EP2467164A2 - Anticorps contre l ectodomaine de erbb3 et leurs utilisations - Google Patents

Anticorps contre l ectodomaine de erbb3 et leurs utilisations

Info

Publication number
EP2467164A2
EP2467164A2 EP10810721A EP10810721A EP2467164A2 EP 2467164 A2 EP2467164 A2 EP 2467164A2 EP 10810721 A EP10810721 A EP 10810721A EP 10810721 A EP10810721 A EP 10810721A EP 2467164 A2 EP2467164 A2 EP 2467164A2
Authority
EP
European Patent Office
Prior art keywords
antibody
erbb3
seq
nos
cdr2
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP10810721A
Other languages
German (de)
English (en)
Inventor
Birgit Schoeberl
Ulrik Nielsen
Michael Feldhaus
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Merrimack Pharmaceuticals Inc
Original Assignee
Merrimack 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
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=43607614&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=EP2467164(A2) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Merrimack Pharmaceuticals Inc filed Critical Merrimack Pharmaceuticals Inc
Publication of EP2467164A2 publication Critical patent/EP2467164A2/fr
Withdrawn legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/32Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against translation products of oncogenes
    • 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
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/21Immunoglobulins specific features characterized by taxonomic origin from primates, e.g. man
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/34Identification of a linear epitope shorter than 20 amino acid residues or of a conformational epitope defined by amino acid residues
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/55Fab or Fab'
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • C07K2317/62Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising only variable region components
    • C07K2317/622Single chain antibody (scFv)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins 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
    • C07K2317/90Immunoglobulins 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

Definitions

  • the ErbB/HER subfamily of polypeptide growth factor receptors include the epidermal growth factor (EGF) receptor (EGFR ErbBl/HERl), the neu oncogene product (ErbB2/HER2), and the more recently identified ErbB3/HER3 and ErbB4/HER4 receptor proteins (see, e.g., Hynes et. al. (1994) Biochim. Biophys. Acta Rev. Cancer 1 198, 165-184).
  • Each of these receptors is predicted to consist of an ectodomain (extracellular ligand-binding domain), a membrane-spanning domain, a cytosolic, protein tyrosine kinase (PTK) domain and a C-terminal phosphorylation domain (see, e.g., Kim et al., (1998) Biochem. J. 334, 189-195).
  • the ectodomains of the ErbB receptors are further characterized as being divided into four domains (I-IV). Domains I and HI of the ErbB ectodomain are involved in ligand binding (see, e.g., Hynes et. al. (2005) Nature Rev. Cancer 5, 341-354).
  • Ligands for these receptors include heregulin (HRG) and betacellulin (BTC).
  • ErbB3 receptor the protein tyrosine kinase activity of the ErbB3 receptor (ErbB3) protein is attenuated significantly relative to that of other ErbB/HER family members and this attenuation has been attributed, in part, to the occurrence of non-conservative amino acid substitutions in the predicted intracellular catalytic domain of ErbB3 (see, e.g., Guy et al. (1994) Proc. Natl. Acad. Sci. USA. 91, 8132-8136; Sierke et al. (1997) Biochem. J. 322, 757-763).
  • the ErbB3 protein has been shown to be phosphorylated in a variety of cellular contexts.
  • ErbB3 is constitutively phosphorylated on tyrosine residues in a subset of human breast cancer cell lines overexpressing this protein (see, e.g., Kraus et al. (1993) Proc. Natl. Acad. Sci. USA. 90, 2900-2904; and Kim et al. Supra; see, also, Schaefer et al. (2006) Neoplasia 8(7):613-22 and Schaefer et al. Cancer Res (2004) 64(10):3395- 405).
  • ErbB3 remains largely unappreciated as a target for clinical intervention.
  • Current immunotherapies primarily focus on inhibiting the action of ErbB2 and, in particular, heterodimerization of ErbB2/ErbB3 complexes (see, e.g., Sliwkowski et al. (1994) J. Biol. Chem.
  • the present invention provides a novel class of antibodies (e.g., monoclonal antibodies) that bind to the ErbB3 receptor and inhibit various ErbB3 functions.
  • the antibodies described herein are capable of binding to ErbB3 and inhibiting EGF-like ligand mediated phosphorylation of the receptor.
  • EGF-like ligands include EGF, TGF-a, betacellulin, heparin-binding epidermal growth factor, biregulin and amphiregulin, which bind to EGFR and induce dimerization of EGFR with ErbB3. This dimerization, in turn, causes phosphorylation of ErbB3, and activates signaling through the receptor.
  • Antibodies of the present invention are thus useful for treating and diagnosing a variety of cancers associated with ErbB3-mediated cellular signaling. Accordingly, in one embodiment, the present invention provides antibodies (including antigen binding portions thereof) which bind to ErbB3 and inhibit EGF-like ligand mediated phosphorylation of ErbB3.
  • the antibodies and fragments thereof are further characterized by one or more of the following properties: (i) inhibition of ErbB3 ligand- mediated signaling, including signaling mediated by binding of ErbB3 ligands, such as heregulin, epiregulin, epigen and biregulin, to ErbB3; (ii) inhibition of proliferation of cells expressing ErbB3; (iii) the ability to decrease levels of ErbB3 on cell surfaces (e.g., by inducing internalization of ErbB3); (iv) inhibition of secretion of VEGF (vascular endothelial growth factor ) by cells expressing ErbB3; (v) inhibition of the migration of cells expressing ErbB3; (vi) inhibition of spheroid growth of cells expressing ErbB3; and (vii) binding to an epitope located on ectodomain (extracellular domain) Domain I, which corresponds to amino acid residues 1 to about 190 of mature ErbB3 (SEQ ID NO:
  • the antibody binds to an epitope involving or spanning residues 93-104 or 92-129 of SEQ ID NO: 73, even more preferably involving residues 92, 93, 99, 101, 102, 104 and 129 of SEQ ID NO: 73 or residues 93, 101, 102, and 104 of SEQ ID NO: 73, with the proviso that the antibody is not: (i) Ab #6 as disclosed herein; (ii) an antibody comprising VH and VL sequences as shown in SEQ ID NOs: 1 and 2, respectively; or (iii) an antibody comprising V H CDR1, CDR2 and CDR3 sequences as shown in SEQ ID NOs: 7, 8 and 9, respectively, and V L CDR1, CDR2 and CDR3 sequences as shown in SEQ ID NOs: 10, 11 and 12, respectively.
  • the antibody binds to an epitope involving or spanning residues 93-104 or 92-129 of SEQ ED NO: 73, even more preferably involving residues 92, 93, 99, 101, 102, 104 and 129 of SEQ ED NO: 73 or residues 93, 101, 102, and 104 of SEQ ID NO: 73, with the proviso that the antibody is not: (i) Ab #6 as disclosed herein; (ii) an antibody comprising VH and VL sequences as shown in SEQ ED NOs: 1 and 2, respectively; (iii) an antibody comprising V H CDR1, CDR2 and CDR3 sequences as shown in SEQ ID NOs: 7, 8 and 9, respectively, and VL CDRl, CDR2 and CDR3 sequences as shown in SEQ ID NOs: 10, 11 and 12, respectively; (iv) Ab #3 as disclosed herein; (v) an antibody comprising VH and VL sequences as shown in SEQ ID NOs: 3
  • Preferred antibodies and antigen binding portions thereof disclosed herein exhibit a KD of 50 nM or less, as measured by a surface plasmon resonance assay or a cell binding assay
  • particular antibodies and antigen binding portions thereof of the present invention include a heavy chain variable region (VH) comprising an amino acid sequence which is at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98% or 99%) identical to the heavy chain variable region amino acid sequence set forth in SEQ ID NO: l, SEQ ID NO:3, SEQ ID NO:5, SEQ ID NO:35, or SEQ ID NO: 37.
  • VH heavy chain variable region
  • V L light chain variable region
  • the antibodies may also include both of the aforementioned heavy chain and light chain variable regions.
  • variable heavy and light chain regions of the antibodies or antigen binding portions thereof typically include one or more complementarity determining regions (CDRs). These include one or more CDRl, CDR2, and CDR3 regions. Accordingly, other particular antibodies and antigen binding portions thereof of the present disclosure include one or more CDR sequences selected from a heavy chain variable region CDRl comprising SEQ ED NO:7; a heavy chain variable region CDR2 comprising SEQ ID NO:8; a heavy chain variable region CDR3 comprising SEQ ID NO:9; a light chain variable region CDRl comprising SEQ ID NO: 10; a light chain variable region CDR2 comprising SEQ ID NO: l 1; a light chain variable region CDR3 comprising SEQ ID NO: 12; and combinations thereof.
  • CDRs complementarity determining regions
  • Still other particular antibodies and antigen binding portions thereof of the present disclosure include one or more CDR sequences selected from a heavy chain variable region CDRl comprising SEQ ID NO: 13; a heavy chain variable region CDR2 comprising SEQ ID NO: 14; a heavy chain variable region CDR3 comprising SEQ ID NO: 15; a light chain variable region CDRl comprising SEQ ID NO: 16; a light chain variable region CDR2 comprising SEQ ID NO: 17; a light chain variable region CDR3 comprising SEQ ID NO: 18; and combinations thereof.
  • Still other particular antibodies and antigen binding portions thereof of the present disclosure include; or one or more CDR sequences selected from a heavy chain variable region CDRl comprising SEQ ID NO: 19; a heavy chain variable region CDR2 comprising SEQ ID NO:20; a heavy chain variable region CDR3 comprising SEQ ID NO:21; a light chain variable region CDRl comprising SEQ ED NO:22; a light chain variable region CDR2 comprising SEQ ID NO:23; a light chain variable region CDR3 comprising SEQ ID NO:24; and combinations thereof.
  • Still other particular antibodies and antigen binding portions thereof of the present disclosure include; or one or more CDR sequences selected from a heavy chain variable region CDRl comprising SEQ ID NO:39; a heavy chain variable region CDR2 comprising SEQ ID NO:40; a heavy chain variable region CDR3 comprising SEQ ID NO:41 ; a light chain variable region CDRl comprising SEQ ID NO:42; a light chain variable region CDR2 comprising SEQ ID NO:43; a light chain variable region CDR3 comprising SEQ ID NO:44; and combinations thereof.
  • Still other particular antibodies and antigen binding portions thereof of the present disclosure include; or one or more CDR sequences selected from a heavy chain variable region CDRl comprising SEQ ID NO:45; a heavy chain variable region CDR2 comprising SEQ ID NO:46; a heavy chain variable region CDR3 comprising SEQ ID NO:47; a light chain variable region CDRl comprising SEQ ID NO:48; a light chain variable region CDR2 comprising SEQ ID NO:49; a light chain variable region CDR3 comprising SEQ ID NO:50; and combinations thereof.
  • the antibodies and antigen binding portions thereof may also comprise one or more CDRs which are at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98% or 99%) identical to any of the aforementioned CDRs, or combinations of CDRs.
  • the antibodies and antibody portions thereof are fully human (i.e., contains human CDR and framework sequences).
  • Particular human antibodies of the present disclosure include those having a heavy chain variable region that is from a human VH3 germ line gene, and/or a light chain variable region from human VL2 germ line gene.
  • antibodies and antigen binding portions thereof that bind to the same or overlapping epitopes bound by any of the antibodies or portions thereof described herein ⁇ e.g., an epitope located on Domain I of the ectodomain of ErbB3), such as an epitope involving or spanning any of residues 1- 183 of the amino acid sequence of mature ErbB3 (SEQ ID NO: 73), more preferably involving or spanning residues 93-104 or 92-129 of SEQ ID NO: 73, even more preferably involving residues 92, 93, 99,101, 102, 104 and 129 of SEQ ID NO: 73 or residues 93, 101, 102, and 104 of SEQ ID NO: 73.
  • an epitope located on Domain I of the ectodomain of ErbB3 such as an epitope involving or spanning any of residues 1- 183 of the amino acid sequence of mature ErbB3 (SEQ ID NO: 73), more preferably involving or spanning residues
  • Antibodies which have the same epitope binding activity as the antibodies described herein e.g., antibodies having the same sequence as Ab #6 or binding epitopes involving residues 93-104 of SEQ ID NO: 73, are also encompassed by the present invention.
  • antibodies and antigen binding portions thereof that bind to the ectodomain of human ErbB3 and comprise:
  • the heavy chain variable region CDRl, CDR2 and CDR3 sequences comprise consensus heavy chain CDRl , CDR2 and CDR3 sequences shown in SEQ ID NOs: 60 or 75 (CDRl), 61 (CDR2) and 62 (CDR3), respectively, and
  • the light chain variable region CDRl, CDR2 and CDR3 sequences comprise consensus light chain CDRl, CDR2 and CDR3 sequences shown in SEQ ID NOs: 66 or 77 (CDRl), 67 (CDR2) and 68 or 79 (CDR3), respectively, with the proviso that the antibody is not: (i) Ab #6 as disclosed herein; (ii) an antibody comprising VH and VL sequences as shown in SEQ ID NOs: 1 and 2, respectively; or (iii) an antibody comprising V H CDRl, CDR2 and CDR3 sequences as shown in SEQ ID NOs: 7, 8 and
  • the isolated antibody, or antigen binding portion thereof binds to an epitope within Domain I of the ectodomain of human ErbB3 comprising residues 92-129 or 93-104 of SEQ ID NO: 73. Even more preferably, the isolated antibody, or antigen binding portion thereof, binds to an epitope within Domain I of the ectodomain of human ErbB3 comprising residues 92-129 or 93-104 of SEQ ID NO: 73 and inhibits proliferation of a cancer cell expressing ErbB3.
  • the cancer cell is a MALME-3M melanoma cell, an AdrR (ADRr) ovarian adenocarcinoma cell or an ACHN renal carcinoma cell and the proliferation is reduced by at least 10% relative to a control (i.e., matched untreated cells).
  • ADRr AdrR
  • antibodies and antigen binding portions thereof that bind to the Domain I of the ectodomain of human ErbB3 and comprise:
  • the heavy chain variable region paratope comprises CDR1, CDR2 and CDR3 sequences shown in SEQ ID NOs: 63 or 76 (CDR1), 64 (CDR2) and 65 (CDR3), respectively, and
  • the light chain variable region paratope comprises CDR1, CDR2 and CDR3 sequences shown in SEQ ID NOs: 69 or 78 (CDR1), 70 (CDR2) and 71 or 80 (CDR3), respectively, with the proviso that the antibody is not: (i) Ab #6 as disclosed herein; (ii) an antibody comprising VH and VL sequences as shown in SEQ ID NOs: 1 and 2, respectively; or (iii) an antibody comprising VH CDRl, CDR2 and CDR3 sequences as shown in SEQ ID NOs: 7, 8 and 9, respectively, and V L CDRl, CDR2 and CDR3 sequences as shown in SEQ ID NOs: 10, 1 1 and 12, respectively. .
  • antibodies and antigen binding portions thereof that bind specifically to Domain I of the ectodomain of human ErbB3, wherein the antibody or antigen binding portion thereof comprises:
  • a heavy chain variable region CDRl comprising SEQ ID NO:7 or conservative amino acid substitutions thereof;
  • a heavy chain variable region CDR2 comprising SEQ ID NO:8 or conservative amino acid substitutions thereof;
  • a heavy chain variable region CDR3 comprising SEQ ID NO:9 or conservative amino acid substitutions thereof
  • a light chain variable region CDR1 comprising SEQ ID NO: 10 or conservative amino acid substitutions thereof
  • a light chain variable region CDR2 comprising SEQ ED NO: 11 or conservative amino acid substitutions thereof;
  • a light chain variable region CDR3 comprising SEQ ED NO: 12 or conservative amino acid substitutions thereof,
  • Antibodies of the present invention include all known forms of antibodies and other protein scaffolds with antibody-like properties.
  • the antibody can be a human antibody, a humanized antibody, a bispecific antibody, a chimeric antibody or a protein scaffold with antibody-like properties, such as fibronectin or ankyrin repeats.
  • the antibody also can be a Fab, Fab'2, ScFv, SMEP, affibody, nanobody, or a domain antibody.
  • the antibody also can have any of the following isotypes: IgGl, IgG2, IgG3, IgG4, IgM, IgAl , IgA2, IgAsec, IgD, and IgE.
  • the present invention further provides compositions comprising combinations of antibodies or antigen binding portions described herein, formulated with an acceptable carrier and/or adjuvant.
  • the composition comprises two or more antibodies that bind different epitopes on ErbB3 or antibodies described herein combined with anti-cancer antibodies which do not bind ErbB3.
  • the present invention provides isolated nucleic acids encoding the antibodies and antigen binding portions thereof described herein.
  • the nucleic acid encodes a heavy chain variable region comprising a nucleotide sequence which is at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98% or 99%) identical to, or which hybridizes under high stringency conditions to, SEQ ED NO:25, SEQ ED NO:27, SEQ ED NO:29, SEQ ED NO:35, or SEQ ED NO:37; or a light chain variable region comprising a nucleotide sequence which is at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98% or 99%) identical to, or which hybridizes under high stringency conditions to, SEQ ED NO:26, SEQ ED NO:28, SEQ ED NO:30, SEQ ED NO:36, or SEQ ED NO:38; or combinations of such heavy
  • the present invention further provides transgenic non-human mammals, hybridomas, and transgenic plants that express and/or produce the antibodies and antigen binding portions described herein.
  • kits comprising one or more isolated antibodies or antigen binding portions thereof described herein and, optionally, instructions for use in treating or diagnosing a disease associated with ErbB3 dependent signaling, such as cancers.
  • the present invention provides method for inhibiting EGF-like ligand mediated phosphorylation of ErbB3 in a subject by administering one or more antibodies or antigen binding portions thereof described herein in an amount sufficient to inhibit EGF-like mediated phosphorylation of ErbB3.
  • the invention further provides methods for treating a variety of cancers in a subject, including, but not limited to, melanoma, breast cancer, ovarian cancer, renal carcinoma, gastrointestinal/colon cancer, lung cancer, clear cell sarcoma, and prostate cancer, by administering one or more antibodies or antigen binding portions thereof described herein in an amount sufficient to treat the cancer.
  • the cancer comprises cells comprising a KRAS mutation.
  • Exemplary KRAS mutations are in either or both of codon 12 and codon 13 of the human KRAS gene.
  • Mutations in codon 12 or codon 13, each of which normally codes for a glycine are activating KRAS mutations that promote oncogenesis, as are mutations in codons 15, 20, 61 and 146 of the human KRAS gene.
  • the cancer comprises cells comprising a PI3K (phosphatidylinositol 3-kinase) mutation.
  • Exemplary PI3K mutations are activating mutations in the human PIK3CA gene in either or both of exon 9 or exon 20.
  • the antibodies or antigen binding portions thereof can be administered alone or in combination with other therapeutic agents, such as anti-cancer agents, e.g., other antibodies, chemotherapeutic agents and/or radiation.
  • an antibody or antigen binding portion thereof is administered in combination with a second agent, which second agent is selected from the group consisting of a protein synthesis inhibitor, a somatostatin analogue, an immunotherapeutic agent, and an enzyme inhibitor.
  • the second agent is selected from: a small molecule targeting IGF1R, a small molecule targeting EGFR, a small molecule targeting ErbB2, a small molecule targeting cMET, an antimetabolite, an alkylating agent, a topoisomerase inhibitor, a microtubule targeting agent, a kinase inhibitor, a hormonal therapy, a glucocorticoid, an aromatase inhibitor, an mTOR inhibitor, a chemotherapeutic agent, a protein kinase B inhibitor, a
  • phosphatidylinositol 3-kinase inhibitor a cyclin dependent kinase inhibitor
  • an MEK inhibitor a cyclin dependent kinase inhibitor
  • Exemplary antibodies for use as second agents in combination therapy include anti-Her2 antibodies, e.g., trastuzumab and anti-EGFR antibodies, e.g., panitumumab or cetuximab.
  • Certain preferred anti-cancer agents for use as second agents in combination therapy include erlotinib, lapatinib, paclitaxel and cisplatin.
  • the present invention provides methods for diagnosing and prognosing diseases (e.g., cancers) associated with ErbB3.
  • this is achieved by contacting antibodies or antigen binding portions of the present disclosure(e.g., ex vivo or in vivo) with cells from the subject, and measuring the level of binding to ErbB3 on the cells, wherein abnormally high levels of binding to ErbB3 indicate that the subject has a cancer associated with ErbB3.
  • Figures 1 A and IB are bar graphs depicting the binding of various anti-ErbB3 antibody candidates to ErbB3 expressed on MALME-3M melanoma cells using a goat anti-human Alexa 647 secondary antibody (GAHu-Alexa 674).
  • the antibodies indicated as “Ab#” are used in the form of Fab fragments and "GAHu-Alexa 674" indicates the fluorescent dye-conjugated secondary antibody used alone as a control, while “unstained” indicates a control in the absence of the secondary antibody .
  • Figures 2A-2D are graphs depicting binding of antibodies to ErbB3 set forth as K D values of various anti-ErbB3 antibody candidates.
  • Figures 2C and 2D are graphs depicting the KD values of Ab #6 and Ab #3, respectively, as measured using a cell binding assay using MALME-3M melanoma cells and the formula
  • Figure 3 is a graph depicting the binding specificity of an anti-ErbB3 antibody (Ab #6) to ErbB3 using ELISA.
  • EGFR extracellular domain, bovine serum albumin (BSA) and TGFa were used as controls.
  • Figure 4 is a graph depicting the ability of an anti-ErbB3 antibody (Ab #6) to decrease total ErbB3 levels in MALME-3M melanoma cells in vitro, as measured using ELISA.
  • Figures 5A and 5B are graphs depicting the ability of an anti-ErbB3 antibody
  • Figures 6A-6D are graphs depicting the timecourse of antibody-mediated ErbB3 downregulation (Ab #6), as measured using FACS analysis.
  • Figure 7 shows the results of a pharmacodynamic study.
  • the a bar graph depicts 24hrs post-injection results of indicated anti-ErbB3 antibodies on total ErbB3 levels in MALME3M melanoma cells in xenografts in vivo. As can be seen, the data demonstrate the ability of Ab #6 to downregulate ErbB3.
  • Figure 8 is a bar graph depicting the ability of an anti-ErbB3 antibody (Ab #6) to downregulate ErbB3 in AdrR xenografts in vivo. Total ErbB3 levels in AdrR xenografts are shown.
  • Figure 9 is a graph depicting the ability of an anti-ErbB3 antibody (Ab #6) to inhibit proliferation of MALME-3M cells in a CellTiter-Glo® assay.
  • Figure 10 is a graph depicting the ability of an anti-ErbB3 antibody (Ab #6) to inhibit cell proliferation of AdrR cells.
  • Figure 11 is a graph depicting the ability of an anti-ErbB3 antibody (Ab #6) to inhibit proliferation of ACHN cells.
  • Figure 12 is a bar graph depicting the ability of an anti-ErbB3 antibody (Ab #6) to inhibit ErbB3 phosphorylation in AdrR xenografts in vivo.
  • Figures 13A-13C are graphs depicting the ability of an anti-ErbB3 antibody (Ab #6) to inhibit betacellulin, heregulin, and TGFa-mediated phosphorylation of ErbB3 in AdrR cells.
  • Figures 14A-14B are graphs depicting the ability of an anti-ErbB3 antibody (Ab #6 IgG2 isotype) to inhibit ErbB3 phosphorylation in ovarian tumor cell lines OVCAR 5 and OVCAR 8.
  • Figures 15A-15C are graphs depicting the ability of betacellulin (BTC) to bind
  • ErbB 1 as shown by a lack of binding to ErbB 1 negative MALME-3M cells (Fig.15 A); binding to ErbBl positive AdrR cells at concentrations of 10 nM ( Figure 15B) and 200 nM (Fig. 15C), respectively, and the inhibition of such binding by cetuximab.
  • Figures 16A-16D are graphs depicting the ability of an anti-ErbB3 antibody (Ab #6, IgG2 isotype) to inhibit heregulin-mediated signaling in MALME-3M cells ( Figures 16A and 16B) and OVCAR8 cells (16C and 16D).
  • Figures 17A-D are graphs depicting the ability of an anti-ErbB3 antibody (Ab #6) to inhibit: (Fig. HA) ovarian (AdrR cells), (Fig. 17B) prostate (Dul45 cells), (Fig. 17C) ovarian (OVCAR8 cells), and (Fig. 17D) pancreatic (Colo-357 cells) tumor growth via xenograft studies.
  • Figures 18A and 18B are graphs depicting the ability of Ab #6 ( Figure 18A) and Fab for Ab #3 ( Figure 18B) to inhibit heregulin binding to ErbB3 on MALME-3M cells, as measured using FACS analysis.
  • Figure 19A depicts the binding of epiregulin to AdrR cells
  • Figure 19B depicts the ability of Ab #6, but not ERBITUX (cetuximab) to inhibit epiregulin binding to AdrR cells.
  • Figure 20 is a graph depicting the ability of heparin binding epidermal growth factor (HB-EGF) to bind to AdrR cells.
  • HB-EGF heparin binding epidermal growth factor
  • Figures 21A-21C show the amino acid sequences of the variable heavy and light chain regions of antibodies: Ab #6, Ab #3, Ab #14, Ab #17, and Ab #19.
  • Figures 22A-22B show the nucleotide sequences of the variable heavy and light chain regions of antibodies: Ab #6, Ab #3, and Ab #14.
  • Figure 23 shows the amino acid sequences of the variable light chain regions of antibodies: Ab #6, Ab #17, and Ab #19, which have been reverted to the corresponding germline amino acid sequence. Amino acid residue changes accomplishing this
  • Figures 24A and B are graphs showing the ability of Ab #6 to inhibit VEGF secretion by tumor cells (see Example 11).
  • Figure 24C shows the correlation between inhibition of VEGF secretion and inhibition of pErbB3 in cancer cells by Ab #6.
  • Figure 25 is a graph showing the effect of Ab #6 on cell migration (see Example 12).
  • Figures 26A-C are graphs showing (Fig. 26A) inhibition of spheroid growth in AdrR cells, (Fig. 26B) inhibition of HRG-induced spheroid growth in AdrR, and (Fig. 26C) inhibition of HRG-induced spheroid growth in Dul45 cells.
  • Figures 27 A and B are graphs showing the effect of Ab #6 on HRG (Fig. 27 A) and BTC (Fig. 27B) binding to AdrR cells.
  • Figure 28 is a graph showing the effect of Ab #6 on HGF (hepatocyte growth factor) induced ErbB3 phosphorylation in AdrR cells.
  • the ICso for HGF was determined to be 2.439e-10.
  • Figures 29 A and B show the effect of Ab #6 on phosphorylation of (Fig. 29A) pErbB l and pErbB3 and (Fig. 29B) HRG induced ErbB2/3 complex formation.
  • Figure 30 is a graph showing the effect of Ab #6 alone, erlotinib alone or Ab #6 plus erlotinib on the growth of ACHN xenograft tumors in nude mice. The data show that after 27 days the combination a dose of 300 ug of Ab #6 (a suboptimal dose when administered alone) plus erlotinib synergistically inhibits tumor growth to a statistically significant extent.
  • Figure 31 is a graph showing the effect of Ab #6 alone, taxol alone or Ab #6 plus taxol on the growth of DU145 xenograft tumors in nude mice. The data show that after 27 days the combination of a dose of 300 ug of Ab #6 (a suboptimal dose when administered alone) plus taxol inhibits tumor growth to a greater extent than does treatment with either drug alone to a statistically significant extent.
  • Figure 32A is a graph showing the effect of Ab #6 treatment alone on the growth of multicellular tumor spheroids of KRAS mutant A549 lung cancer cells. Cells were treated with 0, 0.001, 0.01, 0.1 or 1 ⁇ Ab #6 for seven days. The "-4" on the x axis corresponds to the "0" dose. The results show the Ab #6 dose response effect on KRAS mutant tumor spheroid growth.
  • Figure 32B is a set of photographs of representative A549 spheroids, either untreated or treated with 1 ⁇ Ab #6, on day 1 and day 7 of treatment.
  • Figure 32C is a graph showing the effect of Ab #6 treatment alone on the growth of KRAS mutant A549 subcutaneous xenograft tumors in nude mice. Dosing of Ab #6 (600 ⁇ g every three days) was stopped on day 22.
  • Figure 33A is a graph showing the effect of Ab #6 treatment alone, erlotinib treatment alone or combined treatment with Ab #6 and erlotinib on the growth of KRAS mutant A549 subcutaneous xenograft tumors in nude mice.
  • Figure 33B is a graph showing the effect of Ab #6 treatment alone, taxol treatment alone or combined treatment with Ab #6 and taxol on the growth of KRAS mutant A549 subcutaneous xenograft tumors in nude mice.
  • Figures 34A-34E shows the results of a FACS analysis for the binding of Ab #6 and a control anti-ErbB3 antibody (SGP1) to CHO cells expressing either wild-type ErbB3 (Fig. 34A) or CHO cells expressing ErbB3 having one of the following point mutations: D93A (Fig. 34B), M101A (Fig. 34C), L102A (Fig. 34D) or Y104A (Fig. 34E).
  • Figure 35A is a graph showing the effect of Ab #6 treatment alone on the growth of PI3K mutant SKOV3 xenograft tumors in mice.
  • Figure 35B is a graph showing the showing the effect of Ab #6 treatment alone, or in combination with cisplatin (CDDP), on the growth of PI3K mutant SKOV3 xenograft tumors in mice.
  • CDDP cisplatin
  • Figure 36 presents data from paratope mapping experiments. Shown are the effects of single amino acid mutations (for identity of indicated mutations see example 20, Table 2) on the binding of Ab #6 mutants to ErbB3 as compared to the binding of the same mutants of Ab #6 to protein A.
  • the diagonal bisecting the graph indicates a 1 : 1 correspondence of the effect on ErbB3 and protein A binding.
  • the additional, parallel lines were located created by moving the diagonal down the y axis by 0.33 and 0.66 for convenient grouping of the mutations into three categories (large, small and no effect on binding).
  • the graph shows the effect on binding of the mutation normalized to the wild type Ab #6.
  • Values on the Y axis are ratios of binding of wild type Ab #6 to binding of Ab #6 mutants to ErbB3.
  • a ratio of ⁇ 1 means that the mutant has loss of binding to ErbB3 compared to wild type Ab #6, while a ratio of > 1 means that the mutant exhibits stronger binding to ErbB 3 than wild type Ab #6.
  • Figures 37A and 37B are schematic diagrams of the Ab #6 wild type heavy chain variable region (V H ) CDR1, CDR2 and CDR3 sequences (SEQ ED NOs: 7, 8 and 9, respectively), the consensus V H CDR1, CDR2 and CDR3 sequences (SEQ ED NOs: 60, 61 and 62, respectively, for Fig. 37A and SEQ ED NOs: 75, 61 and 62, respectively, for Fig. 37B), the V H CDR1, CDR2 and CDR3 paratope sequences (SEQ ED NOs: 63, 64 and 65, respectively, for Fig. 37A and SEQ ED NOs: 76, 64, 65, respectively, for Fig.
  • V H Ab #6 wild type heavy chain variable region
  • V L Ab #6 wild type light chain variable region
  • CDR1, CDR2 and CDR3 sequences SEQ ED NOs: 10, 11 and 12, respectively
  • consensus V L CDR1, CDR2 and CDR3 sequences SEQ ED NOs: 66, 67 and 68, respectively, for Fig. 37A and SEQ ED NOs: 77, 67 and 79, respectively, for Fig. 37B
  • V L CDR1, CDR2 and CDR3 paratope sequences SEQ ED NOs: 69, 70 and 71, respectively, for Fig. 37 A and SEQ ED NOs: 78, 70 and 80, respectively, for Fig. 37B).
  • the standard single letter amino acid abbreviations are used and the separation of single letters indicating amino acids by dashes indicates sequential residues in the sequence, while any group of two or more adjacent single letters indicating amino acids separated by one or more slashes indicates that any of the grouped adjacent amino acids so separated may be substituted for any of the others at that position in the sequence.
  • the notation "(Xaa) 7 -W-T/A/G/S-L-(Xaa)7” indicates a sequence running from amino to carboxy terminus as follows: seven unspecified amino acids followed by tryptophan, followed by (threonine or alanine or glycine or serine), followed by leucine, followed by seven unspecified amino acids.
  • Unspecified amino acid is used here to indicate that any amino acid may appear independently at each position designated Xaa, even where several such positions appear together.
  • the repetition of Xaa or of any group designated as Xaa # — e.g., "(Xaa) 7 " -- does not indicate any correspondence between the designated group of unspecified amino acids at one position in these sequences and designated group of unspecified amino acids at any other position.
  • the repetition of (Xaa) 7 at the beginning and end of the sequence "(Xaa) 7 -W-T/A/G/S-L-(Xaa) 7 " does not indicate any correspondence between the sequence of each (Xaa) other than that each contains seven unspecified amino acids.
  • Figures 38 A-D show the effects of Ab #6 (decreasing concentrations along the
  • Figure 39 is a FACS profile demonstrating that Ab #6 binds to Domain I of the ectodomain of ErbB3.
  • ErbB3 refers to human ErbB3 protein, as described in U.S. Pat. No. 5,480,968 and Plowman et al, Proc. Natl. Acad. Sci. USA, 87:4905-4909 (1990); see, also, Kani et al., Biochemistry 44: 15842-857 (2005), Cho and Leahy, Science 297: 1330- 1333 (2002)).
  • the full-length, mature human ErbB3 protein sequence (without leader sequence) is shown in SEQ ID NO: 73. This sequence corresponds to the sequence shown in Figure 4 and SEQ ID NO: 4 of U.S. Pat. No. 5,480,968, minus the 19 amino acid leader sequence that is cleaved from the mature protein.
  • EGF-like ligand refers to ligands of epidermal growth factor receptor (EGFR), including epidermal growth factor (EGF) and closely related proteins, such as transforming growth factor-a (TGFa), betacellulin (BTC), heparin-binding epidermal growth factor (HB-EGF), biregulin (BIR) and amphiregulin (AR), which bind to EGFR on the surface of cells and stimulate the receptor's intrinsic protein-tyrosine kinase activity.
  • GGFa transforming growth factor-a
  • BTC betacellulin
  • HB-EGF heparin-binding epidermal growth factor
  • BIR biregulin
  • AR amphiregulin
  • EGF-like ligands induce formation of EGFR and ErbB3 protein complex (see e.g., Kim et al., (1998) Biochem J., 334: 189-195), which results in phosphorylation of tyrosine residues in the complex.
  • Preferred antibodies and antigen binding portions thereof disclosed herein inhibit EGF-like ligand mediated phosphorylation of ErbB3 and, in certain embodiments, exhibit one or more of the following additional properties: (i) inhibition of one or more of heregulin, epiregulin, epigen and biregulin-mediated signaling through ErbB3; (ii) inhibition of proliferation of cells expressing ErbB3; (iii) the ability to decrease levels of ErbB3 on cell surfaces; (iv) inhibition of VEGF secretion of cells expressing ErbB3; (v) inhibition of the migration of cells expressing ErbB3; (vi) inhibition of spheroid growth of cells expressing ErbB3; and/or (vii) specific binding to an epitope located on Domain I of the ectodomain of ErbB3, e.g., an epitope which involves or spans residues 1-183 of the amino acid sequence of mature ErbB3 (SEQ ID NO: 73, more preferably involving or spanning or containing
  • inhibitor refers to any statistically significant decrease in biological activity, including full blocking of the activity.
  • “inhibition” can refer to a decrease of about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or 100% in biological activity.
  • the phrase "inhibition of EGF-like ligand mediated phosphorylation of ErbB3,” as used herein, refers to the ability of an antibody or antigen binding portion to statistically significantly decrease the phosphorylation of ErbB3 induced by an EGF- like ligand, relative to the phosphorylation in an untreated (control) cell.
  • the cell which expresses ErbB3 can be a naturally occurring cell or cell line or can be recombinantly produced by introducing nucleic acid encoding ErbB3 into a host cell.
  • the antibody or antigen binding portion thereof inhibits EGF-like ligand mediated phosphorylation of ErbB3 by at least 10%, or at least 20%, or at least 30%, or at least 40%, or at least 50%, or at least 60%, or at least 70%, or at least 80%, or at least 90%, or about 100%, as determined, for example, by Western blotting followed by probing with an anti-phosphotyrosine antibody as described in Kim et al., (1998) Biochem J., 334: 189-195 and the Examples infra.
  • the phrase "inhibition of heregulin, epiregulin, epigen or biregulin-mediated signaling through ErbB3,” as used herein, refers to the ability of an antibody or an antigen-binding portion thereof to statistically significantly decrease signaling mediated by an ErbB3 ligand (e.g., heregulin, epiregullin, epigen and biregulin) through ErbB3, relative to the signaling in the absence of the antibody (control).
  • an ErbB3 ligand e.g., heregulin, epiregullin, epigen and biregulin
  • ErbB3-ligands are also referred to herein as "heregulin-like ligands.” This means that, in the presence of the antibody or antigen binding portion thereof, a signal mediated in a cell expressing ErbB3 by one or more of heregulin, epiregulin, epigen and biregulin, relative to a control (no antibody), is statistically significantly decreased.
  • An ErbB3-ligand mediated signal can be measured by assaying for the level or activity of an ErbB3 substrate, and/or a protein which is present in a cellular cascade involving ErbB3.
  • the antibody or antigen binding portion thereof decreases the level or activity of an ErbB3 substrate and/or that of a protein in a cellular cascade involving ErbB3, by at least 10%, or at least 20%, or at least 30%, or at least 40%, or at least 50%, or at least 60%, or at least 70%, or at least 80%, or at least 90%, or 100% relative to the level or activity in the absence of such antibody or antigen binding portion thereof (control).
  • ErbB3- ligand mediated signaling can be measured using art recognized techniques which measure the level or activity of a substrate of ErbB3 (e.g., SHC or PI3K) or a protein in a cellular cascade involving ErbB3 (e.g., the AKT pathway - AKT refers to a set of serine/threonine kinases also referred to as protein kinases B or PKB) using kinase assays for such proteins (see, e.g., Horst et al. supra, Sudo et al. (2000) Methods Enzymol, 322:388-92; and Morgan et al. (1990) Eur. J. Biochem., 191:761-767).
  • a substrate of ErbB3 e.g., SHC or PI3K
  • AKT pathway - AKT refers to a set of serine/threonine kinases also referred to as protein kinases B
  • the antibody or antigen binding portion thereof inhibits ErbB3-ligand (e.g., heregulin, epiregulin, epigen or biregulin) mediated signaling through ErbB3 by inhibiting the binding of the ErbB3-ligand (e.g., one or more of heregulin, epiregulin, epigen or biregulin) to ErbB3.
  • ErbB3-ligand e.g., heregulin, epiregulin, epigen or biregulin
  • Some ligands function both as EGF-like ligands (i.e., bind to EGFR/ErbBl) as well as ErbB3-like ligands (i.e., bind to ErbB3).
  • ErbB3 refers to the ability of an antibody or an antigen-binding portion thereof to statistically significantly decrease the binding of an ErbB3 ligand (e.g., one or more of heregulin, epiregulin, epigen or biregulin) to ErbB3, relative to the binding in the absence of the antibody (control). This means that, in the presence of the antibody or antigen binding portion thereof, the amount of the ErbB 3 -ligand (e.g., heregulin, epiregulin, epigen or biregulin) which binds to ErbB3 relative to a control (no antibody), is statistically significantly decreased.
  • an ErbB3 ligand e.g., one or more of heregulin, epiregulin, epigen or biregulin
  • the amount of an ErbB3 ligand which binds ErbB3 may be decreased in the presence of an antibody or antigen binding portion thereof of the present disclosure by at least 10%, or at least 20%, or at least 30%, or at least 40%, or at least 50%, or at least 60%, or at least 70%, or at least 80%, or at least 90%, or 100% relative to the amount in the absence of the antibody or antigen binding portion thereof (control).
  • a decrease in ErbB3-ligand binding can be measured using art recognized techniques which measure the level of binding of labeled ErbB3-ligand (e.g., radiolabeled heregulin, epiregulin, epigen or biregulin) to cells expressing ErbB3 in the presence or absence (control) of the antibody or antigen binding portion thereof.
  • labeled ErbB3-ligand e.g., radiolabeled heregulin, epiregulin, epigen or biregulin
  • the phrase "inhibition of proliferation of a cell expressing ErbB3,” as used herein, refers to the ability of an antibody or an antigen-binding portion thereof to statistically significantly decrease proliferation of a cell expressing ErbB3 relative to the proliferation in the absence of the antibody.
  • the proliferation of a cell expressing ErbB3 (e.g., a cancer cell) may be decreased by at least 10%, or at least 20%, or at least 30%, or at least 40%, or at least 50%, or at least 60%, or at least 70%, or at least 80%, or at least 90%, or 100% when the cells are contacted with an antibody or antigen binding portion thereof of the present disclosure, relative to the proliferation measured in the absence of the antibody or antigen binding portion thereof (control).
  • Cellular proliferation can be assayed using art recognized techniques which measure rate of cell division, the fraction of cells within a cell population undergoing cell division, and/or rate of cell loss from a cell population due to terminal differentiation or cell death (e.g., using a CellTiter-Glo® assay or thymidine incorporation).
  • the phrase "the ability to decrease levels of ErbB3 on cell surfaces," as used herein, refers to the ability of an antibody or antigen binding portion thereof to statistically significantly reduce the amount of ErbB3 found on the surface of a cell which has been exposed to the antibody relative to an untreated (control) cell.
  • a decrease in levels of ErbB3 on cell surfaces may result from increased internalization of ErbB3 (or increased ErbB3 endocytosis).
  • the antibody or antigen binding portion thereof decreases cell surface expression of ErbB3 by at least 10%, or at least 20%, or at least 30%, or at least 40%, or at least 50%, or at least 60%, or at least 70%, or at least 80%, or at least 90%, or 100% and/or increases internalization of the ErbB3 receptor by at least 10%, or at least 20%, or at least 30%, or at least 40%, or at least 50%, or at least 60%, or at least 70%, or at least 80%, or at least 90%, or 100% relative to the cell surface expression or internalization in the absence of the antibody or antigen binding portion thereof (control).
  • the levels of ErbB3 on surfaces of cells and/or internalization of the ErbB3 receptor in the absence and the presence of an antibody or antigen-binding portion thereof can be readily measured using art recognized techniques, such as those described in Horst et al., supra and in the examples herein.
  • VEGF secretion of cells expressing ErbB3 refers to the ability of an antibody or an antigen-binding portion thereof to statistically significantly decrease VEGF secretion of a cell expressing ErbB3 relative to the VEGF secretion in the absence of the antibody.
  • the VEGF secretion of a cell expressing ErbB3 may be decreased by at least 10%, or at least 20%, or at least 30%, or at least 40%, or at least 50%, or at least 60%, or at least 70%, or at least 80%, or at least 90%, or 100% when the cells are contacted with an antibody or antigen binding portion thereof of the present disclosure, relative to the VEGF secretion measured in the absence of the antibody or antigen binding portion thereof (control).
  • VEGF secretion can be assayed using art recognized techniques, such as those described herein.
  • the phrase "inhibition of the migration of cells expressing ErbB3,” as used herein, refers to the ability of an antibody or an antigen-binding portion thereof to statistically significantly decrease the migration of a cell expressing ErbB3 relative to the migration of the cell in the absence of the antibody.
  • the migration of a cell expressing ErbB3 (e.g., a cancer cell) may be decreased by at least 10%, or at least 20%, or at least 30%, or at least 40%, or at least 50%, or at least 60%, or at least 70%, or at least 80%, or at least 90%, or 100% when the cells are contacted with an antibody or antigen binding portion thereof of the present disclosure, relative to cell migration measured in the absence of the antibody or antigen binding portion thereof (control).
  • Cell migration can be assayed using art recognized techniques, such as those described herein.
  • inhibitortion of spheroid growth of cells expressing ErbB3 refers to the ability of an antibody or an antigen-binding portion thereof to statistically significantly decrease the migration of a cell expressing ErbB3 relative to the migration of the cell in the absence of the antibody.
  • the migration of a cell expressing ErbB3 may be decreased by at least 10%, or at least 20%, or at least 30%, or at least 40%, or at least 50%, or at least 60%, or at least 70%, or at least 80%, or at least 90%, or 100% when the cells are contacted with an antibody or antigen binding portion thereof of the present disclosure, relative to cell migration measured in the absence of the antibody or antigen binding portion thereof (control).
  • Cell migration can be assayed using art recognized techniques, such as those described herein.
  • antibody or “immunoglobulin,” as used interchangeably herein, includes whole antibodies and any antigen binding fragment (i.e., "antigen-binding portion") or single chains thereof.
  • a typical antibody comprises at least two heavy (H) chains and two light (L) chains inter-connected by disulfide bonds.
  • Each heavy chain is comprised of a heavy chain variable region (abbreviated herein as VH) and a heavy chain constant region.
  • the heavy chain constant region is comprised of three domains, CHI, CH2 and CH3.
  • Each light chain is comprised of a light chain variable region
  • VL a light chain constant region
  • the light chain constant region is comprised of one domain, CL.
  • CL complementarity determining regions
  • FR framework regions
  • Each VH and V L is composed of three CDRs and four FRs, arranged from amino- terminus to carboxy-terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4.
  • the variable regions of the heavy and light chains contain a binding domain that interacts with an antigen.
  • the constant regions of the antibodies may mediate the binding of the immunoglobulin to host tissues or factors, including various cells of the immune system (e.g., effector cells) and the first component (Clq) of the classical complement system.
  • exemplary antibodies of the present disclosure include antibodies#l, 3, 6 and 14, and antigen-binding portions thereof.
  • antibody portion refers to one or more fragments of an antibody that retain the ability to specifically bind to an antigen (e.g., ErbB3). It has been shown that the antigen-binding function of an antibody can be performed by fragments of a full-length antibody.
  • binding fragments encompassed within the term "antigen- binding portion" of an antibody include (i) a Fab fragment, a monovalent fragment consisting of the V L , V H , CL and CHI domains; (ii) a F(ab')2 fragment, a bivalent fragment comprising two Fab fragments linked by a disulfide bridge at the hinge region; (iii) a Fd fragment consisting of the V H and CHI domains; (iv) a Fv fragment consisting of the V L and V H domains of a single arm of an antibody, (v) a dAb including VH and VL domains; (vi) a dAb fragment (Ward et al.
  • V L and VH are coded for by separate genes, they can be joined, using recombinant methods, by a synthetic linker that enables them to be made as a single protein chain in which the V L and VH regions pair to form monovalent molecules (known as single chain Fv (scFv); see e.g., Bird et al.
  • scFv single chain Fv
  • Such single chain antibodies are also intended to be encompassed within the term "antigen-binding portion" of an antibody.
  • antibody fragments are obtained using conventional techniques known to those with skill in the art, and the fragments are screened for utility in the same manner as are intact antibodies.
  • Antigen-binding portions can be produced by recombinant DNA techniques, or by enzymatic or chemical cleavage of intact immunoglobulins.
  • monoclonal antibody refers to an antibody obtained from or prepared as a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical except for possible naturally occurring mutations that may be present in minor amounts. Monoclonal antibodies are highly specific, being directed against a single antigenic site.
  • each monoclonal antibody is typically directed against a single determinant on the antigen.
  • Monoclonal antibodies can be prepared using any art recognized technique and those described herein such as, for example, a hybridoma method, as described by Kohler et al. (1975) Nature, 256:495, a transgenic animal, as described by, for example, (see e.g., Lonberg, et al. (1994) Nature 368(6474): 856-859), recombinant DNA methods (see, e.g., U.S. Pat. No.
  • Monoclonal antibodies include chimeric antibodies, human antibodies and humanized antibodies and may occur naturally or be recombinantly produced.
  • recombinant antibody refers to antibodies that are prepared, expressed, created or isolated by recombinant means, such as (a) antibodies isolated from an animal (e.g., a mouse) that is transgenic or transchromosomal for
  • immunoglobulin genes e.g., human immunoglobulin genes
  • a hybridoma prepared therefrom antibodies isolated from a host cell transformed to express the antibody, e.g., from a transfectoma
  • antibodies isolated from a recombinant, combinatorial antibody library e.g., containing human antibody sequences
  • combinatorial antibody library e.g., containing human antibody sequences
  • Such recombinant antibodies may have variable and constant regions derived from human germline immunoglobulin sequences.
  • such recombinant human antibodies can be subjected to in vitro mutagenesis and thus the amino acid sequences of the VH and VL regions of the recombinant antibodies are sequences that, while derived from and related to human germline V H and VL sequences, may not naturally exist within the human antibody germline repertoire in vivo.
  • chimeric immunoglobulin or “chimeric antibody” refers to an immunoglobulin or antibody whose variable regions derive from a first species and whose constant regions derive from a second species. Chimeric immunoglobulins or antibodies can be constructed, for example by genetic engineering, from
  • immunoglobulin gene segments belonging to different species belonging to different species.
  • human antibody is intended to include antibodies having variable regions in which both the framework and CDR regions are derived from human germline immunoglobulin sequences as described, for example, by Kabat et al. (See Kabat, et al. (1991) Sequences of proteins of Immunological Interest, Fifth Edition, U.S. Department of Health and Human Services, NIH Publication No. 91-3242).
  • the constant region also is derived from human germline immunoglobulin sequences.
  • the human antibodies may include amino acid residues not encoded by human germline immunoglobulin sequences (e.g., mutations' introduced by random or site-specific mutagenesis in vitro or by somatic mutation in vivo).
  • the term "human antibody”, as used herein, is not intended to include antibodies in which CDR sequences derived from the germline of another mammalian species, such as a mouse, have been grafted onto human framework sequences.
  • the human antibody can have at least one ore more amino acids replaced with an amino acid residue, e.g., an activity enhancing amino acid residue which is not encoded by the human germline immunoglobulin sequence.
  • the human antibody can have up to twenty positions replaced with amino acid residues which are not part of the human germline immunoglobulin sequence. In a particular embodiment, these replacements are within the CDR regions as described in detail below.
  • the term "humanized immunoglobulin” or “humanized antibody” refers to an immunoglobulin or antibody that includes at least one humanized immunoglobulin or antibody chain (i.e., at least one humanized light or heavy chain).
  • humanized immunoglobulin chain or “humanized antibody chain” (i.e., a "humanized
  • immunoglobulin light chain refers to an immunoglobulin or antibody chain (i.e., a light or heavy chain, respectively) having a variable region that includes a variable framework region substantially from a human immunoglobulin or antibody and complementarity determining regions (CDRs) (e.g., at least one CDR, preferably two CDRs, more preferably three CDRs) substantially from a non-human immunoglobulin or antibody, and further includes constant regions (e.g., at least one constant region or portion thereof, in the case of a light chain, and preferably three constant regions in the case of a heavy chain).
  • CDRs complementarity determining regions
  • humanized variable region refers to a variable region that includes a variable framework region substantially from a human immunoglobulin or antibody and complementarity determining regions (CDRs) substantially from a non-human immunoglobulin or antibody.
  • CDRs complementarity determining regions
  • bispecific or “bifunctional” antibody is an artificial hybrid antibody having two different heavy/light chain pairs and two different binding sites.
  • Bispecific antibodies can be produced by a variety of methods including fusion of hybridomas or linking of Fab' fragments. See, e.g., Songsivilai & Lachmann, (1990) Clin. Exp.
  • a bispecific antibody according to the present invention includes binding sites for both ErbB3 and IGF1-R (i.e., insulin-like growth factor 1-receptor). In another embodiment, a bispecific antibody according to the present invention includes binding sites for both ErbB3 and C-MET.
  • a bispecific antibody includes a binding site for ErbB3 and a binding site for ErbB2, ERbB3, ErbB4, EGFR, Lewis Y, MUC-1, EpCAM, CA125, prostate specific membrane antigen, PDGFR-a, PDGFR- ⁇ , C- ⁇ , or any of the FGF receptors.
  • a heterologous antibody is defined in relation to the transgenic non-human organism or plant producing such an antibody.
  • an “isolated antibody,” as used herein, is intended to refer to an antibody which is substantially free of other antibodies having different antigenic specificities (e.g., an isolated antibody that specifically binds to ErbB3 is substantially free of antibodies that specifically bind antigens other than ErbB3).
  • an isolated antibody is typically substantially free of other cellular material and/or proteins.
  • a combination of "isolated” antibodies having different ErbB3 binding specificities are combined in a well defined composition.
  • “isotype” refers to the antibody class (e.g., IgM or IgGl) that is encoded by heavy chain constant region genes.
  • an antibody or antigen binding portion thereof is of an isotype selected from an IgGl, an IgG2, an IgG3, an IgG4, an IgM, an IgAl, an IgA2, an IgAsec, an IgD, or an IgE antibody isotype.
  • an antibody is of the IgGl isotype.
  • an antibody is of the IgG2 isotype.
  • isotype switching refers to the phenomenon by which the class, or isotype, of an antibody changes from one Ig class to one of the other Ig classes.
  • nonswitched isotype refers to the isotypic class of heavy chain that is produced when no isotype switching has taken place; the CH gene encoding the nonswitched isotype is typically the first CH gene immediately downstream from the functionally rearranged VDJ gene. Isotype switching has been classified as classical or non-classical isotype switching.
  • Classical isotype switching occurs by recombination events which involve at least one switch sequence regions in a gene encoding an antibody.
  • Non-classical isotype switching may occur by, for example, homologous recombination between human ⁇ ⁇ and human ⁇ ⁇ ( ⁇ -associated deletion).
  • Alternative non-classical switching mechanisms such as intertransgene and/or interchromosomal recombination, among others, may occur and effectuate isotype switching.
  • switch sequence refers to those DNA sequences responsible for switch recombination.
  • a "switch donor” sequence typically a ⁇ switch region, will be 5' (i.e., upstream) of the construct region to be deleted during the switch recombination.
  • the "switch acceptor” region will be between the construct region to be deleted and the replacement constant region (e.g., , ⁇ , etc.). As there is no specific site where recombination always occurs, the final gene sequence will typically not be predictable from the construct.
  • an “antigen” is an entity (e.g., a proteinaceous entity or peptide) to which an antibody or antigen-binding portion thereof binds.
  • the antigen is ErbB3 or a ErbB3-like molecule.
  • the antigen is human ErbB3.
  • epitopes refers to a site on an antigen to which an immunoglobulin or antibody specifically binds.
  • Epitopes can be formed both from contiguous amino acids or noncontiguous amino acids juxtaposed by tertiary folding of a protein. Epitopes formed from contiguous amino acids are typically retained on exposure to denaturing solvents, whereas epitopes formed by tertiary folding are typically lost on treatment with denaturing solvents.
  • An epitope typically includes at least 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14 or 15 amino acids in a unique spatial conformation.
  • Methods of determining spatial conformation of epitopes include techniques in the art and those described herein, for example, x-ray crystallography and 2 -dimensional nuclear magnetic resonance. See, e.g., Epitope Mapping Protocols in Methods in Molecular Biology, Vol. 66, G. E. Morris, Ed. (1996).
  • antibodies that bind the same or an overlapping epitope as the antibodies for which amino acid sequences are disclosed herein i.e., antibodies that compete for binding to ErbB3, or bind epitopes which overlap with epitopes bound by the antibodies described herein, i.e., an epitope located on ectodomain of ErbB3, preferably on Domain I of the ectodomain of ErbB3.
  • Antibodies that recognize the same epitope can be identified using routine techniques such as an immunoassay, for example, by showing the ability of one antibody to block the binding of another antibody to a target antigen, i.e., a competitive binding assay.
  • Competitive binding is determined in an assay in which the immunoglobulin under test inhibits specific binding of a reference antibody to a common antigen, such as ErbB3.
  • such an assay involves the use of purified antigen (e.g., ErbB3) bound to a solid surface or cells bearing either of these, an unlabeled test immunoglobulin and a labeled reference immunoglobulin.
  • Purified antigen e.g., ErbB3
  • an unlabeled test immunoglobulin e.g., an unlabeled test immunoglobulin and a labeled reference immunoglobulin.
  • Competitive inhibition is measured by determining the amount of label bound to the solid surface or cells in the presence of the test immunoglobulin.
  • the test immunoglobulin is present in excess.
  • a competing antibody is present in excess, it will inhibit specific binding of a reference antibody to a common antigen by at least 50-55%, 55-60%, 60- 65%, 65-70% 70-75% or more.
  • paratope refers to the parts, or amino acid residues, of an antibody that appear to be directly involved in recognizing and contacting the epitope on an antigen to which the antibody specifically binds.
  • Paratopes typically comprise some but not all amino acid residues within the complementarity determining regions (CDRs) of the heavy and light chains.
  • CDRs complementarity determining regions
  • a paratope may involve amino acid residues in all of the VH and V L CDRs or only some of the CDRs (e.g., certain CDRs may not be involved in binding antigen).
  • the paratope for a particular antigen can be defined, for example, by scanning mutagenesis (e.g., alanine scanning mutagenesis) of amino acid residues within the antibody, particularly the CDRs, that are thought to be surface exposed (e.g., as determined by crystallographic modeling) and possibly involved in antigen binding.
  • scanning mutagenesis e.g., alanine scanning mutagenesis
  • Evaluation of the binding of the mutants to the antigen then can determine whether the mutated amino acid position is involved in antigen binding and thus forms part of the paratope of the antibody. Methods for determining an antibody paratope are described in further detail in Example 20.
  • an anti-ErbB3 antibody comprises a heavy chain paratope comprising CDRl, CDR2 and CDR3 sequences as shown in SEQ ID NOs: 63 or 76 (CDRl), 64 (CDR2) and 65 (CDR3), respectively, with the proviso that the antibody is not: (i) Ab #6 as disclosed herein; (ii) an antibody comprising VH and VL sequences as shown in SEQ ID NOs: 1 and 2, respectively; or (iii) an antibody
  • V H CDRl comprising V H CDRl, CDR2 and CDR3 sequences as shown in SEQ ID NOs: 7, 8 and
  • an anti-ErbB3 antibody comprises a light chain > - paratope comprising CDRl, CDR2 and CDR3 sequences as shown in SEQ ID NOs: 69 or 78 (CDRl), 70 (CDR2) and 71 or 80 (CDR3), respectively, with the proviso that the antibody is not: (i) Ab #6 as disclosed herein; (ii) an antibody comprising VH and VL sequences as shown in SEQ ID NOs: 1 and 2, respectively; or (iii) an antibody
  • the anti-ErbB3 antibody comprises a heavy chain paratope comprising CDRl , CDR2 and CDR3 sequences as shown in SEQ ID NOs: 63, 64 and 65, respectively, and a light chain paratope comprising CDRl, CDR2 and CDR3 sequences as shown in SEQ ID NOs: 69, 70 and 71, respectively, with the proviso that the antibody is not: (i) Ab #6 as disclosed herein; (ii) an antibody comprising VH and VL sequences as shown in SEQ ID NOs: 1 and 2, respectively; or (iii) an antibody
  • the anti-ErbB3 antibody comprises a heavy chain paratope comprising CDRl, CDR2 and CDR3 sequences as shown in SEQ ID NOs: 76, 64 and 65, respectively, and a light chain paratope comprising CDRl, CDR2 and CDR3 sequences as shown in SEQ ID NOs: 78, 70 and 80, respectively, with the proviso that the antibody is not: (i) Ab #6 as disclosed herein; (ii) an antibody comprising VH and VL sequences as shown in SEQ ID NOs: 1 and 2, respectively; or (iii) an antibody
  • CDRs complementarity determining regions
  • a CDR complementarity determining regions
  • certain amino acid positions are occupied by one of multiple possible amino acid residues at that position. For example, within a CDR, if antigen binding has been found to be unaffected by the presence of either a tyrosine or a phenylalanine at a particular position, then that particular position within the consensus sequence can be either tyrosine or phenylalanine (T/F).
  • Consensus sequences for CDRs can be defined, for example, by scanning mutagenesis (e.g., alanine scanning mutagenesis) of amino acid residues within the antibody CDRs, followed by evaluation of the binding of the mutants to the antigen to determine whether the mutated amino acid position affects antigen binding. Methods for determining antibody CDR consensus sequences are described in further detail in Example 20.
  • an anti-ErbB3 antibody comprises a heavy chain variable region comprising consensus heavy chain CDRl , CDR2 and CDR3 sequences shown in SEQ ID NOs: 60 or 75 (CDRl), 61 (CDR2) and 62 (CDR3), respectively, with the proviso that the antibody is not: (i) Ab #6 as disclosed herein; (ii) an antibody comprising VH and V L sequences as shown in SEQ ID NOs: 1 and 2, respectively; or (iii) an antibody comprising VH CDRl, CDR2 and CDR3 sequences as shown in SEQ ID NOs: 7, 8 and 9, respectively, and V L CDRl, CDR2 and CDR3 sequences as shown in SEQ ID NOs: 10, 1 1 and 12, respectively.
  • an anti-ErbB3 antibody comprises a light chain variable region comprising consensus light chain CDRl, CDR2 and CDR3 sequences shown in SEQ ID NOs: 66 or 77 (CDRl), 67 (CDR2) and 68 or 79 (CDR3), respectively, with the proviso that the antibody is not: (i) Ab #6 as disclosed herein; (ii) an antibody comprising V H and V L sequences as shown in SEQ ID NOs: 1 and 2, respectively; or (iii) an antibody comprising V H CDRl , CDR2 and CDR3 sequences as shown in SEQ ID NOs: 7, 8 and 9, respectively, and V L CDRl , CDR2 and CDR3 sequences as shown in SEQ ED NOs: 10, 1 1 and 12, respectively.
  • the anti-ErbB3 antibody comprises a heavy chain variable region comprising consensus heavy chain CDRl , CDR2 and CDR3 sequences shown in SEQ ED NOs: 60, 61 and 62, respectively, and a light chain variable region comprising consensus light chain CDRl, CDR2 and CDR3 sequences shown in SEQ ED NOs: 66, 67 and 68, respectively, with the proviso that the antibody is not: (i) Ab #6 as disclosed herein; (ii) an antibody comprising V H and V L sequences as shown in SEQ ED NOs: 1 and 2, respectively; or (iii) an antibody comprising V H CDRl, CDR2 and CDR3 sequences as shown in SEQ ID NOs: 7, 8 and 9, respectively, and V L CDRl, CDR2 and CDR3 sequences as shown in SEQ ID NOs: 10, 11 and 12, respectively.
  • the anti-ErbB3 antibody comprises a heavy chain variable region comprising consensus heavy chain CDRl, CDR2 and CDR3 sequences shown in SEQ ID NOs: 75, 61 and 62, respectively, and a light chain variable region comprising consensus light chain CDRl, CDR2 and CDR3 sequences shown in SEQ ID NOs: 77, 67 and 79, respectively, with the proviso that the antibody is not: (i) Ab #6 as disclosed herein; (ii) an antibody comprising VH and VL sequences as shown in SEQ ID NOs: 1 and 2, respectively; or (iii) an antibody comprising V H CDRl, CDR2 and CDR3 sequences as shown in SEQ ID NOs: 7, 8 and 9, respectively, and VL CDRl, CDR2 and CDR3 sequences as shown in SEQ ID NOs: 10, 1 1 and 12, respectively.
  • telomere binding means that an antibody or antigen-binding portion thereof, exhibits appreciable affinity for a particular antigen or epitope and, generally, does not exhibit significant cross-reactivity with other antigens and epitopes.
  • “Appreciable” or preferred binding includes binding with an affinity of at least 10 6 , 10 7 , 10 8 , 10 9 M “1 , or 10 10 M “1 . Affinities greater than 10 7 M “ ', preferably greater than 10 8 M “1 are more preferred. Values intermediate of those set forth herein are also intended to be within the scope of the present invention and a preferred binding affinity can be indicated as a range of affinities, for example, 10 6 to 10 10 M “1 , preferably 10 7 to 10 10 M “ ', more preferably 10 8 to 10 10 M “1 .
  • An antibody that "does not exhibit significant cross- reactivity" is one that will not appreciably bind to an undesirable entity (e.g. , an undesirable proteinaceous entity).
  • an antibody or antigen-binding portion thereof that specifically binds to ErbB3 will appreciably bind that ErbB3 molecule but will not significantly react with other ErbB molecules and non- ErbB proteins or peptides.
  • Specific or selective binding can be determined according to any art-recognized means for determining such binding, including, for example, according to Scatchard analysis and/or competitive binding assays.
  • KD is intended to refer to the dissociation equilibrium constant of a particular antibody-antigen interaction or the affinity of an antibody for an antigen, preferably as measured using a surface plasmon resonance assay (e.g., as determined in a BIACORE 3000 instrument (GE Healthcare) using recombinant ErbB3 as the analyte and the antibody as the ligand) or a cell binding assay. Both such assays are detailed in Example 3, below.
  • the antibody or antigen binding portion thereof according to the present invention binds an antigen (e.g., ErbB3) with an affinity (K D ) of 50 nM or better (i.e., or less) (e.g., 40 nM or 30 nM or 20 nM or 10 nM or less),.
  • an antibody or antigen binding portion thereof according to the present invention binds ErbB3 with an affinity (K D ) of 8 nM or better (e.g., 7 nM, 6 nM, 5 nM, 4 nM, 2 nM, 1.5 nM, 1.4 nM, 1.3 nM, InM or less.
  • an antibody or antigen binding portion thereof binds an antigen (e.g., ErbB3) with an affinity (K D ) of approximately less than 10 "7 M, such as approximately less than 10 8 M, 10 "9 M or 10 '10 M or even lower, and binds to the predetermined antigen with an affinity that is at least two-fold greater than its affinity for binding to a non-specific antigen (e.g., BSA, casein) other than the predetermined antigen or a closely-related antigen.
  • an antigen e.g., ErbB3
  • K D affinity
  • a non-specific antigen e.g., BSA, casein
  • K o m is intended to refer to the off rate constant for the dissociation of an antibody from the antibody/antigen complex.
  • EC50 refers to the concentration of an antibody or an antigen-binding portion thereof, which induces a response, either in an in vitro or an in vivo assay, which is 50% of the maximal response, i.e., halfway between the maximal response and the baseline.
  • glycosylation pattern is defined as the pattern of carbohydrate units that are covalently attached to a protein, more specifically to an immunoglobulin protein.
  • naturally-occurring refers to the fact that an object can be found in nature.
  • a polypeptide or polynucleotide sequence that is present in an organism (including viruses) that can be isolated from a source in nature and which has not been intentionally modified by man in the laboratory is naturally-occurring.
  • a rearranged immunoglobulin gene locus can be identified by comparison to germline DNA; a rearranged locus will have at least one recombined heptamer/nonamer homology element.
  • V segment configuration refers to the configuration wherein the V segment is not recombined so as to be immediately adjacent to a D or J segment.
  • nucleic acid molecule is intended to include DNA molecules and RNA molecules.
  • a nucleic acid molecule may be single-stranded or double-stranded, but preferably is double-stranded DNA.
  • isolated nucleic acid molecule as used herein in reference to nucleic acids encoding antibodies or antibody portions (e.g., V H , V L , CDR3) that bind to ErbB3, is intended to refer to a nucleic acid molecule in which the nucleotide sequences encoding the antibody or antibody portion are free of other nucleotide sequences encoding antibodies that bind antigens other than ErbB3, which other sequences may naturally flank the nucleic acid in human genomic DNA.
  • modifying is intended to refer to changing one or more amino acids in the antibodies or antigen-binding portions thereof.
  • the change can be produced by adding, substituting or deleting an amino acid at one or more positions.
  • the change can be produced using known techniques, such as PCR mutagenesis.
  • an antibody or an antigen-binding portion thereof identified using the methods of the present disclosure can be modified, to thereby modify the binding affinity of the antibody or antigen-binding portion thereof to ErbB3.
  • the present invention also encompasses "conservative amino acid substitutions" in the sequences of the antibodies or fragments therof of the invention, i.e., nucleotide and amino acid sequence modifications which do not abrogate the binding of the antibody encoded by the nucleotide sequence or containing the amino acid sequence, to the antigen, i.e., ErbB3.
  • Conservative amino acid substitutions include the substitution of an amino acid in one class by an amino acid of the same class, where a class is defined by common physicochemical amino acid side chain properties and high substitution frequencies in homologous proteins found in nature, as determined, for example, by a standard Dayhoff frequency exchange matrix or BLOSUM matrix.
  • non-conservative amino acid substitution refers to the substitution of an amino acid in one class with an amino acid from another class; for example, substitution of an Ala, a class ⁇ residue, with a class ⁇ residue such as Asp, Asn, Glu, or Gin.
  • mutations can be introduced randomly along all or part of an anti-ErbB3 antibody coding sequence, such as by saturation mutagenesis, and the resulting modified anti- ErbB3 antibodies can be screened for binding activity.
  • a "consensus sequence” is a sequence formed from the most frequently occurring amino acids (or nucleotides) in a family of related sequences (See e.g., Winnaker, From Genes to Clones (Verlagsgesellschaft, Weinheim, Germany 1987). In a family of proteins, each position in the consensus sequence is occupied by the amino acid occurring most frequently at that position in the family. If two amino acids occur equally frequently, either can be included in the consensus sequence.
  • a "consensus framework" of an immunoglobulin refers to a framework region in the consensus immunoglobulin sequence.
  • the consensus sequence for the CDRs of can be derived by optimal alignment of the CDR amino acid sequences of ErbB3 antibodies for which CDR ⁇ amino acid sequences are disclosed herein.
  • nucleic acids For nucleic acids, the term "substantial homology" indicates that two nucleic acids, or designated sequences thereof, when optimally aligned and compared, are identical, with appropriate nucleotide insertions or deletions, in at least about 80% of the nucleotides, usually at least about 90% to 95%, and more preferably at least about 98% to 99.5% of the nucleotides. Alternatively, substantial homology exists when the segments will hybridize under selective hybridization conditions, to the complement of the strand.
  • the comparison of sequences and determination of percent identity between two sequences can be accomplished using a mathematical algorithm, as described in the non-limiting examples below.
  • the percent identity between two nucleotide sequences can be determined using the GAP program in the GCG software, using a NWSgapdna.CMP matrix and a gap weight of 40, 50, 60, 70, or 80 and a length weight of 1, 2, 3, 4, 5, or 6.
  • the percent identity between two nucleotide or amino acid sequences can also be determined using the algorithm of E. Meyers and W. Miller (CABIOS, 4: 11-17 (1989)) which has been incorporated into the ALIGN program (version 2.0), using a PAM120 weight residue table, a gap length penalty of 12 and a gap penalty of 4.
  • the percent identity between two amino acid sequences can be determined using the Needleman and Wunsch (/. Mol. Biol.
  • nucleic acid and protein sequences of the present disclosure can further be used as a "query sequence" to perform a search against public databases to, for example, identify related sequences. Such searches can be performed using the NBLAST and XBLAST programs (version 2.0) of Altschul, et al. (1990) J. Mol. Biol. 215:403-10.
  • Gapped BLAST can be utilized as described in Altschul et al, (1997) Nucleic Acids Res. 25(17):3389-3402.
  • the default parameters of the respective programs ⁇ e.g., XBLAST and NBLAST) can be used.
  • the nucleic acids may be present in whole cells, in a cell lysate, or in a partially purified or substantially pure form.
  • a nucleic acid is "isolated” or “rendered substantially pure” when purified away from other cellular components or other contaminants, e.g., other cellular nucleic acids or proteins, by standard techniques, including alkaline/SDS treatment, CsCl banding, column chromatography, agarose gel electrophoresis and others well known in the art. See, F. Ausubel, et al, ed. Current Protocols in Molecular Biology, Greene Publishing and Wiley Interscience, New York (1987).
  • nucleic acid compositions of the present disclosure while often in a native sequence (except for modified restriction sites and the like), from either cDNA, genomic or mixtures thereof may be mutated, in accordance with standard techniques to provide gene sequences. For coding sequences, these mutations, may affect amino acid sequence as desired.
  • DNA sequences substantially homologous to or derived from native V, D, J, constant, switches and other such sequences described herein are contemplated (where "derived" indicates that a sequence is identical or modified from another sequence).
  • operably linked refers to a nucleic acid sequence placed into a functional relationship with another nucleic acid sequence.
  • DNA for a presequence or secretory leader is operably linked to DNA for a polypeptide if it is expressed as a preprotein that participates in the secretion of the polypeptide;
  • a promoter or enhancer is operably linked to a coding sequence if it affects the transcription of the sequence; or
  • a ribosome binding site is operably linked to a coding sequence if it is positioned so as to facilitate translation.
  • “operably linked” means that the DNA sequences being linked are contiguous, and, in the case of a secretory leader, contiguous and in reading phase.
  • a nucleic acid is "operably linked" when it is placed into a functional relationship with another nucleic acid sequence.
  • a promoter or enhancer is operably linked to a coding sequence if it affects the transcription of the sequence.
  • operably linked means that the DNA sequences being linked are contiguous and, where necessary to join two protein coding regions, contiguous and in reading frame.
  • operably linked indicates that the sequences are capable of effecting switch
  • vector is intended to refer to a nucleic acid molecule capable of transporting another nucleic acid to which it has been linked.
  • plasmid refers to a circular double stranded DNA loop into which additional DNA segments may be ligated.
  • viral vector Another type of vector is a viral vector, wherein additional DNA segments may be ligated into the viral genome.
  • Certain vectors are capable of autonomous replication in a host cell into which they are introduced (e.g. , bacterial vectors having a bacterial origin of replication and episomal mammalian vectors). Other vectors (e.g.
  • non-episomal mammalian vectors can be integrated into the genome of a host cell upon introduction into the host cell, and thereby are replicated along with the host genome.
  • certain vectors are capable of directing the expression of genes to which they are operatively linked.
  • Such vectors are referred to herein as "recombinant expression vectors"(or simply, "expression vectors”).
  • expression vectors of utility in recombinant DNA techniques are often in the form of plasmids.
  • plasmid and vector may be used interchangeably.
  • the invention is intended to include such other forms of expression vectors, such as viral vectors (e.g. , replication defective retroviruses, adenoviruses and adeno- associated viruses), which serve equivalent functions.
  • recombinant host cell (or simply “host cell”), as used herein, is intended to refer to a cell into which a recombinant expression vector has been introduced. It should be understood that such terms are intended to refer not only to the particular subject cell but to the progeny of such a cell. Because certain modifications may occur in succeeding generations due to either mutation or environmental influences, such progeny may not, in fact, be identical to the parent cell, but are still included within the scope of the term “host cell” as used herein.
  • treat refers to therapeutic or preventative measures described herein.
  • the methods of “treatment” employ administration to a subject, an antibody or antigen binding portion disclosed herein, for example, a subject having a disease or disorder associated with ErbB3 dependent signaling or predisposed to having such a disease or disorder, in order to prevent, cure, delay, reduce the severity of, or ameliorate one or more symptoms of the disease or disorder or recurring disease or disorder, or in order to prolong the survival of a subject beyond that expected in the absence of such treatment.
  • disease associated with ErbB3 dependent signaling includes disease states and/or symptoms associated with a disease state, where increased levels of ErbB3 and/or activation of cellular cascades involving ErbB3 are found. It is understood that ErbB3 heterodimerizes with other ErbB proteins such as, EGFR and ErbB2, when increased levels of ErbB3 are found. Accordingly, the term “disease associated with ErbB3 dependent signaling,” also includes disease states and/or symptoms associated with disease states where increased levels of EGFR/ErbB3 and/or ErbB2 ErbB3 heterodimers are found.
  • disease associated with ErbB3 dependent signaling refers to any disorder, the onset, progression or the persistence of the symptoms of which requires the participation of ErbB3.
  • Exemplary ErbB3-mediated disorders include, but are not limited to, for example, cancer.
  • cancer and “cancerous” refer to or describe the physiological condition in mammals that is typically characterized by unregulated cell growth.
  • cancer examples include but are not limited to, carcinoma, lymphoma, blastoma, sarcoma, and leukemia. More particular examples of such cancers include squamous cell cancer, small-cell lung cancer, non-small cell lung cancer, gastric cancer, pancreatic cancer, glial cell tumors such as glioblastoma and neurofibromatosis, cervical cancer, ovarian cancer, liver cancer, bladder cancer, hepatoma, breast cancer, colon cancer, melanoma, colorectal cancer, endometrial carcinoma, salivary gland carcinoma, kidney cancer, renal cancer, prostate cancer, vulval cancer, thyroid cancer, hepatic carcinoma and various types of head and neck cancer.
  • a cancer treated or diagnosed using the methods disclosed herein is selected from melanoma, breast cancer, ovarian cancer, renal carcinoma, gastrointestinal/colon cancer, lung cancer, and prostate cancer.
  • KRAS mutation refers to mutations found in certain cancers in a human homolog of the v-Ki-ras2 Kirsten rat sarcoma viral oncogene.
  • Non- limiting examples of human KRAS gene mRNA sequences include Genbank Accession Nos. NM_004985 and NM_033360. It has been reported that KRAS mutations are found in 73% of pancreatic tumors, 35% of colorectal tumors, 16% of ovarian tumors and 17% of lung tumors.
  • PI3K mutation refers to mutations found in certain cancers in a phosphatidyl-inositol-3-kinase gene, typically leading to activation of PDK in the cancer cells. It has been reported that PI3K mutations are found in 12% of colorectal tumors, 15% of head and neck tumors and 26% of breast tumors.
  • an effective amount refers to that amount of an antibody or an antigen binding portion thereof that binds ErbB3, which is sufficient to effect treatment, prognosis or diagnosis of a disease associated with ErbB3 dependent signaling, as described herein, when administered to a subject.
  • a therapeutically effective amount will vary depending upon the subject and disease condition being treated, the weight and age of the subject, the severity of the disease condition, the manner of administration and the like, which can readily be determined by one of ordinary skill in the art.
  • the dosages for administration can range from, for example, about 1 ng to about 10,000 mg, about 5 ng to about 9,500 mg, about 10 ng to about 9,000 mg, about 20 ng to about 8,500 mg, about 30 ng to about 7,500 mg, about 40 ng to about 7,000 mg, about 50 ng to about 6,500 mg, about 100 ng to about 6,000 mg, about 200 ng to about 5,500 mg, about 300 ng to about 5,000 mg, about 400 ng to about 4,500 mg, about 500 ng to about 4,000 mg, about 1 ⁇ g to about 3,500 mg, about 5 ⁇ g to about 3,000 mg, about 10 ⁇ g to about 2,600 mg, about 20 ⁇ g to about 2,575 mg, about 30 ⁇ g to about 2,550 mg, about 40 ⁇ g to about 2,500 mg, about 50 ⁇ g to about 2,475 mg, about 100 ⁇ g to about 2,450 mg, about 200 ⁇ g to about 2,425 mg, about 300 ⁇ g to about 2,000, about 400 ⁇ g to about 1,175 mg
  • Dosage regimen may be adjusted to provide the optimum therapeutic response.
  • An effective amount is also one in which any toxic or detrimental effects (i.e., side effects) of an antibody or antigen binding portion thereof are minimized and/or outweighed by the beneficial effects. Additional preferred dosages regimens are described further below in the section pertaining to pharmaceutical compositions.
  • patient includes human and other mammalian subjects that receive either prophylactic or therapeutic treatment.
  • the term "subject” or “patient” includes any human or non-human animal.
  • the methods and compositions disclosed herein can be used to treat a subject having cancer.
  • the subject is a human.
  • non-human animal includes all vertebrates, e.g., mammals and non-mammals, such as non-human primates, sheep, dog, cow, etc.
  • sample refers to tissue, body fluid, or a cell from a patient or a subject. Normally, the tissue or cell will be removed from the patient, but in vivo diagnosis is also contemplated.
  • a tissue sample can be taken from a surgically removed tumor and prepared for testing by conventional techniques.
  • lymphomas and leukemias lymphocytes, leukemic cells, or lymph tissues can be obtained and appropriately prepared.
  • Other patient samples including urine, tear drops, serum, cerebrospinal fluid, feces, sputum, cell extracts etc. can also be useful for particular tumors.
  • anti-cancer agent and “antineoplastic agent” refer to drugs used to treat malignancies, such as cancerous growths. Drug therapy may be used alone, or in combination with other treatments such as surgery or radiation therapy. Several classes of drugs may be used in cancer treatment, depending on the nature of the organ involved. For example, breast cancers are commonly stimulated by estrogens, and may be treated with drugs which inactive the sex hormones. Similarly, prostate cancer may be treated with drugs that inactivate androgens, the male sex hormone.
  • Anti-cancer agents for use in certain methods of the present invention include, among others, the following agents:
  • Antibodies which bind A12 (fully humanized mAb)
  • anti-ErbB3 growth factor type 1 CP751-871 (fully humanized mAb) antibodies; and receptor), which is H7C10 (humanized mAb)
  • mAb 806 result in cancer nimotuzumab (TheraCIM)
  • panitumumab (Vectibix®; Amgen)
  • Anti-ErbB3 antibodies Ab #14 described herein which bind different 1B4C3; 2D1D12 (U3 Pharma AG) epitopes U3-1287/AMG888 (U3 Pharma/Amgen)
  • Anti-ErbB2 Herceptin® (trastuzumab; Genentech/Roche) antibodies binds ectodomain Domain II of ErbB2;
  • IGF-1R insulin-like NVP-AEW541 - A
  • IGF1R growth factor type 1 BMS-536,924 1 H-benzoimidazol-2-yl- 1 H- receptor, which is pyridin-2-one
  • Antimetabolites An antimetabolite is a flourouracil (5-FU)
  • 6- mercaptopurine (Mercaptopurine, 6-MP) azathioprine / Azasan® (AAIPHARMA LLC) 6-thioguanine (6-TG) / Purinethol® (TEVA) pentostatin / Nipent® (Hospira Inc.) fludarabine phosphate / Fludara® (Bayer Health Care)
  • cladribine / Leustatin® (2-CdA, 2- chlorodeoxyadenosine) (Ortho Biotech) floxuridine (5-fluoro-2'-deoxyuridine) / FUDR® (Hospira, Inc,)
  • RNR Ribonucleotide Reductase Inhibitor
  • BMS Cytoxan®
  • TEVA Neosar®
  • chlorambucil / Leukaran® Smith line Beecham
  • Topoisomerase Topoisomerase inhibitors doxorubicin HCL / Doxil® (Alza) inhibitors are chemotherapy agents daunorubicin citrate / Daunoxome® (Gilead) designed to interfere with mitoxantrone HCL Novantrone (EMD the action of Serono)
  • camptothecin CPT
  • Microtubule Microtubules are one of vincristine / Oncovin® (Lilly)
  • docetaxel / Taxotere® (Sanofi Aventis US) Microtubules serve as Nanoparticle paclitaxel (AB 1-007) / structural components Abraxane® (Abraxis Bioscience, Inc.) within cells and are ixabepilone / DCEMPRATM (BMS) involved in many cellular larotaxel
  • Tyrosine kinases are imatinib mesylate / Gleevec (Novartis) enzymes within the cell sunitinib malate / Sutent® (Pfizer) that function to attach sorafenib tosylate / Nexavar® (Bayer) phosphate groups to the nilotinib hydrochloride monohydrate / amino acid tyrosine.
  • Tasigna® Novartis
  • AMG 386 Amgen
  • protein tyrosine kinases axitinib (AG-013736; Pfizer, Inc.)
  • BMS-582664 BMS
  • BMS cancerous cediranib
  • AZD2171 Recentin, AstraZeneca
  • dasatinib BMS-354825: Sprycel®; BMS
  • lestaurtinib CEP-701; Cephalon
  • AMG-706 AMG-706
  • pazopanib HCL (GW786034; Armala, GSK) semaxanib (SU5416; Pharmacia) vandetanib (AZD647; Zactima; AstraZeneca) vatalanib (PTK-787; Novartis, Bayer Schering Pharma)
  • Hormonal therapies Hormonal therapies Ttoremifene citrate / Fareston® (GTX, Inc.) associated with fulvestrant / Faslodex® (AstraZeneca) menopause and aging raloxifene HCL / Evista® (Lilly) seek to increase the anastrazole / Arimidex® (AstraZeneca) amount of certain letrozole / Femara® (Novartis)
  • Hormonal leuprolide acetate / Eligard® (QTL USA) therapy as a cancer Lupron® (TAP Pharm.) treatment generally either goserelin acetate / Zoladex® (AstraZeneca) reduces the level of one triptorelin pamoate / Trelstar® (Watson Labs) or more specific buserelin / Suprefact® (Sanofi Aventis) hormones, blocks a nafarelin
  • hormone abiraterone acetate CB7630; BTG pic
  • hormone afimoxifene TamoGel; Ascend Therapeutics, synthesis inhibitors. In Inc.
  • hormone aromatase inhibitor (Atamestane plus agonists may also be used toremifene; Intarcia Therapeutics, Inc.) as anticancer hormonal arzoxifene (Eli Lilly & Co)
  • letrozole (CGS20267) (Femara®, Chugai; Estrochek®, (Jagsonpal Pharmaceuticals Ltd;) Delestrogen®, estradiol valerate (Jagsonpal) magestrol acetate / Megace®
  • MT206 Medisyn Technologies, Inc.
  • Zestabolin® Mankind Pharma Ltd
  • tamoxifen (Taxifen®, Yung Shin
  • tamoxifen citrate Nolvadex, AstraZeneca; soltamox, EUSA Pharma Inc;
  • Aromatase inhibitors Includes imidazoles ketoconazole mTOR inhibitors
  • the mTOR signaling sirolimus (Rapamycin) /Rapamune® (Wyeth) pathway was originally Temsirolimus (CCI-779) / Torisel® (Wyeth) discovered during studies Deforolimus (AP23573) (Ariad Pharm.) of the Everolimus (RADOOl) /Certican® (Novartis) immunosuppressive agent
  • PI-3K phosphoinositide-3- kinase
  • PKT Inhibitor Astex® Astex Therapeutics
  • NERVIANO Neviano
  • AKT Kinase Inhibitor TELIK (Telik Inc) AKT DECIPHERA (Deciphera Pharmaceuticals, LLC)
  • VQD002 (VioQuest Pharmaceuticals Inc)
  • ETERNA Erk/PI3K Inhibitors
  • GDC0941 Genentech Inc/Piramed Limited/Roche Holdings Ltd
  • enzastaurin HCL LY317615; Enzastaurin; Eli Lilly
  • VMD-8000 (VM Discovery, Inc.)
  • AMG-655 (Aeterna Zentaris, Keryx Biopharma)
  • APOMAB fully humanized mAb
  • Protein Kinase Kinase 1 ARRY704 (Array BioPharma Inc) (MAP2K1); Mitogen- ARRY886 (Array BioPharma Inc) Activated Protein Kinase AS703026 (Merck Serono S.A)
  • Inhibitors CHR-2797 (AminopeptidaseMl inhibitor
  • apricoxib (TP2001; COX-2 Inhibitor, Daiichi).
  • One or more anti-cancer agents may be administered either simultaneously or before or after administration of an antibody or antigen binding portion thereof disclosed herein.
  • Monoclonal antibodies of the present disclosure can be produced using a variety of known techniques, such as the standard somatic cell hybridization technique described by Kohler and Milstein (1975) Nature 256: 495, viral or oncogenic
  • the antibodies are fully human monoclonal antibodies.
  • a hybridoma method is used for producing an antibody that binds ErbB3.
  • a mouse or other appropriate host animal can be immunized with a suitable antigen in order to elicit lymphocytes that produce or are capable of producing antibodies that will specifically bind to the antigen used for immunization.
  • lymphocytes may be immunized in vitro. Lymphocytes can then be fused with myeloma cells using a suitable fusing agent, such as polyethylene glycol, to form a hybridoma cell (Goding, Monoclonal Antibodies: Principles and
  • Culture medium in which hybridoma cells are growing is assayed for production of monoclonal antibodies directed against the antigen.
  • the clones may be subcloned by limiting dilution procedures and grown by standard methods (Goding, 1986 supra). Suitable culture media for this purpose include, for example, D-MEM or RPMI-1640 medium.
  • the hybridoma cells may be grown in vivo as ascites tumors in an animal.
  • the monoclonal antibodies secreted by the subclones can be separated from the culture medium, ascites fluid, or serum by conventional immunoglobulin purification procedures such as, for example, protein A-SEPHAROSE, hydroxylapatite
  • antibodies and antibody portions that bind ErbB3 can be isolated from antibody phage libraries generated using the techniques described in, for example, McCafferty et al, Nature, 348:552-554 (1990). Clackson et al, Nature, 352:624-628 (1991), Marks et al, J. Mol. Biol, 222:581-597 (1991) and Hoet et al (2005) Nature Biotechnology 23, 344-348 ; U.S. Patent Nos. 5,223,409; 5,403,484; and 5,571,698 to Ladner et al ; U.S. Patent Nos. 5,427,908 and 5,580,717 to Dower et al; U.S. Patent Nos.
  • the antibody or antigen binding portion thereof that binds ErbB3 is produced using the phage display technique described by Hoet et al, supra. This technique involves the generation of a human Fab library having a unique combination of immunoglobulin sequences isolated from human donors and having synthetic diversity in the heavy-chain CDRs is generated. The library is then screened for Fabs that bind to ErbB3.
  • human monoclonal antibodies directed against ErbB3 can be generated using transgenic or transchromosomic mice carrying parts of the human immune system rather than the mouse system (see e.g., Lonberg, et al. (1994) Nature 368(6474): 856-859; Lonberg, N. et al. (1994), supra; reviewed in Lonberg, N. (1994) Handbook of Experimental Pharmacology 113:49-101; Lonberg, N. and Huszar, D. (1995) Intern. Rev. Immunol. 13: 65-93, and Harding, F. and Lonberg, N. (1995) Ann. N. Y. Acad. Sci. 764:536-546. See further, U.S. Patent Nos. 5,545,806; 5,569,825;
  • human antibodies disclosed herein can be raised using a mouse that carries human immunoglobulin sequences on transgenes and
  • transchomosomes such as a mouse that carries a human heavy chain transgene and a human light chain transchromosome (see e.g., PCT Publication WO 02/43478 to Ishida et al).
  • immunoglobulin genes are available in the art and can be used to raise anti-ErbB3 antibodies or fragments therof of the invention.
  • an alternative transgenic system referred to as the XENOMOUSE (Abgenix, Inc.) can be used; such mice are described in, for example, U.S. Pat. Nos. 5,939,598; 6,075,181; 6,114,598; 6, 150,584 and 6,162,963 to Kucherlapati et al
  • transchromosomic animal systems expressing human immunoglobulin genes are available in the art and can be used to raise anti-ErbB3 antibodies or fragments therof of the invention.
  • mice carrying both a human heavy chain transchromosome and a human light chain transchromosome can be used; as described in Tomizuka et al (2000) Proc. Natl. Acad. Sci. USA 97:722-727.
  • cows carrying human heavy and light chain transchromosomes have been described in the art (Kuroiwa et al. (2002) Nature Biotechnology 20:889-894) and can be used to raise anti-ErbB3 antibodies or fragments therof of the invention.
  • antibodies disclosed herein can be prepared using a transgenic plant and/or cultured plant cells (such as, for example, tobacco, maize and duckweed) that produce such antibodies.
  • transgenic tobacco leaves expressing antibodies or antigen binding portions thereof can be used to produce such antibodies by, for example, using an inducible promoter (see, e.g., Cramer et al, Curr. Top. Microbol. Immunol. 240:95 118 (1999)).
  • transgenic maize can be used to express such antibodies and antigen binding portions thereof (see, e.g., Hood et al, Adv. Exp. Med. Biol. 464: 127 147 (1999)).
  • Antibodies can also be produced in large amounts from transgenic plant seeds including antibody portions, such as single chain antibodies (e.g., scFvs), for example, using tobacco seeds and potato tubers (see, e.g., Conrad et al, Plant Mol. Biol. 38: 101 109 (1998)).
  • Methods of producing antibodies or antigen binding portions in plants can also be found in, e.g., Fischer et al, Biotechnol. Appl. Biochem. 30:99 108 (1999), Ma et al, Trends Biotechnol. 13:522 7 (1995); Ma et al, Plant Physiol. 109:341 6 (1995); Whitelam et al, Biochem. Soc. Trans. 22:940 944 (1994) and U.S. Pat. Nos. 6,040,498 and 6,815,184.
  • the binding specificity of antibodies or portions thereof that bind ErbB3 prepared using any technique including those disclosed here, can be determined by immunoprecipitation or by an in vitro binding assay, such as radioimmunoassay (RIA) or enzyme-linked immunoabsorbent assay (ELISA).
  • RIA radioimmunoassay
  • ELISA enzyme-linked immunoabsorbent assay
  • the binding affinity of a antibody or portion thereof also can be determined by the Scatchard analysis of Munson et al, Anal. Biochem., 107:220 (1980).
  • an ErbB3 antibody or portion thereof produced using any of the methods discussed above may be further altered or optimized to achieve a desired binding specificity and/or affinity using art recognized techniques, such as those described herein.
  • partial antibody sequences derived from an ErbB3 antibody may be used to produce structurally and functionally related antibodies.
  • antibodies interact with target antigens predominantly through amino acid residues that are located in the six heavy and light chain complementarity determining regions (CDRs). For this reason, the amino acid sequences within CDRs are more diverse between individual antibodies than sequences outside of CDRs. Because CDR sequences are responsible for most antibody-antigen interactions, it is possible to express recombinant antibodies that mimic the properties of specific naturally occurring antibodies by constructing expression vectors that include CDR sequences from the specific naturally occurring antibody grafted onto framework sequences from a different antibody with different properties (see, e.g., Riechmann, L.
  • Such framework sequences can be obtained from public DNA databases that include germline antibody gene sequences.
  • an anti-ErbB3 antibody or fragment therof of the invention can be used to create structurally related anti- ErbB3 antibodies that retain at least one functional property of other antibodies or fragments therof of the invention, e.g., inhibiting EGF-like ligand mediated
  • ErbB3 phosphorylation of ErbB3; inhibiting one or more of heregulin, epiregulin, epigen or biregulin-mediated signaling through ErbB3; inhibiting proliferation or cells expressing ErbB3; and/or decreasing levels of ErbB3 on cell surfaces.
  • one or more CDR regions selected from SEQ ID NO: 1 selected from SEQ ID NO: 1
  • antibodies are generated that include the heavy and/or light chain CDR3s of the particular antibodies described herein ⁇ e.g., SEQ ED NOs:9, 15, 21, 41 , 47 and/or SEQ ED NOs: 12, 18, 24, 44, 50).
  • the antibodies can further include the heavy and/or light chain CDR1 and/or CDR2s of the antibodies specifically disclosed herein ⁇ e.g., SEQ ED NOs:7-8 and/or SEQ ED NOs: 10-l 1 ; SEQ ED NOs: 13-14 and/or SEQ ED NOs: 16-17; SEQ ED NOs:20-21 and/or SEQ ED NOs:22-23; SEQ ED NOs:39-40 and/or SEQ ED NOs:42-43; or SEQ ED NOs:45-46 and/or SEQ ED NOs:48-49).
  • the CDR1, 2, and/or 3 regions of the engineered antibodies described above can comprise the exact amino acid sequence(s) as those disclosed herein (e.g., CDRs of Ab #6, Ab #3, Ab #14, Ab #17, or Ab #19, set forth in SEQ ED NOs:7-12, 13-18, 19-24, 39- 44, and 45-50, respectively).
  • the engineered antibody may be composed of one or more CDRs that are, for example, 90%, 95%, 98%, 99% or 99.5% identical to one or more CDRs of Ab #6, Ab #3 or Ab #14.
  • one or more residues of a CDR may be altered to modify binding to achieve a more favored on-rate of binding.
  • an antibody having ultra high binding affinity of, for example, 10 10 M "1 or more can be achieved.
  • Affinity maturation techniques well known in the art and those described herein, can be used to alter the CDR region(s) followed by screening of the resultant binding molecules for the desired change in binding. Accordingly, as CDR(s) are altered, changes in binding affinity as well as immunogenicity can be monitored and scored such that an antibody optimized for the best combined binding and low immunogenicity are achieved.
  • mutagenesis ⁇ e.g., alanine scanning mutagenesis
  • Ab #6 the heavy and light chain CDRs of Ab #6 to identify amino acid residues involved in binding to ErbB3 to thereby define the paratope of the antibody.
  • an anti- ErbB3 antibody or fragment therof of the invention can comprise a heavy chain paratope comprising CDR1, CDR2 and CDR3 sequences as shown in SEQ ED NOs: 63, 64 and 65, respectively, and a light chain paratope comprising CDRl, CDR2 and CDR3 sequences as shown in SEQ ID NOs: 69, 70 and 71 , respectively, with the proviso that the antibody is not: (i) Ab #6 as disclosed herein; (ii) an antibody comprising VH and VL sequences as shown in SEQ ID NOs: 1 and 2, respectively; or (iii) an antibody comprising V H CDRl, CDR2 and CDR3 sequences as shown in SEQ ID NOs: 7, 8 and
  • an anti-ErbB3 antibody or fragment therof of the invention can comprise a heavy chain paratope comprising CDRl , CDR2 and CDR3 sequences as shown in SEQ ID NOs: 76, 64 and 65, respectively, and a light chain paratope comprising CDRl, CDR2 and CDR3 sequences as shown in SEQ ID NOs: 78, 70 and 80, respectively, with the proviso that the antibody is not: (i) Ab #6 as disclosed herein; (ii) an antibody comprising VH and VL sequences as shown in SEQ ID NOs: 1 and 2, respectively; or (iii) an antibody comprising VH CDRl, CDR2 and CDR3 sequences as shown in SEQ ID NOs: 7, 8 and 9, respectively, and V L CDRl, CDR2 and CDR3 sequences as shown in SEQ ID NOs: 10, 11 and 12, respectively.
  • the invention provides an anti-ErbB3 antibody comprising a heavy chain paratope comprising CDRl, CDR2 and CDR3 sequences as shown in SEQ ID NOs: 63 or 76 (CDRl), 64 (CDR2) and 65 (CDR3), respectively, with the proviso that the antibody is not: (i) Ab #6 as disclosed herein; (ii) an antibody comprising V H and V L sequences as shown in SEQ ED NOs: 1 and 2, respectively; or (iii) an antibody comprising VH CDRl, CDR2 and CDR3 sequences as shown in SEQ ID NOs: 7, 8 and 9, respectively, and VL CDRl, CDR2 and CDR3 sequences as shown in SEQ ID NOs: 10, 11 and 12, respectively.
  • the invention provides an anti-ErbB3 antibody comprising a light chain paratope comprising CDRl, CDR2 and CDR3 sequences as shown in SEQ ID NOs: 69 or 78 (CDRl), 70 (CDR2) and 71 or 80 (CDR3), respectively, with the proviso that the antibody is not: (i) Ab #6 as disclosed herein; (ii) an antibody comprising VH and VL sequences as shown in SEQ ID NOs: 1 and 2, respectively; or (iii) an antibody comprising VH CDRl, CDR2 and CDR3 sequences as shown in SEQ ID NOs: 7, 8 and 9, respectively, and V L CDRl, CDR2 and CDR3 sequences as shown in SEQ ID NOs: 10, 11 and 12, respectively.
  • an anti-ErbB3 antibody or fragment therof of the invention can comprise a heavy chain variable region comprising CDRl, CDR2 and CDR3 sequences as shown in SEQ ID NOs: 60, 61 and 62, respectively, and a light chain variable region comprising CDRl, CDR2 and CDR3 sequences as shown in SEQ ID NOs: 66, 67 and 68, respectively, with the proviso that the antibody is not: (i) Ab #6 as disclosed herein; (ii) an antibody comprising V H and VL sequences as shown in SEQ ID NOs: 1 and 2, respectively; or (iii) an antibody comprising V H CDRl, CDR2 and CDR3 sequences as shown in SEQ ID NOs: 7, 8 and 9, respectively, and V L CDRl, CDR2
  • an anti-ErbB3 antibody or fragment therof of the invention can comprise a heavy chain variable region comprising CDRl, CDR2 and CDR3 sequences as shown in SEQ ID NOs: 75, 61 and 62, respectively, and a light chain variable region comprising CDRl, CDR2 and CDR3 sequences as shown in SEQ ID NOs: 77, 67 and 79, respectively, with the proviso that the antibody is not: (i) Ab #6 as disclosed herein; (ii) an antibody comprising V H and V L sequences as shown in SEQ ID NOs: 1 and 2, respectively; or (iii) an antibody comprising VH CDRl, CDR2 and CDR3 sequences as shown in SEQ ID NOs: 7, 8 and 9, respectively, and V L CDRl, CDR2 and CDR3 sequences as shown in SEQ ID NOs: 10, 11 and 12, respectively.
  • the invention provides an anti-ErbB3 antibody comprising a heavy chain variable region comprising CDRl, CDR2 and CDR3 sequences as shown in SEQ ID NOs: 60 or 75 (CDRl), 61 (CDR2) and 62 (CDR3), respectively, with the proviso that the antibody is not: (i) Ab #6 as disclosed herein; (ii) an antibody comprising VH and VL sequences as shown in SEQ ID NOs: 1 and 2, respectively; or (iii) an antibody comprising VH CDRl, CDR2 and CDR3 sequences as shown in SEQ ID NOs: 7, 8 and 9, respectively, and V L CDRl, CDR2 and CDR3 sequences as shown in SEQ ID NOs: 10, 11 and 12, respectively.
  • the invention provides an anti-ErbB3 antibody comprising a light chain variable region comprising CDRl, CDR2 and CDR3 sequences as shown in SEQ ID NOs: 66 or 77 (CDRl), 67 (CDR2) and 68 or 79 (CDR3), respectively, with the proviso that the antibody is not: (i) Ab #6 as disclosed herein; (ii) an antibody comprising VH and V L sequences as shown in SEQ ID NOs: 1 and 2, respectively; or (iii) an antibody comprising VH CDRl, CDR2 and CDR3 sequences as shown in SEQ ID NOs: 7, 8 and 9, respectively, and V L CDRl, CDR2 and CDR3 sequences as shown in SEQ ID NOs: 10, 11 and 12, respectively.
  • modifications can also be made within one or more of the framework regions, FRl, FR2, FR3 and FR4, of the heavy and/or the light chain variable regions of an antibody, so long as these modifications do not eliminate the binding affinity of the antibody.
  • the antibody is further modified with respect to effector function, so as to enhance the effectiveness of the antibody in treating cancer, for example.
  • cysteine residue(s) may be introduced in the Fc region, thereby allowing interchain disulfide bond formation in this region.
  • the homodimeric antibody thus generated may have improved internalization capability and/or increased complement-mediated cell killing and antibody-dependent cellular cytotoxicity (ADCC). See Caron et al, J. Exp Med. 176: 1191-1195 (1992) and Shopes, B. J. Immunol.
  • Homodimeric antibodies with enhanced anti-tumor activity may also be prepared using heterobifunctional cross-linkers as described in Wolff et al.
  • an antibody can be engineered which has dual Fc regions and may thereby have enhanced complement lysis and ADCC capabilities. See Stevenson et al. Anti-Cancer Drug Design 3:219-230 (1989).
  • Mutagenesis of one or more residues in the CDRs, framework regions, Fc regions, or other antibody regions as disclosed herein can be accomplished using standard recombinant DNA techniques, including but not limited to site-directed mutagenesis and PCR-mediated mutagenesis. Screening of the effects of mutation on antigen binding ⁇ e.g., binding to ErbB3) and other functional characteristics also can be accomplished using standard methods.
  • the antibody e.g., an scFv version
  • the mutated CDRs can be expressed on the surface of cells, such as mammalian cells, yeast cells or bacterial cells ⁇ e.g., using a phage display system
  • binding of the antigen to the cells can be determined using standard methods, for example flow cytometry in which antigen bound to the cells is detected, e.g., using a labeled secondary antibody.
  • the antibody can be expressed in soluble form and the binding of the antibody to the antigen can be assessed using a standard binding assay such as ELISA or BIACORE analysis.
  • ELISA ELISA
  • BIACORE analysis Detailed descriptions of the foregoing and related techniques for such purposes may be found in numerous well known textbooks and laboratory manuals, for example: Handbook of Therapeutic Antibodies Vols. 1-3, Stefan Dubel, ed., Wiley-VCH 2007; Making and Using
  • Antibodies A Practical Handbook, Gary C. Howard, CRC 2006; Antibody Engineering: Methods and Protocols, Benny K. C. Lo, Humana Press 2003; Therapeutic Antibodies: Methods and Protocols, Antony S. Dimitrov, ed., Humana Press 2009; Antibody Phage Display: Methods and Protocols, 2nd ed. Robert Aitken, ed., Humana Press 2009; Flow Cytometry Protocols, 2nd ed. Maria S. Hawley and Robert G. Hawley, eds., Humana Press 2004; Flow Cytometry: Principles and Applications, Marion G. Macey, ed., Humana Press 2007. Also see “Selecting and Screening Recombinant Antibody Libraries," H.R. Hoogenboom, Nature Biotechnol. 23: 1105-1116; 2005.
  • bispecific antibodies and immunoconjugates are also encompassed by the present invention.
  • Bispecific antibodies of the present disclosure include at least one binding specificity for ErbB3 and at least one binding specificity for another antigen, such as the product of an oncogene.
  • Bispecific antibodies can be prepared as full length antibodies or antibody fragments (e.g. F(ab')2 bispecific antibodies).
  • bispecific antibodies are well known in the art (see, e.g., WO 05117973 and WO 06091209).
  • production of full length bispecific antibodies can be based on the coexpression of two immunoglobulin heavy chain-light chain pairs, where the two chains have different specificities (see, e.g., Millstein et al, Nature, 305:537-539 (1983)).
  • Further details of generating bispecific antibodies can be found, for example, in Suresh et al., Methods in Enzymology, 121:210 (1986) and in Brennan et al., Science, 229: 81 (1985), which describes a chemical linkage process for making bispecific antibodies.
  • bispecific antibodies have been produced using leucine zippers (see, e.g., Kostelny et al, J. Immunol, 148(5): 1547-1553 (1992)).
  • scFv single-chain Fv dimers
  • the bispecific antibody comprises a first antibody or binding portion thereof which binds to ErbB3 and a second antibody or binding portion thereof which binds to ErbB2, ERbB3, ErbB4, EGFR, IGF1-R, C-MET, Lewis Y, MUC-1, EpCAM, CA125, prostate specific membrane antigen, PDGFR-a, PDGFR- ⁇ , C-KIT, or any of the FGF receptors.
  • Immunoconjugates of the present disclosure can be formed by conjugating the antibodies or antigen binding portions thereof described herein to another therapeutic agent.
  • Suitable agents include, for example, a cytotoxic agent (e.g., a chemotherapeutic agent), a toxin (e.g. an enzymatically active toxin of bacterial, fungal, plant or animal origin, or fragments thereof), and/or a radioactive isotope (i.e., a radioconjugate).
  • Enzymatically active toxins and fragments thereof which can be used include diphtheria A chain, nonbinding active fragments of diphtheria toxin, exotoxin A chain (from Pseudomonas aeruginosa), ricin A chain, abrin A chain, modeccin A chain, alpha-sarcin, Aleurites fordii proteins, dianthin proteins, Phytolaca americana proteins (PAPI, ⁇ , and PAP-S), Momordica charantia inhibitor, curcin, crotin, Sapaonaria officinalis inhibitor, gelonin, mitogellin, restrictocin, phenomycin, enomycin and the tricothecenes.
  • a variety of radionuclides are available for the production of
  • radioconjugated anti-ErbB3 antibodies examples include 212 Bi, 131 1, 131 In, 90 Y and 186 Re.
  • Immunoconjugates of the invention can be made using a variety of bifunctional protein coupling agents such as N-succinimidyl-3-(2-pyridyldithiol) propionate (SPDP), 2- iminothiolane ( ⁇ ), bifunctional derivatives of imidoesters (such as dimethyl adipimidate HCL), active esters (such as disuccinimidyl suberate), aldehydes (such as
  • a ricin immunotoxin can be prepared as described in Vitetta et ai, Science 238: 1098 (1987).
  • Carbon- 14-labeled 1- isothiocyanatobenzyl-3-methyldiethylene triaminepentaacetic acid is an exemplary chelating agent for conjugation of radionucleotide to the antibody (see, e.g., WO94/11026).
  • monoclonal and monospecific polyclonal antibodies having various of the desireable properties of Ab #6 can be readily obtained using conventional immunization methods using peptides (e.g., synthetic peptides), or conjugates thereof (e.g., KLH conjugates), as immunogens.
  • Peptide for use as immunogens in this embodiment are those comprising any 10 or more contiguous amino acid residues from residuesl-183 of SEQ ID NO: 73.
  • the 10 or more contiguous amino acid residues are from Domain I of the ectodomain of ErbB3, preferably the residues at least partially fall within or span residues 92-104 of SEQ ID NO: 73.
  • antibodies or antigen binding portions that bind ErbB3 can be screened for various properties, such as those described herein, using a variety of assays that are well known in the art.
  • the antibodies or antigen binding portions thereof are screened for the ability to inhibit EGF-like ligand mediated phosphorylation of ErbB3.
  • This can be done by treating cells expressing ErbB3 with an EGF-like ligand in the presence and absence of the antibody or antigen binding portion thereof. The cells can then be lysed and the crude lysates can be centrifuged to remove insoluble material. ErbB3 phosphorylation can be measured, for example, by Western blotting followed by probing with an anti-phosphotyrosine antibody as described in Kim et ah, supra and the Examples below.
  • the antibodies and antigen binding portions are further screened for one or more of the following properties: (1) inhibition of ErbB3-ligand (e.g., heregulin, epiregulin, epigen or biregulin) mediated signaling through ErbB3; (2) inhibition of proliferation of cells expressing ErbB3; (3) the ability to decrease levels of ErbB3 on cell surface (e.g., by inducing internalization of ErbB3), (4) inhibition of VEGF secretion of cells expressing ErbB3; (5) inhibition of the migration of cells expressing ErbB3; (6) inhibition of spheroid growth of cells expressing ErbB3; and/or (7) binding to an epitope located on Domain I of the ectodomain of ErbB3, each of which can be readily measured using art recognized techniques and those discussed herein.
  • ErbB3-ligand e.g., heregulin, epiregulin, epigen or biregulin
  • Inhibition of one or more of heregulin, epiregulin, epigen or biregulin-mediated signaling through ErbB3 can be readily measured using routine assays, such as, described in Horst et al. supra.
  • the ability of an antibody or antigen binding portion thereof to inhibit heregulin, epiregulin, epigen or biregulin-mediated signaling through ErbB3 can be measured by kinase assays for known substrates of ErbB3 such as, for example, SHC and PI3K, as described in, for example, Horst et al. supra, Sudo et al., (2000) Methods Enzymol, 322:388-92; and Morgan et al. (1990) Eur. J.
  • Biochem., 191:761-767 following stimulation by one or more of heregulin, epiregulin; epigen or biregulin. Accordingly, cells expressing ErbB3 can be stimulated with one or more of heregulin, epiregulin, epigen or biregulin, and incubated with a candidate antibody or antigen-binding portion thereof.
  • Cell lysates subsequently prepared from such cells can be immunoprecipitated with an antibody for a substrate of ErbB3 (or a protein in a cellular pathway involving ErbB3) such as, for example, an anti-JNK-1 antibody, and assayed for kinase activity (e.g., JNK kinase activity or PI3- kinase activity) using art recognized techniques.
  • an antibody for a substrate of ErbB3 or a protein in a cellular pathway involving ErbB3
  • kinase activity e.g., JNK kinase activity or PI3- kinase activity
  • the antibody or antigen binding portion thereof inhibits ErbB3-ligand (e.g., heregulin, epiregulin, epigen or biregulin) mediated signaling by decreasing the binding of one or more of heregulin, epiregulin, epigen or biregulin to ERbB3.
  • ErbB3-ligand e.g., heregulin, epiregulin, epigen or biregulin
  • cells which express ErbB3 e.g. MALME-3M cells, as described in the Examples infra
  • a labeled ErbB3-ligand e.g., radiolabeled heregulin, epiregulin, epigen or biregulin
  • the antibody or antigen binding portion thereof inhibits heregulin, epiregulin, epigen or biregulin binding to ErbB3, then a statistically significantly decrease in the amount of label recovered (e.g., radiolabeled heregulin, epiregulin, epigen or biregulin), relative to the amount in the absence of the antibody or antigen binding portion thereof, will be observed.
  • label recovered e.g., radiolabeled heregulin, epiregulin, epigen or biregulin
  • the antibody or antigen binding portion thereof may inhibit the binding of the
  • ErbB3-ligand e.g., heregulin, epiregulin, epigen or biregulin
  • the antibody or antigen binding portion thereof may inhibit binding of the ErbB3 ligand (e.g., one or more of heregulin, epiregulin, epigen or biregulin) to ErbB3 by binding to the same site or an overlapping site on ErbB3 as the ErbB3 ligand.
  • the antibody or antigen binding portion thereof may inhibit binding of an ErbB3 ligand by altering or distorting the conformation of ErbB3, such that it is unable to bind to the ErbB3 ligand.
  • Antibodies and antigen binding portions thereof that decrease levels of ErbB3 on cell surfaces can be identified by their ability to downregulate ErbB3 on tumor cells.
  • the antibodies or antigen binding portions thereof decrease ErbB3 cell surface expression by inducing internalization (or increasing endocytosis) of Erbb3.
  • ErbB3 can be biotinylated and the number of ErbB3 molecules on the cell surface can be readily determined, for example, by measuring the amount of biotin on a monolayer of cells in culture in the presence or absence of an antibody or antigen binding portion thereof, for example, as described in, e.g., Waterman et al., J. Biol. Chem.
  • Antibodies or antigen binding portions thereof of the present disclosure can also be tested for their ability to inhibit proliferation of cells expressing ErbB3, for example, tumor cells, using art recognized techniques, such as the CellTiter-Glo® Assay described in the Examples below (also see, e.g., Macallan et al., Proc. Natl. Acad. Sci. (1998) 20;95(2):708-13; Perez et al. (1995) Cancer Research 55, 392-398).
  • the antibodies or antigen binding portions thereof are screened for the ability to inhibit VEGF secretion of cells expressing ErbB3. This can be done by using well-known assays, such as the VEGF ELISA kit available from R&D Systems, Minneapolis, MN, # DY293B. Similarly, the antibodies or portions can be screened for the ability to inhibit the migration of cells expressing ErbB3 (e.g., MCF-7 cells) using a trans-well assay (Millipore Corp., Billerica, MA, # ECM552) as described herein.
  • the antibodies or antigen binding portions thereof are screened for the ability to inhibit spheroid growth of cells expressing ErbB3. This can be done by using an assay which approximates conditions of a developing tumor growth (see, e.g., Herman et al. (2007) Journal of Biomolecular Screening Electronic publication) as described herein.
  • Antibodies or antigen binding portions thereof that bind to the same or overlapping epitopes as one or more antibodies specifically disclosed herein can also be identified using standard techniques known in the art and described herein. For example, in order to screen for antibodies which bind to the same or an overlapping epitope on ErbB3 bound by an antibody of interest, a cross-blocking assay, such as that described in Antibodies, A Laboratory Manual, Cold Spring Harbor Laboratory, Ed Harlow and David Lane (1988), can be performed.
  • the present invention provides a composition, e.g., a pharmaceutical composition, containing one or a combination of antibodies, or antigen- binding portion(s) thereof disclosed herein, formulated together with a pharmaceutically acceptable carrier.
  • the compositions include a combination of multiple (e.g., two or more) isolated antibodies, which bind different epitopes on ErbB3.
  • pharmaceutically acceptable carrier includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like that are physiologically compatible.
  • the carrier is suitable for intravenous, intramuscular, subcutaneous, parenteral, spinal or epidermal administration (e.g., by injection or infusion).
  • the active agent i.e., antibody, antibody fragment, bispecific and multispecific molecule, may be coated in a material to protect the agent from the action of acids and other natural conditions that may inactivate it.
  • a “pharmaceutically acceptable salt” refers to a salt that retains the desired biological activity of the parent compound and does not impart any undesired
  • Acid addition salts include those derived from nontoxic inorganic acids, such as hydrochloric, nitric, phosphoric, sulfuric, hydrobromic, hydroiodic, phosphorous and the like, as well as from nontoxic organic acids such as aliphatic mono- and dicarboxylic acids, phenyl- substituted alkanoic acids, hydroxy alkanoic acids, aromatic acids, aliphatic and aromatic sulfonic acids and the like.
  • Base addition salts include those derived from alkaline earth metals, such as sodium, potassium, magnesium, calcium and the like, as well as from nontoxic organic amines, such as ⁇ , ⁇ '-dibenzylethylenediamine, N- methylglucamine, chloroprocaine, choline, diethanolamine, ethylenediamine, procaine and the like.
  • Pharmaceutical compositions of the invention can comprise other agents.
  • the composition can include at least one or more additional therapeutic agents, such as the anti-cancer agents described infra.
  • the pharmaceutical compositions can also be administered in conjunction with radiation therapy and/or surgery. Alternately a composition of the invention can be separately co-administered with at least one or more additional therapeutic agents, such as the anti-cancer agents described infra.
  • composition of the present disclosure can be administered by a variety of methods known in the art. As will be appreciated by the skilled artisan, the route and/or mode of administration will vary depending upon the desired results.
  • the active agents can be prepared with carriers that will protect the agents against rapid release, such as a controlled release formulation, including implants, transdermal patches, and
  • Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid. Many methods for the preparation of such formulations are patented or generally known to those skilled in the art. See, e.g., Sustained and Controlled Release Drug Delivery Systems, J.R. Robinson, ed., Marcel Dekker, Inc., New York, 1978.
  • an antibody or fragment thereof of the invention may be necessary to coat it with, or co-administer it with, a material to prevent its inactivation.
  • a material to prevent its inactivation.
  • it may be administered to a subject in an appropriate carrier, for example, liposomes, or a diluent.
  • Pharmaceutically acceptable diluents include saline and aqueous buffer solutions.
  • Liposomes include water-in-oil-in- water CGF emulsions as well as conventional liposomes (Strejan et al. (1984) J.
  • Pharmaceutically acceptable carriers include sterile aqueous solutions or dispersions and sterile powders or lyophylysates for the extemporaneous preparation of sterile injectable solutions or dispersion.
  • sterile aqueous solutions or dispersions and sterile powders or lyophylysates for the extemporaneous preparation of sterile injectable solutions or dispersion.
  • the use of such media and agents for pharmaceutically active antibodies is known in the art. Except insofar as any conventional media or agent is incompatible with the active agent, use thereof in the pharmaceutical compositions of the invention is contemplated. Supplementary active compounds can also be incorporated into the compositions.
  • compositions typically must be sterile and stable under the conditions of manufacture and storage.
  • the composition can be formulated as a solution, microemulsion, liposome, or other ordered structure suitable to high drug concentration.
  • the carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), and suitable mixtures thereof.
  • the proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants.
  • isotonic agents for example, sugars, polyalcohols such as mannitol, sorbitol, or sodium chloride in the composition.
  • Prolonged absorption of the injectable compositions can be brought about by including in the composition an agent that delays absorption, for example, monostearate salts and gelatin.
  • Sterile injectable solutions can be prepared by incorporating the active agent in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by sterilization microfiltration.
  • dispersions are prepared by incorporating the active agent into a sterile vehicle that contains a basic dispersion medium and the required other ingredients from those enumerated above.
  • the preferred methods of preparation are vacuum drying and freeze-drying (lyophilization) that yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.
  • Dosage regimens are adjusted to provide the optimum desired response ⁇ e.g., a therapeutic response). For example, a single bolus may be administered, several divided doses may be administered over time or the dose may be proportionally reduced or increased as indicated by the exigencies of the therapeutic situation.
  • the human antibodies disclosed herein may be administered once or twice weekly by subcutaneous injection or once or twice monthly by subcutaneous injection.
  • Non-limiting examples of suitable dosage ranges and regimens include 2-50 mg/kg (body weight of the subject) administered once a week, or twice a week or once every three days, or once every two weeks, and 1-100 mg/kg administered once a week, or twice a week or once every three days, or once every two weeks.
  • an antibody is administered at a dosage of 3.2 mg/kg, 6 mg/kg, 10 mg/kg, 15 mg/kg, 20 mg kg, 25 mg/kg, 30 mg/kg, 35 mg/kg or 40 mg/kg at a timing of once a week, or twice a week or once every three days, or once every two weeks.
  • Additional dosage ranges include: 1-1000 mg/kg, 1-500 mg/kg, 1-400 mg/kg, 1-300 mg/kg and 1- 200 mg/kg.
  • Suitable dosage schedules include once every three days, once every five days, once every seven days (i.e., once a week), once every 10 days, once every 14 days (i.e., once every two weeks), once every 21 days (i.e., once every three weeks), once every 28 days (i.e., once every four weeks) and once a month.
  • an antibody disclosed herein in combination with an additional therapeutic agent can lead to an additive effect for antitumor activity.
  • suboptimal dosages of the antibody or the second therapeutic agent, or both can be used to achieve a desired therapeutic outcome due to the additive effects of the agents.
  • an antibody or fragment therof of the invention may be administered at a dosage that is 90 %, or 80 %, or 70 % or 60 % or 50 % of the dosage used when the antibody is administered alone.
  • Dosage unit form refers to physically discrete units suited as unitary dosages for the subjects to be treated; each unit contains a predetermined quantity of active agent calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier.
  • the specification for the dosage unit forms of the invention are dictated by and directly dependent on (a) the unique characteristics of the active agent and the particular therapeutic effect to be achieved, and (b) the limitations inherent in the art of compounding such an active agent for the treatment of sensitivity in individuals.
  • antioxidants examples include: (1) water soluble antioxidants, such as ascorbic acid, cysteine hydrochloride, sodium bisulfate, sodium metabisulfite, sodium sulfite and the like; (2) oil-soluble antioxidants, such as ascorbyl palmitate, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT), lecithin, propyl gallate, alpha-tocopherol, and the like; and (3) metal chelating agents, such as citric acid, ethylenediamine tetraacetic acid (EDTA), sorbitol, tartaric acid, phosphoric acid, and the like.
  • water soluble antioxidants such as ascorbic acid, cysteine hydrochloride, sodium bisulfate, sodium metabisulfite, sodium sulfite and the like
  • oil-soluble antioxidants such as ascorbyl palmitate, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT), le
  • formulations of the present disclosure include those suitable for oral, nasal, topical (including buccal and sublingual), rectal, vaginal and/or parenteral administration.
  • the formulations may conveniently be presented in unit dosage form and may be prepared by any methods known in the art of pharmacy.
  • the amount of active ingredient which can be combined with a carrier material to produce a single dosage form will vary depending upon the subject being treated, and the particular mode of administration.
  • the amount of active ingredient which can be combined with a carrier material to produce a single dosage form will generally be that amount of the composition which produces a therapeutic effect. Generally, out of one hundred per cent, this amount will range from about 0.001 per cent to about ninety percent of active ingredient, preferably from about 0.005 per cent to about 70 per cent, most preferably from about 0.01 per cent to about 30 per cent.
  • Formulations of the present disclosure which are suitable for vaginal administration also include pessaries, tampons, creams, gels, pastes, foams or spray formulations containing such carriers as are known in the art to be appropriate.
  • Dosage forms for the topical or transdermal administration of compositions of this invention include powders, sprays, ointments, pastes, creams, lotions, gels, solutions, patches and inhalants.
  • the active agent may be mixed under sterile conditions with a pharmaceutically acceptable carrier, and with any preservatives, buffers, or propellants which may be required.
  • parenteral administration and “administered parenterally” as used herein means modes of administration other than enteral and topical administration, usually by injection, and includes, without limitation, intravenous, intramuscular, intraarterial, intrathecal, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticular, subcapsular, subarachnoid, intraspinal, epidural and intrasternal injection and infusion.
  • aqueous and nonaqueous carriers examples include water, ethanol, polyols (such as glycerol, propylene glycol, polyethylene glycol, and the like), and suitable mixtures thereof, vegetable oils, such as olive oil, and injectable organic esters, such as ethyl oleate.
  • polyols such as glycerol, propylene glycol, polyethylene glycol, and the like
  • vegetable oils such as olive oil
  • injectable organic esters such as ethyl oleate.
  • Proper fluidity can be maintained, for example, by the use of coating materials, such as lecithin, by the maintenance of the required particle size in the case of dispersions, and by the use of surfactants.
  • compositions may also contain adjuvants such as preservatives, wetting agents, emulsifying agents and dispersing agents.
  • adjuvants such as preservatives, wetting agents, emulsifying agents and dispersing agents.
  • adjuvants which are well-known in the art include, for example, inorganic adjuvants (such as aluminum salts, e.g., aluminum phosphate and aluminum hydroxide), organic adjuvants (e.g., squalene), oil-based adjuvants, virosomes (e.g., virosomes which contain a membrane-bound hemagglutinin and neuraminidase derived from the influenza virus).
  • inorganic adjuvants such as aluminum salts, e.g., aluminum phosphate and aluminum hydroxide
  • organic adjuvants e.g., squalene
  • oil-based adjuvants e.g., virosomes which contain a membrane-
  • Prevention of presence of microorganisms may be ensured both by sterilization procedures, supra, and by the inclusion of various antibacterial and antifungal agents, for example, paraben, chlorobutanol, phenol sorbic acid, and the like. It may also be desirable to include isotonic agents, such as sugars, sodium chloride, and the like into the compositions. In addition, prolonged absorption of the injectable pharmaceutical form may be brought about by the inclusion of agents which delay absorption such as aluminum monostearate and gelatin.
  • compositions containing, for example, 0.001 to 90% (more preferably, 0.005 to 70%, such as 0.01 to 30%) of active ingredient in combination with a pharmaceutically acceptable carrier.
  • the antibodies of the present disclosure which may be used in a suitable hydrated form, and/or the pharmaceutical compositions of the present disclosure, are formulated into pharmaceutically acceptable dosage forms by conventional methods known to those of skill in the art.
  • Actual dosage levels of the active ingredients in the pharmaceutical compositions of the present disclosure may be varied so as to obtain an amount of the active ingredient which is effective to achieve the desired therapeutic response for a particular patient, composition, and mode of administration, without being toxic to the patient.
  • the selected dosage levels will depend upon a variety of pharmacokinetic factors including the activity of the particular compositions employed, or, for compounds co-administered with antibodies or fragments therof provided herein, the ester, salt or amide thereof, the route of administration, the time of administration, the rate of excretion of the particular agent being employed, the duration of the treatment, other drugs, compounds and/or materials used in combination with the particular compositions employed, the age, sex, weight, condition, general health and prior medical history of the patient being treated, and like factors well known in the medical arts.
  • a physician or veterinarian having ordinary skill in the art can readily determine and prescribe the effective amount of the pharmaceutical composition required.
  • a suitable daily dose of a composition of the invention will be that amount which provides the lowest dose effective to produce a therapeutic effect.
  • Such an effective dose will generally depend upon the factors described above. It is preferred that administration be intravenous, intramuscular, intraperitoneal, or subcutaneous, preferably administered proximal to the site of the target.
  • the effective daily dose of a therapeutic composition may be administered as two, three, four, five, six or more sub-doses administered separately at appropriate intervals throughout the day, optionally, in unit dosage forms. While it is possible for a antibody of the present disclosure to be administered alone, it is preferable to administer the antibody as a pharmaceutical formulation (composition).
  • compositions can be administered with medical devices known in the art.
  • a therapeutic composition of the invention can be administered with a needleless hypodermic injection device, such as the devices disclosed in U.S. Pat. Nos. 5,399,163, 5,383,851, 5,312,335, 5,064,413, 4,941,880, 4,790,824, or 4,596,556.
  • a needleless hypodermic injection device such as the devices disclosed in U.S. Pat. Nos. 5,399,163, 5,383,851, 5,312,335, 5,064,413, 4,941,880, 4,790,824, or 4,596,556.
  • Examples of well-known implants and modules useful in the present invention include: U.S. Pat. No. 4,487,603, which discloses an implantable micro-infusion pump for dispensing medication at a controlled rate; U.S. Pat. No. 4.,486,194, which discloses a therapeutic device for administering medications through the skin; U.S. Pat. No.
  • compositions disclosed herein can be formulated to ensure proper distribution in vivo.
  • the blood-brain barrier excludes many highly hydrophilic compounds.
  • therapeutic compounds in compositions of the invention cross the BBB (if desired)
  • they can be formulated, for example, in liposomes.
  • liposomes For methods of manufacturing liposomes, see, e.g., U.S. Pat. Nos. 4,522,811; 5,374,548; and 5,399,331.
  • the liposomes may comprise one or more moieties which are selectively transported into specific cells or organs, thus enhance targeted drug delivery (see, e.g., V.V. Ranade (1989) J. Clin. Pharmacol. 29:685).
  • Exemplary targeting moieties include folate or biotin (see, e.g., U.S. Pat. No. 5,416,016 to Low et al.); mannosides (Umezawa et al., (1988) Biochem. Biophys. Res. Commun. 153: 1038); antibodies (P.G. Bloeman et al. (1995) FEBS Lett. 357: 140; M. Owais et al. (1995) Antimicrob. Agents Chemother. 39: 180); surfactant protein A receptor (Briscoe et al. (1995) Am. J. Physiol.
  • the present invention also provides methods of using antibodies and antigen- binding portions thereof that bind ErbB3 in a variety of ex vivo and in vivo diagnostic and therapeutic applications.
  • antibodies disclosed herein can be used for treating a disease associated with ErbB3 dependent signaling, including a variety of cancers.
  • the present invention provides a method for treating a disease associated with ErbB3 dependent signaling by administering to a subject an antibody or antigen binding portion thereof of the invention in an amount effective to treat the disease.
  • Suitable diseases include, for example, a variety of cancers including, but not limited to, melanoma, breast cancer, ovarian cancer, renal carcinoma, gastrointestinal cancer, colon cancer, lung cancer (e.g., non-small cell lung cancer), and prostate cancer.
  • a tumor sample obtained from the patient is tested and treatment is provided in accordance with the methods disclosed in International Application No. PCT/US09/054051 , filed August 17, 2009, titled "Methods, Systems And Products For Predicting Response Of Tumor Cells To A Therapeutic Agent And Treating A Patient According To The Predicted Response” which is incorporated herein by reference.
  • the patient is to be treated for a malignant tumor, a sample of the tumor is obtained, a level of phosoho-ErbB3 (pErbB3) in the sample is determined, and at least one antineoplastic therapeutic agent is subsequently
  • the at least one antineoplastic therapeutic agent subsequently administered to the patient comprises an anti-ErbB3 antibody of the present invention, e.g., Ab #6, and if the level of pErbB3 determined in the sample is lower than 50% of the level of pErbB3 measured in the culture of ACHN cells then the at least one antineoplastic therapeutic agent subsequently administered to the patient does not comprise an anti-ErbB3 antibody.
  • an anti-ErbB3 antibody of the present invention e.g., Ab #6
  • the at least one antineoplastic therapeutic agent subsequently administered to the patient does not comprise an anti-ErbB3 antibody.
  • the cancer comprises a KRAS mutation.
  • antibodies disclosed herein(£.g., Ab #6) are capable of inhibiting the growth of tumor cells that comprise a KRAS mutation, either when used as a single agent (monotherapy) or in combination with another therapeutic agent.
  • the cancer comprises a PI3K mutation.
  • antibodies disclosed herein(e.g., Ab #6) are capable of inhibiting the growth of tumor cells that comprise a PI3K mutation, either when used as a single agent (monotherapy) or in combination with another therapeutic agent.
  • the antibody can be administered alone or with another therapeutic agent which acts in conjunction with or synergistically with the antibody to treat the disease associated with ErbB3 mediated signaling.
  • therapeutic agents include, for example, the anticancer agents described infra (e.g., cytotoxins, chemotherapeutic agents, small molecules and radiation).
  • antibodies disclosed herein(e.g., Ab #6) when used in combination with another therapeutic agent can exhibit increased tumor growth inhibition as compared to when used in monotherapy.
  • Preferred therapeutic agents for combination therapy include erlotinib (Tarceva®), paclitaxel (Taxol®) and cisplatin (CDDP).
  • antibodies disclosed herein are administered to patients
  • the present invention provides a method for diagnosing a disease (e.g., a cancer) associated with ErbB3 upregulation in a subject, by contacting antibodies or antigen binding portions disclosed herein(e.g., ex vivo or in vivo) with cells from the subject, and measuring the level of binding to ErbB3 on the cells. Abnormally high levels of binding to ErbB3 indicate that the subject has a disease associated with ErbB3 upregulation.
  • a disease e.g., a cancer
  • antibodies or antigen binding portions disclosed herein(e.g., ex vivo or in vivo) with cells from the subject, and measuring the level of binding to ErbB3 on the cells.
  • Abnormally high levels of binding to ErbB3 indicate that the subject has a disease associated with ErbB3 upregulation.
  • kits comprising antibodies and antigen binding portions thereof of the invention.
  • the kits may include a label indicating the intended use of the contents of the kit and optionally including instructions for use of the kit in treating or diagnosing a disease associated with ErbB3 upregulation and/or ErbB3 dependent signaling, e.g., treating a tumor.
  • the term label includes any writing, marketing materials or recorded material supplied on or with the kit, or which otherwise accompanies the kit.
  • HRG refers to the isoform of heregulin variously known as heregulin 1 beta 1, HRG1-B, HRG- ⁇ , neuregulin 1, NRG1 , neuregulin 1 beta 1, NRGl-bl, HRG ECD, and the like.
  • HRG is commercially available, e.g., R&D Systems # 377-HB-050/CF.
  • a cryopulverizer (Covaris Inc) is used for the pulverization of tumors. Tumors are stored in special bags (pre-weighed before the addition of the tumor) and placed in liquid nitrogen while handling them. For small tumors, 200 uL of Lysis buffer is first added to the bag containing the tumor, frozen in liquid nitrogen and then pulverized to improve the recovery of the tumor from the bag. Pulverized tumors are transferred to 2 mL EPPENDORF tubes and placed in liquid nitrogen until ready for further processing
  • Tumors are lysed in Lysis buffer supplemented with protease and phosphatase inhibitors. Lysis Buffer is added to the tumor aliquots in a final concentration of about 62.5 mg/mL. Tumor samples are homogenized by vortexing for 30 sec and incubating on ice for about 30 min. The lysates are spun for about 10 min in QIAGEN
  • BCA assay (Pierce) is performed following the manufacturer's protocol on all tumor samples. The total protein concentration (in mg/mL) of each tumor sample is later used in the normalization of the ELISA results
  • ELISA reagents for the total and phospho-ErbB3 ELISAs are purchased from R&D Systems as DUOSET kits.
  • 96-well NUNC MAXISORB plates are coated with 50 uL of an antibody and incubated overnight at room temperature. Next morning, plates are washed 3 times with 1000 ⁇ /well in the BIOTE plate washer with Dulbecco's phosphate buffered saline without calcium or magnesium (PBS) with added Tween detergent (PBST) (0.05% Tween-20). Plates are subsequently blocked for about an 1 hr at room temperature with 2 % BSA in PBS.
  • PBS Dulbecco's phosphate buffered saline without calcium or magnesium
  • PBST Tween detergent
  • the plates are washed 3 times with 1000 ⁇ /well in the BIOTEK plate washer with PBST (0.05%Tween-20). 50 ⁇ L ⁇ of cell lysates and standards diluted in 50% Lysis buffer and 1%BSA are used in duplicates for further processing. Samples are incubated for 2hrs at 4°C on a plate shaker and washed as before. About 50 ⁇ of a detection antibody diluted in 2% BSA, PBST is added and incubated for about 1 hr at room temperature. For phosphor-ErbB3, the detection antibody is directly conjugated to horseradish peroxidase (HRP) and incubated for 2 hrs at room temperature. The plate is washed as before.
  • HRP horseradish peroxidase
  • a human Fab-phage library including a unique combination of immunoglobulin sequences obtained from human donors is initially screened for ErbB3 binders.
  • the 73 Fabs without the phage are spotted on a chip surface and the binding kinetics and epitope blocking to a ErbB3-his fusion target protein or a ErbB3-Fc protein (R & D Systems) are measured. The equilibrium binding constant and on/off rates for the Fabs are calculated from the data obtained.
  • Binding of the various Fabs to MALME-3M cells is next examined using about 500 nM of the Fabs and a 1 :750 dilution of a goat anti-human Alexa 647 secondary antibody. As shown in Figures 1 A and IB, data obtained using the methods described above or minor variations thereof indicate that several candidate Fabs exhibited appreciable staining of MALME-3M cells.
  • Example 2 Optimization of anti-ErbB3 Fabs
  • VH and VL sequences of the Fabs are codon-optimized as follows.
  • the VH and VL regions are reformatted using expression constructs for expression as an IgGl or IgG2 isotype.
  • the constructs include a SELEXIS backbone which has a cassette designed for substitution of the appropriate heavy and light chain sequences.
  • the SELEXIS vectors include a CMV promoter and a matching poly-A signal.
  • the dissociation constants of the anti-ErbB3 antibodies are measured using two independent techniques, i.e., a Surface Plasmon Resonance Assay and a cell binding assay using MALME-3M cells.
  • Surface Plasmon Resonance Assay a Surface Plasmon Resonance Assay
  • the Surface Plasmon Resonance Assay (e.g., a FLEXCHIP assay) is performed substantially as described in Wassaf et al. (2006) Analytical Biochem., 351 :241-253, in a BIACORE 3000 instrument or the like using recombinant ErbB3 as the analyte and the subject antibody as the ligand.
  • the cell binding assay for determining the KD values of Ab #6 and Ab #3 is performed as follows.
  • MALME-3M cells are detached with 2 mLs trypsin-EDTA + 2 mLs RMPI +
  • a 150 ⁇ solution of 200 nM anti-ErbB3 antibody (Ab #6 or Ab #3) in BD stain buffer is prepared in an EPPENDORF tube and serially diluted 2-fold into 75 ⁇ BD stain buffer.
  • the concentrations of the diluted antibody ranged from 200 nM to 0.4 nM.
  • 50 ⁇ aliquots of the different protein dilutions are then added directly to the 50 ul cell suspension giving the final concentrations of 100 nM, 50 nM, 25 nM, 12 nM, 6 nM, 3 nM, 1.5 nM, 0.8 nM, 0.4 nM and 0.2 nM of the antibody.
  • MFI values and the corresponding concentrations of the anti-ErbB3-antibodies are plotted on the y-axis and x-axis, respectively.
  • the KD of the molecule is determined using GraphPad PRISM software using the one-site binding model for a non-linear regression curve.
  • Ab #6 and Ab #3 had K D values of about 4 nM and 1.3 nM, respectively, were obtained (using the methods described above or minor variations thereof) in a cell binding assay using MALME-3M cells.
  • a Kinetic Exclusion Assay (KinExA®) is used to determine the association and dissociation rates for Ab #6.
  • the binding specificity of an IgG2 isotype of Ab #6 to ErbB3 is assayed using ELISA as follows. Identification of the epitope bound by Ab #6 is also analyzed.
  • 96-well NUNC M AXIS ORB plates are coated by overnight incubation at room temperature with 50 ⁇ /well of 5 ⁇ of individual proteins.
  • the proteins are recombinant human EGFR ectodomain, BSA, recombinant human ErbB3 ectodomain and TGF-a.
  • PBST 0.05% Tween-20
  • the wells are blocked for 1 hr at room temperature with 2 % BSA in PBS and washed again as before.
  • About 50 ⁇ of the Ab #6 is added at several dilutions (1 ⁇ and serial 2 fold dilutions) in 2% BSA, PBST.
  • a DNA fragment encoding an ErbB3 ectodomain fragment corresponding to amino acid residues 1-183 of mature ErbB3 (SEQ ID NO: 73) is cloned into the yeast display vector pYD2 (a modified version of pYDl (Invitrogen) with a stop codon engineered in front of the His tag) between the Nhe and BsiWI restriction sites.
  • the plasmid is transformed into the yeast strain EBY100 (Invitrogen) and clones containing the plasmid selected on Trp- selective medium.
  • the clone is grown in glucose containing medium overnight at 30°C and expression of the ErbB3 truncation mutant is induced by transfer to a galactose-containing medium for 2 days at 18°C.
  • Yeast displaying the ErbB3 truncation mutant are stained with 50 nM of Ab #6, followed by a goat anti-human antibody labeled with Alexa dye-647.
  • a separate sample is stained with the goat anti-human antibody only to show that there is no non-specific binding to yeast of the secondary antibody. Analysis is performed by flow cytometry on the FACSCALIBUR cell sorter (BD Biosciences).
  • MALME-3M cells are seeded in 96 well tissue culture plates and grown in
  • RPMI-1640 media supplemented with antibiotics, 2mM L-glutamine and 10% fetal bovine serum (FBS) for 24 hours at 37°C and 5% carbon dioxide. Media are then switched to the same medium without FBS and with and without the antibody at concentrations of luM, 250nM, 63nM, 16nM, 4.0nM, l.OnM, 240pM, 61pM and 15pM. Medium containing no FBS or antibody is used as control.
  • FBS fetal bovine serum
  • Cells are grown for 24 hours at 37°C and 5% carbon dioxide, washed with cold PBS, then harvested with mammalian protein extract (MPER) lysis buffer (Pierce, 78505) to which 150mM NaCl, 5mM sodium pyrophosphate, lOuM bpV (phen), 50uM phenylarsine, ImM sodium orthovanadate, and protease inhibitor cocktail (Sigma, P2714) is added.
  • MPER mammalian protein extract
  • lysis buffer Pieris, 78505
  • Cell lysates are diluted two-fold with 4% bovine serum albumin in PBST (0.1% tween-20), then analyzed by ELISA with mouse anti-human ErbB
  • MALME-3M cells are trypsinized from a 15 cm dish and washed once with RPMI + 10% FBS. Cell pellets are resuspended at a density of 1 x 10 6 cells per ml. Two aliquots of 2 x 10 5 cells are added to separate wells in a 12-well tissue culture plate and each resuspended in a final volume of 800 ul RPMI + 10% FBS. To one well, Ab #6 IgGl or Ab #6IgG2 isotype is added to a final concentration of 100 nM (treated sample) and to the other well, an equivalent volume of PBS (untreated sample) is added.
  • treated and untreated cells are trypsinized, washed and incubated with 100 nM of Ab #6 in BD stain buffer for 30 minutes on ice.
  • Cells are washed twice with 1 ml BD stain buffer and incubated with 100 ul of a 1 :500 dilution of Alexa 647-labeled goat anti-human Alexa 647 for 45 minutes on ice.
  • Cells are then washed and resuspended in 300 ul BD stain buffer + 0.5 ug/ml propidium iodide.
  • MALME-3M cells are plated overnight on 6-well plates (0.2 x 10 6 per well) in RPMI+10% FBS. The following day, old medium is removed and cells are preincubated for 60 minutes on ice in RPMI + 2% FBS containing 100 nM of Ab#6 conjugated to the fluorescent dye Alexa 488. Cells are then returned to a 37°C incubator and incubated for 0.5h, 2h or 24h.
  • T-cell deficient nu/nu mice (3-4 week old female mice originated at NIH;
  • MALME-3M cells for implantation were grown in culture (RPMI media, 10% FBS, L-glutamine and antibiotics, 37°C, 5% C02) to about 80% confluency before harvesting. Cells were kept on ice until implantation. Mice were implanted via subcutaneous injection with lOOul MALME-3M cells (3.5 x 10 6 cells in PBS) via subcutaneous injection on the right flank and allowed to recover while being monitored for initial tumor growth.
  • mice were pretreated with murine IgG2a (Sigma, M7769 - 5mg) by intravenous injection to block in vivo depletion of tested antibodies by murine Fc receptors.
  • Mice were dosed intra- peritoneally every other day with either 15 ⁇ g or 100 ⁇ of Ab # 1, Ab #6, Ab #11, and Ab #13, each separately in both IgGl and IgG2 form, and tumors were measured three times per week and the measurements recorded in a Microsoft EXCEL spreadsheet.
  • each tumor in each group was divided by the initial tumor size determined by caliper measurement.
  • the samples were pulverized in a cryopulverizer (Covaris Inc). Tumors were stored in special bags (pre-weighed before the addition of the tumor) and placed in liquid nitrogen while handling them. For small tumors, 200 of Lysis buffer was first added to the bag with the tumor, frozen in liquid nitrogen and then pulverized to improve the recovery of the tumor from the bag. Pulverized tumors were transferred to 2 ml EPPENDORF tubes and placed in liquid nitrogen until lysed. Tumors were lysed in Lysis buffer supplemented with protease and phosphatase inhibitors. Lysis Buffer was added to the tumor aliquots in a final concentration of 62.5 mg/ml.
  • Tumor samples were homogenized by vortexing for 30 seconds and letting them sit on ice for 30 min. The lysates were spun for 10 minutes in QIAGEN QIASHREDDER columns for further homogenization of the samples. Cleared lysates were aliquoted into fresh tubes.
  • the BCA assay is performed as set forth substantially as described in the materials and methods section supra.
  • the total levels of ErbB3 were determined by ELISA.
  • the ELISA reagents were purchased from R&D Systems as DUOSET kits.
  • 96-well NUNC MAXISORB plates were coated with 50 ⁇ of respective capture antibody and incubated overnight at room temperature. The next morning, the plates were washed 3 times with 1000 ⁇ /well in a BIOTEK plate washer with PBST (0.05% Tween-20) and then blocked for 1 hour at room temperature with 2 % BSA in PBS. The plates were then washed again as before. Lysates (50 ⁇ ) and standards were diluted in 50% Lysis buffer and 1 %BSA; all samples were run in duplicate.
  • MALME3M, ACHN and NCI/AdrR cells are seeded in 96 well tissue culture plates and grown in RPMI-1640 media supplemented with antibiotics, 2mM L- glutamine and 10% FBS for 24 hours at 37 degrees Celsius and 5% carbon dioxide. Media are then switched to RPMI-1640 media with antibiotics and 2mM L-glutamine without FBS in the presence or absence of Ab #6 at luM, 250nM, 63nM, 16nM, 4.0nM, l .OnM, 240pM, 61pM and 15pM concentrations.
  • Cells are grown for 96 hours at 37°C and 5% carbon dioxide, then harvested with CellTiter-Glo® Luminescent Cell Viability Assay (Promega, G7573) and analyzed on a luminometer. Media containing no serum and antibody is used as control.
  • MALME-3M cells (Figure 9), AdrR ovarian cancer cells ( Figure 10) and ACHN cells ( Figure 11) which express ErbB3.
  • Ab #6 inhibited proliferation of MALME-3M cells by about 19.6%, as measured using the CellTiter-Glo® assay, and inhibited proliferation of AdrR ovarian cancer cells by about 30.5%.
  • Ab #6 inhibited proliferation of ACHN cells by about 25.4%.
  • the samples are pulverized substantially as described in Example 5 supra, with respect to Figure 8.
  • the BCA assay is performed substantially as set forth in the Materials and Methods section supra, and the ELISA assay is performed substantially as described in Example 5 supra with respect to Figure 8.
  • Ovarian AdrR cells are preincubated with Ab #6 for 30 minutes prior to stimulation with 50 mM BTC, 10 mM HRG or 333 nM TGF-a. Following pre incubation, the media are removed and the cells are stimulated for 5 minutes at 37°C, 5% C02 with 50nM BTC or 333nM TGF-a. HRG controls (5 minutes, 5nM), 10% serum and 0% serum controls are also used.
  • OVCAR 5 and OVCAR 8 cell lines are obtained from the National Cancer Institute, Division of Cancer Treatment and Diagnostics ("DCTD").
  • DCTD Cancer Treatment and Diagnostics
  • the ELISA is performed as substantially as described in the Materials and Methods section supra.
  • AdrR cells or MALME-3M cells (1 x 10 s ) are pre-incubated with 25 ⁇ of anti- ErbB3 Ab #6 or 25 ⁇ of cetuximab (anti-ErbBl) in 50 ⁇ 1 BD stain buffer for 30 minutes on ice. After 30 minutes, 50 ⁇ of 400 nM biotinylated BTC is added to the cells and incubated for another 30 minutes on ice. This yields a final concentration of 12.5 ⁇ antibodies and 200 nM BTC. Cells are then washed twice with 500 ⁇ BD stain buffer and incubated with 100 ⁇ of a 1 :200 dilution of streptavidin-PE
  • FACSCALEBUR flow cytometer As a positive control, 1 x 10 s AdrR or MALME-3M cells are incubated with 200 nM BTC for 30 minutes on ice, washed twice and incubated with a 1 :200 dilution of streptavidin-PE for 45 minutes. To assess background staining from the streptavidin-PE conjugate, cells are incubated with 100 ⁇ of a 1 :200 dilution of streptavidin-PE only for 45 minutes.
  • MALME-3M cells and OVCAR8 cells are separately seeded in 96 well tissue culture plates (35,000 cells per well) and grown in RPMI-1640 media supplemented with antibiotics, 2mM L-glutamine and 10% FBS for 24 hours at 37°C and 5% carbon dioxide.
  • Cells are serum-starved in RPMI-1640 media with antibiotics and 2mM L- glutamine for 24 hours at 37°C and 5% carbon dioxide.
  • Cells are pre-treated with and without the anti-ErbB3 antibody (IgG2 isotype of Ab #6) at ⁇ , 250nM, 63nM, 16nM, 4.0nM, l .OnM, 240pM, 61pM and 15pM concentrations for 30 minutes then stimulated with 5nM HRG for 10 minutes at 37°C and 5% carbon dioxide. Controls are HRG without added antibody and untreated cells (no HRG and no Ab).
  • IgG2 isotype of Ab #6 the anti-ErbB3 antibody
  • M-PER mammalian protein extract
  • Cell lysates are diluted two-fold with 4% bovine serum albumin in PBST (0.2% tween-20), then analyzed by ELISA for phosphorylation of either ErbB3, or AKT (a downstream effector of ErbB3).
  • lysates are run on an ELISA plate with a capture antibody specific for ErbB3 and an anti-phosphotyrosine detection antibody conjugated to horseradish- peroxidase. This is then reacted with the same chemiluminescent substrate.
  • Luminescent signal on the ELISAs is measured using a luminometer.
  • Ab #6 is a potent inhibitor of heregulin- mediated signaling in MALME-3M cells and OVCAR8 cells, as measured by decreased phosphorylation of ErbB3 ( Figure 16A) and AKT ( Figure 16B). Notably, essentially complete inhibiton of the phosphorylation of each of AKT and ErbB3 by Ab #6 is observed.
  • T-cell deficient nu nu mice (3-4 week old female mice originated at NIH; outbred; albino background) are purchased from Charles River Labs (Wilmington, MA) for xenograft studies.
  • AdrR cells for implantation are grown in culture (RPMI media, 10% FBS, L-glutamine and antibiotics, 37°C, 5% C02) to about 85% confluency before harvesting. Cells are kept on ice until implantation. Mice are implanted via subcutaneous injection with ⁇ AdrR cells (6 x 10 6 cells in PBS) on the right flank and allowed to recover while being monitored for initial tumor growth.
  • Tumors are measured (length by width) by digital caliper and the mice are dosed with IgG2a (Sigma, M7769-5MG) by intravenous injection. Mice are dosed intra- peritoneally every third day with either 30 ⁇ g or 300 ⁇ g of Ab #6 and tumors are measured three times per week and recorded in a Microsoft Excel spreadsheet.
  • ErbB3 ligands e.g., heregulin and epiregulin
  • Ab #6 and Ab Fab #3 to inhibit the binding of heregulin to ErbB3.
  • MALME3M cells (1 x 10 5 ) are incubated with 10 ⁇ of an anti-ErbB3 antibody (e.g., Ab #6 or Ab Fab #3) in 50 ⁇ BD stain buffer for 30 minutes on ice. After 30 minutes, 50 ⁇ of 40 nM biotinylated heregulin EGF is added to the cells and incubated for another 10 minutes on ice. This yields a final concentration of 5 ⁇ antibody and 20 nM heregulin EGF.
  • an anti-ErbB3 antibody e.g., Ab #6 or Ab Fab #3
  • AdrR cells (1 x 10 5 ) are pre-incubated with 25 ⁇ of the anti-ErbB3 antibody, Ab #6, or 25 ⁇ of the anti-ErbBl antibody cetuximab, or with no added antibody (as control) in 50 ⁇ BD stain buffer for 30 minutes on ice. After 30 minutes, 50 ⁇ of 2 ⁇ biotinylated Epi is added to the cells and incubated for another 30 minutes on ice. This yields a final concentration of 12.5 ⁇ antibodies and 1 ⁇ Epi.
  • PE phycoerythrin
  • HB-EGF binds to AdrR cells, presumably to either or both of ErbB 1 and ErbB4 on AdrR cells.
  • Ab #6 does not inhibit this binding, evidencing that Ab #6 is specific for inhibiting the binding of ErbB3 ligands (e.g., heregulin and epiregulin) to ErbB3.
  • ErbB3 ligands e.g., heregulin and epiregulin
  • VEGF secretion assay VEGF ELISA, R&D Systems DY293B
  • ELISA Assay VEGF secretion assay
  • HRG is able to induce more than a doubling of VEGF secretion in MCF-7 and T47D cells.
  • Ab #6 also shows a similar effect in vivo by inhibiting VEGF secretion in three different xenografts, the highest being in COLO-357 xenograft (Fig. 24B). Inhibition of VEGF correlates with inhibition of ErbB3 phosphorylation (Fig. 24 C). It has been demonstrated that myeloma cell-secreted factors, such as VEGF and bFGF, trigger angiogenesis (see, e.g., Leung et al. (1989) Science 246(4935): 1306-9; Yen et al. (2000) Oncogene 19(31):3460-9). Inhibition of VEGF secretion is also known in the art to correlate with inhibition of tumor induced angiogenesis, a facilitator of tumor growth.
  • MCF-7 cells e.g., MCF-7 cells
  • a trans-well assay (Millipore Corp., Billerica, MA, # ECM552).
  • MCF-7 cells are serum-starved overnight and then incubated in the presence or absence of Ab #6 (8uM final concentration) for 15 minutes at room temperature.
  • the cells are then transferred to an upper chamber that is separated from a lower chamber by a collagen type I-coated membrane through which the cells can migrate.
  • 10%FBS is added to media in the lower chamber to act as a chemoattractant in the presence of absence of Ab #6.
  • the chambers are incubated at 37°C for 16 hours and then the cells that migrate through the membrane into the lower chamber are removed using a detachment buffer and incubated with a cell-binding fluorescent dye.
  • Fluorescence is quantitated using a fluorescent plate reader.
  • 10% FBS stimulates cell migration (lane 3) as compared to untreated control (lane 1) and 8uM Ab #6 inhibits the FBS induced cell migration (lane 4).
  • Ab #6 Ab #6 to inhibit the spheroid growth of cells expressing ErbB3 is examined using an assay which approximates conditions of developing tumor growth (Herrmann et al. J Biomol Screen. 2008 Jan; 13( 1 ): 1 -8. Epub 2007 Nov 26)
  • AdrR an ovarian cancer cell line
  • DU145 a prostate cancer cell line
  • Subconfluent cells are trypsinized, counted, and resuspended in filtered medium.
  • the cell concentration is adjusted to 100,000 cells/ml, and one 20 ul drop (containing 2000 cells) is added to each ring on the underside of the lid of a 96-well plate.
  • the lids with these hanging droplets are then placed back on the original 96-well plate, which contained 100 ul of PBS in each well to maintain moisture.
  • the hanging droplets containing the spheroids are replated into new 96- well plates. This replating involves transferring the lid containing the hanging droplets to a fresh 1% agarose/RPMI-coated 96-well plate that contains 150 ul of medium per well.
  • the plate and lid are then centrifuged together at 500 rpm for 1 minute to transfer the spheroid-containing drops from the lids to the wells. Plates are then further incubated at 37° C in a humidified C02 incubator. Spheroids are photographed in an inverted phase contrast microscope. A micrometer scale is also photographed at the same magnification to determine spheroid size. The diameter of the spheroids is determined using Metamorph Analysis software (MDS Analytical Technologies).
  • Multicellular tumor spheroids are generated as before.
  • the antibody is added to a final concentration of 25 ⁇ / ⁇ 1 on days 1 and 4 of spheroid formation.
  • Data obtained using the methods described above or minor variations thereof showed that Ab #6 caused a 30-40% decrease in OVCAR8 spheroid area.
  • Ab #6 The ability of Ab #6 to inhibit the signaling induced by different ligands is examined. For example, the effect of Ab #6 on HRG and BTC binding to AdrR cells expressing ErbB3 receptor is tested. As shown by the FACS analysis results presented in Figures 27A and B, Ab #6 competes with HRG but not BTC for binding to AdrR cells. Accordingly, since Ab #6 does not activate HRG-induced signaling (as is indicated by experiments below) blocking of HRG binding to ErbB3 by Ab #6 will prevent signaling induced by HRG.
  • ligands are tested for inducement of ErBb3 phosphorylation. At least three ligands, HRG, BTC, and HGF, are able to stimulate ErbB3 induced
  • HGF Hepatocyte Growth Factor
  • ErbB3 phosphorylation in AdrR cells carrying the wild type EGFR and is not dependent on amplified c-MET.
  • HGF Hepatocyte Growth Factor
  • ErbB3 phosphorylation in AdrR cells in a dose dependent manner as shown in Figure 28.
  • Ab #6 inhibits HGF induced erbB3 phosphorylation.
  • HRG and BTC are found to induce phosphorylation of both ErbBl and ErbB3.
  • HRG is found to be a more potent inducer of ErbB3 phosphorylation while BTC is a potent inducer of ErbB 1 phosphorylation (Fig. 29A).
  • AdrR cells are plated into 96 well plate at a density of 30,000 cells/well/ 100 uL in RPMI medium containing 10% FBS and allowed to grow overnight;
  • Cells are pre-treated with different concentrations of Ab #6 (from 0.01 nM to ⁇ ⁇ ⁇ ), or buffer (un-pretreated, "0"), for 2hours;
  • Pretreated and un-pretreated cells are then stimulated with 10 nM HRG or HGF for 10 minutes, or 10 nM BTC or EGF for 5 minutes, and separate wells of un- pretreated cells are left unstimulated ("Control");
  • the reaction is stopped by removing the culture medium and washing the cells once with ice cold PBS;
  • the cells are then lysed in 25 mM Tris, pH+7.5, 150 mM NaCl, lmM EDTA, 1.0% Triton X-100, 1.0% CHAPS, 10% v/v glycerol, containing IX protease inhibitor and IX phosphatase inhibitor; and
  • ErbB3 phosphorylation is measured in cell lysates using Human Phospho-ErbB3 ELISA kit (R&D Systems, DYC1769) according to manufacturer's instructions.
  • AdrR cells are pre-incubated with buffer (control), or 250 nM Ab #6 for 60 minutes at room temperature, then treated with 10 nM HRG or 10 nM
  • the cells are lysed in 25 mM Tris,
  • the immunoprecipitates are then washed with ice-cold lysis buffer 3 times,
  • Example 15 Inhibition Profile of Ab #6 is Distinct from cetuximab, lapatinib and pertuzumab.
  • AdrR cells stimulated with either HRG or BTC in the presence of either Ab #6, cetuximab, lapatinib or pertuzumab.
  • Serum starved AdrR cells are pre-incubated for 30 minutes with 4-fold serial dilutions of Ab #6, lapatinib or cetuximab from 2 mM to 7.6 pM or of pertuzumab from 100 nM to 1.5 pM and then stimulated for 10 minutes with 25 nM of HRG or BTC.
  • Phosphorylation of ErbB3 is measured by ELISA (R&D Systems, DYC1769-5, per manufacturers protocol) and IC50 values are determined using Prism (GraphPad
  • Lapatinib (a reversible tyrosine kinase inhibitor of ErbBl and ErbB2) inhibited HRG and BTC-induced ErbB3 phosphorylation with IC 50 s of 150 nM and 360 nM, respectively (with 95% confidence intervals of 46-504 nM and 119- 1069 nM, respectively).
  • Cetuximab (an anti-ErbB l antibody) inhibited BTC-induced ErbB3 phosphorylation with an IC50 of 1.8 nM (with a 95% confidence interval of 0.9- 3.8 nM). but cetuximab did not inhibit HRG-induced ErbB3-induced ErbB3 phosphorylation, confirming earlier observations that HRG signaling is mediated primarily by ErbB2/ErbB3 and not ErbB l/ErbB3 heterodimers.
  • Pertuzumab (a monoclonal antibody that sterically hinders ErbB2's recruitment into ErbB ligand complexes) inhibited HRG-induced ErbB3 phosphorylation with an IC50 of 3.6 nM (with a 95% confidence interval of 1.0-
  • Ab #6 was the only inhibitor tested that is capable of potently inhibiting ErbB3 phosphorylation with low nanomolar IC50 values for both HRG and BTC stimulation.
  • the efficacy of Ab #6 in inhibiting tumor growth in xenograft tumor models is assessed in combination with other therapeutic agents, namely erlotinib or taxol.
  • mice bearing ACHN a renal tumor cell line
  • xenograft tumors are prepared by establishing tumors subcutaneously in the flanks of nude mice (Charles River Laboratories).
  • the tumor-bearing mice are dosed intraperitoneally (EP) every three days with either a suboptimal dose of 300 ⁇ g Ab #6 or vehicle.
  • Erlotinib 25 mg/kg is administered orally once daily, either alone or in combination with the Ab #6 treatment.
  • Tumor size length x width
  • p/6 L x W2
  • mice bearing DU145 a prostate tumor cell line
  • xenograft tumors are prepared by establishing tumors subcutaneously in the flanks of nude mice (Charles River Laboratories). The tumor-bearing mice are dosed
  • A549 is a lung cancer cell line that comprises a G12S KRAS mutation, a mutation in codon 12 of the human KRAS gene, in which the codon is changed from one coding for glycine (G) to one coding for serine (S).
  • This cell line is insensitive to treatment with erlotinib or taxol alone.
  • the cells are grown as multicellular tumor spheroids (2000 cells/spheroid/well of a 96 well plate) and then treated with 0, 0.001, 0.01, 0.1 or 1 uM Ab #6 for seven days.
  • the area of the spheroid is measured under 4x magnification using METAMORPH software on day 1 and day 7 and the percent change in spheroid area is calculated ((Initial Area - Final Area/ Initial Area x 100).
  • the results (obtained using the methods described above or minor variations thereof) are shown in the graph of Figure 32A ("-4" on the x axis of the graph corresponds to the "0" dose), and the photographs of Figure 32B.
  • the data of Figure 32A demonstrate that Ab #6 treatment alone is sufficient to inhibit the growth of the KRAS mutant A549 tumor cells in vitro and that there is a linear dose response result regarding the percent decrease in spheroid area with the indicated ten-fold increases in Ab #6 concentration.
  • Photographs of representative spheroids are shown in Figure 32B. Untreated spheroids grew
  • nude mice bearing A549 subcutaneous xenograft tumors are treated with 600 ⁇ g of Ab #6 every three days, followed by measurement of tumor volume.
  • the results are shown in the graph of Figure 32C, which demonstrates significant tumor growth inhibition by Ab #6 treatment alone, which growth inhibition is retained after dosing stopped on day 22.
  • Ab #6 alone was able to inhibit the growth of KRAS mutant tumor cells in vivo.
  • nude mice bearing A549 subcutaneous xenograft tumors are treated with either: (i) 300 g of Ab #6 alone every three day; (ii) 25 mg/kg erlotinib alone every day; (iii) 20 mg kg taxol alone every 7 days; (iv) Ab #6 and erlotinib in combination (at the same dosing); or (v) Ab #6 and taxol in combination (at the same dosing).
  • alanine scanning mutagenesis is used to map the epitope within ErbB3 to which Ab #6 binds.
  • ErbB3 point mutations are made: L14A, N15A, L17A, S 18A, VI 9 A, T20A, N25A, K32A, L33A, V47A, L48A, M72A, Y92A, Y92P, D93A, M101A, L102A, Y104A, Y104P, N105A, T106A, Y129A, Y129P, 132A, Q59A, T77A, Q90A, Ql 14A, Ql 19A, R145A, R151A, V156A, H168A, K172A and L186A.
  • L14A indicates that leucine (L) at amino acid position 14 of mature ErbB3 (SEQ ID NO: 73) is specifically mutated to alanine (A), with other substitution mutations indicated in the same fashion.
  • Mutagenesis is accomplished using standard methods known in the art and the ErbB3 mutants, within the context of the ErbB3 ectodomain Domain I, are expressed on the surface of yeast cells using standard methods known in the art. All mutants expressed well on the surface of the yeast at levels identical to those of the wild type ErbB3 Domain I. Expression of the ErbB3 Domain I mutants on yeast allows for the examination of binding of Ab #6 to mutants using FACS without first purifying the recombinant proteins.
  • Binding of Ab #6 to ErbB3 ectodomain Domain I fragments comprising single mutations is determined by standard FACS analysis.
  • a second anti-ErbB3 antibody (SGP1 ; LabVision), which binds an epitope on ErbB3 which does not overlap with the epitope bound by Ab #6 (as evidenced by lack of competition for binding to ErbB3), is used as a control to ensure that the mutations are not just generally destabilizing the ErbB3 structure.
  • SGP1 Second anti-ErbB3 antibody
  • yeast cells expressing the different mutant version of ErbB3 Domain I on their surface are labeled simultaneously with Ab #6 (followed by a goat anti-human Ab-Alexa 488 as secondary antibody) and with SGPl-Alexa 647.
  • Data obtained using the methods described above or minor variations thereof) indicate that four of the alanine point mutations describned above inhibit Ab #6 binding to ErbB3 ectodomain Domain I while not inhibiting the binding of the control anti-ErbB3 antibody (SGP1). These four point mutations are: D93A, M101A, L102A and Y104A.
  • Ab #6 epitope comprises residues 93, 101, 102 and 104 of mature ErbB3 (SEQ ID NO: 73)
  • these four mutations are expressed in the context of the full length mature ErbB3 following stable transfection into CHO Kl cells in order to examine their effects in this context.
  • CHO Kl cells expressing wild type full-length ErbB3 serve as a positive control.
  • High expressing clones are selected for each mutant and labeled for standard FACS analysis either with Ab #6 or SGP1 (to confirm proper folding of ErbB3).
  • SGP1 is directly labeled with Alexa 647 dye and Ab #6 is directly labeled with Alexa 488 dye.
  • Figures 34A-34E show the binding of SGP1 or Ab #6 to wild type ErbB3, the D93A mutant, the M101A mutant, the L102A mutant or the Y104A mutant, respectively.
  • SGP1 control anti-ErbB3 antibody
  • Ab #6 displays essentially no binding to three of the mutants (D93A, L102A and Y104A) and only very minimal binding to the fourth mutant (M101A).
  • the epitope bound by Ab #6 may be viewed as comprising the sequence spanning residues 92-104 and 129 (which lies adjacent to 92-104 in the folded protein,), of the mature, human ErbB3 sequence shown in SEQ ID NO: 73.
  • SKOV3 is an ovarian cancer cell line that comprises a mutation that upregulates PI3K expression.
  • nude mice bearing SKOV3 subcutaneous xenograft tumors (tumor volume of 150-200 mm 3 ) are treated with 600 ⁇ g of Ab #6 every three days, followed by measurement of tumor volume.
  • nude mice bearing SKOV3 subcutaneous xenograft tumors are treated with either: (i) 300 ⁇ g of Ab #6 alone every three day (a suboptimal dose of Ab #6); (ii) 1.5 mg kg cisplatin (CDDP) alone; or (iii) Ab #6 and cisplatin in combination (at the same dosing).
  • a suboptimal dose of Ab #6 a suboptimal dose of Ab #6
  • CDDP 1.5 mg kg cisplatin
  • Ab #6 and cisplatin in combination at the same dosing.
  • paratope mapping of Ab #6 is performed using a single chain (scFv) version of the antibody.
  • scFv version SEQ ID NO: 72
  • the VH region SEQ ID NO: 1
  • V L region SEQ ID NO: 2
  • this scFv still retains the ability to specifically bind ErbB3.
  • This format is used to investigate the effect on ErbB3 binding of alanine (and in some cases phenylalanine) substitution mutations in the CDR loops.
  • Table 1 Also shown in Table 1 are the sequences of the VH CDRS of the lmhp antibody and of the V L CDRs of the lmco antibody, with the number of amino acid mismatches compared to Ab #6 shown in parentheses following the SEQ ID NO for each CDR and the surface residues highlighted in italics, boldface and underlined.
  • Table 1 CDR Loops in Antibody #6
  • the mutations made in Ab #6 are summarized below in Table 2.
  • the amino acid residue numbering for the mutations corresponds to the linear numbering of the V H and V L residues within the scFv format, not to Kabat numbering.
  • the notation Thr28Ala indicates that threonine (Thr) at amino acid position 28 of the scFv is specifically mutated to alanine (Ala), with the rest of the substitution mutations indicated in the same fashion.
  • Table 2 Ab #6 Mutations
  • consensus sequences for each of the heavy and light chain CDRs were derived. These consensus sequences are shown in Figures 37A and 37B, below the wild type CDR sequences. The consensus sequences differ from the wild type CDR sequences in that those amino acid residues that are mutated to alanine and found not to substantially affect binding have been genericized to allow any amino acid (Xaa) at those positions.
  • any of the aromatic amino acids tyrosine, histidine, tryptophan and phenylalanine are allowed at that position in the consensus sequences shown in Figure 37A, while in the consensus sequences shown in Figure 37B those amino acid positions are allowed to be either tyrosine or phenylalanine.
  • paratope sequences for each of the heavy and light chain CDRs were derived. These paratope sequences are shown in Figures 37A and 37B, below the wild type and consensus CDR sequences. The paratope sequences correspond to those amino acid residue positions within the CDRs that were shown to substantially decrease binding to ErbB3 when mutated to alanine, thereby suggesting a substantial role for these positions in antigen binding.
  • any of the aromatic amino acids tyrosine, histidine, tryptophan and phenylalanine in Figure 37A are allowed at each such position in the paratope.
  • the other amino acid positions within the CDRs that do not appear to be directly involved in antigen binding are allowed to be any amino acid residue (Xaa) within the paratope.
  • the wild-type heavy chain CDRl , CDR2 and CDR3 sequences for Ab #6 are also shown in SEQ ID NOs: 7, 8 and 9, respectively, and the wild type light chain CDRl, CDR2 and CDR3 sequences for Ab #6 are also shown in SEQ ID NOs: 10, 11 and 12, respectively.
  • the consensus heavy chain CDRl, CDR2 and CDR3 sequences for Ab #6 are also shown in SEQ ID NOs: 60 and 75 (CDRl), 61 (CDR2) and 62 (CDR3), respectively, and the consensus light chain CDRl, CDR2 and CDR3 sequences for Ab #6 are also shown in SEQ ID NOs: 66 and 77 (CDRl), 67 (CDR2) and 68 and 79 (CDR3), respectively.
  • the heavy chain CDRl, CDR2 and CDR3 paratope sequences for Ab #6 are also shown in SEQ ID NOs: 63 and 76 (CDRl), 64 (CDR2) and 65 (CDR3), respectively, and the light chain CDRl, CDR2 and CDR3 paratope sequences for Ab #6 are also shown in SEQ ID NOs: 69 and 77 (CDRl), 70 (CDR2) and 71 and 80 (CDR3), respectively.

Landscapes

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

Abstract

La présente invention a pour objet une nouvelle classe d’anticorps et leurs fragments de liaison à l’antigène qui se lient au domaine extracellulaire du récepteur ErbB3 et inhibent différentes fonctions de ErbB3. Par exemple, les anticorps et les fragments de liaison à l’antigène selon la présente invention sont capables de se lier au récepteur désigné ErbB3 et d’inhiber la phosphorylation du récepteur médiée par un ligand de type EGF. De tels anticorps et leurs fragments de liaison à l’antigène ont la caractéristique utile d’inhiber la prolifération des cellules cancéreuses exprimant ErbB3.
EP10810721A 2009-08-21 2010-08-23 Anticorps contre l ectodomaine de erbb3 et leurs utilisations Withdrawn EP2467164A2 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US23579909P 2009-08-21 2009-08-21
PCT/US2010/046364 WO2011022727A2 (fr) 2009-08-21 2010-08-23 Anticorps contre l’ectodomaine de erbb3 et leurs utilisations

Publications (1)

Publication Number Publication Date
EP2467164A2 true EP2467164A2 (fr) 2012-06-27

Family

ID=43607614

Family Applications (1)

Application Number Title Priority Date Filing Date
EP10810721A Withdrawn EP2467164A2 (fr) 2009-08-21 2010-08-23 Anticorps contre l ectodomaine de erbb3 et leurs utilisations

Country Status (21)

Country Link
EP (1) EP2467164A2 (fr)
JP (1) JP5752687B2 (fr)
KR (1) KR20120059568A (fr)
CN (1) CN103002912A (fr)
AU (1) AU2010284018C1 (fr)
BR (1) BR112012003809A2 (fr)
CA (1) CA2771744A1 (fr)
CR (1) CR20120108A (fr)
DO (1) DOP2012000044A (fr)
EA (1) EA201200195A1 (fr)
EC (1) ECSP12011740A (fr)
IL (1) IL218097A0 (fr)
IN (1) IN2012DN01518A (fr)
MA (1) MA33582B1 (fr)
MX (1) MX336091B (fr)
NI (1) NI201200027A (fr)
PE (1) PE20121585A1 (fr)
SG (1) SG178509A1 (fr)
TN (1) TN2012000057A1 (fr)
WO (1) WO2011022727A2 (fr)
ZA (1) ZA201201195B (fr)

Families Citing this family (33)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2129396B1 (fr) 2007-02-16 2013-08-21 Merrimack Pharmaceuticals, Inc. Anticorps contre erbb3 et leur utilisation
DK3351558T3 (da) * 2009-11-13 2020-06-02 Daiichi Sankyo Europe Gmbh Materiale og fremgangsmåder til behandling eller forebyggelse af her-3-relaterede sygdomme
EP2516469B1 (fr) 2009-12-22 2016-03-30 Roche Glycart AG Anticorps anti-HER3 et leurs utilisations
CN102858335B (zh) 2010-03-11 2015-04-15 梅里麦克制药股份有限公司 Erbb3抑制剂在三阴性乳腺癌和基底样乳腺癌治疗中的用途
BR112012025730B1 (pt) 2010-04-09 2020-12-08 Aveo Pharmaceuticals, Inc anticorpo isolado que se liga ao erbb3 humano, seus usos, seu processo de produção e vetor de expressão
KR102027011B1 (ko) 2010-08-20 2019-09-30 노파르티스 아게 표피 성장 인자 수용체 3 (her3)에 대한 항체
TW201302793A (zh) 2010-09-03 2013-01-16 Glaxo Group Ltd 新穎之抗原結合蛋白
IT1402149B1 (it) * 2010-10-04 2013-08-28 Ist Fisioterap Ospitalroma Uso di un fosfopeptide in grado di bloccare l interazione her3/p85 per il trattamento dei tumori iperesprimenti her2.
JP6033783B2 (ja) * 2010-11-01 2016-11-30 シムフォゲン・アクティーゼルスカブSymphogen A/S Pan−her抗体組成物
US8476409B2 (en) 2011-04-19 2013-07-02 Merrimack Pharmaceuticals, Inc. Bispecific anti-IGF-1R and anti-ErbB3 antibodies
JPWO2012176779A1 (ja) * 2011-06-20 2015-02-23 協和発酵キリン株式会社 抗erbB3抗体
US9273143B2 (en) 2011-09-30 2016-03-01 Regeneron Pharmaceuticals, Inc. Methods and compositions comprising a combination of an anti-ErbB3 antibody and an anti-EGFR antibody
IN2014CN03042A (fr) 2011-09-30 2015-07-03 Regeneron Pharma
AU2012318541B2 (en) 2011-10-06 2018-04-12 Aveo Pharmaceuticals, Inc. Predicting tumor response to anti-ERBB3 antibodies
EP2788377B1 (fr) * 2011-11-09 2019-01-23 The UAB Research Foundation Anticorps her3 et utilisations de ceux-ci
EP3590538A1 (fr) 2011-12-05 2020-01-08 Novartis AG Anticorps dirigés contre le récepteur 3 du facteur de croissance épidermique (her3)
WO2013124297A1 (fr) * 2012-02-22 2013-08-29 U3 Pharma Gmbh Composition d'une protéine de liaison d'hb-egf et d'un inhibiteur de l'egfr
AR094403A1 (es) 2013-01-11 2015-07-29 Hoffmann La Roche Terapia de combinación de anticuerpos anti-her3
US20160232293A1 (en) * 2013-10-17 2016-08-11 Sanford-Burnham Medical Research Institute Drug sensitivity biomarkers and methods of identifying and using drug sensitivity biomarkers
EP3087394A2 (fr) 2013-12-27 2016-11-02 Merrimack Pharmaceuticals, Inc. Profils de biomarqueur pour prédire les résultats d'une thérapie cancéreuse utilisant des inhibiteurs d'erbb3 et/ou des chimiothérapies
NZ724013A (en) * 2014-02-28 2019-11-29 Merus Nv Antibodies that bind egfr and erbb3
CN110642952B (zh) * 2014-08-14 2021-05-25 上海生物制品研究所有限责任公司 抗her3抗体、其制法及其应用
WO2016038610A1 (fr) 2014-09-08 2016-03-17 Yeda Research And Development Co. Ltd. Compositions et procédés de traitement d'un cancer résistant à un inhibiteur de la tyrosine kinase (tki)
WO2016038609A1 (fr) * 2014-09-08 2016-03-17 Yeda Research And Development Co. Ltd. Anticorps anti-her3 et leurs utilisations
US10184006B2 (en) 2015-06-04 2019-01-22 Merrimack Pharmaceuticals, Inc. Biomarkers for predicting outcomes of cancer therapy with ErbB3 inhibitors
WO2017069628A2 (fr) 2015-10-23 2017-04-27 Merus N.V. Molécules de liaison inhibant la croissance de cancer
US11780925B2 (en) 2017-03-31 2023-10-10 Merus N.V. ErbB-2 and ErbB3 binding bispecific antibodies for use in the treatment of cells that have an NRG1 fusion gene
EP3665200A4 (fr) 2017-08-09 2021-04-28 University of Saskatchewan Agents de liaison à her3 et utilisations associées
BR112020002695A2 (pt) 2017-08-09 2020-08-25 Merus N.V. anticorpos que se ligam à egfr e cmet
GB201804094D0 (en) * 2018-03-14 2018-04-25 Ultrahuman Thirteen Ltd ERBB3 Binding agents
JP2021534811A (ja) * 2018-06-22 2021-12-16 シーアールディー ファーマシューティカルズ インコーポレイティド 抗her3抗体およびその用途
AU2021360625A1 (en) * 2020-10-14 2023-06-01 Jiangsu Hengrui Pharmaceuticals Co., Ltd. Anti-her3 antibody and anti-her3 antibody-drug conjugate and medical use thereof
WO2023198138A1 (fr) * 2022-04-13 2023-10-19 上海翰森生物医药科技有限公司 Anticorps ou fragment de liaison à l'antigène de celui-ci et son utilisation médicale

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2129396B1 (fr) * 2007-02-16 2013-08-21 Merrimack Pharmaceuticals, Inc. Anticorps contre erbb3 et leur utilisation

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO2011022727A3 *

Also Published As

Publication number Publication date
AU2010284018C1 (en) 2015-10-15
CR20120108A (es) 2012-06-05
KR20120059568A (ko) 2012-06-08
TN2012000057A1 (en) 2013-09-19
ECSP12011740A (es) 2013-02-28
CA2771744A1 (fr) 2011-02-24
AU2010284018A1 (en) 2012-03-22
ZA201201195B (en) 2015-07-29
JP2013506622A (ja) 2013-02-28
MX336091B (es) 2016-01-08
PE20121585A1 (es) 2012-11-29
SG178509A1 (en) 2012-04-27
IN2012DN01518A (fr) 2015-06-05
AU2010284018B2 (en) 2014-06-05
CN103002912A (zh) 2013-03-27
EA201200195A1 (ru) 2012-12-28
DOP2012000044A (es) 2012-06-30
WO2011022727A3 (fr) 2013-06-27
IL218097A0 (en) 2012-04-30
WO2011022727A2 (fr) 2011-02-24
MX2012002172A (es) 2012-05-29
MA33582B1 (fr) 2012-09-01
BR112012003809A2 (pt) 2019-09-24
NI201200027A (es) 2013-01-29
JP5752687B2 (ja) 2015-07-22

Similar Documents

Publication Publication Date Title
US20220204642A1 (en) Antibodies against the ectodomain of erbb3 and uses thereof
AU2010284018C1 (en) Antibodies against the ectodomain of ErbB3 and uses thereof
AU2012279132B2 (en) Antibodies against epidermal growth factor receptor (EGFR) and uses thereof
AU2011248088B2 (en) Antibodies against epidermal growth factor receptor (EGFR) and uses thereof

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20120316

AK Designated contracting states

Kind code of ref document: A2

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO SE SI SK SM TR

AX Request for extension of the european patent

Extension state: BA ME RS

REG Reference to a national code

Ref country code: HK

Ref legal event code: DE

Ref document number: 1166273

Country of ref document: HK

R17D Deferred search report published (corrected)

Effective date: 20130627

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION HAS BEEN WITHDRAWN

18W Application withdrawn

Effective date: 20160629