped. In contrast, anti—VEGF antibody bevacizumab was not able to t regrowth of the tumor after irinotecan had been stopped. The combination of anti-DLL4 antibody 21M18 and anti-VEGF antibody bevacizumab resulted in greater inhibition of tumor regrowth than either agent alone. Furthermore, the anti-VEGF/anti-DLL4 bispecific dy 219R45-MB-21M18 was more effective at inhibiting tumor regrowth than the mixture of the two antibodies.
Example 10 Reduction in tumorigenicity of OMP—C8 colon tumors Single cell sions of OMP—C8 colon tumor xenografts (20,000 cells) were injected subcutaneously into the flanks of 6-8 week old NOD/SCID mice. Tumors were allowed to grow for 33 days until they d an average volume of . The mice were randomized (n = 5 per group) and treated with anti-DLL4 antibody 21M18, anti—VEGF antibody bevacizumab, a combination of antibodies 21M18 and zumab, anti-VEGF/anti—DLL4 bispecific antibody 2l9R45-MB-21M18, anti- VEGF/anti-DLL4 bispecific antibody 219R45—MB—21R79, or control antibody, either in combination with irinotecan or without irinotecan. Antibodies and irinotecan were dosed weekly by injection into the intraperitoneal cavity. dies 21Ml 8 and bevacizumab were dosed at 7.5mg/kg, bispecific antibodies 219R45-MB-21M18 and 219R45-MB-21R79 were dosed at 15mg/kg, and irinotecan was dosed at 45mg/kg. Tumors were harvested after 4 weeks, processed into single cell suspensions, and the human tumor cells were isolated. 150 tumor cells from each experimental group were injected subcutaneously into a new cohort of mice (n = 10 per group) and tumors were allowed to grow without treatment. Tumor growth was monitored and tumor s were measured with electronic calipers.
Individual tumor volumes at day 68 are shown in Figure 10. Anti-DLL4 antibody 21M18, the combination of 21M18 with anti—VEGF antibody bevacizumab, bispecific dies 219R45-MB-21M18 and 219R45-MB-21R79, and irinotecan all reduced tumor growth frequency as single agents. In st, anti-VEGF bevacizumab as a single agent had no effect on tumor growth frequency as compared to the control dy. In the groups treated with a combination of irinotecan and antibodies, the bispecific dy 219R45-MB-21M18 had the greatest effect in reducing tumor growth frequency.
Example 11 Inhibition of OMP-C8 colon tumor growth in vivo Single cell suspensions of OMP-C8 colon tumor xenografts (50,000 cells) were injected subcutaneously into the flanks of 6-8 week old NOD/SCID mice. Tumors were allowed to grow for 21 days until they reached an average volume of 80mm3. The mice were randomized (n = 8 per group) and treated with anti-DLL4 antibody 21M18, anti—VEGF antibody bevacizumab, anti-VEGF/anti-DLL4 bispecific dies 219R45-MB-21M18, 219R45—MB—21R?5, 2 l 9R45-MB-2 1 R79, 2 l 9R45-MB- 21R83, or control dy, either alone or in combination with ecan. Antibodies and irinotecan were dosed weekly by injection into the intraperitoneal cavity. Bevacizumab and bispecific antibodies 219R45-MB-21M18, 219R45-MB-21R75, 219R45—MB—21R79, and 219R45—MB—21R83 were dosed at 15mg/kg, and irinotecan was dosed at 7.5mg/kg. Tumor growth was monitored and tumor volumes were measured with electronic calipers at the indicated time points. Data are expressed as mean :I: S.E.M.
As single agents, all four anti-VEGF/anti-DLL4 ific antibodies showed enhanced anti- tumor activity relative to anti—VEGF antibody bevacizumab. In combination with irinotecan, treatment with anti-VEGF/anti-DLL4 bispecific antibodies 219R45—MB—21M1 8 and 219R45-MB-21R83 resulted in the greatest tion of tumor growth (Figure 1 1).
Following the treatment phase, tumor sections were prepared and analyzed by xylin and eosin (H&E) staining. The tumors treated with —MB-21M18 and 219R45-MB-21R83 in combination with irinotecan showed dark pink ng regions providing evidence of extensive calcification. This is characteristic of highly necrotic tumor tissue.
Example 12 Non-GLP toxicity study of bispecific antibodies in cynomolgus monkeys A non-GLP ty study in cynomolgus monkeys was initiated to evaluate and e the toxicity profile of some of the bispecific antibodies. The animals were dosed with 0 mg/kg (control), 5 mg/kg (low dose), or 30 mg/kg (high dose) of anti-DLL4/anti-VEGF bispecific antibody 5-MB-21M18, 219R45-MB-21R83, or 219R45—MB—21R79) every 2 weeks Via IV infusion. 3 males and 3 females were dosed in each group. After 15 weeks, mean body weights were lower in animals receiving the high dose of 219R45-MB-21R79 than in animals that received the high dose of either 219R45-MB-21R18 or 219R45-MB-21R83. In addition, mean serum albumin levels were lower in animals that received 219R45-MB-21R79 than in those that ed either 219R45—MB-21R18 or 219R45-MB-21R83.
Although preliminary in nature, these early data suggest that 219R45-MB-21R18 and 219R45-MB-21R83 may have a superior toxicity profile compared to 219R45—MB-21R79.
It is understood that the examples and embodiments described herein are for illustrative es only and that various modifications or changes in light thereof will be suggested to persons skilled in the art and are to be included within the spirit and purview of this application.
All ations, s, patent applications, intemet sites, and accession numbers/database ces including both polynucleotide and polypeptide sequences cited herein are hereby incorporated by reference herein in their entirety for all purposes to the same extent as if each individual publication, patent, patent application, intemet site, or accession number/database sequence was specifically and individually indicated to be so incorporated by reference.
SEQUENCES 21M] 8 Heavy chain with signal sequence (underlined) (SEQ ID NO:1) MKHLWFFLLLVAAPRWVLSQVQLVQSGAEVKKPGASVKISCKASGYSFTAYYIHWVKQAP GQG.flW GY SSYNGATNYNQKFKGRVTFTTDTSTSTAYMELRSLRSDDTAVYYCARDYD YDVGWDYWGQGTLVTVSSASTKGPSVFPLAPCSRSTSESTAALGCLVKDYFPEPVTVSWN SGA_4TSGVHTFPAVLQSSGLYSLSSVVTVPSSNFGTQTYTCNVDHKPSNTKVDKTVERKC CVECPPCPAPPVAGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVQFNWYVDGVE VHNAKTKPREEQFNSTFRVVSVLTVVHQDWLNGKEYKCKVSNKGLPAPIEKTISKTKGQP REPQVYTLPPSREEMTKNQVSLTCLVEGFYPSDIAVEWESNGQPENNYKTTPPMLDSDGS FFLYSELTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK 21R79 Heavy chain with signal sequence (underlined) (SEQ ID N022) MKHLWFFLLLVAAPRWVLSQVQLVQSGAEVKKPGASVKISCKASGYSFTAYYIHWVKQAP GQGLEWIGYIANYNRATNYNQKFKGRVTFTTDTSTSTAYMELRSLRSDDTAVYYCARDYD YDVGMDYWGQGTLVTVSSASTKGPSVFPLAPCSRSTSESTAALGCLVKDYFPEPVTVSWN SGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSNFGTQTYTCNVDHKPSNTKVDKTVERKC CVECPPCPAPPVAGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVQFNWYVDGVE VHNAKTKPREEQFNSTFRVVSVLTVVHQDWLNGKEYKCKVSNKGLPAPIEKTISKTKGQP REPQVYTLPPSREEMTKNQVSLTCLVEGFYPSDIAVEWESNGQPENNYKTTPPMLDSDGS FFLYSELTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK 219R45 Heavy chain with signal sequence lined) (SEQ ID NO: 3) E {HIWFFIuIVAAPRWVLSQVQLVQSGAEVKKPGASVKVSCKASGYTFTNYWMHWVRQAP MGD NPSNGRTSYKEKFKRRVTLSVDKSSSTAYMELSSLRSEDTAVYFCT__HYD U {YYPLMDYWGQGTLVTVSSASTKGPSVFPLAPCSRSTSESTAALGCLVKDYFPEPVTVS 2 SGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSNFGTQTYTCNVDHKPS-TKVDKTVE? NCCVECPPCPAPPVAGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVQF WYVDG <EVI AKTKPR.flQhNSThRVVSVLTVVHQDWLNGKEYKCKVSNKGLPAP.|. flKT SKT<G QPREPQVYTLPPSREKMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENVY(TTPPMLKSD GSFFLYSKLTVD(SRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK Light chain With signal sequence (underlined) (SEQ ID NO:4) MVLQTQVFSIUIW SGAYGDIVMTQSPDSLAVSLGERATISCRASESVDNYGISFMKWF QQKPGQPP<LH'YAASNQGSGVPDRFSGSGSGTDFTLTISSLQAEDVAVYYCQQSKEVPW TFGGGTKVfl KRTVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQS VTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQGLSSPVTKSFNRGEC 21M18 Heavy chain without predicted signal sequence (SEQ ID NO:5) QVQLVQSGAEVKKPGASVKISCKASGYSFTAYYIHWVKQAPGQGLEWIGYISSYNGATNY NQKFKGRVTFTTDTSTSTAYMELRSLRSDDTAVYYCARDYDYDVGMDYWGQGTLVTVSSA STKGPSVFPLAPCSRSTSESTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSG LYSLSSVVTVPSSNFGTQTYTCNVDHKPSNTKVDKTVERKCCVECPPCPAPPVAGPSVFL FPPKPKDTLMISRTPEVTCVVVDVSHEDPEVQFNWYVDGVEVHNAKTKPREEQFNSTFRV VSVLTVVHQDWLNGKEYKCKVSNKGLPAPIEKTISKTKGQPREPQVYTLPPSREEMTKNQ VSLTCLVEGFYPSDIAVEWESNGQPENNYKTTPPMLDSDGSFFLYSELTVDKSRWQQGNV HEALHNHYTQKSLSLSPGK 21R79 Heavy chain without ted signal sequence (SEQ ID NO:6) QVQLVQSGAEVKKPGASVKISCKASGYSFTAYYIHWVKQAPGQGLEWIGYTANYNRATNY NQKFKGRVTFTTDTSTSTAYMELRSLRSDDTAVYYCARDYDYDVGMDYWGQGTLVTVSSA STKGPSVFPLAPCSRSTSESTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSG LYSLSSVVTVPSSNFGTQTYTCNVDHKPSNTKVDKTVERKCCVECPPCPAPPVAGPSVFL FPPKPKDTLMTSRTPEVTCVVVDVSHEDPEVQFNWYVDGVEVHNAKTKPREEQFNSTFRV VSVLTVViQDWLNGKEYKCKVSNKGLPAPIEKTISKTKGQPREPQVYTLPPSRflflMTKNQ VSLTCLVLGEYPSDHAVEWESNGQPENNYKTTPPMLDSDGSFFLYSELTVDKSRWQQGNV FSCSVMHHAHHNHYTQKSLSLSPGK 219R45 Heavy chain without predicted signal sequence (SEQ ID NO:7) QVQLVQSGAEVKKPGASVKVSCKASGYTFTNYWMHWVRQAPGQGLEWMGDINPSNGRTSY KEKFKRRVTLSVDKSSSTAYMELSSLRSEDTAVYFCTIHYDDKYYPLMDYWGQGTLVTVS SASTKGPSVFPLAPCSRSTSESTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQS SSVVTVPSSNFGTQTYTCNVDHKPSNTKVDKTVERKCCVECPPCPAPPVAGPSV LFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVQFNWYVDGVEVHNAKTKPREEQFNSTF RVVSVLTVVHQDWLNGKEYKCKVSNKGLPAPIEKTISKTKGQPREPQVYTLPPSREKMTK NQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPMLKSDGSFFLYSKLTVDKSRWQQG VMHEALHNHYTQKSLSLSPGK Light chain without predicted signal sequence (SEQ ID NO:8) DIVMTQSPDSLAVSLGERATISCRASESVDNYGISFMKWFQQKPGQPPKLLIYAASNQGS GVPDRFSGSGSGTDFTLTISSLQAEDVAVYYCQQSKEVPWTFGGGTKVEIKRTVAAPSVI FPPSDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQSGNSQESVTEQDSKDSTYSLSS TLT;SKADYEKHKVYACEVTHQGLSSPVTKSFNRGEC 21M18 Heavy chain variable region (SEQ ID NO:9) QVQLVQSGAEVKKPGASVK:SCKASGYSFTAYYIHWVKQAPGQGLEWIGYISSYNGATNY NQKFKGRVTFTT3TSTSTAYMELRSLRSDDTAVYYCARDYDYDVGMDYWGQGTLVTVSS 21R79 Heavy chain variable region (SEQ ID NO:10) QVQLVQSGAEVK{PGASVKISCKASGYSFTAYYIHWVKQAPGQGLEWIGYIANYNRATNY NQKFKGRVTFTT3TSTSTAYMELRSLRSDDTAVYYCARDYDYDVGMDYWGQGTLVTVSS 219R45 Heavy chain variable region (SEQ ID NO:1 l) QVQLVQSGA.EVK{PGASVKVSCKASGYTFTNYWMHWVRQAPGQGLEWMGD:NPSNGRTSY KEKFKRRVT_JSVDKSSSTAYMELSSLRSEDTAVYFCTIHYDDKYYPLMDYWGQGTLVTVSS Light chain variable region (SEQ ID NO: 12) QSPDSLAVSLGERATISCRASESVDNYGISFMKWFQQKPGQPPKLLIYAASNQGS GVPDRFSGSGSGTDFTLTISSLQAEDVAVYYCQQSKEVPWTFGGGTKVEIK 21R75, 21R79 21R83 and 21Ml8 Heavy chain CDRl (SEQ ID NO:13) TAYY..H Alternative 21R75, 21R79, 21R83, and 21M18 Heavy chain CDRl (SEQ ID NO:79) AYYIH 21R79 Heavy chain CDR2 (SEQ ID NO:14) YIANYNRATNYNQKFKG 21M18 Heavy chain CDR2 (SEQ ID NO:15) YISSYNGATNYNQKFKG 21R75, 21R79, 21R83, and 21M18 Heavy chain CDR3 (SEQ ID NO:16) RDYDYDVGMDY 219R45 Heavy chain CDRl (SEQ ID NO:17) NYWMH 219R45 Heavy chain CDR2 (SEQ ID NO:18) DL PSNGRTSYK_3KFKR 219R45 Heavy chain CDR3 (SEQ ID NO: 19) HYDDKYYPLMDY Light chain CDRl (SEQ ID NO:20) RASESVDNYGISFMK Light chain CDR2 (SEQ ID NO:21) AASNQGS Light chain CDR3 (SEQ ID NO:22) QQSKEVPWTFGG Human DLL4 with signal sequence (underlined) (SEQ ID N0223) MAAASRSASGWALLLLVALWQQRAAGSGVFQLQLQEFINERGVLASGRPCEPGCRTFFRV CLKHFQAVVSPGPCTFGTVSTPVLGTNSFAVRDDSSGGGRNPLQLPFNFTWPGTFSLILE AWHAPGDDLRPEALPPDALISKTAIQGSLAVGQNWLLDEQTSTLTRLRYSYRVLCSD-YY GDNCSQLCKKRNDHFGHYVCQPDGNLSCLPGWTGEYCQQPICLSGCHEQNGYCSKPAECL CRPGWQGRLCNECIPHNGCRHGTCSTPWQCTCDEGWGGLFCDQDLNYCTHHSPCK-GATC S TCTCRPGYTGVDCELELSECDSNPCRNGGSCKDQEDGYHCLCPPGYYGL{CE HSTLSCADSPCFNGGSCRERNQGANYACECPPNFTGSNCEKKVDRCTSNPCA-GGQCLN N GPSRMCRCRPGFTGTYCELHVSDCARNPCAHGGTCHDLENGLMCTCPAGFSGRRCEVQTS LDACASSPCFNRATCYTDLSTDTFVCNCPYGFVGSRCEFPVG Human DLL4 without predicted signal sequence (SEQ ID NO:24) SGVFQIQIQ4b SGQPCEPGCRTFFRVCLKHFQAVVSPGPCTFGTVSTPVLGT NSFAVQDDSSGGGRNPLQIPF FTWPGTFSLIIEAWHAPGDDLRPEALPPDAL S< A Q GSLAVGQ WITDTQTSTLTRLRYSYRVICSDNYYGDNCSRLCKKRNDHFGHYVCQPDGNL SCLPGWTGTYCQQPC ISGCHEQNGYCSKPAECLCRPGWQGRLCNECIPHNGCRHGTCST PWQCTCDHGWGGIFCDQ HSPCKNGATCSNSGQRSYTCTCRPGYTGVDCflLflLS ECDSNPC? GGSCKDQEDGYHCLCPPGYYGLHCEHSTLSCADSPCFNGGSCRERNQGANY ACECPP FTGSNCEKKVDRCTSNPCANGGQCLNRGPSRMCRCRPGFTGTYCELdVSDCAR PCAHGGTCHDLENGLMCTCPAGFSGRRCEVRTSIDACASSPCFNRATCYTDLSTDTPVC CPYGFVGSRCEFPVG Human DLL4 inal Region (SEQ ID NO:25) SGVFQLQLQEFINERGVLASGRPCEPGCRTFFRVCLKHFQAVVSPGPCTFGTVSTPVLGT NSFAVRDDSSGGGRNPLQLPFNFTWPGTFSLIIEAWHAPGDDLRPEALPPDALISKIAIQ GSLAVGQW Human DLL4 DSL Domain (SEQ ID NO:26) WLLDEQTSTLTRLRYSYRVICSDNYYGDNCSRLCKKRNDHFGHYVCQPDGNLSCLPGWTG Human VEGF-A with signal sequence (underlined)(SEQ ID N022?) MNFLLSWVHWSLALLLYLHHAKWSQAAPMAEGGGQNHHEVVKFMDVYQRSYCHPIETLVD IFQEYPDEIEYIFKPSCVPLMRCGGCCNDEGLECVPTEESNITMQIMRIKPHQGQHLGEM SFLQHNKCECRPKKDRARQEKKSVRGKGKGQKRKRKKSRYKSWSVYVGARCCLMPWSLPG PHPCGPCSERRKHLFVQDPQTCKCSCKNTDSRCKARQLELNERTCRCDKPRR Human VEGF-A without predicted signal sequence (SEQ ID NO:28) APMAEGGGQNH{EVVKFMDVYQRSYCHPIETLVDIFQEYPDEIEYIFKPSCVPLMRCGGC CNDflGHflCVPT.flSNTTMQTMRTKPHQGQHIGEMSFLQHNKCECRPKKDRARQEKKSVRG.|.
KGKGQ(RKRKKSRYKSWSVYVGARCCLMPWSLPGPHPCGPCSERRKHLFVQDPQTCKCSC KNTDSRCKARQLELNERTCRCDKPRR 21M18 Heavy chain nucleotide sequence (13B Version 1) (SEQ ID NO:29) ATGAAGCACCTGTGGTTCTTTCTGCTGCTGGTGGCCGCTCCCAGATGGGTGCTGTCCCAG GTGCAGCTGGTGCAGTCTGGCGCCGAAGTGAAGAAACCTGGCGCCTCCGTGAAGATCTCC TGCAAGGCCTCCGGCTACTCCTTCACCGCTTACTACATCCACTGGGTCAAGCAGGCCCCT GGGCAGGGCCTGGAATGGATCGGCTACATCTCCTCCTACAACGGCGCCACCAACTACAAC CAGAAATTCAAGGGCCGCGTGACCTTCACCACCGACACCTCCACCTCCACCGCCTACATG CGGTCCCTGCGGAGCGACGACACCGCCGTGTACTACTGCGCCAGAGACTACGAC GTGGGCATGGACTACTGGGGCCAGGGCACCCTGGTCACCGTGTCCTCTGCCTCC ACCAAGGGCCCATCCGTGTTCCCTCTGGCCCCTTGCTCCCGGTCCACCTCTGAGTCTACC GCCGCTCTGGGCTGCCTGGTGAAGGACTACTTCCCTGAGCCTGTGACCGTGTCCTGGAAC TCTGGCGCCCTGACCTCTGGCGTGCACACCTTCCCTGCCGTGCTGCAGTCCTCCGGCCTG TACTCCCTGTCTAGCGTGGTGACCGTGCCTTCCTCCAACTTCGGCACCCAGACCTACACC TGTAACGTGGACCACAAGCCTTCCAACACCAAGGTGGACAAGACCGTGGAGCGGAAGTGC TGCGTGGAGTGCCCTCCTTGTCCTGCTCCTCCTGTGGCTGGCCCTTCTGTGTTCCTGTTC CCTCCAAAGCCTAAGGACACCCTGATGATCTCCCGGACCCCTGAAGTGACCTGCGTGGTG GTGGACGTGTCCCACGAGGACCCTGAGGTGCAGTTCAATTGGTACGTGGACGGCGTGGAG GTGCACAACGCCAAGACCAAGCCTCGGGAGGAACAGTTCAACTCCACCTTCCGGGTGGTG TCTGTGCTGACCGTGGTGCACCAGGACTGGCTGAACGGCAAAGAATACAAGTGCAAGGTG TCCAACAAGGGCCTGCCTGCCCCTATCGAAAAGACCATCAGCAAGACCAAGGGCCAGCCT CGCGAGCCTCAGGTGTACACCCTGCCTCCCAGCCGGGAAGAAATGACCAAGAACCAGGTG TCCCTGACCTGTCTGGTGGAGGGCTTCTACCCTTCCGATATCGCCGTGGAGTGGGAGTCT AACGGCCAGCCTGAGAACAACTACAAGACCACCCCTCCTATGCTGGACTCCGACGGCTCC TTCTTCCTGTACTCCGAACTGACCGTGGACAAGTCCCGGTGGCAGCAGGGCAACGTGTTC TCCGTGATGCACGAGGCCCTGCACAACCACTACACCCAGAAGTCCCTGTCCCTG TCTCCTGGCAAGTAG 21R79 Heavy chain nucleotide sequence (13B Version 1) (SEQ ID NO:30) ATGAAGCACCTGTGGTTCTTTCTGCTGCTGGTGGCCGCTCCCAGATGGGTGCTGTCCCAG GTGCAGCTGGTGCAGTCTGGCGCCGAAGTGAAGAAACCTGGCGCCTCCGTGAAGATCTCC TGCAAGGCCTCCGGCTACTCCTTCACCGCCTACTACATCCACTGGGTGAAACAGGCACCA GGCCAGGGACTGGAATGGATCGGCTATATCGCCAACTACAACCGGGCCACCAACTACAAC CAGAAATTCAAGGGCCGCGTGACCTTCACCACCGACACCTCCACCTCCACAGCCTACATG GAACTGCGGTCCCTGCGGAGCGACGACACCGCCGTGTACTACTGCGCCAGAGACTACGAC GTGGGCATGGACTACTGGGGCCAGGGCACCCTGGTGACAGTGTCCTCCGCCTCC ACCAAGGGCCCCTCCGTGTTCCCTCTGGCCCCTTGCTCCCGGTCCACCTCTGAGTCTACC GCCGCTCTGGGCTGCCTGGTGAAGGACTACTTCCCTGAGCCTGTGACCGTGTCCTGGAAC TCTGGCGCCCTGACCTCTGGCGTGCACACCTTCCCTGCCGTGCTGCAGTCCTCCGGCCTG TACTCCCTGTCTAGCGTGGTGACCGTGCCTTCCTCCAACTTCGGCACCCAGACCTACACC GTGGACCACAAGCCTTCCAACACCAAGGTGGACAAGACCGTGGAGCGGAAGTGC TGCGTGGAGTGCCCTCCTTGTCCTGCTCCTCCTGTGGCTGGCCCTTCTGTGTTCCTGTTC CCTCCAAAGCCTAAGGACACCCTGATGATCTCCCGGACCCCTGAAGTGACCTGCGTGGTG GTGGACGTGTCCCACGAGGACCCTGAGGTGCAGTTCAATTGGTACGTGGACGGCGTGGAG GTGCACAACGCCAAGACCAAGCCTCGGGAGGAACAGTTCAACTCCACCTTCCGGGTGGTG TCTGTGCTGACCGTGGTGCACCAGGACTGGCTGAACGGCAAAGAATACAAGTGCAAGGTG TCCAACAAGGGCCTGCCTGCCCCTATCGAAAAGACCATCAGCAAGACCAAGGGCCAGCCT CGCGAGCCTCAGGTGTACACCCTGCCTCCCAGCCGGGAAGAAATGACCAAGAACCAGGTG TCCCTGACCTGTCTGGTGGAGGGCTTCTACCCTTCCGATATCGCCGTGGAGTGGGAGTCT AACGGCCAGCCTGAGAACAACTACAAGACCACCCCTCCTATGCTGGACTCCGACGGCTCC TTCTTCCTGTACTCCGAACTGACCGTGGACAAGTCCCGGTGGCAGCAGGGCAACGTGTTC TCCTGCTCCGTGATGCACGAGGCCCTGCACAACCACTACACCCAGAAGTCCCTGTCCCTG TCTCCTGGCAAGTAG 21R79 Heavy chain nucleotide sequence (13B Version 2) (SEQ ID NO:31) ATGAAGCACCTATGGTTCTTTCTATTATTAGTGGCCGCTCCCCGTTGGGTGTTATCGCAG GTTCAGCTAGTTCAGTCTGGAGCGGAAGTTAAGAAACCTGGAGCATCCGTGAAAATAAGT TGCAAGGCATCCGGTTACTCGTTCACCGCATACTATATCCACTGGGTTAAACAGGCACCA GGACAGGGACTTGAATGGATCGGATATATCGCTAATTATAATAGAGCTACAAACTATAAC CAAAAATTCAAAGGACGCGTGACTTTCACAACTGACACCTCAACCTCGACAGCATACATG GAATTACGGTCCCTACGGTCTGACGACACTGCCGTTTACTATTGCGCTAGAGATTATGAT TATGATGTTGGAATGGACTATTGGGGCCAGGGAACACTGGTGACAGTGTCTTCTGCATCC ACTAAGGGACCATCCGTGTTCCCTTTGGCCCCTTGCTCTCGTTCGACCTCTGAATCGACT GCCGCTCTGGGATGCCTCGTGAAAGATTACTTCCCTGAGCCTGTGACCGTTTCCTGGAAC TCGGGCGCCCTAACCTCTGGCGTGCACACATTCCCTGCCGTGCTACAGTCTTCTGGCCTA TACTCTTTATCTTCGGTTGTTACCGTACCTTCTTCTAACTTCGGAACCCAAACTTACACC TGTAACGTAGACCACAAGCCTTCGAACACCAAGGTGGACAAGACTGTTGAGCGAAAGTGC TGCGTTGAGTGCCCTCCATGTCCTGCACCTCCTGTGGCTGGCCCTTCTGTGTTCCTGTTC AAACCTAAGGACACTCTAATGATCTCTCGGACTCCTGAGGTGACTTGCGTGGTT GTGGACGTGTCCCACGAGGACCCTGAGGTGCAGTTCAATTGGTACGTGGACGGAGTCGAG AATGCAAAGACCAAGCCTCGGGAGGAACAGTTCAACTCCACCTTCCGGGTGGTT TTGACCGTTGTGCACCAAGACTGGCTGAACGGCAAAGAATACAAGTGCAAGGTG TCCAACAAGGGCCTGCCTGCCCCTATCGAAAAGACCATCAGCAAGACCAAGGGCCAGCCT CGCGAGCCTCAGGTGTACACCCTGCCTCCCAGCCGGGAAGAAATGACCAAGAACCAGGTG TCCCTGACCTGTCTGGTGGAGGGCTTCTACCCTTCCGACATCGCCGTTGAGTGGGAGTCT AACGGACAGCCGGAGAACAACTACAAGACTACGCCTCCAATGCTGGACTCCGACGGCTCC TTCTTCCTGTACTCCGAACTGACCGTGGACAAGTCCCGGTGGCAGCAGGGCAACGTGTTC TCATGCTCCGTAATGCACGAAGCCTTGCACAATCACTACACTCAAAAGTCCCTATCCTTA TCTCCTGGCAAGTAG 219R45 Heavy chain nucleotide sequence (13A Version 1) (SEQ ID NO:32) ATGAAGCATCTGTGGTTTTTCCTGTTGCTCGTGGCGGCACCCAGATGGGTGTTGTCCCAA GTGCAGCTGGTCCAGAGCGGGGCTGAGGTGAAGAAACCCGGAGCAAGCGTAAAAGTATCG TGTAAGGCCTCGGGGTACACGTTTACAAACTACTGGATGCATTGGGTGCGGCAGGCTCCG GGACAGGGGTTGGAATGGATGGGTGACATTAACCCCTCAAATGGCAGAACATCATATAAG GAAAAGTTCAAACGCCGCGTCACACTCTCCGTGGACAAGTCAAGCTCGACTGCGTACATG GAACTTTCGTCGCTGAGGTCGGAGGACACGGCAGTGTACTTTTGCACCATCCATTATGAT GACAAGTATTACCCTCTGATGGATTATTGGGGTCAGGGTACGTTGGTCACCGTCTCCAGC GCGTCGACGAAAGGTCCCTCGGTATTTCCCCTCGCCCCCTGCTCGAGGTCGACATCCGAA TCAACAGCTGCCCTCGGCTGCCTGGTCAAAGACTACTTCCCAGAGCCGGTAACGGTGTCG TGGAACTCGGGAGCGCTTACGTCCGGAGTCCACACATTTCCGGCGGTACTGCAATCCTCG GGACTGTATTCGTTGTCGTCAGTGGTGACTGTCCCGTCCTCCAATTTCGGGACTCAGACC TATACGTGCAACGTCGACCACAAACCCTCAAACACCAAGGTGGATAAGACAGTGGAGCGC AAGTGCTGCGTGGAGTGTCCCCCGTGTCCGGCACCCCCTGTCGCCGGACCCTCAGTCTTT CCGCCGAAGCCCAAAGATACACTCATGATCTCAAGAACGCCCGAGGTAACATGC GTGGTGGTCGATGTAAGCCACGAGGATCCAGAAGTACAATTCAATTGGTATGTAGACGGG GTCGAGGTCCATAACGCAAAGACGAAACCGAGGGAAGAGCAGTTCAATTCGACTTTCCGG GTGGTGTCGGTGCTTACAGTCGTACATCAGGACTGGTTGAACGGGAAGGAGTACAAGTGT AAAGTATCGAATAAGGGCCTTCCAGCGCCGATTGAAAAGACCATCTCCAAGACCAAAGGA CAGCCACGAGAGCCGCAAGTCTATACGCTTCCTCCCAGCCGAGAAAAGATGACTAAAAAC CAGGTATCGCTTACGTGTCTCGTCAAGGGTTTCTACCCTTCGGACATCGCGGTGGAATGG GAGAGCAATGGACAACCGGAAAACAACTACAAGACGACACCGCCTATGTTGAAAAGCGAT GGATCGTTTTTCCTCTATTCGAAACTCACGGTCGATAAGTCACGGTGGCAGCAGGGGAAT GTGTTCTCCTGTTCAGTGATGCACGAGGCGCTCCACAATCACTATACCCAGAAAAGCCTG TCACTTTCCCCGGGAAAATGA 219R45 Heavy chain nucleotide sequence (13A Version 2) (SEQ ID NO:33) ATGAAGCACCTCTGGTTCTTCCTGCTCCTCGTGGCTGCTCCTCGGTGGGTCCTCTCCCAA GTGCAGCTGGTCCAGAGCGGGGCTGAGGTGAAGAAACCCGGAGCTTCCGTCAAAGTCTCC TGTAAGGCTTCCGGATACACCTTTACCAACTATTGGATGCACTGGGTGCGGCAGGCTCCT GGACAAGGGCTGGAATGGATGGGAGACATCAATCCTTCCAATGGCAGAACCTCCTACAAG TTCAAACGGCGGGTCACACTCTCCGTGGACAAGTCTAGCTCCACAGCTTACATG GAACTCTCCTCCCTGCGGTCCGAAGACACAGCTGTCTACTTCTGCACCATCCACTACGAC GACAAGTACTACCCTCTGATGGACTACTGGGGCCAGGGAACCCTGGTCACCGTGTCCAGC ACAAAAGGACCCTCCGTCTTTCCCCTCGCCCCCTGCTCCCGGTCCACATCCGAA TCAACAGCTGCCCTCGGCTGCCTGGTCAAAGACTACTTCCCAGAGCCTGTCACAGTGTCC TGGAACTCCGGAGCTCTCACATCCGGAGTCCACACATTTCCTGCTGTGCTCCAATCCTCC GGACTGTATTCCCTCTCCTCCGTGGTGACAGTGCCTTCCTCCAATTTCGGGACACAGACC TATACATGCAACGTGGACCACAAACCCTCCAACACCAAAGTCGATAAGACAGTGGAGCGC TGCGTGGAGTGTCCCCCTTGTCCTGCTCCCCCTGTGGCTGGACCTTCCGTCTTT CTGTTTCCTCCTAAACCTAAAGACACCCTCATGATCTCCCGGACCCCCGAGGTCACATGC GTCGATGTGAGCCACGAGGACCCCGAAGTCCAATTTAATTGGTATGTGGACGGG GTGGAGGTCCATAACGCTAAGACCAAACCTAGGGAAGAGCAGTTCAATTCCACTTTCCGG GTGGTGTCCGTGCTGACCGTCGTTCATCAGGACTGGCTCAACGGGAAAGAATACAAATGC AAAGTCTCTAATAAGGGCCTCCCTGCTCCTATTGAAAAAACAATTTCCAAAACAAAAGGA CAACCTCGGGAGCCTCAAGTCTACACACTGCCACCTTCCCGGGAAAAAATGACAAAAAAT CAAGTCTCCCTCACATGTCTCGTCAAGGGATTCTACCCTTCCGACATTGCTGTGGAATGG GAATCCAATGGACAACCTGAAAACAACTACAAGACAACACCTCCTATGCTCAAAAGCGAT GGGTCCTTTTTCCTCTATTCCAAACTCACAGTCGATAAGTCTCGGTGGCAGCAGGGGAAT GTGTTCTCCTGTTCCGTGATGCACGAGGCTCTCCACAATCACTATACCCAGAAAAGCCTG TCCCTCTCCCCTGGAAAATGA Light chain nucleotide sequence (SEQ ID NO:34) ATGGTGCTGCAGACCCAGGTGTTCATCTCCCTGCTGCTGTGGATCTCCGGCGCCTACGGC GACATCGTGATGACCCAGTCCCCAGACTCCCTGGCTGTGTCTCTGGGAGAGCGGGCCACC ATCTCTTGCAGAGCCTCCGAGTCCGTGGACAACTACGGCATCTCCTTCATGAAGTGGTTC CAGCAGAAGCCCGGCCAGCCCCCAAAGCTGCTGATCTACGCCGCCTCCAACCAGGGATCT CCCGACCGGTTCTCTGGATCCGGCTCTGGCACCGACTTTACCCTGACCATCAGC TCCCTGCAGGCCGAGGACGTGGCCGTGTACTACTGCCAGCAGTCCAAAGAGGTGCCCTGG ACCTTCGGCGGAGGCACCAAGGTGGAAATCAAGCGGACCGTGGCCGCTCCCTCCGTGTTC ATCTTCCCACCCTCCGACGAGCAGCTGAAGTCCGGAACCGCCTCCGTCGTGTGCCTGCTG AACAACTTCTACCCCCGCGAGGCCAAGGTGCAGTGGAAGGTGGACAACGCCCTGCAGTCC GGCAACTCCCAGGAATCCGTCACCGAGCAGGACTCCAAGGACAGCACCTACTCCCTGTCC TCCACCCTGACCCTGTCCAAGGCCGACTACGAGAAGCACAAGGTGTACGCCTGCGAAGTG ACCCACCAGGGCCTGTCCAGCCCCGTGACCAAGTCCTTCAACCGGGGCGAGTGTTAG 21M18 Heavy chain variable region nucleotide sequence (SEQ ID NO:35) CAGGTGCAGCTGGTGCAGTCTGGCGCCGAAGTGAAGAAACCTGGCGCCTCCGTGAAGATC TCCTGCAAGGCCTCCGGCTACTCCTTCACCGCTTACTACATCCACTGGGTCAAGCAGGCC CCTGGGCAGGGCCTGGAATGGATCGGCTACATCTCCTCCTACAACGGCGCCACCAACTAC AACCAGAAATTCAAGGGCCGCGTGACCTTCACCACCGACACCTCCACCTCCACCGCCTAC ATGGAACTGCGGTCCCTGCGGAGCGACGACACCGCCGTGTACTACTGCGCCAGAGACTAC GACTACGACGTGGGCATGGACTACTGGGGCCAGGGCACCCTGGTCACCGTGTCCTCT 21R79 Heavy chain variable region nucleotide sequence (13B) (SEQ ID NO:36) CAGGTGCAGCTGGTGCAGTCTGGCGCCGAAGTGAAGAAACCTGGCGCCTCCGTGAAGATC TCCTGCAAGGCCTCCGGCTACTCCTTCACCGCCTACTACATCCACTGGGTGAAACAGGCA CCAGGCCAGGGACTGGAATGGATCGGCTATATCGCCAACTACAACCGGGCCACCAACTAC AACCAGAAATTCAAGGGCCGCGTGACCTTCACCACCGACACCTCCACCTCCACAGCCTAC ATGGAACTGCGGTCCCTGCGGAGCGACGACACCGCCGTGTACTACTGCGCCAGAGACTAC GACTACGACGTGGGCATGGACTACTGGGGCCAGGGCACCCTGGTGACAGTGTCCTCC 21R79 Heavy chain variable region nucleotide sequence (13B Version 2 ) (SEQ ID NO:37) CAGGTTCAGCTAGTTCAGTCTGGAGCGGAAGTTAAGAAACCTGGAGCATCCGTGAAAATA AGTTGCAAGGCATCCGGTTACTCGTTCACCGCATACTATATCCACTGGGTTAAACAGGCA CAGGGACTTGAATGGATCGGATATATCGCTAATTATAATAGAGCTACAAACTAT AACCAAAAATTCAAAGGACGCGTGACTTTCACAACTGACACCTCAACCTCGACAGCATAC ATGGAATTACGGTCCCTACGGTCTGACGACACTGCCGTTTACTATTGCGCTAGAGATTAT GATTATGATGTTGGAATGGACTATTGGGGCCAGGGAACACTGGTGACAGTGTCTTCT 219R45 Heavy chain variable region nucleotide sequence (13A n 1) (SEQ ID NO:38) CAAGTGCAGCTGGTCCAGAGCGGGGCTGAGGTGAAGAAACCCGGAGCAAGCGTAAAAGTA TCGTGTAAGGCCTCGGGGTACACGTTTACAAACTACTGGATGCATTGGGTGCGGCAGGCT CCGGGACAGGGGTTGGAATGGATGGGTGACATTAACCCCTCAAATGGCAGAACATCATAT AAGGAAAAGTTCAAACGCCGCGTCACACTCTCCGTGGACAAGTCAAGCTCGACTGCGTAC ATGGAACTTTCGTCGCTGAGGTCGGAGGACACGGCAGTGTACTTTTGCACCATCCATTAT GATGACAAGTATTACCCTCTGATGGATTATTGGGGTCAGGGTACGTTGGTCACCGTCTCC 219R45 Heavy chain variable region nucleotide sequence (13A Version 2) (SEQ ID NO:39) CAAGTGCAGCTGGTCCAGAGCGGGGCTGAGGTGAAGAAACCCGGAGCTTCCGTCAAAGTC TCCTGTAAGGCTTCCGGATACACCTTTACCAACTATTGGATGCACTGGGTGCGGCAGGCT CCTGGACAAGGGCTGGAATGGATGGGAGACATCAATCCTTCCAATGGCAGAACCTCCTAC AAGGAAAAATTCAAACGGCGGGTCACACTCTCCGTGGACAAGTCTAGCTCCACAGCTTAC ATGGAACTCTCCTCCCTGCGGTCCGAAGACACAGCTGTCTACTTCTGCACCATCCACTAC GACGACAAGTACTACCCTCTGATGGACTACTGGGGCCAGGGAACCCTGGTCACCGTGTCC Light chain variable region nucleotide ce (SEQ ID NO:40) GACATCGTGATGACCCAGTCCCCAGACTCCCTGGCTGTGTCTCTGGGAGAGCGGGCCACC ATCTCTTGCAGAGCCTCCGAGTCCGTGGACAACTACGGCATCTCCTTCATGAAGTGGTTC CAGCAGAAGCCCGGCCAGCCCCCAAAGCTGCTGATCTACGCCGCCTCCAACCAGGGATCT GGCGTGCCCGACCGGTTCTCTGGATCCGGCTCTGGCACCGACTTTACCCTGACCATCAGC TCCCTGCAGGCCGAGGACGTGGCCGTGTACTACTGCCAGCAGTCCAAAGAGGTGCCCTGG ACCTTCGGCGGAGGCACCAAGGTGGAAATCAAG Human IgGl Heavy chain constant region (SEQ ID NO:41) ASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSS GLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKVEPKSCDKTHTCPPCPAPELLGG PSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYN STYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDE LTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRW QQGNVFSCSVMHEALHNHYTQKSLSLSPGK Human IgG2 Heavy chain nt region (SEQ ID NO:42) ASTKGPSVFPLAPCSRSTSESTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSS GLYSLSSVVTVPSSNFGTQTYTCNVDHKPSNTKVDKTVERKCCVECPPCPAPPVAGPSVF LFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVQFNWYVDGVEVHNAKTKPREEQFNSTFR VVSVLTVVHQDWLNGKEYKCKVSNKGLPAPIEKTISKTKGQPREPQVYTLPPSREEMTKN LVKGFYPSD:AVEWESNGQPENNYKTTPPMLDSDGSFFLYSKLTVDKSRWQQGN VFSCSVMHEALHNHYTQKSLSLSPGK —1o4— Human IgG3 Heavy chain nt region (SEQ ID NO:43) ASTKGPSVFPLAPCSRSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSS G.YSUSSVVTVPSSSLGTQTYTCNVNHKPSNTKVDKRVELKTPLGDTTHTCPRCPEPKSC DTPPPCPQCPEPKSC3TPPPCPRCPEPKSCDTPPPCPRCPAPELLGGPSVFLFPPKPKDT U SRTPfiVTCVVVDVSHEDPEVQFKWYVDGVEVHNAKTKPREEQYNSTFRVVSVLTVLH QDWL‘GKEYKCKVSNKALPAPIEKTISKTKGQPREPQVYTLPPSREEMTKNQVSLTCLVK GFYPSDIAVEWESSGQPENNYNTTPPMLDSDGSFFLYSKLTVDKSRWQQGNIFSCSVMHE LHNRFTQKSLSLSPGK Human IgG4 Heavy chain constant region (SEQ ID NO:44) ASTKGPSVFPLAPCSRSTSESTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSS GLYSLSSVVTVPSSSLGTKTYTCNVDHKPSNTKVDKRVESKYGPPCPSCPAPEFLGGPSV FLFPPKPKDTLMISRTPEVTCVVVDVSQEDPEVQFNWYVDGVEVHNAKTKPREEQFNSTY RVVSVLTVLHQDWLNGKEYKCKVSNKGLPSSIEKTISKAKGQPREPQVYTLPPSQEEMTK NQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSRLTVDKSRWQEG VMHEALHNHYTQKSLSLSLGK FLAG peptide (SEQ ID NO:45) Parental 21R79 Heavy chain With signal sequence underlined unmodified chain (SEQ ID NO:46) (HLWFFLLLVAAPRWVLSQVQLVQSGAEVKKPGASVKISCKASGYSFTAYYIHWVKQA? QGUfiW GY ANYNRATNYNQKFKGRVTFTTDTSTSTAYMELRSLRSDDTAVYYCARDYD DVG DYWGQGTLVTVSSASTKGPSVFPLAPCSRSTSESTAALGCLVKDYFPEPVTVSW GALTSGVHTFPAVLQSSGLYSLSSVVTVPSSNFGTQTYTCNVDHKPS_TKVDKTV_3 VECPPCPAPPVAGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVQFNWYVDGVE Vi A<TKPRflfiQh STERVVSVLTVVHQDWLNGKEYKCKVSNKGLPA?IEKTTSKT{GQP R.EPQVYTPPPSRflflMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPMLDSDGS F "11LYSKLTV){SRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK Parental 219R45 Heavy chain with signal sequence underlined (SEQ ID NO:47) M<iIWFFIIIVAAPQWVLSQVQLVQSGAEVKKPGASVKVSCKASGYTFTNYWMHWVRQAP GQGUflWMGD NPSNGRTSYKEKFKRRVTLSVDKSSSTAYMELSSLRSEDTAVYFCT__HYD D{YYPLMDYWGQGTLVTVSSASTKGPSVFPLAPCSRSTSESTAALGCLVKDYFPEPVTVS W GVHTFPAVLQSSGLYSLSSVVTVPSSNFGTQTYTCNVDHKPSNTKVDKTV.ER KCCVZCPPCPAPPVAGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVQFNWYVDG VEVH AKTKPREEQFNSTFRVVSVLTVVHQDWLNGKEYKCKVSNKGLPAPIEKTISKTKG VYTLPPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQ?ENNYKTTPPMLDSD GSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK Parental 21R79 Heavy chain without predicted signal sequence (SEQ ID NO:48) QVQLVQSGAEVKKPGASVKISCKASGYSFTAYYIHWVKQAPGQGLEWIGYIANYNRATNY NQKFKGRVTFTTDTSTSTAYMELRSLRSDDTAVYYCARDYDYDVGMDYWGQGTLVTVSSA STKGPSVFPLAPCSRSTSESTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSG LYSLSSVVTVPSSNFGTQTYTCNVDHKPSNTKVDKTVERKCCVECPPCPAPPVAGPSVFL FPPKPKDTLMISRTPEVTCVVVDVSHEDPEVQFNWYVDGVEVHNAKTKPREEQFNSTFRV VSVLTVVHQDWLNGKEYKCKVSNKGLPAPIEKTISKTKGQPREPQVYTLPPSREEMTKNQ VSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPMLDSDGSFFLYSKLTVDKSRWQQGNV FSCSVMHEALHNHYTQKSLSLSPGK Parental 219R45 Heavy chain t signal sequence (SEQ ID NO:49) QVQLVQSGAEVKKPGASVKVSCKASGYTFTNYWMHWVRQAPGQGLEWMGDINPSNGRTSY KLKFKRRVTLSVDKSSSTAYMELSSLRSEDTAVYFCTIHYDDKYYPLMDYWGQGTLVTVS SASTKGPSVFPLAPCSRSTSESTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQS SGLYSLSSVVTVPSSNFGTQTYTCNVDHKPSNTKVDKTVERKCCVECPPCPAPPVAGPSV FLFPP<PKDTL ISRTPEVTCVVVDVSHEDPEVQFNWYVDGVEVHNAKTKPREEQFNSTF RVVSVLTVVHQDWLNGKEYKCKVSNKGLPAPIEKTISKTKGQPREPQVYTLPPSREEMTK CLVKGFYPSDIAVEWESNGQPENNYKTTPPMLDSDGSFFLYSKLTVDKSRWQQG NVFSCSVMHEALHNHYTQKSLSLSPGK Parental 21R79 Heavy chain variable region nucleotide sequence (SEQ ID NO:50) CAAGTGCAGCTCGTGCAGTCAGGGGCGGAGGTCAAGAAGCCGGGAGCATCGGTCAAAATC AAGGCCTCGGGGTACTCCTTTACTGCGTATTACATCCATTGGGTAAAGCAGGCG CCAGGGCAGGGATTGGAGTGGATTGGGTATATCGCCAATTACAATCGCGCGACGAACTAT AACCAGAAATTCAAGGGAAGGGTGACCTTCACAACGGATACATCGACATCGACGGCCTAC ATGGAACTTCGCAGCCTGCGATCAGATGACACGGCGGTATACTATTGCGCAAGAGATTAC GACTATGATGTGGGAATGGACTATTGGGGTCAAGGTACTCTGGTCACAGTCTCCTCC Parental 219R45 Heavy chain variable region nucleotide sequence (SEQ ID NO:51) CAGGTACAGCTCGTGCAATCGGGGGCAGAGGTCAAAAAGCCCGGTGCGTCGGTAAAGGTC AGCTGCAAAGCGTCAGGTTATACATTCACGAATTACTGGATGCATTGGGTCAGACAGGCC CCTGGACAAGGGCTTGAATGGATGGGAGATATCAATCCGTCGAACGGACGGACTAGCTAT AAGGAGAAGTTTAAGAGGCGCGTAACACTGTCGGTGGACAAATCGTCCTCAACGGCCTAC TTGTCATCCCTGCGGTCGGAAGATACGGCGGTCTACTTCTGTACTATCCACTAT GACGATAAGTACTACCCGCTTATGGACTACTGGGGTCAGGGAACATTGGTAACCGTGAGC Parental 21R79 Heavy chain nucleotide sequence with signal sequence (SEQ ID NO:52) ATGAAACACTTGTGGTTTTTCCTCTTGCTCGTGGCAGCTCCTCGGTGGGTACTTTCACAA GTGCAGCTCGTGCAGTCAGGGGCGGAGGTCAAGAAGCCGGGAGCATCGGTCAAAATCTCG TGTAAGGCCTCGGGGTACTCCTTTACTGCGTATTACATCCATTGGGTAAAGCAGGCGCCA GGGCAGGGATTGGAGTGGATTGGGTATATCGCCAATTACAATCGCGCGACGAACTATAAC CAGAAATTCAAGGGAAGGGTGACCTTCACAACGGATACATCGACATCGACGGCCTACATG GAACTTCGCAGCCTGCGATCAGATGACACGGCGGTATACTATTGCGCAAGAGATTACGAC TATGATGTGGGAATGGACTATTGGGGTCAAGGTACTCTGGTCACAGTCTCCTCCGCCAGC ACCAAGGGCCCTAGCGTCTTCCCTCTGGCTCCCTGCAGCAGGAGCACCAGCGAGAGCACA GCCGCCCTGGGCTGCCTGGTCAAGGACTACTTCCCCGAACCGGTGACGGTGTCGTGGAAC TCAGGCGCTCTGACCAGCGGCGTGCACACCTTCCCAGCTGTCCTACAGTCCTCAGGACTC TACTCCCTCAGCAGCGTGGTGACCGTGCCCTCCAGCAACTTCGGCACCCAGACCTACACC TGCAACGTAGATCACAAGCCCAGCAACACCAAGGTGGACAAGACAGTTGAGCGCAAATGT TGTGTCGAGTGCCCACCGTGCCCAGCACCACCTGTGGCAGGACCGTCAGTCTTCCTCTTC CCCCCAAAACCCAAGGACACCCTCATGATCTCCCGGACCCCTGAGGTCACGTGCGTGGTG GTGAGCCACGAAGACCCCGAGGTCCAGTTCAACTGGTACGTGGACGGCGTGGAG GTGCATAATGCCAAGACAAAGCCACGGGAGGAGCAGTTCAACAGCACGTTCCGTGTGGTC AGCGTCCTCACCGTTGTGCACCAGGACTGGCTGAACGGCAAGGAGTACAAGTGCAAGGTC TCCAACAAAGGCCTCCCAGCCCCCATCGAGAAAACCATCTCCAAAACCAAAGGGCAGCCC CGAGAACCACAGGTGTACACCCTGCCCCCATCCCGGGAGGAGATGACCAAGAACCAGGTC AGCCTGACCTGCCTGGTCAAAGGCTTCTACCCCAGCGACATCGCCGTGGAGTGGGAGAGC AATGGGCAGCCGGAGAACAACTACAAGACCACACCTCCCATGCTGGACTCCGACGGCTCC CTCTACAGCAAGCTCACCGTGGACAAGAGCAGGTGGCAGCAGGGGAACGTCTTC TCATGCTCCGTGATGCATGAGGCTCTGCACAACCACTACACGCAGAAGAGCCTCTCCCTG TCTCCGGGTAAA Parental 219R45 Heavy chain nucleotide sequence with signal ce (SEQ ID NO:53) ATGAAACACCTCTGGTTCTTTTTGCTCCTGGTGGCAGCTCCCCGATGGGTGCTTAGCCAG GTACAGCTCGTGCAATCGGGGGCAGAGGTCAAAAAGCCCGGTGCGTCGGTAAAGGTCAGC TGCAAAGCGTCAGGTTATACATTCACGAATTACTGGATGCATTGGGTCAGACAGGCCCCT GGACAAGGGCTTGAATGGATGGGAGATATCAATCCGTCGAACGGACGGACTAGCTATAAG GAGAAGTTTAAGAGGCGCGTAACACTGTCGGTGGACAAATCGTCCTCAACGGCCTACATG GAGTTGTCATCCCTGCGGTCGGAAGATACGGCGGTCTACTTCTGTACTATCCACTATGAC GATAAGTACTACCCGCTTATGGACTACTGGGGTCAGGGAACATTGGTAACCGTGAGCAGC GCGTCCACAAAGGGCCCTAGCGTCTTCCCTCTGGCTCCCTGCAGCAGGAGCACCAGCGAG GCCGCCCTGGGCTGCCTGGTCAAGGACTACTTCCCCGAACCGGTGACGGTGTCG TGGAACTCAGGCGCTCTGACCAGCGGCGTGCACACCTTCCCAGCTGTCCTACAGTCCTCA GGACTCTACTCCCTCAGCAGCGTGGTGACCGTGCCCTCCAGCAACTTCGGCACCCAGACC TACACCTGCAACGTAGATCACAAGCCCAGCAACACCAAGGTGGACAAGACAGTTGAGCGC AAATGTTGTGTCGAGTGCCCACCGTGCCCAGCACCACCTGTGGCAGGACCGTCAGTCTTC CTCTTCCCCCCAAAACCCAAGGACACCCTCATGATCTCCCGGACCCCTGAGGTCACGTGC GTGGTGGTGGACGTGAGCCACGAAGACCCCGAGGTCCAGTTCAACTGGTACGTGGACGGC GTGCATAATGCCAAGACAAAGCCACGGGAGGAGCAGTTCAACAGCACGTTCCGT GTGGTCAGCGTCCTCACCGTTGTGCACCAGGACTGGCTGAACGGCAAGGAGTACAAGTGC AAGGTCTCCAACAAAGGCCTCCCAGCCCCCATCGAGAAAACCATCTCCAAAACCAAAGGG CGAGAACCACAGGTGTACACCCTGCCCCCATCCCGGGAGGAGATGACCAAGAAC CAGGTCAGCCTGACCTGCCTGGTCAAAGGCTTCTACCCCAGCGACATCGCCGTGGAGTGG GAGAGCAATGGGCAGCCGGAGAACAACTACAAGACCACACCTCCCATGCTGGACTCCGAC GGCTCCTTCTTCCTCTACAGCAAGCTCACCGTGGACAAGAGCAGGTGGCAGCAGGGGAAC GTCTTCTCATGCTCCGTGATGCATGAGGCTCTGCACAACCACTACACGCAGAAGAGCCTC TCCCTGTCTCCGGGTAAA Parental 21R79 and 219R45 light chain variable region nucleotide sequence (SEQ ID NO:54) GACATCGTGATGACCCAGTCCCCTGACTCCCTGGCTGTGTCCCTGGGCGAGAGGGCCACC ATCTCCTGCAGAGCCAGCGAATCCGTCGATAATTATGGCATTTCCTTTATGAAGTGGTTC CAGCAGAAACCAGGACAGCCTCCTAAGCTGCTCATTTACGCTGCATCCAACCAAGGGTCC GGGGTCCCTGACAGGTTCTCCGGCAGCGGGTCCGGAACAGATTTCACTCTCACCATCAGC AGCCTGCAGGCTGAAGATGTGGCTGTCTATTACTGTCAGCAAAGCAAGGAGGTGCCTTGG ACATTCGGAGGAGGGACCAAGGTGGAAATCAAA Parental 21R79 and 219R45 light chain nucleotide sequence (SEQ ID NO:55) ATGGTGCTCCAGACCCAGGTCTTCATTTCCCTGCTGCTCTGGATCAGCGGAGCCTACGGG GACATCGTGATGACCCAGTCCCCTGACTCCCTGGCTGTGTCCCTGGGCGAGAGGGCCACC ATCTCCTGCAGAGCCAGCGAATCCGTCGATAATTATGGCATTTCCTTTATGAAGTGGTTC CAGCAGAAACCAGGACAGCCTCCTAAGCTGCTCATTTACGCTGCATCCAACCAAGGGTCC GGGGTCCCTGACAGGTTCTCCGGCAGCGGGTCCGGAACAGATTTCACTCTCACCATCAGC AGCCTGCAGGCTGAAGATGTGGCTGTCTATTACTGTCAGCAAAGCAAGGAGGTGCCTTGG ACATTCGGAGGAGGGACCAAGGTGGAAATCAAACGTACGGTGGCTGCCCCCTCCGTCTTC CCCCCCAGCGATGAGCAGCTGAAAAGCGGCACTGCCAGCGTGGTGTGCCTGCTG AATAACTTCTATCCCCGGGAGGCCAAAGTGCAGTGGAAGGTGGATAACGCCCTCCAAAGC GGCAACTCCCAGGAGAGCGTCACAGAGCAGGACAGCAAGGACAGCACCTACAGCCTCAGC AGCACCCTGACCCTGAGCAAAGCCGACTACGAGAAACACAAAGTCTACGCCTGCGAAGTC ACCCATCAGGGCCTGAGCAGCCCCGTCACAAAGAGCTTCAACAGGGGCGAGTGTTGA 21R75 Heavy chain without predicted signal sequence (SEQ ID NO:56) QVQLVQSGAEVKKPGASVKISCKASGYSFTAYYIHWVKQAPGQGLEWIGYIAGYKDATNY NQKFKGRVTFTTDTSTSTAYMELRSLRSDDTAVYYCARDYDYDVGMDYWGQGTLVTVSSA VFPLAPCSRSTSESTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSG LYSLSSVVTVPSSNFGTQTYTCNVDHKPSNTKVDKTVERKCCVECPPCPAPPVAGPSVFL FPPKPKDTLMISRTPLVTCVVVDVSHEDBEVQFNWYVDGVEVHNAKTKPREEQFNSTFRV VSVLTVVHQDWLNGKEYKCKVSNKGLPAPIEKTISKTKGQPREPQVYTL?PSREEMTKNQ VSLTCLVEGEYPSD-AVLWESNGQBLNNYKTTPPMLDSDGSFFLYSELTVDKSRWQQGNV FSCSVMHEALHNHYTQKSLSLSPGK 21R75 Heavy chain with predicted signal sequence (underlined) (SEQ ID NO:57) MKHLWFFLLLVAAPRWVLSQVQLVQSGAEVKKPGASVKISCKASGYSFTAYYTHWVKQAP GQGHflW GY AGYKDATNYNQKFKGRVTFTTDTSTSTAYMELRSLRSDDTAVYYCARDYD YDVG DYWGQGTLVTVSSASTKGPSVFPLAPCSRSTSESTAALGCLVKDYFPEPVTVSWN SGALTSGVHTFPAVTQSSGLYSTSSVVTVPSSNFGTQTYTCNVDHKPSNTKVDKTVERKC CVECPPCPAPPVAGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVQFNWYVDGVE VH A(TKPREEQFNSTFRVVSVLTVVHQDWLNGKEYKCKVSNKGLPAPIEKTISKTKGQP REPQVYTLPPSREEMTKNQVSLTCLVEGFYPSDIAVEWESNGQPENNYKTTPPMLDSDGS FFLYSELTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK 21R75 Heavy chain le region (SEQ ID NO:58) QVQLVQSGAEVKKPGASVKISCKASGYSFTAYYIHWVKQAPGQGLEWIGYIAGYKDATNY NQKFKGRVTFTTDTSTSTAYMELRSLRSDDTAVYYCARDYDYDVGMDYWGQGTLVTVSS 21R75 Heavy chain CDR2 (SEQ ID NO:59) Y-AGYKDATNYNQKFKG 21R75 Heavy chain nucleotide sequence with signal sequence (13B Version 1) (SEQ ID NO: 60) ATGAAGCACCTGTGGTTCTTTCTGCTGCTGGTGGCCGCTCCCAGATGGGTGCTGTCCCAG GTGCAGCTGGTGCAGTCTGGCGCCGAAGTGAAGAAACCTGGCGCCTCCGTGAAGATCTCC TGCAAGGCCTCCGGCTACTCCTTCACCGCCTACTACATCCACTGGGTCAAGCAGGCCCCT GGACAGGGCCTGGAATGGATCGGCTATATCGCCGGCTACAAGGACGCCACCAACTACAAC TTCAAGGGCAGAGTGACCTTCACCACCGACACCTCCACCTCTACCGCCTACATG GAACTGCGGTCCCTGCGGAGCGACGACACCGCCGTGTACTACTGCGCCAGAGACTACGAC TACGACGTGGGCATGGACTACTGGGGCCAGGGCACACTCGTGACCGTGTCCTCTGCTTCC ACCAAGGGCCCCTCCGTGTTTCCTCTGGCCCCTTGCTCCAGATCCACCTCCGAGTCTACC GCCGCTCTGGGCTGCCTCGTGAAGGACTACTTCCCCGAGCCCGTGACAGTGTCTTGGAAC TCTGGCGCCCTGACCTCCGGCGTGCACACCTTTCCAGCTGTGCTGCAGTCCTCCGGCCTG TACTCCCTGTCCTCCGTCGTGACTGTGCCCTCCTCCAACTTCGGCACCCAGACCTACACC TGTAACGTGGACCACAAGCCCTCCAACACCAAGGTGGACAAGACCGTGGAACGGAAGTGC TGCGTGGAATGCCCCCCTTGTCCTGCCCCTCCTGTGGCTGGCCCTAGCGTGTTCCTGTTC CCCCCAAAGCCCAAGGACACCCTGATGATCTCCCGGACCCCCGAAGTGACCTGCGTGGTG GTGGATGTGTCCCACGAGGACCCCGAGGTGCAGTTCAATTGGTACGTGGACGGCGTGGAA GTGCACAACGCCAAGACCAAGCCCAGAGAGGAACAGTTCAACTCCACCTTCCGGGTGGTG TCCGTGCTGACCGTGGTGCATCAGGACTGGCTGAACGGCAAAGAGTACAAGTGCAAGGTG TCCAACAAGGGCCTGCCTGCCCCCATCGAAAAGACCATCTCTAAGACCAAGGGACAGCCC CCCCAGGTGTACACACTGCCTCCATCCCGGGAAGAGATGACCAAGAACCAGGTG TCCCTGACCTGTCTGGTGGAAGGCTTCTACCCCTCCGATATCGCCGTGGAATGGGAGTCC AACGGCCAGCCCGAGAACAACTACAAGACCACCCCCCCCATGCTGGACTCCGACGGCTCA TTCTTCCTGTACAGCGAGCTGACAGTGGACAAGTCCCGGTGGCAGCAGGGCAACGTGTTC TCCTGCTCCGTGATGCACGAGGCCCTGCACAACCACTACACCCAGAAGTCCCTGTCCCTG AGCCCCGGCAAG 21R75 Heavy chain nucleotide sequence with signal sequence (13B Version 1T) (SEQ ID NO:77) ATGAAGCACCTGTGGTTCTTTCTGCTGCTGGTGGCCGCTCCCAGATGGGTGCTGTCTCAG GTGCAGCTGGTGCAGTCTGGCGCCGAAGTGAAGAAACCTGGCGCCTCCGTGAAGATCTCC TGCAAGGCCTCCGGCTACTCCTTCACCGCCTACTACATCCACTGGGTCAAGCAGGCCCCT GGACAGGGCCTGGAATGGATCGGCTATATCGCCGGCTACAAGGACGCCACCAACTACAAC CAGAAATTCAAGGGCAGAGTGACCTTCACCACCGACACCTCCACCTCTACCGCCTACATG GAACTGCGGTCCCTGCGGAGCGACGACACCGCCGTGTACTACTGCGCCAGAGACTACGAC TACGACGTGGGCATGGACTACTGGGGCCAGGGCACACTCGTGACCGTGTCCTCTGCTTCC ACCAAGGGCCCCTCCGTGTTTCCTCTGGCCCCTTGCTCCAGATCCACCTCCGAGTCTACC GCCGCTCTGGGCTGCCTCGTGAAGGACTACTTCCCCGAGCCCGTGACAGTGTCTTGGAAC GCCCTGACCTCCGGCGTGCACACCTTTCCAGCTGTGCTGCAGTCCTCCGGCCTG TACTCCCTGTCCTCCGTCGTGACTGTGCCCTCCTCCAACTTCGGCACCCAGACCTACACC TGTAACGTGGACCACAAGCCCTCCAACACCAAGGTGGACAAGACCGTGGAACGGAAGTGC TGCGTGGAATGCCCCCCTTGTCCTGCCCCTCCTGTGGCTGGCCCTAGCGTGTTCCTGTTC AAGCCCAAGGACACCCTGATGATCTCCCGGACCCCCGAAGTGACCTGCGTGGTG GTGGATGTGTCCCACGAGGACCCCGAGGTGCAGTTCAATTGGTACGTGGACGGCGTGGAA GTGCACAACGCCAAGACCAAGCCCAGAGAGGAACAGTTCAACTCCACCTTCCGGGTGGTG TCCGTGCTGACCGTGGTGCATCAGGACTGGCTGAACGGCAAAGAGTACAAGTGCAAGGTG TCCAACAAGGGCCTGCCTGCCCCCATCGAAAAGACCATCTCTAAGACCAAGGGACAGCCC CGCGAGCCCCAGGTGTACACACTGCCTCCATCCCGGGAAGAGATGACCAAGAACCAGGTG TCCCTGACCTGTCTGGTGGAAGGCTTCTACCCCTCCGATATCGCCGTGGAATGGGAGTCC AACGGCCAGCCCGAGAACAACTACAAGACCACCCCCCCCATGCTGGACTCCGACGGCTCA TTCTTCCTGTACAGCGAGCTGACAGTGGACAAGTCCCGGTGGCAGCAGGGCAACGTGTTC TCCTGCTCCGTGATGCACGAGGCCCTGCACAACCACTACACCCAGAAGTCCCTGTCCCTG AGCCCCGGCAAG 21R75 Heavy chain nucleotide sequence with signal sequence (13B n Sl-2) (SEQ ID NO:61) ATGAAGCACCTGTGGTTCTTTCTGCTGCTGGTGGCCGCTCCCAGATGGGTGCTGTCCCAG GTTCAGCTAGTTCAGTCTGGAGCGGAAGTTAAGAAACCTGGAGCATCCGTGAAAATAAGT TGCAAGGCATCCGGTTACTCGTTCACCGCATACTATATCCACTGGGTTAAACAGGCACCA GGACAGGGACTTGAATGGATCGGATATATCGCTGGATATAAAGATGCTACAAACTATAAC TTCAAAGGACGCGTGACTTTCACAACTGACACCTCAACCTCGACAGCATACATG CGGTCCCTACGGTCTGACGACACTGCCGTTTACTATTGCGCTAGAGATTATGAT TATGATGTTGGAATGGACTATTGGGGCCAGGGAACACTGGTGACAGTGTCTTCTGCATCC ACTAAGGGACCATCCGTGTTCCCTTTGGCCCCTTGCTCTCGTTCGACCTCTGAATCGACT GCCGCTCTGGGATGCCTCGTGAAAGATTACTTCCCTGAGCCTGTGACCGTTTCCTGGAAC TCGGGCGCCCTAACCTCTGGCGTGCACACATTCCCTGCCGTGCTACAGTCTTCTGGCCTA TACTCTTTATCTTCGGTTGTTACCGTACCTTCTTCTAACTTCGGAACCCAAACTTACACC TGTAACGTAGACCACAAGCCTTCGAACACCAAGGTGGACAAGACTGTTGAGCGAAAGTGC TGCGTTGAGTGCCCTCCATGTCCTGCACCTCCTGTGGCTGGCCCTTCTGTGTTCCTGTTC CCTCCAAAACCTAAGGACACTCTAATGATCTCTCGGACTCCTGAGGTGACTTGCGTGGTT GTGGACGTGTCCCACGAGGACCCTGAGGTGCAGTTCAATTGGTACGTGGACGGAGTCGAG GTGCACAATGCAAAGACCAAGCCTCGGGAGGAACAGTTCAACTCCACCTTCCGGGTGGTT TCTGTGTTGACCGTTGTGCACCAAGACTGGCTGAACGGCAAAGAATACAAGTGCAAGGTG TCCAACAAGGGCCTGCCTGCCCCTATCGAAAAGACCATCAGCAAGACCAAGGGCCAGCCT CGCGAGCCTCAGGTGTACACCCTGCCTCCCAGCCGGGAAGAAATGACCAAGAACCAGGTG TCCCTGACCTGTCTGGTGGAGGGCTTCTACCCTTCCGACATCGCCGTTGAGTGGGAGTCT AACGGACAGCCGGAGAACAACTACAAGACTACGCCTCCAATGCTGGACTCCGACGGCTCC TTCTTCCTGTACTCCGAACTGACCGTGGACAAGTCCCGGTGGCAGCAGGGCAACGTGTTC TCATGCTCCGTAATGCACGAAGCCTTGCACAATCACTACACTCAAAAGTCCCTATCCTTA TCTCCTGGCAAG 21R83 Heavy chain without predicted signal sequence (SEQ ID NO:62) QVQLVQSGAEVKKPGASVKISCKASGYSFTAYYIHWVKQAPGQGLEWIGYISNYNRATNY NQKFKGRVTFTTDTSTSTAYMELRSLRSDDTAVYYCARDYDYDVGMDYWGQGTLVTVSSA STKGPSVFPLAPCSRSTSESTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSG LYSLSSVVTVPSSNFGTQTYTCNVDHKPSNTKVDKTVERKCCVECPPCPAPPVAGPSVFL FPPKPKDTLMISRTPEVTCVVVDVSHEDPEVQFNWYVDGVEVHNAKTKPREEQFNSTFRV VSVLTVVHQDWLNGKEYKCKVSNKGLPAPIEKTISKTKGQPREPQVYTLPPSREEMTKNQ VSLTCLVEGFYPSDIAVEWESNGQPENNYKTTPPMLDSDGSFFLYSELTVDKSRWQQGNV FSCSVMHEALHNHYTQKSLSLSPGK 21R83 Heavy chain with predicted signal sequence (underlined) (SEQ ID NO:63) MKHLWFFLLLVAAPRWVLSQVQLVQSGAEVKKPGASVKISCKASGYSFTAYYIHWVKQA? GQGLEWIGYISNYNRATNYNQKFKGRVTFTTDTSTSTAYMELRSLRSDDTAVYYCARDYD YDVG DYWGQGTLVTVSSASTKGPSVFPLAPCSRSTSESTAALGCLVKDYFPEPVTVSW- SGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSNFGTQTYTCNVDHKPSNTKVDKTVERKC CVECPPCPAPPVAGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVQFNWYVDGVE Vi AKTKPRflflQhNSTFRVVSVLTVVHQDWLNGKEYKCKVSNKGLPAPIEKTISKTKGQP REPQVYTLPPSREEMTKNQVSLTCLVEGFYPSDIAVEWESNGQPENNYKTTPPMLDSDGS FFHYS?LTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK 21R83 Heavy chain le region (SEQ ID NO:64) QVQLVQSGAEVKKPGASVKISCKASGYSFTAYYIHWVKQAPGQGLEWIGYISNYNRATNY RVTFTTDTSTSTAYMELRSLRSDDTAVYYCARDYDYDVGMDYWGQGTLVTVSS 21R83 Heavy chain CDR2 (SEQ ID NO:65) YISNYNRATNYNQKFKG 21R83 Heavy chain nucleotide sequence with signal sequence underlined (13B n 1) (SEQ ID NO:66) ATGAAGCACCTGTGGTTCTTTCTGCTGCTGGTGGCCGCTCCCAGATGGGTGCTGTCCCAG GTGCAGCTGGTGCAGTCTGGCGCCGAAGTGAAGAAACCTGGCGCCTCCGTGAAGATCTCC TGCAAGGCCTCCGGCTACTCCTTCACCGCCTACTACATCCACTGGGTCAAGCAGGCCCCT GGACAGGGCCTGGAATGGATCGGCTACATCTCCAACTACAACCGGGCCACCAATTACAAC CAGAAATTCAAGGGCCGCGTGACCTTCACCACCGACACCTCTACCTCTACCGCCTACATG GAACTGCGGTCCCTGCGGAGCGACGACACCGCCGTGTACTACTGCGCCAGAGACTACGAC TACGACGTGGGCATGGACTACTGGGGCCAGGGCACACTCGTGACCGTGTCTAGCGCTTCC ACCAAGGGCCCCTCCGTGTTTCCTCTGGCCCCTTGCTCCAGATCCACCTCCGAGTCTACC GCCGCTCTGGGCTGCCTCGTGAAGGACTACTTCCCCGAGCCCGTGACAGTGTCCTGGAAC TCTGGCGCTCTGACCTCCGGCGTGCACACCTTTCCAGCTGTGCTGCAGTCCTCCGGCCTG CTGTCCTCCGTCGTGACTGTGCCCTCCTCCAACTTCGGCACCCAGACCTACACC TGTAACGTGGACCACAAGCCCTCCAACACCAAGGTGGACAAGACCGTGGAACGGAAGTGC TGCGTGGAATGCCCCCCTTGTCCTGCCCCTCCTGTGGCTGGCCCTAGCGTGTTCCTGTTC CCCCCAAAGCCCAAGGACACCCTGATGATCTCCCGGACCCCCGAAGTGACCTGCGTGGTG GTGGATGTGTCCCACGAGGACCCCGAGGTGCAGTTCAATTGGTACGTGGACGGCGTGGAA GTGCACAACGCCAAGACCAAGCCCAGAGAGGAACAGTTCAACTCCACCTTCCGGGTGGTG TCCGTGCTGACCGTGGTGCATCAGGACTGGCTGAACGGCAAAGAGTACAAGTGCAAGGTG AAGGGCCTGCCTGCCCCCATCGAAAAGACCATCTCTAAGACCAAGGGACAGCCC CGCGAGCCCCAGGTGTACACACTGCCTCCATCCCGGGAAGAGATGACCAAGAACCAGGTG TCCCTGACCTGTCTGGTGGAAGGCTTCTACCCCTCCGATATCGCCGTGGAATGGGAGTCC AACGGCCAGCCCGAGAACAACTACAAGACCACCCCCCCCATGCTGGACTCCGACGGCTCA TTCTTCCTGTACAGCGAGCTGACAGTGGACAAGTCCCGGTGGCAGCAGGGCAACGTGTTC TCCTGCTCCGTGATGCACGAGGCCCTGCACAACCACTACACCCAGAAGTCCCTGTCCCTG AGCCCCGGCAAG 21R83 Heavy chain nucleotide sequence with signal sequence underlined (13B Version 1T) (SEQ ID NO:78) ATGAAGCACCTGTGGTTCTTTCTGCTGCTGGTGGCCGCTCCCAGATGGGTGCTGTCTCAG GTGCAGCTGGTGCAGTCTGGCGCCGAAGTGAAGAAACCTGGCGCCTCCGTGAAGATCTCC TGCAAGGCCTCCGGCTACTCCTTCACCGCCTACTACATCCACTGGGTCAAGCAGGCCCCT GGACAGGGCCTGGAATGGATCGGCTACATCTCCAACTACAACCGGGCCACCAATTACAAC CAGAAATTCAAGGGCCGCGTGACCTTCACCACCGACACCTCTACCTCTACCGCCTACATG GAACTGCGGTCCCTGCGGAGCGACGACACCGCCGTGTACTACTGCGCCAGAGACTACGAC TACGACGTGGGCATGGACTACTGGGGCCAGGGCACACTCGTGACCGTGTCTAGCGCTTCC ACCAAGGGCCCCTCCGTGTTTCCTCTGGCCCCTTGCTCCAGATCCACCTCCGAGTCTACC GCCGCTCTGGGCTGCCTCGTGAAGGACTACTTCCCCGAGCCCGTGACAGTGTCCTGGAAC TCTGGCGCTCTGACCTCCGGCGTGCACACCTTTCCAGCTGTGCTGCAGTCCTCCGGCCTG TACTCCCTGTCCTCCGTCGTGACTGTGCCCTCCTCCAACTTCGGCACCCAGACCTACACC TGTAACGTGGACCACAAGCCCTCCAACACCAAGGTGGACAAGACCGTGGAACGGAAGTGC GAATGCCCCCCTTGTCCTGCCCCTCCTGTGGCTGGCCCTAGCGTGTTCCTGTTC CCCCCAAAGCCCAAGGACACCCTGATGATCTCCCGGACCCCCGAAGTGACCTGCGTGGTG GTGGATGTGTCCCACGAGGACCCCGAGGTGCAGTTCAATTGGTACGTGGACGGCGTGGAA GTGCACAACGCCAAGACCAAGCCCAGAGAGGAACAGTTCAACTCCACCTTCCGGGTGGTG TCCGTGCTGACCGTGGTGCATCAGGACTGGCTGAACGGCAAAGAGTACAAGTGCAAGGTG AAGGGCCTGCCTGCCCCCATCGAAAAGACCATCTCTAAGACCAAGGGACAGCCC CGCGAGCCCCAGGTGTACACACTGCCTCCATCCCGGGAAGAGATGACCAAGAACCAGGTG TCCCTGACCTGTCTGGTGGAAGGCTTCTACCCCTCCGATATCGCCGTGGAATGGGAGTCC AACGGCCAGCCCGAGAACAACTACAAGACCACCCCCCCCATGCTGGACTCCGACGGCTCA TTCTTCCTGTACAGCGAGCTGACAGTGGACAAGTCCCGGTGGCAGCAGGGCAACGTGTTC TCCTGCTCCGTGATGCACGAGGCCCTGCACAACCACTACACCCAGAAGTCCCTGTCCCTG AGCCCCGGCAAG 21R75 Heavy chain nucleotide sequence with signal sequence underlined (13B Version S1-2) (SEQ ID NO:67) ATGAAGCACCTGTGGTTCTTTCTGCTGCTGGTGGCCGCTCCCAGATGGGTGCTGTCCCAG GTTCAGCTAGTTCAGTCTGGAGCGGAAGTTAAGAAACCTGGAGCATCCGTGAAAATAAGT TGCAAGGCATCCGGTTACTCGTTCACCGCATACTATATCCACTGGGTTAAACAGGCACCA GGACAGGGACTTGAATGGATCGGATATATCGCTGGATATAAAGATGCTACAAACTATAAC CAAAAATTCAAAGGACGCGTGACTTTCACAACTGACACCTCAACCTCGACAGCATACATG GAATTACGGTCCCTACGGTCTGACGACACTGCCGTTTACTATTGCGCTAGAGATTATGAT TATGATGTTGGAATGGACTATTGGGGCCAGGGAACACTGGTGACAGTGTCTTCTGCATCC ACTAAGGGACCATCCGTGTTCCCTTTGGCCCCTTGCTCTCGTTCGACCTCTGAATCGACT GCCGCTCTGGGATGCCTCGTGAAAGATTACTTCCCTGAGCCTGTGACCGTTTCCTGGAAC TCGGGCGCCCTAACCTCTGGCGTGCACACATTCCCTGCCGTGCTACAGTCTTCTGGCCTA TACTCTTTATCTTCGGTTGTTACCGTACCTTCTTCTAACTTCGGAACCCAAACTTACACC TGTAACGTAGACCACAAGCCTTCGAACACCAAGGTGGACAAGACTGTTGAGCGAAAGTGC GAGTGCCCTCCATGTCCTGCACCTCCTGTGGCTGGCCCTTCTGTGTTCCTGTTC CCTCCAAAACCTAAGGACACTCTAATGATCTCTCGGACTCCTGAGGTGACTTGCGTGGTT GTGGACGTGTCCCACGAGGACCCTGAGGTGCAGTTCAATTGGTACGTGGACGGAGTCGAG GTGCACAATGCAAAGACCAAGCCTCGGGAGGAACAGTTCAACTCCACCTTCCGGGTGGTT TCTGTGTTGACCGTTGTGCACCAAGACTGGCTGAACGGCAAAGAATACAAGTGCAAGGTG TCCAACAAGGGCCTGCCTGCCCCTATCGAAAAGACCATCAGCAAGACCAAGGGCCAGCCT CCTCAGGTGTACACCCTGCCTCCCAGCCGGGAAGAAATGACCAAGAACCAGGTG TCCCTGACCTGTCTGGTGGAGGGCTTCTACCCTTCCGACATCGCCGTTGAGTGGGAGTCT AACGGACAGCCGGAGAACAACTACAAGACTACGCCTCCAATGCTGGACTCCGACGGCTCC TTCTTCCTGTACTCCGAACTGACCGTGGACAAGTCCCGGTGGCAGCAGGGCAACGTGTTC TCATGCTCCGTAATGCACGAAGCCTTGCACAATCACTACACTCAAAAGTCCCTATCCTTA TCTCCTGGCAAG 21R75 Heavy chain variable region nucleotide sequence (13B Version 1) (SEQ ID NO:68) CAGGTGCAGCTGGTGCAGTCTGGCGCCGAAGTGAAGAAACCTGGCGCCTCCGTGAAGATC TCCTGCAAGGCCTCCGGCTACTCCTTCACCGCCTACTACATCCACTGGGTCAAGCAGGCC CCTGGACAGGGCCTGGAATGGATCGGCTATATCGCCGGCTACAAGGACGCCACCAACTAC AAATTCAAGGGCAGAGTGACCTTCACCACCGACACCTCCACCTCTACCGCCTAC ATGGAACTGCGGTCCCTGCGGAGCGACGACACCGCCGTGTACTACTGCGCCAGAGACTAC GACTACGACGTGGGCATGGACTACTGGGGCCAGGGCACACTCGTGACCGTGTCCTCT 21R75 Heavy chain variable region nucleotide sequence (13B Version 2) (SEQ ID NO:69) CAGGTTCAGCTAGTTCAGTCTGGAGCGGAAGTTAAGAAACCTGGAGCATCCGTGAAAATA AGTTGCAAGGCATCCGGTTACTCGTTCACCGCATACTATATCCACTGGGTTAAACAGGCA CCAGGACAGGGACTTGAATGGATCGGATATATCGCTGGATATAAAGATGCTACAAACTAT AACCAAAAATTCAAAGGACGCGTGACTTTCACAACTGACACCTCAACCTCGACAGCATAC ATGGAATTACGGTCCCTACGGTCTGACGACACTGCCGTTTACTATTGCGCTAGAGATTAT -1ll- GATTATGATGTTGGAATGGACTATTGGGGCCAGGGAACACTGGTGACAGTGTCTTCT 21R83 Heavy chain variable region tide sequence (133 Version 1) (SEQ ID NO:70) CAGGTGCAGCTGGTGCAGTCTGGCGCCGAAGTGAAGAAACCTGGCGCCTCCGTGAAGATC TCCTGCAAGGCCTCCGGCTACTCCTTCACCGCCTACTACATCCACTGGGTCAAGCAGGCC CCTGGACAGGGCCTGGAATGGATCGGCTACATCTCCAACTACAACCGGGCCACCAATTAC AACCAGAAATTCAAGGGCCGCGTGACCTTCACCACCGACACCTCTACCTCTACCGCCTAC ATGGAACTGCGGTCCCTGCGGAGCGACGACACCGCCGTGTACTACTGCGCCAGAGACTAC GACTACGACGTGGGCATGGACTACTGGGGCCAGGGCACACTCGTGACCGTGTCTAGC 21R75 Heavy chain variable region nucleotide ce (13B n 2) (SEQ ID NO:71) CAGGTTCAGCTAGTTCAGTCTGGAGCGGAAGTTAAGAAACCTGGAGCATCCGTGAAAATA AGTTGCAAGGCATCCGGTTACTCGTTCACCGCATACTATATCCACTGGGTTAAACAGGCA CCAGGACAGGGACTTGAATGGATCGGATATATCGCTGGATATAAAGATGCTACAAACTAT AACCAAAAATTCAAAGGACGCGTGACTTTCACAACTGACACCTCAACCTCGACAGCATAC ATGGAATTACGGTCCCTACGGTCTGACGACACTGCCGTTTACTATTGCGCTAGAGATTAT GATTATGATGTTGGAATGGACTATTGGGGCCAGGGAACACTGGTGACAGTGTCTTCT 21R83 Heavy chain nucleotide sequence with signal sequence underlined (13B Version 2) (SEQ ID NO:72) ATGAAGCACCTATGGTTCTTTCTATTATTAGTGGCCGCTCCCCGTTGGGTGTTATCGCAG GTTCAGCTAGTTCAGTCTGGAGCGGAAGTTAAGAAACCTGGAGCATCCGTGAAAATAAGT TGCAAGGCATCCGGTTACTCGTTCACCGCATACTATATCCACTGGGTTAAACAGGCACCA GGACAGGGACTTGAATGGATCGGATATATCTCCAATTATAATAGAGCTACAAACTATAAC CAAAAATTCAAAGGACGCGTGACTTTCACAACTGACACCTCAACCTCGACAGCATACATG GAATTACGGTCCCTACGGTCTGACGACACTGCCGTTTACTATTGCGCTAGAGATTATGAT TATGATGTTGGAATGGACTATTGGGGCCAGGGAACACTGGTGACAGTGTCTTCTGCATCC ACTAAGGGACCATCCGTGTTCCCTTTGGCCCCTTGCTCTCGTTCGACCTCTGAATCGACT GCCGCTCTGGGATGCCTCGTGAAAGATTACTTCCCTGAGCCTGTGACCGTTTCCTGGAAC TCGGGCGCCCTAACCTCTGGCGTGCACACATTCCCTGCCGTGCTACAGTCTTCTGGCCTA TACTCTTTATCTTCGGTTGTTACCGTACCTTCTTCTAACTTCGGAACCCAAACTTACACC GTAGACCACAAGCCTTCGAACACCAAGGTGGACAAGACTGTTGAGCGAAAGTGC TGCGTTGAGTGCCCTCCATGTCCTGCACCTCCTGTGGCTGGCCCTTCTGTGTTCCTGTTC AAACCTAAGGACACTCTAATGATCTCTCGGACTCCTGAGGTGACTTGCGTGGTT GTGGACGTGTCCCACGAGGACCCTGAGGTGCAGTTCAATTGGTACGTGGACGGAGTCGAG GTGCACAATGCAAAGACCAAGCCTCGGGAGGAACAGTTCAACTCCACCTTCCGGGTGGTT TCTGTGTTGACCGTTGTGCACCAAGACTGGCTGAACGGCAAAGAATACAAGTGCAAGGTG AAGGGCCTGCCTGCCCCTATCGAAAAGACCATCAGCAAGACCAAGGGCCAGCCT CGCGAGCCTCAGGTGTACACCCTGCCTCCCAGCCGGGAAGAAATGACCAAGAACCAGGTG TCCCTGACCTGTCTGGTGGAGGGCTTCTACCCTTCCGACATCGCCGTTGAGTGGGAGTCT AACGGACAGCCGGAGAACAACTACAAGACTACGCCTCCAATGCTGGACTCCGACGGCTCC TTCTTCCTGTACTCCGAACTGACCGTGGACAAGTCCCGGTGGCAGCAGGGCAACGTGTTC TCATGCTCCGTAATGCACGAAGCCTTGCACAATCACTACACTCAAAAGTCCCTATCCTTA TCTCCTGGCAAGTAG 21R83 Heavy chain variable region nucleotide sequence (13B Version 2) (SEQ ID NO:73) CAGGTTCAGCTAGTTCAGTCTGGAGCGGAAGTTAAGAAACCTGGAGCATCCGTGAAAATA AGTTGCAAGGCATCCGGTTACTCGTTCACCGCATACTATATCCACTGGGTTAAACAGGCA CCAGGACAGGGACTTGAATGGATCGGATATATCTCCAATTATAATAGAGCTACAAACTAT AACCAAAAATTCAAAGGACGCGTGACTTTCACAACTGACACCTCAACCTCGACAGCATAC ATGGAATTACGGTCCCTACGGTCTGACGACACTGCCGTTTACTATTGCGCTAGAGATTAT GATTATGATGTTGGAATGGACTATTGGGGCCAGGGAACACTGGTGACAGTGTCTTCT -ll2- 21R75 Heavy chain nucleotide sequence with signal sequence underlined (13B Version 2) (SEQ ID NO:74) ATGAAGCACCTATGGTTCTTTCTATTATTAGTGGCCGCTCCCCGTTGGGTGTTATCGCAG GTTCAGCTAGTTCAGTCTGGAGCGGAAGTTAAGAAACCTGGAGCATCCGTGAAAATAAGT TGCAAGGCATCCGGTTACTCGTTCACCGCATACTATATCCACTGGGTTAAACAGGCACCA GGACAGGGACTTGAATGGATCGGATATATCGCTGGATATAAAGATGCTACAAACTATAAC CAAAAATTCAAAGGACGCGTGACTTTCACAACTGACACCTCAACCTCGACAGCATACATG GAATTACGGTCCCTACGGTCTGACGACACTGCCGTTTACTATTGCGCTAGAGATTATGAT TATGATGTTGGAATGGACTATTGGGGCCAGGGAACACTGGTGACAGTGTCTTCTGCATCC ACTAAGGGACCATCCGTGTTCCCTTTGGCCCCTTGCTCTCGTTCGACCTCTGAATCGACT GCCGCTCTGGGATGCCTCGTGAAAGATTACTTCCCTGAGCCTGTGACCGTTTCCTGGAAC TCGGGCGCCCTAACCTCTGGCGTGCACACATTCCCTGCCGTGCTACAGTCTTCTGGCCTA TACTCTTTATCTTCGGTTGTTACCGTACCTTCTTCTAACTTCGGAACCCAAACTTACACC TGTAACGTAGACCACAAGCCTTCGAACACCAAGGTGGACAAGACTGTTGAGCGAAAGTGC TGCGTTGAGTGCCCTCCATGTCCTGCACCTCCTGTGGCTGGCCCTTCTGTGTTCCTGTTC CCTCCAAAACCTAAGGACACTCTAATGATCTCTCGGACTCCTGAGGTGACTTGCGTGGTT GTGTCCCACGAGGACCCTGAGGTGCAGTTCAATTGGTACGTGGACGGAGTCGAG GTGCACAATGCAAAGACCAAGCCTCGGGAGGAACAGTTCAACTCCACCTTCCGGGTGGTT TCTGTGTTGACCGTTGTGCACCAAGACTGGCTGAACGGCAAAGAATACAAGTGCAAGGTG TCCAACAAGGGCCTGCCTGCCCCTATCGAAAAGACCATCAGCAAGACCAAGGGCCAGCCT CGCGAGCCTCAGGTGTACACCCTGCCTCCCAGCCGGGAAGAAATGACCAAGAACCAGGTG TCCCTGACCTGTCTGGTGGAGGGCTTCTACCCTTCCGACATCGCCGTTGAGTGGGAGTCT AACGGACAGCCGGAGAACAACTACAAGACTACGCCTCCAATGCTGGACTCCGACGGCTCC TTCTTCCTGTACTCCGAACTGACCGTGGACAAGTCCCGGTGGCAGCAGGGCAACGTGTTC TCATGCTCCGTAATGCACGAAGCCTTGCACAATCACTACACTCAAAAGTCCCTATCCTTA TCTCCTGGCAAGTAG 21M18 Heavy chain nucleotide sequence on 2) (SEQ ID NO:75) ATGAAGCACCTATGGTTCTTTCTATTATTAGTGGCCGCTCCCCGTTGGGTGTTATCGCAG GTTCAGCTAGTTCAGTCTGGAGCGGAAGTTAAGAAACCTGGAGCATCCGTGAAAATAAGT TGCAAGGCATCCGGTTACTCGTTCACCGCATACTATATCCACTGGGTTAAACAGGCACCA GGACAGGGACTTGAATGGATCGGATATATCTCCTCTTATAATGGAGCTACAAACTATAAC CAAAAATTCAAAGGACGCGTGACTTTCACAACTGACACCTCAACCTCGACAGCATACATG GAATTACGGTCCCTACGGTCTGACGACACTGCCGTTTACTATTGCGCTAGAGATTATGAT TATGATGTTGGAATGGACTATTGGGGCCAGGGAACACTGGTGACAGTGTCTTCTGCATCC ACTAAGGGACCATCCGTGTTCCCTTTGGCCCCTTGCTCTCGTTCGACCTCTGAATCGACT GCCGCTCTGGGATGCCTCGTGAAAGATTACTTCCCTGAGCCTGTGACCGTTTCCTGGAAC TCGGGCGCCCTAACCTCTGGCGTGCACACATTCCCTGCCGTGCTACAGTCTTCTGGCCTA TACTCTTTATCTTCGGTTGTTACCGTACCTTCTTCTAACTTCGGAACCCAAACTTACACC TGTAACGTAGACCACAAGCCTTCGAACACCAAGGTGGACAAGACTGTTGAGCGAAAGTGC GAGTGCCCTCCATGTCCTGCACCTCCTGTGGCTGGCCCTTCTGTGTTCCTGTTC CCTCCAAAACCTAAGGACACTCTAATGATCTCTCGGACTCCTGAGGTGACTTGCGTGGTT GTGGACGTGTCCCACGAGGACCCTGAGGTGCAGTTCAATTGGTACGTGGACGGAGTCGAG AATGCAAAGACCAAGCCTCGGGAGGAACAGTTCAACTCCACCTTCCGGGTGGTT TCTGTGTTGACCGTTGTGCACCAAGACTGGCTGAACGGCAAAGAATACAAGTGCAAGGTG TCCAACAAGGGCCTGCCTGCCCCTATCGAAAAGACCATCAGCAAGACCAAGGGCCAGCCT CGCGAGCCTCAGGTGTACACCCTGCCTCCCAGCCGGGAAGAAATGACCAAGAACCAGGTG TCCCTGACCTGTCTGGTGGAGGGCTTCTACCCTTCCGACATCGCCGTTGAGTGGGAGTCT AACGGACAGCCGGAGAACAACTACAAGACTACGCCTCCAATGCTGGACTCCGACGGCTCC TTCTTCCTGTACTCCGAACTGACCGTGGACAAGTCCCGGTGGCAGCAGGGCAACGTGTTC TCATGCTCCGTAATGCACGAAGCCTTGCACAATCACTACACTCAAAAGTCCCTATCCTTA GGCAAGTAG 21M] 8 Heavy chain variable region (version 2) (SEQ ID N0276) CAGCTAGTTCAGTCTGGAGCGGAAGTTAAGAAACCTGGAGCATCCGTGAAAATAAGTTGC AAGGCATCCGGTTACTCGTTCACCGCATACTATATCCACTGGGTTAAACAGGCACCAGGA CAGGGACTTGAATGGATCGGATATATCTCCTCTTATAATGGAGCTACAAACTATAACCAA AAATTCAAAGGACGCGTGACTTTCACAACTGACACCTCAACCTCGACAGCATACATGGAA TTACGGTCCCTACGGTCTGACGACACTGCCGTTTACTATTGCGCTAGAGATTATGATTAT GATGTTGGAATGGACTATTGGGGCCAGGGAACACTGGTGACAGTGTCTTCT LL4 heavy chain CDR2 consensus sequence (SEQ ID NO:80): YIX1X2YX3X4ATNYNQKFKG, where X1 is serine or alanine, X2 is , asparagine, or glycine, X3 is asparagine or lysine, and X4 is glysine, arginine, or aspartic acid

Claims (17)

1. A bispecific antibody comprising: a) a first antigen-binding site that specifically binds human vascular endothelial growth factor (VEGF), and b) a second antigen-binding site that specifically binds human delta-like 4 ligand (DLL4), wherein the first antigen-binding site comprises a heavy chain CDR1 comprising NYWMH (SEQ ID NO:17), a heavy chain CDR2 comprising DINPSNGRTSYKEKFKR (SEQ ID NO:18), and a heavy chain CDR3 sing HYDDKYYPLMDY (SEQ ID NO:19); n the second antigen-binding site comprises a heavy chain CDR1 comprising TAYYIH (SEQ ID NO:13) or AYYIH (SEQ ID NO:79), a heavy chain CDR2 comprising YIX1X2YX3X4ATNYNQKFKG (SEQ ID NO:80), wherein X1 is serine or alanine, X2 is serine, asparagine, or glycine, X3 is asparagine or lysine, and X4 is glycine, arginine, or aspartic acid , and a heavy chain CDR3 comprising RDYDYDVGMDY (SEQ ID NO:16); and wherein both the first and second antigen-binding sites comprise a light chain CDR1 comprising RASESVDNYGISFMK (SEQ ID NO:20), a light chain CDR2 sing AASNQGS (SEQ ID NO:21), and a light chain CDR3 sing QQSKEVPWTFGG (SEQ ID NO:22).
2. The bispecific antibody of claim 1, wherein the second antigen-binding site ses a heavy chain CDR1 comprising TAYYIH (SEQ ID NO:13), a heavy chain CDR2 comprising RATNYNQKFKG (SEQ ID NO:14), YISSYNGATNYNQKFKG (SEQ ID NO:15), YIAGYKDATNYNQKFKG (SEQ ID NO:59), or YISNYNRATNYNQKFKG (SEQ ID NO:65), and a heavy chain CDR3 comprising RDYDYDVGMDY (SEQ ID NO:16).
3. The ific antibody of claim 2, n the second antigen-binding site comprises a heavy chain CDR1 comprising TAYYIH (SEQ ID NO:13), a heavy chain CDR2 11283960_1 comprising YISNYNRATNYNQKFKG (SEQ ID NO:65), and a heavy chain CDR3 comprising RDYDYDVGMDY (SEQ ID NO:16).
4. The bispecific antibody of claim 1 or claim 2, which comprises: (a) a first heavy chain variable region having at least 90% ce identity to SEQ ID NO:11; (b) a second heavy chain variable region having at least 90% sequence ty to SEQ ID NO:9, SEQ ID NO:10, SEQ ID NO:58, or SEQ ID NO:64; and (c) a first and a second light chain variable region having at least 90% sequence identity to SEQ ID NO:12.
5. The bispecific antibody of claim 1 or claim 2, which comprises: (a) a first heavy chain variable region sing SEQ ID NO:11; (b) a second heavy chain variable region comprising SEQ ID NO:9, SEQ ID NO:10, SEQ ID NO:58, or SEQ ID NO:64; and (c) a first and a second light chain variable region comprising SEQ ID NO:12.
6. The bispecific antibody of claim 5, which comprises: (a) a first heavy chain variable region comprising SEQ ID NO:11; (b) a second heavy chain variable region comprising SEQ ID NO:64; and (c) a first and a second light chain variable region comprising SEQ ID NO:12.
7. The bispecific antibody of any one of claims 1-6, which is a monoclonal antibody, a recombinant antibody, a ic antibody, a humanized antibody, a human antibody, an IgG1 antibody, or an IgG2 antibody.
8. The bispecific antibody of any one of claims 1-6, which comprises a first human IgG2 constant region with amino acid substitutions at positions corresponding to positions 249 and 288 of SEQ ID NO:42, wherein the amino acids are ed with glutamate or aspartate, and a second human IgG2 constant region with amino acid tutions at 11283960_1 positions corresponding to positions 236 and 278 of SEQ ID NO:42, wherein the amino acids are replaced with lysine.
9. The bispecific antibody of any one of claims 1-8, which: (i) inhibits binding of VEGF to at least one VEGF receptor; (ii) inhibits binding of DLL4 to at least one Notch receptor; (iii) inhibits Notch signaling; and/or (iv) modulates angiogenesis.
10. A bispecific dy that specifically binds human VEGF and human DLL4, which comprises: (a) a heavy chain of SEQ ID NO:7; (b) a heavy chain of SEQ ID NO:5, SEQ ID NO:6, SEQ ID NO:56, or SEQ ID NO:62; and (c) two light chains of SEQ ID NO:8.
11. A bispecific antibody of claim 10, which comprises: (a) a heavy chain of SEQ ID NO:7; (b) a heavy chain of SEQ ID NO:62; and (c) two light chains of SEQ ID NO:8.
12. A bispecific antibody that specifically binds human VEGF and human DLL4, which ses: (a) a heavy chain encoded by the DNA sing SEQ ID NO:75, a heavy chain encoded by the DNA comprising SEQ ID NO:33, and a light chain d by the DNA comprising SEQ ID NO:34; (b) a heavy chain encoded by the DNA comprising SEQ ID NO:31, a heavy chain encoded by the DNA comprising SEQ ID NO:33, and a light chain encoded by the DNA comprising SEQ ID NO:34; 11283960_1 (c) a heavy chain encoded by the DNA comprising SEQ ID NO:72, a heavy chain d by the DNA comprising SEQ ID NO:33, and a light chain encoded by the DNA comprising SEQ ID NO:34; or (d) a heavy chain encoded by the DNA comprising SEQ ID NO:74, a heavy chain encoded by the DNA comprising SEQ ID NO:33, and a light chain encoded by the DNA comprising SEQ ID NO:34.
13. An isolated antibody that specifically binds human VEGF, which comprises: (a) a heavy chain CDR1 comprising NYWMH (SEQ ID NO:17), a heavy chain CDR2 comprising DINPSNGRTSYKEKFKR (SEQ ID NO:18), and a heavy chain CDR3 sing YPLMDY (SEQ ID NO:19); and (b) a light chain CDR1 comprising RASESVDNYGISFMK (SEQ ID NO:20), a light chain CDR2 comprising AASNQGS (SEQ ID NO:21), and a light chain CDR3 comprising PWTFGG (SEQ ID NO:22).
14. The antibody of claim 13, which comprises: (a) a heavy chain variable region having at least 90% sequence identity to SEQ ID NO:11; and (b) a light chain variable region having at least 90% sequence identity to SEQ ID NO:12.
15. The antibody of claim 13, which comprises: (a) a heavy chain variable region sing SEQ ID NO:11; and (b) a light chain variable region comprising SEQ ID NO:12.
16. The antibody of claim 13, which comprises: (a) a heavy chain comprising SEQ ID NO:49 or SEQ ID NO:7; and (b) a light chain comprising SEQ ID NO:8.
17. The antibody of any one of claims 13-16, which inhibits binding of VEGF to at least one VEGF receptor. 11283960
NZ623724A 2011-09-23 2012-09-24 Vegf/dll4 binding agents and uses thereof NZ623724B2 (en)

Applications Claiming Priority (7)

Application Number Priority Date Filing Date Title
US201161538454P 2011-09-23 2011-09-23
US61/538,454 2011-09-23
US201261597409P 2012-02-10 2012-02-10
US61/597,409 2012-02-10
US201261692978P 2012-08-24 2012-08-24
US61/692,978 2012-08-24
PCT/US2012/056886 WO2013044215A1 (en) 2011-09-23 2012-09-24 Vegf/dll4 binding agents and uses thereof

Publications (2)

Publication Number Publication Date
NZ623724A NZ623724A (en) 2016-09-30
NZ623724B2 true NZ623724B2 (en) 2017-01-05

Family

ID=

Similar Documents

Publication Publication Date Title
US11512128B2 (en) VEGF/DLL4 binding agents and uses thereof
AU2016326609B2 (en) Methods and compositions for treatment of cancer
CA2878868A1 (en) Rspo3 binding agents and uses thereof
US20160319034A1 (en) Met-binding agents and uses thereof
US20170157245A1 (en) Treatment of gastric cancer
AU2017210673B2 (en) VEGF/DLL4 binding agents and uses thereof
NZ623724B2 (en) Vegf/dll4 binding agents and uses thereof