WO2011161119A1 - Antibodies against insulin-like growth factor i receptor and uses thereof - Google Patents

Antibodies against insulin-like growth factor i receptor and uses thereof Download PDF

Info

Publication number
WO2011161119A1
WO2011161119A1 PCT/EP2011/060376 EP2011060376W WO2011161119A1 WO 2011161119 A1 WO2011161119 A1 WO 2011161119A1 EP 2011060376 W EP2011060376 W EP 2011060376W WO 2011161119 A1 WO2011161119 A1 WO 2011161119A1
Authority
WO
WIPO (PCT)
Prior art keywords
antibody
igf
seq
antibodies
region
Prior art date
Application number
PCT/EP2011/060376
Other languages
French (fr)
Inventor
Erhard Kopetzki
Georg Tiefenthaler
Original Assignee
F. Hoffmann-La Roche Ag
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by F. Hoffmann-La Roche Ag filed Critical F. Hoffmann-La Roche Ag
Publication of WO2011161119A1 publication Critical patent/WO2011161119A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2863Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against receptors for growth factors, growth regulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/34Identification 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
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/565Complementarity determining region [CDR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value

Definitions

  • the present invention relates to antibodies against human insulin-like growth factor I receptor (IGF-IR), methods for their production, pharmaceutical compositions containing said antibodies, and uses thereof.
  • IGF-IR insulin-like growth factor I receptor
  • Insulin-like growth factor I receptor (IGF-IR, EC 2.7.10.1, CD 221 antigen) belongs to the family of transmembrane protein tyrosine kinases (LeRoith, D., et al, Endocrin. Rev. 16 (1995) 143-163; and Adams, T.E., et al, Cell. Mol. Life Sci. 57 (2000) 1050-1093).
  • IGF IR binds IGF I with high affinity and initiates the physiological response to this ligand in vivo.
  • IGF IR also binds to IGF II, however with slightly lower affinity.
  • the IGF-1 system including IGF-IR, plays an important role during proliferation of (normal and neoplastic) cells.
  • IGF-IR is found on normal human tissues e. g. from placenta, prostate, bladder, kidney, duodenum, small bowel, gallbladder, common bile duct, intrahepatic bile duct, bronchi, tonsil, thymus, breast, sebaceous gl, salivary gland, uterine cervix, and salpinx.
  • IGF IR overexpression promotes the neoplastic transformation of cells and there exists evidence that IGF IR is involved in malignant transformation of cells and is therefore a useful target for the development of therapeutic agents for the treatment of cancer (Adams, T.E., et al, Cell. Mol. Life Sci. 57 (2000) 1050-1093).
  • Antibodies against IGF IR are well-known in the state of the art and investigated for their antitumor effects in vitro and in vivo (Benini, S., et al, Clin. Cancer Res. 7 (2001) 1790-1797; Scotlandi, K., et al, Cancer Gene Ther. 9 (2002) 296-307; Scotlandi, K., et al., Int. J. Cancer 101 (2002) 11-16; Brunetti, A., et al., Biochem. Biophys. Res. Commun. 165 (1989) 212-218; Prigent, S.A., et al, J. Biol. Chem.
  • Antibodies against IGF-IR are also described in a lot of further publications, e.g., Arteaga, C.L., et al, Breast Cancer Res. Treatment 22 (1992) 101-106; and Hailey, J., et al, Mol. Cancer Ther. 1 (2002) 1349-1353.
  • aIR3 the monoclonal antibody against IGF IR called aIR3 is widely used in the investigation of studying IGF IR mediated processes and IGF I mediated diseases such as cancer.
  • Alpha IR 3 was described by Kull, F.C., et al, J. Biol. Chem. 258 (1983) 6561-6566.
  • aIR3 is a murine monoclonal antibody which is known to inhibit IGF I binding to IGF receptor but not IGF II binding to IGF-IR.
  • aIR3 stimulates at high concentrations tumor cell proliferation and IGF-IR phosphorylation (Bergmann, U., et al, Cancer Res. 55 (1995) 2007-2011; Kato, FL, et al, J. Biol. Chem. 268 (1993) 2655-2661).
  • There exist other antibodies e.g., 1H7, Li, S.L., et al, Cancer Immunol. Immunother. 49 (2000) 243-252
  • IGF II binding to IGF IR more potently than IGF I binding.
  • Examples of human antibodies against IGF-IR are described in WO 2002/053596, WO 2004/071529, WO 2005/016967, WO 2006/008639, US 2005/0249730, US 2005/0084906, WO 2005/058967, WO 2006/013472, US 2003/0165502, WO 2005/082415, WO 2005/016970, WO 2003/106621, WO 2004/083248, WO 2003/100008, WO 2004/087756, WO 2005/005635 and WO 2005/094376.
  • WO 2004/087756 describes antibodies binding to IGF-IR and inhibiting the binding of IGF-I and IGF-II to IGF-IR characterized in being of human IgGl isotype, and showing a ratio of inhibition of the binding of IGF-I to IGF-IR to the inhibition of binding of IGF-II to IGF-IR of 1 :3 to 3: 1, and induces cell death of 20% or more cells of a preparation of IGF-IR expressing cells after 24 hours at a concentration of said antibody of 100 nM by ADCC.
  • WO 2005/005635 describes an anti-IGF-lR antibody characterized in comprising a CDRH3 region of SEQ ID NO: 1, a CDRH2 region of SEQ ID NO:2 and a CDRH1 region of SEQ ID NO: 3 and in that the light chain variable domain a CDRL3 region of SEQ ID NO: 4, a CDRL2 region of SEQ ID NO:5 and a CDRLl region of SEQ ID NO: 6 and an antibody characterized in that the heavy chain variable domain comprises SEQ ID NO:7 and the light chain variable domain comprises SEQ ID NO:8.
  • the invention comprises an antibody specifically binding to human IGF-IR, characterized in comprising as heavy chain variable domain CDRH3 region a CDRH3 region of SEQ ID NO: 1 with a mutation selected from the group consisting of E1Q, L2E, L2F, L2H, L2M, L2T, G3K, G3L, G3Q, R4H, R4K, Y6F, Y6H, Y6W, D8A, D8G, D8Q, D8S, L9G, L9I, L9N, L9P, L9Q, L9R, L9T, or L9V.
  • the antibodies according to the invention differ from the antibodies described in WO 2005/005635 by mutations in the heavy chain CDRH3 region.
  • the antibody according to the invention is characterized in that the heavy chain variable domain comprises a CDRH3 region of SEQ ID NO: 1 with a mutation selected from the group consisting of E1Q, L2E, L2F, L2H, L2M, L2T, G3K, G3L, G3Q, R4H, R4K, Y6F, Y6H, Y6W, D8A, D8G, D8Q, D8S, L9G, L9I, L9N, L9P, L9Q, L9R, L9T, or L9V, a CDRH2 region of SEQ ID NO:2 and a CDRH1 region of SEQ ID NO:3.
  • the antibody according to the invention is characterized in that the heavy chain variable domain comprises a CDRH3 region of SEQ ID NO: 1 with a mutation selected from the group consisting of EIQ, L2E, L2F, L2H, L2M, L2T, G3K, G3L, G3Q, R4H, R4K, Y6F, Y6H, Y6W, D8A, D8G, D8Q, D8S, L9G, L9I, L9N, L9P, L9Q, L9R, L9T, or L9V, a CDRH2 region of SEQ ID NO:2 and a CDRH1 region of SEQ ID NO: 3 and the light chain variable domain comprises a CDRL3 region of SEQ ID NO: 4, a CDRL2 region of SEQ ID NO:5 and a CDRL1 region of SEQ ID NO:6.
  • the antibody according to the invention is characterized in that the heavy chain variable domain comprises SEQ ID NO: 7 comprising a mutation selected from the group consisting of EIQ, L2E, L2F, L2H, L2M, L2T, G3K, G3L, G3Q, R4H, R4K, Y6F, Y6H, Y6W, D8A, D8G, D8Q, D8S, L9G, L9I, L9N, L9P, L9Q, L9R, L9T, or L9V.
  • SEQ ID NO: 7 comprising a mutation selected from the group consisting of EIQ, L2E, L2F, L2H, L2M, L2T, G3K, G3L, G3Q, R4H, R4K, Y6F, Y6H, Y6W, D8A, D8G, D8Q, D8S, L9G, L9I, L9N, L9P, L9
  • the antibody according to the invention is characterized in that the heavy chain variable domain comprises SEQ ID NO: 7 comprising a double mutation selected from the group consisting of E99Q EIQ, L100E L2E, LI OOF L2F, L100H L2H, L100M L2M, L100T L2T, G101K G3K, G101L G3L, G101Q G3Q, R102H R4H, R102K R4K, Y104F Y6F, Y104H Y6H, Y104W Y6W, D106A D8A, D106G D8G, D106Q D8Q, D106S D8S, L107G L9G, L107I L9I, L107N L9N, L107P L9P, L107Q L9Q, L107R L9R, L107T L9T, or L107V L9V.
  • SEQ ID NO: 7 comprising a double mutation selected from the group consisting of E99Q EIQ, L100
  • the antibody according to the invention is characterized in that the heavy chain variable domain comprises SEQ ID NO: 7 comprising a mutation selected from the group consisting of EIQ, L2E, L2F, L2H, L2M, L2T, G3K, G3L, G3Q, R4H, R4K, Y6F, Y6H, Y6W, D8A, D8G, D8Q, D8S, L9G, L9I, L9N, L9P, L9Q, L9R, L9T, or L9V and the light chain variable domain comprises SEQ ID NO:8.
  • the heavy chain variable domain comprises SEQ ID NO: 7 comprising a mutation selected from the group consisting of EIQ, L2E, L2F, L2H, L2M, L2T, G3K, G3L, G3Q, R4H, R4K, Y6F, Y6H, Y6W, D8A, D8G, D8Q, D8S
  • the antibody according to the invention is characterized in that the heavy chain variable domain comprises SEQ ID NO: 7 comprising a double mutation selected from the group consisting of E99Q EIQ, L100E L2E, LI OOF L2F, L100H L2H, L100M L2M, L100T L2T, G101K G3K, G101L G3L, G101Q G3Q, R102H R4H, R102K R4K, Y104F Y6F, Y104H Y6H, Y104W Y6W, D106A D8A, D106G D8G, D106Q D8Q, D106S D8S, L107G L9G, L107I L9I, L107N L9N, L107P L9P, L107Q L9Q, L107R L9R, L107T L9T, or LI 07V L9V and the light chain variable domain comprises SEQ ID NO:8.
  • the light chain variable domain comprises SEQ ID NO:8.
  • Heavy chain variable domain SEQ ID NO: 7 and light chain variable domain SEQ ID NO:8 are preferably of human lambda isotype.
  • the antibody according to the invention is characterized by the above mentioned amino acid sequences or amino acid sequence fragments and properties.
  • the antibody according to the invention preferably comprises a Fc part derived from human origin.
  • the antibody according to the invention is of human IgGl or IgG4 isotype.
  • the antibody is of human IgGl class.
  • the antibody is humanized or is a human antibody.
  • the antibody is specifically binding to IGF-IR and inhibiting the binding of IGF-I and IGF -II to IGF-IR.
  • the antibody according to the invention is preferably of IgGl isotype, partially fucosylated, and/or is not activating the IGF-IR signaling even in IGF-IR overexpressing cells at a 200-fold concentration or even 20.000-fold concentration of its IC50 value.
  • the antibodies according to the invention completely inhibit IGF-I mediated signal transduction of IGF-IR in tumor cells.
  • the antibody is preferably a monoclonal antibody and, in addition, a chimeric antibody (human constant chain), a humanized antibody and especially preferably a human antibody.
  • the antibody preferably binds to IGF-IR human (EC 2.7.1.10.1, SwissProt P08069) in competition to antibody 18, which is described in WO 2005/005635 and comprises the heavy chain variable domain of SEQ ID NO: 7 and the light chain variable domain of SEQ ID NO:8.
  • the antibody is preferably further characterized by an affinity of 10 "8 M (KD) or less, preferably of about 10 "9 to 10 "13 M.
  • the antibody shows preferably no detectable concentration dependent inhibition of insulin binding to the insulin receptor.
  • the antibody is preferably of IgGl type.
  • the antibody according to the invention considerably prolongates the time to progression in relevant xenograft tumor models in comparison with vehicle treated animals and reduces tumor growth.
  • the antibody inhibits the binding of IGF I and IGF II to IGF IR in vitro and in vivo, preferably in about an equal manner for IGF-I and IGF-II.
  • the invention further provides methods for the recombinant production of such antibodies.
  • the invention further provides a nucleic acid encoding a heavy chain variable domain characterized in that the heavy chain variable domain comprises SEQ ID NO:7 with a mutation selected from the group consisting of EIQ, L2E, L2F, L2H, L2M, L2T, G3K, G3L, G3Q, R4H, R4K, Y6F, Y6H, Y6W, D8A, D8G, D8Q, D8S, L9G, L9I, L9N, L9P, L9Q, L9R, L9T, or L9V.
  • a mutation selected from the group consisting of EIQ, L2E, L2F, L2H, L2M, L2T, G3K, G3L, G3Q, R4H, R4K, Y6F, Y6H, Y6W, D8A, D8G, D8Q, D8S, L9G, L9I, L9N, L9P
  • the invention further provides a nucleic acid encoding a heavy chain variable domain characterized in that the heavy chain variable domain comprises SEQ ID NO:7 with a double mutation selected from the group consisting of E99Q EIQ, L100E L2E, LI OOF L2F, L100H L2H, L100M L2M, L100T L2T, G101K G3K, G101L G3L, G101Q G3Q, R102H R4H, R102K R4K, Y104F Y6F, Y104H Y6H, Y104W Y6W, D106A D8A, D106G D8G, D106Q D8Q, D106S D8S, L107G L9G, LI 071 L9I, L107N L9N, L107P L9P, L107Q L9Q, L107R L9R, L107T L9T, or LI 07V L9V.
  • a double mutation selected from the group consisting of E99Q EIQ
  • the invention further provides a nucleic acid encoding a heavy chain variable domain characterized in that the heavy chain variable domain comprises SEQ ID NO:7 with a mutation selected from the group consisting of EIQ, L2E, L2F, L2H, L2M, L2T, G3K, G3L, G3Q, R4H, R4K, Y6F, Y6H, Y6W, D8A, D8G, D8Q, D8S, L9G, L9I, L9N, L9P, L9Q, L9R, L9T, or L9V and the light chain variable domain comprises SEQ ID NO:8.
  • the invention further provides a nucleic acid encoding a heavy chain variable domain characterized in that the heavy chain variable domain comprises SEQ ID NO:7 with a double mutation selected from the group consisting of E99Q EIQ, L100E L2E, LI OOF L2F, L100H L2H, L100M L2M, L100T L2T, G101K G3K, G101L G3L, G101Q G3Q, R102H R4H, R102K R4K, Y104F Y6F, Y104H Y6H, Y104W Y6W, D106A D8A, D106G D8G, D106Q D8Q, D106S D8S, L107G L9G, LI 071 L9I, L107N L9N, L107P L9P, L107Q L9Q, L107R L9R, L107T L9T, or LI 07V L9V and the light chain variable domain comprises SEQ ID NO:8.
  • the invention further
  • the invention further comprises an immunoconjugate comprising an antibody according to the invention and a cytotoxic agent.
  • the invention further comprises a pharmaceutical formulation comprising an antibody according to the invention and a pharmaceutically acceptable carrier.
  • the invention further comprises an antibody according to the invention for use as a medicament.
  • the invention further comprises an antibody according to the invention for use in treating tumor diseases.
  • the invention further comprises an antibody according to the invention for use in inhibiting the binding of human IGF I and human IGF-II to human IGF-IR.
  • the invention further comprises an antibody according to the invention for use in the manufacture of a medicament.
  • the invention further comprises an antibody according to the invention for use in the manufacture of a medicament, wherein the medicament is for treatment of a tumor disease.
  • the invention further comprises the use of an according to the invention in the manufacture of a medicament for the treatment of a tumor disease.
  • the invention further comprises a method for inhibiting the binding of human IGF I and human IGF-II to human IGF-IR in an individual comprising administering to the individual an effective amount of the antibody according to the invention.
  • the invention further comprises the use of an antibody according to the invention for the manufacture of a medicament for the treatment of diseases, preferably of tumor diseases.
  • the invention further comprises a method for the manufacture of a medicament for the treatment of diseases, preferably of tumor diseases (especially breast cancer, primary glioma, pancreatic cancers, bladder papilloma, colon adenocarcinoma, melanoma, medulloblastoma, pediatric tumors, fibrosarcoma), characterized in comprising an antibody according to the invention.
  • tumor diseases especially breast cancer, primary glioma, pancreatic cancers, bladder papilloma, colon adenocarcinoma, melanoma, medulloblastoma, pediatric tumors, fibrosarcoma
  • Antibodies according to the invention show benefits for patients in need of antitumor therapy and provide reduction of tumor growth and a significant prolongation of the time to progression.
  • the antibodies according to the invention have new and inventive properties causing a benefit for a patient suffering from a disease associated with an IGF deregulation, especially a tumor disease.
  • the antibodies according to the invention are characterized by the above mentioned properties.
  • the invention further provides methods for treating cancer, preferably breast cancer, pancreatic cancer, prostate cancer, bladder cancer, malignal melanoma, Ewing's sarcoma, Neuroblastoma, Osteosarcoma, Rhabdomyosarcoma, and/or NSCLC, comprising administering to a patient diagnosed as having cancer (and therefore being in need of an antitumor therapy) an antibody against IGF IR according to the invention.
  • the antibody may be administered alone, in a pharmaceutical composition, or alternatively in combination with other inhibitors of cancer related signaling pathways such as EGFR, Her2/neu or estrogen receptor or in combination with a cytotoxic treatment such as radiotherapy or a cytotoxic agent or a prodrug thereof.
  • the antibody is administered in a pharmaceutically effective amount.
  • the invention further comprises the use of an antibody according to the invention for cancer treatment, preferably breast cancer, pancreatic cancer and/or NSCLC, and for the manufacture of a pharmaceutical composition according to the invention.
  • the invention comprises a method for the manufacture of a pharmaceutical composition according to the invention.
  • the invention further comprises an antibody according to the invention for cancer treatment, preferably breast cancer, pancreatic cancer, prostate cancer, bladder cancer, malignal melanoma, Ewing's sarcoma, Neuroblastoma, Osteosarcoma, Rhabdomyosarcoma and/or NSCLC.
  • the invention further comprises a pharmaceutical composition containing an antibody according to the invention, optionally together with a buffer and/or an adjuvant useful for the formulation of antibodies for pharmaceutical purposes.
  • the invention further comprises a pharmaceutical composition comprising an antibody according to the invention.
  • the invention further provides pharmaceutical compositions comprising such antibodies in a pharmaceutically acceptable carrier.
  • the pharmaceutical composition may be included in an article of manufacture or kit.
  • the invention further provides the use of an antibody according to the invention for the manufacture of a pharmaceutical composition for the treatment of cancer.
  • the antibody is used in a pharmaceutically effective amount.
  • the invention further comprises the use of an antibody according to the invention for the manufacture of a pharmaceutical composition for the treatment of cancer, preferably breast cancer, pancreatic cancer and/or NSCLC.
  • the antibody is used in a pharmaceutically effective amount.
  • the invention further comprises a method for the production of a recombinant human antibody according to the invention, characterized by expressing a nucleic acid encoding an antibody binding to IGF-IR in a CHO host cell and recovering said antibody from said cell.
  • the invention further comprises a process for the production of an antibody against IGF-IR according to the invention, comprising the steps of transforming a host cell, preferably a CHO cell.
  • acceptor human framework for the purposes herein is a framework comprising the amino acid sequence of a light chain variable domain (VL) framework or a heavy chain variable domain (VH) framework derived from a human immunoglobulin framework or a human consensus framework, as defined below.
  • An acceptor human framework "derived from” a human immunoglobulin framework or a human consensus framework may comprise the same amino acid sequence thereof, or it may contain amino acid sequence changes. In some embodiments, the number of amino acid changes are 10 or less, 9 or less, 8 or less, 7 or less, 6 or less, 5 or less, 4 or less, 3 or less, or 2 or less.
  • the VL acceptor human framework is identical in sequence to the VL human immunoglobulin framework sequence or human consensus framework sequence.
  • Binding affinity refers to the strength of the sum total of noncovalent interactions between a single binding site of a molecule (e.g., an antibody) and its binding partner (e.g., an antigen). Unless indicated otherwise, as used herein, "binding affinity” refers to intrinsic binding affinity which reflects a 1 : 1 interaction between members of a binding pair (e.g., antibody and antigen).
  • the affinity of a molecule X for its partner Y can generally be represented by the dissociation constant (Kd). Affinity can be measured by common methods known in the art, including those described herein. Specific illustrative and exemplary embodiments for measuring binding affinity are described in the following.
  • anti-IGF-lR antibody and "an antibody that binds to IGF-1R” refer to an antibody that is capable of binding IGF-1R with sufficient affinity such that the antibody is useful as a diagnostic and/or therapeutic agent in targeting IGF-1R.
  • the extent of binding of an anti-IGF-lR antibody to an unrelated, non-IGF-lR protein is less than about 10% of the binding of the antibody to IGF-1R as measured, by surface Plasmon resonance.
  • an antibody that binds to IGF-1R has a dissociation constant (Kd) of 10 "8 M or less, e.g. from 10 "8 M to 10 "13 M, e.g., from 10 "9 M to 10 "13 M).
  • antibody herein is used in the broadest sense and encompasses various antibody structures, including but not limited to monoclonal antibodies, polyclonal antibodies, multispecific antibodies (e.g., bispecific antibodies), and antibody fragments so long as they exhibit the desired antigen-binding activity.
  • antibody fragment refers to a molecule other than an intact antibody that comprises a portion of an intact antibody that binds the antigen to which the intact antibody binds.
  • antibody fragments include but are not limited to Fv, Fab, Fab', Fab'-SH, F(ab') 2 ; diabodies; linear antibodies; single-chain antibody molecules (e.g. scFv); and multispecific antibodies formed from antibody fragments.
  • chimeric antibody refers to an antibody in which a portion of the heavy and/or light chain is derived from a particular source or species, while the remainder of the heavy and/or light chain is derived from a different source or species.
  • the "class” of an antibody refers to the type of constant domain or constant region possessed by its heavy chain. There are five major classes of antibodies: IgA, IgD, IgE, IgG, and IgM, and several of these may be further divided into subclasses (isotypes), e.g., IgGi, IgG 2 , IgG 3 , IgG 4 , IgA ls and IgA 2 .
  • the heavy chain constant domains that correspond to the different classes of immunoglobulins are called a, ⁇ , ⁇ , ⁇ , and ⁇ , respectively.
  • cytotoxic agent refers to a substance that inhibits or prevents a cellular function and/or causes cell death or destruction.
  • 21 1 131 125 agents include, but are not limited to, radioactive isotopes (e.g., At , 1 , 1 , Y 90 , Re 186 , Re 188 , Sm 153 , Bi 212 , P 32 , Pb 212 and radioactive isotopes of Lu); chemotherapeutic agents or drugs (e.g., methotrexate, adriamicin, vinca alkaloids (vincristine, vinblastine, etoposide), doxorubicin, melphalan, mitomycin C, chlorambucil, daunorubicin or other intercalating agents); growth inhibitory agents; enzymes and fragments thereof such as nucleolytic enzymes; antibiotics; toxins such as small molecule toxins or enzymatically active toxins of bacterial, fungal, plant or animal origin, including fragments and/or variants thereof; and the various antitumor or anticancer agents disclosed below.
  • Antibody effector functions refer to those biological activities attributable to the Fc region of an antibody, which vary with the antibody isotype. Examples of antibody effector functions include: Clq binding and complement dependent cytotoxicity (CDC); Fc receptor binding; antibody-dependent cell-mediated cytotoxicity (ADCC); phagocytosis; down regulation of cell surface receptors (e.g. B cell receptor); and B cell activation.
  • an "effective amount" of an agent refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired therapeutic or prophylactic result.
  • Fc region herein is used to define a C-terminal region of an immunoglobulin heavy chain that contains at least a portion of the constant region.
  • the term includes native sequence Fc regions and variant Fc regions.
  • a human IgG heavy chain Fc region extends from Cys226, or from Pro230, to the carboxyl-terminus of the heavy chain.
  • the C-terminal lysine (Lys447) of the Fc region may or may not be present.
  • numbering of amino acid residues in the Fc region or constant region is according to the EU numbering system, also called the EU index, as described in Kabat et al, Sequences of Proteins of Immunological Interest, 5th ed. Public Health Service, NIH Publication 91-3242, National Institutes of Health, Bethesda, MD (1991).
  • FR Framework or "FR” refers to variable domain residues other than hypervariable region (HVR) residues.
  • the FR of a variable domain generally consists of four FR domains: FR1, FR2, FR3, and FR4. Accordingly, the CDR and FR sequences generally appear in the following sequence in VH (or VL): FR1-CDR1(L1)-FR2- CDR2(L2)-FR3-CDR3(L3)-FR4.
  • full length antibody “intact antibody,” and “whole antibody” are used herein interchangeably to refer to an antibody having a structure substantially similar to a native antibody structure or having heavy chains that contain an Fc region as defined herein.
  • host cell refers to cells into which exogenous nucleic acid has been introduced, including the progeny of such cells.
  • Host cells include “transformants” and “transformed cells,” which include the primary transformed cell and progeny derived therefrom without regard to the number of passages. Progeny may not be completely identical in nucleic acid content to a parent cell, but may contain mutations. Mutant progeny that have the same function or biological activity as screened or selected for in the originally transformed cell are included herein.
  • a "human antibody” is one which possesses an amino acid sequence which corresponds to that of an antibody produced by a human or a human cell or derived from a non-human source that utilizes human antibody repertoires or other human antibody-encoding sequences. This definition of a human antibody specifically excludes a humanized antibody comprising non-human antigen-binding residues.
  • a "human consensus framework” is a framework which represents the most commonly occurring amino acid residues in a selection of human immunoglobulin VL or VH framework sequences.
  • the selection of human immunoglobulin VL or VH sequences is from a subgroup of variable domain sequences.
  • the subgroup of sequences is a subgroup as in Kabat et al, Sequences of Proteins of Immunological Interest, 5th ed., Public Health Service, NIH Publication 91-3242, National Institutes of Health, Bethesda, MD (1991).
  • the subgroup is subgroup kappa I as in Kabat et al, supra.
  • the subgroup is subgroup III as in Kabat et al., supra.
  • a “humanized” antibody refers to a chimeric antibody comprising amino acid residues from non-human CDRs and amino acid residues from human FRs.
  • a humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the CDRs correspond to those of a non-human antibody, and all or substantially all of the FRs correspond to those of a human antibody.
  • a humanized antibody optionally may comprise at least a portion of an antibody constant region derived from a human antibody.
  • a "humanized form" of an antibody, e.g., a non-human antibody refers to an antibody that has undergone humanization.
  • CDR complementarity determining regions
  • an “immunoconjugate” is an antibody conjugated to one or more heterologous molecule(s), including but not limited to a cytotoxic agent.
  • mammals include, but are not limited to, domesticated animals (e.g., cows, sheep, cats, dogs, and horses), primates (e.g., humans and non-human primates such as monkeys), rabbits, and rodents (e.g., mice and rats).
  • domesticated animals e.g., cows, sheep, cats, dogs, and horses
  • primates e.g., humans and non-human primates such as monkeys
  • rabbits e.g., mice and rats
  • rodents e.g., mice and rats.
  • the individual or subject is a human.
  • Inhibiting binding of IGF I and II to IGF-1R can be determined using a IGF-1R expressing cell line (HT29) and radioactive labeled IGF-I or IGF-II. The amount of radioactive peptide bound to the cell surface receptors is measured.
  • An antibody according to the invention should inhibit the binding like antibody 18 described in WO 2005/005635 with an IC 50 value of 0.5 nM or lower.
  • an “isolated” antibody is one which has been separated from a component of its natural environment.
  • an antibody is purified to greater than 95% or 99% purity as determined by, for example, electrophoretic (e.g., SDS- PAGE, isoelectric focusing (IEF), capillary electrophoresis) or chromatographic (e.g., ion exchange or reverse phase HPLC).
  • electrophoretic e.g., SDS- PAGE, isoelectric focusing (IEF), capillary electrophoresis
  • chromatographic e.g., ion exchange or reverse phase HPLC
  • nucleic acid refers to a nucleic acid molecule that has been separated from a component of its natural environment.
  • An isolated nucleic acid includes a nucleic acid molecule contained in cells that ordinarily contain the nucleic acid molecule, but the nucleic acid molecule is present extrachromosomally or at a chromosomal location that is different from its natural chromosomal location.
  • isolated nucleic acid encoding an anti-IGF-lR antibody refers to one or more nucleic acid molecules encoding antibody heavy and light chains (or fragments thereof), including such nucleic acid molecule(s) in a single vector or separate vectors, and such nucleic acid molecule(s) present at one or more locations in a host cell.
  • monoclonal antibody refers to an antibody obtained from a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical and/or bind the same epitope, except for possible variant antibodies, e.g., containing naturally occurring mutations or arising during production of a monoclonal antibody preparation, such variants generally being present in minor amounts.
  • polyclonal antibody preparations typically include different antibodies directed against different determinants (epitopes)
  • each monoclonal antibody of a monoclonal antibody preparation is directed against a single determinant on an antigen.
  • the modifier "monoclonal” indicates the character of the antibody as being obtained from a substantially homogeneous population of antibodies, and is not to be construed as requiring production of the antibody by any particular method.
  • the monoclonal antibodies to be used in accordance with the present invention may be made by a variety of techniques, including but not limited to the hybridoma method, recombinant DNA methods, phage-display methods, and methods utilizing transgenic animals containing all or part of the human immunoglobulin loci, such methods and other exemplary methods for making monoclonal antibodies being described herein.
  • mutation refers to one or more amino acid substitutions in a CDR and/or variable region of an antibody according to the invention.
  • EIQ, L2E, L2F, L2H, L2M, L2T, G3K, G3L, G3Q, R4H, R4K, Y6F, Y6H, Y6W, D8A, D8G, D8Q, D8S, L9G, L9I, L9N, L9P, L9Q, L9R, L9T, or L9V relate to substitutions in the heavy chain CDRH3 (SEQ ID NO: l).
  • the numbering 1 to 9 relates therefore to amino acids 1 to 9 of SEQ ID NO: l .
  • EIQ means therefore that the first amino acid of SEQ ID NO: l, which is E (Glu), is replaced by amino acid Q (Gin).
  • E99Q means therefore that amino acid 99 of SEQ ID NO:7, which is E (Glu), is replaced by amino acid Q (Gin).
  • E99Q EIQ describes therefore an antibody according to the invention wherein E99 of SEQ ID NO:7 is replaced by Q and El of SEQ ID NO: l is replaced by Q.
  • naked antibody refers to an antibody that is not conjugated to a heterologous moiety (e.g., a cytotoxic moiety) or radiolabel.
  • the naked antibody may be present in a pharmaceutical formulation.
  • Native antibodies refer to naturally occurring immunoglobulin molecules with varying structures.
  • native IgG antibodies are heterotetrameric glycoproteins of about 150,000 daltons, composed of two identical light chains and two identical heavy chains that are disulfide-bonded. From N- to C-terminus, each heavy chain has a variable region (VH), also called a variable heavy domain or a heavy chain variable domain, followed by three constant domains (CHI, CH2, and CH3).
  • VH variable region
  • VL variable region
  • the light chain of an antibody may be assigned to one of two types, called kappa ( ⁇ ) and lambda ( ⁇ ), based on the amino acid sequence of its constant domain.
  • package insert is used to refer to instructions customarily included in commercial packages of therapeutic products, that contain information about the indications, usage, dosage, administration, combination therapy, contraindications and/or warnings concerning the use of such therapeutic products.
  • Percent (%) amino acid sequence identity with respect to a reference polypeptide sequence is defined as the percentage of amino acid residues in a candidate sequence that are identical with the amino acid residues in the reference polypeptide sequence, after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent sequence identity, and not considering any conservative substitutions as part of the sequence identity. Alignment for purposes of determining percent amino acid sequence identity can be achieved in various ways that are within the skill in the art, for instance, using publicly available computer software such as BLAST, BLAST-2, ALIGN or Megalign (DNASTAR) software. Those skilled in the art can determine appropriate parameters for aligning sequences, including any algorithms needed to achieve maximal alignment over the full length of the sequences being compared.
  • % amino acid sequence identity values are generated using the sequence comparison computer program ALIGN-2.
  • the ALIGN -2 sequence comparison computer program was authored by Genentech, Inc., and the source code has been filed with user documentation in the U.S. Copyright Office, Washington D.C., 20559, where it is registered under U.S. Copyright Registration No. TXU510087.
  • the ALIGN-2 program is publicly available from Genentech, Inc., South San Francisco, California, or may be compiled from the source code.
  • the ALIGN-2 program should be compiled for use on a UNIX operating system, including digital UNIX V4.0D. All sequence comparison parameters are set by the ALIGN-2 program and do not vary.
  • % amino acid sequence identity of a given amino acid sequence A to, with, or against a given amino acid sequence B is calculated as follows:
  • % amino acid sequence identity refers to a preparation which is in such form as to permit the biological activity of an active ingredient contained therein to be effective, and which contains no additional components which are unacceptably toxic to a subject to which the formulation would be administered.
  • a “pharmaceutically acceptable carrier” refers to an ingredient in a pharmaceutical formulation, other than an active ingredient, which is nontoxic to a subject.
  • a pharmaceutically acceptable carrier includes, but is not limited to, a buffer, excipient, stabilizer, or preservative.
  • IGF-IR refers to human insulin- like growth factor I receptor (IGF-IR, EC 2.7.10.1, CD 221 antigen) which belongs to the family of transmembrane protein tyrosine kinases (LeRoith, D., et al, Endocrin. Rev. 16 (1995) 143-163; and Adams, T.E., et al, Cell. Mol. Life Sci. 57 (2000) 1050-1093.
  • the IGF-I receptor consists of an alpha subunit (a 130-135 kD protein that is involved in ligand binding) and a beta subunit (a 95-kD transmembrane protein with kinase activity).
  • treatment refers to clinical intervention in an attempt to alter the natural course of the individual being treated, and can be performed either for prophylaxis or during the course of clinical pathology. Desirable effects of treatment include, but are not limited to, preventing occurrence or recurrence of disease, alleviation of symptoms, diminishment of any direct or indirect pathological consequences of the disease, preventing metastasis, decreasing the rate of disease progression, amelioration or palliation of the disease state, and remission or improved prognosis.
  • antibodies of the invention are used to delay development of a disease or to slow the progression of a disease.
  • variable region refers to the domain of an antibody heavy or light chain that is involved in binding the antibody to antigen.
  • the variable domains of the heavy chain and light chain (VH and VL, respectively) of a native antibody generally have similar structures, with each domain comprising four conserved framework regions (FRs) and three hypervariable regions (HVRs).
  • FRs conserved framework regions
  • HVRs hypervariable regions
  • antibodies that bind a particular antigen may be isolated using a VH or VL domain from an antibody that binds the antigen to screen a library of complementary VL or VH domains, respectively. See, e.g., Portolano, S., et al, J. Immunol. 150 (1993) 880-887; Clackson, T., et al, Nature 352 (1991) 624-628.
  • vector refers to a nucleic acid molecule capable of propagating another nucleic acid to which it is linked.
  • the term includes the vector as a self-replicating nucleic acid structure as well as the vector incorporated into the genome of a host cell into which it has been introduced.
  • Certain vectors are capable of directing the expression of nucleic acids to which they are operatively linked. Such vectors are referred to herein as "expression vectors.”
  • compositions and methods II. Compositions and methods
  • the invention is based on antibodies that bind to IGF-1R.
  • Antibodies of the invention are useful, e.g., for the diagnosis or treatment of tumor diseases.
  • an anti-IGF-lR antibody is a monoclonal antibody, including a chimeric, humanized or human antibody.
  • an anti-IGF-lR antibody is an antibody fragment, e.g., a Fv, Fab, Fab', scFv, diabody, or F(ab') 2 fragment.
  • the antibody is a full length antibody, e.g., an intact IgGl antibody or other antibody class or isotype as defined herein.
  • an anti-IGF-lR antibody may incorporate any of the features, singly or in combination, as described in the sections below:
  • an antibody provided herein has a dissociation constant (Kd) of 10 "8 M or less, e.g. from 10 "8 M to 10 "13 M, e.g., from 10 "9 M to 10 "13 M).
  • Kd is measured using surface plasmon resonance assays using a BIACORE ® -2000 or a BIACORE ® -3000 (BIAcore, Inc., Piscataway, NJ) at 25°C with immobilized antigen CM5 chips at ⁇ 10 response units (RU).
  • CM5 carboxymethylated dextran biosensor chips
  • EDC N-ethyl-N'- (3-dimethylaminopropyl)-carbodiimide hydrochloride
  • NHS N-hydroxysuccinimide
  • Antigen is diluted with 10 mM sodium acetate, pH 4.8, to 5 ⁇ g/ml (-0.2 ⁇ ) before injection at a flow rate of 5 ⁇ /minute to achieve approximately 10 response units (RU) of coupled protein. Following the injection of antigen, 1 M ethanolamine is injected to block unreacted groups. For kinetics measurements, two-fold serial dilutions of Fab (0.78 nM to 500 nM) are injected in PBS with 0.05% polysorbate 20 (TWEEN®-20TM) surfactant (PBST) at 25°C at a flow rate of approximately 25 ⁇ /min.
  • TWEEN®-20TM polysorbate 20
  • Association rates (k on ) and dissociation rates (k 0 ff) are calculated using a simple one-to-one Langmuir binding model (BIACORE Evaluation Software version 3.2) by simultaneously fitting the association and dissociation sensorgrams.
  • the equilibrium dissociation constant (Kd) is calculated as the ratio k off k on See, e.g., Chen, Y., et al, J. Mol. Biol. 293 (1999) 865-881.
  • an antibody provided herein is an antibody fragment.
  • Antibody fragments include, but are not limited to, Fab, Fab', Fab'-SH, F(ab') 2 , Fv, and scFv fragments, and other fragments described below.
  • Fab fragment-specific antibody fragment
  • Fab' fragment-specific Fab
  • Fab'-SH fragment-specific Fab
  • F(ab') 2 fragment-specific Fab
  • Fv fragment antigen V-binding Function
  • scFv fragments include, but are not limited to, Fab, Fab', Fab'-SH, F(ab') 2 , Fv, and scFv fragments, and other fragments described below.
  • scFv fragments see, e.g., Plueckthun, A., in The Pharmacology of Monoclonal Antibodies, vol. 113, Rosenburg and Moore eds., (Springer-Verlag, New York), (1994) pp.
  • Diabodies are antibody fragments with two antigen-binding sites that may be bivalent or bispecific. See, for example, EP 404,097; WO 1993/01161; Hudson, P.J., et al, Nat. Med. 9 (2003) 129-134; and Holliger, P., et al, Proc. Natl. Acad. Sci. USA 90 (1993) 6444-6448. Triabodies and tetrabodies are also described in Hudson, P.J., et al, Nat. Med. 9 (2003) 129-134.
  • Single-domain antibodies are antibody fragments comprising all or a portion of the heavy chain variable domain or all or a portion of the light chain variable domain of an antibody. In certain embodiments, a single-domain antibody is a human single-domain antibody (Domantis, Inc., Waltham, MA; see, e.g., U.S. Patent No. 6,248,516 Bl).
  • Antibody fragments can be made by various techniques, including but not limited to proteolytic digestion of an intact antibody as well as production by recombinant host cells (e.g. E. coli or phage), as described herein.
  • recombinant host cells e.g. E. coli or phage
  • an antibody provided herein is a multispecific antibody, e.g. a bispecific antibody.
  • Multispecific antibodies are monoclonal antibodies that have binding specificities for at least two different sites. In certain embodiments, one of the binding specificities is for IGF-1R and the other is for any other antigen.
  • bispecific antibodies may bind to two different epitopes of IGF-1R. Bispecific antibodies may also be used to localize cytotoxic agents to cells which express IGF-1R. Bispecific antibodies can be prepared as full length antibodies or antibody fragments.
  • Techniques for making multispecific antibodies include, but are not limited to, recombinant co-expression of two immunoglobulin heavy chain-light chain pairs having different specificities (see Milstein, C, and Cuello, A.C., Nature 305 (1983) 537-540), WO 93/08829, and Traunecker, A., et al, EMBO J. 10 (1991) 3655- 3659), and "knob-in-hole” engineering (see, e.g., U.S. Patent No. 5,731,168).
  • Multi-specific antibodies may also be made by engineering electrostatic steering effects for making antibody Fc-heterodimeric molecules (WO 2009/089004A1); cross-linking two or more antibodies or fragments (see, e.g., US Patent No. 4,676,980, and Brennan, M., et al, Science 229 (1985) 81-83); using leucine zippers to produce bi-specific antibodies (see, e.g., Kostelny, S.A., et al, J. Immunol. 148 (1992) 1547-1553); using "diabody” technology for making bispecific antibody fragments (see, e.g., Holliger, P., et al, Proc. Natl. Acad. Sci.
  • the antibody or fragment herein also includes a "Dual Acting FAb” or “DAF” comprising an antigen binding site that binds to IGF-1R as well as another, different antigen (see, US 2008/0069820, for example).
  • an antibody provided herein is altered to increase or decrease the extent to which the antibody is glycosylated.
  • Addition or deletion of glycosylation sites to an antibody may be conveniently accomplished by altering the amino acid sequence such that one or more glycosylation sites is created or removed.
  • the carbohydrate attached thereto may be altered.
  • Native antibodies produced by mammalian cells typically comprise a branched, biantennary oligosaccharide that is generally attached by an N-linkage to Asn297 of the CH2 domain of the Fc region. See, e.g., Wright, A., et al, TIBTECH 15 (1997) 26-32.
  • the oligosaccharide may include various carbohydrates, e.g., mannose, N-acetyl glucosamine (GlcNAc), galactose, and sialic acid, as well as a fucose attached to a GlcNAc in the "stem" of the biantennary oligosaccharide structure.
  • modifications of the oligosaccharide in an antibody of the invention may be made in order to create antibody variants with certain improved properties.
  • antibody variants having a carbohydrate structure that lacks fucose attached (directly or indirectly) to an Fc region.
  • the amount of fucose in such antibody may be from 1% to 80%, from 1% to 65%, from 5%> to 65%o or from 20%> to 40%>.
  • the amount of fucose is determined by calculating the average amount of fucose within the sugar chain at Asn297, relative to the sum of all glycostructures attached to Asn 297 (e. g. complex, hybrid and high mannose structures) as measured by MALDI-TOF mass spectrometry, as described in WO 2008/077546, for example.
  • Asn297 refers to the asparagine residue located at about position 297 in the Fc region (Eu numbering of Fc region residues); however, Asn297 may also be located about ⁇ 3 amino acids upstream or downstream of position 297, i.e., beTween® positions 294 and 300, due to minor sequence variations in antibodies. Such fucosylation variants may have improved ADCC function. See, e.g., US Patent Publication Nos. US 2003/0157108 (Presta, L.); US 2004/0093621 (Kyowa Hakko Kogyo Co., Ltd).
  • Examples of publications related to "defucosylated” or “fucose-deficient” antibody variants include: US 2003/0157108; WO 2000/61739; WO 2001/29246; US 2003/0115614; US 2002/0164328; US 2004/0093621; US 2004/0132140; US 2004/0110704; US 2004/0110282; US 2004/0109865; WO 2003/085119; WO 2003/084570; WO 2005/035586; WO 2005/035778; WO2005/053742; WO2002/031140; Okazaki, A., et al, J. Mol. Biol.
  • Antibodies variants are further provided with bisected oligosaccharides, e.g., in which a biantennary oligosaccharide attached to the Fc region of the antibody is bisected by GlcNAc. Such antibody variants may have reduced fucosylation and/or improved ADCC function. Examples of such antibody variants are described, e.g., in WO 2003/011878 (Jean-Mairet, et al); US Patent No. 6,602,684 (Umana, et al); and US 2005/0123546 (Umana, et al.). Antibody variants with at least one galactose residue in the oligosaccharide attached to the Fc region are also provided.
  • Such antibody variants may have improved CDC function.
  • Such antibody variants are described, e.g., in WO 1997/30087 (Patel, et al); WO 1998/58964 (Raju, S.); and WO 1999/22764 (Raju, S.).
  • one or more amino acid modifications may be introduced into the Fc region of an antibody provided herein, thereby generating an Fc region variant.
  • the Fc region variant may comprise a human Fc region sequence ⁇ e.g., a human IgGl, IgG2, IgG3 or IgG4 Fc region) comprising an amino acid modification ⁇ e.g. a substitution) at one or more amino acid positions.
  • the invention contemplates an antibody variant that possesses some but not all effector functions, which make it a desirable candidate for applications in which the half life of the antibody in vivo is important yet certain effector functions (such as complement and ADCC) are unnecessary or deleterious.
  • Fc receptor (FcR) binding assays can be conducted to ensure that the antibody lacks FcyR binding (hence likely lacking ADCC activity), but retains FcRn binding ability.
  • FcR expression on hematopoietic cells is summarized in Table 3 on page 464 of Ravetch, J.V., and Kinet, Annu. Rev. Immunol. 9 (1991) 457-492.
  • Non-limiting examples of in vitro assays to assess ADCC activity of a molecule of interest is described in U.S. Patent No. 5,500,362 (see, e.g. Hellstrom, I., et al, Proc. Nat'l Acad. Sci. USA 83 (1986) 7059-7063) and Hellstrom, I., et al, Proc. Nat'l Acad. Sci. USA 82 (1985) 1499- 1502; US 5,821,337 (see Bruggemann, M., et al, J. Exp. Med. 166 (1987) 1351- 1361).
  • non-radioactive assays methods may be employed (see, for example, ACTITM non-radioactive cytotoxicity assay for flow cytometry (CellTechnology, Inc. Mountain View, CA; and CytoTox 96® non-radioactive cytotoxicity assay (Promega, Madison, WI).
  • Useful effector cells for such assays include peripheral blood mononuclear cells (PBMC) and Natural Killer (NK) cells.
  • PBMC peripheral blood mononuclear cells
  • NK Natural Killer
  • ADCC activity of the molecule of interest may be assessed in vivo, e.g., in a animal model such as that disclosed in Clynes, R., et al, Proc. Natl. Acad. Sci. USA 95 (1998) 652-656.
  • Clq binding assays may also be carried out to confirm that the antibody is unable to bind Clq and hence lacks CDC activity. See, e.g., Clq and C3c binding ELISA in WO 2006/029879 and WO 2005/100402.
  • a CDC assay may be performed (see, for example, Gazzano-Santoro, H., et al., J. Immunol. Methods 202 (1997) 163ff; Cragg, M.S., et al, Blood 101 (2003) 1045-1052; and Cragg, M.S. and Glenni, M.J., Blood 103 (2004) 2738-2743).
  • FcRn binding and in vivo clearance/half life determinations can also be performed using methods known in the art (see, e.g., Petkova, S.B., et al, Int'l. Immunol. 18 (2006) 1759-1769).
  • Antibodies with reduced effector function include those with substitution of one or more of Fc region residues 238, 265, 269, 270, 297, 327 and 329 (U.S. Patent No. 6,737,056).
  • Fc mutants include Fc mutants with substitutions at two or more of amino acid positions 265, 269, 270, 297 and 327, including the so-called "DANA" Fc mutant with substitution of residues 265 and 297 to alanine (US Patent No. 7,332,581).
  • an antibody variant comprises an Fc region with one or more amino acid substitutions which improve ADCC, e.g., substitutions at positions 298, 333, and/or 334 of the Fc region (EU numbering of residues).
  • alterations are made in the Fc region that result in altered (i.e., either improved or diminished) Clq binding and/or Complement Dependent Cytotoxicity (CDC), e.g., as described in US Patent No. 6,194,551, WO 99/51642, and Idusogie, E.E., et al, J. Immunol. 164 (2000) 4178-4184.
  • CDC Complement Dependent Cytotoxicity
  • Antibodies with increased half lives and improved binding to the neonatal Fc receptor (FcRn), which is responsible for the transfer of maternal IgGs to the fetus are described in US 2005/0014934A1 (Hinton et al.). Those antibodies comprise an Fc region with one or more substitutions therein which improve binding of the Fc region to FcRn.
  • Such Fc variants include those with substitutions at one or more of Fc region residues: 238, 256, 265, 272, 286, 303, 305, 307, 311, 312, 317, 340, 356, 360, 362, 376, 378, 380, 382, 413, 424 or 434, e.g., substitution of Fc region residue 434 (US Patent No. 7,371,826).
  • cysteine engineered antibodies e.g., "thioMAbs”
  • one or more residues of an antibody are substituted with cysteine residues.
  • the substituted residues occur at accessible sites of the antibody.
  • reactive thiol groups are thereby positioned at accessible sites of the antibody and may be used to conjugate the antibody to other moieties, such as drug moieties or linker-drug moieties, to create an immunoconjugate, as described further herein.
  • any one or more of the following residues may be substituted with cysteine: V205 (Kabat numbering) of the light chain; Al 18 (EU numbering) of the heavy chain; and S400 (EU numbering) of the heavy chain Fc region.
  • Cysteine engineered antibodies may be generated as described, e.g., in U.S. Patent No. 7,521,541. d) Antibody Derivatives
  • an antibody provided herein may be further modified to contain additional nonproteinaceous moieties that are known in the art and readily available.
  • the moieties suitable for derivatization of the antibody include but are not limited to water soluble polymers.
  • water soluble polymers include, but are not limited to, polyethylene glycol (PEG), copolymers of ethylene glycol/propylene glycol, carboxymethylcellulose, dextran, polyvinyl alcohol, polyvinyl pyrrolidone, poly-1, 3-dioxolane, poly-l,3,6-trioxane, ethylene/maleic anhydride copolymer, polyaminoacids (either homopolymers or random copolymers), and dextran or poly(n-vinyl pyrrolidone)poly ethylene glycol, propropylene glycol homopolymers, prolypropylene oxide/ethylene oxide co-polymers, polyoxyethylated polyols (e.g.,
  • Polyethylene glycol propionaldehyde may have advantages in manufacturing due to its stability in water.
  • the polymer may be of any molecular weight, and may be branched or unbranched.
  • the number of polymers attached to the antibody may vary, and if more than one polymer are attached, they can be the same or different molecules. In general, the number and/or type of polymers used for derivatization can be determined based on considerations including, but not limited to, the particular properties or functions of the antibody to be improved, whether the antibody derivative will be used in a therapy under defined conditions, etc.
  • conjugates of an antibody and nonproteinaceous moiety that may be selectively heated by exposure to radiation are provided.
  • the nonproteinaceous moiety is a carbon nanotube (Kam, N.W., et al, Proc. Natl. Acad. Sci. USA 102 (2005) 11600-11605).
  • the radiation may be of any wavelength, and includes, but is not limited to, wavelengths that do not harm ordinary cells, but which heat the nonproteinaceous moiety to a temperature at which cells proximal to the antibody-nonproteinaceous moiety are killed.
  • Antibodies may be produced using recombinant methods and compositions, e.g., as described in U.S. Patent No. 4,816,567.
  • isolated nucleic acid encoding an anti-IGF-lR antibody described herein is provided.
  • Such nucleic acid may encode an amino acid sequence comprising the VL and/or an amino acid sequence comprising the VH of the antibody (e.g., the light and/or heavy chains of the antibody).
  • one or more vectors e.g., expression vectors
  • a host cell comprising such nucleic acid is provided.
  • a host cell comprises (e.g., has been transformed with): (1) a vector comprising a nucleic acid that encodes an amino acid sequence comprising the VL of the antibody and an amino acid sequence comprising the VH of the antibody, or (2) a first vector comprising a nucleic acid that encodes an amino acid sequence comprising the VL of the antibody and a second vector comprising a nucleic acid that encodes an amino acid sequence comprising the VH of the antibody.
  • the host cell is eukaryotic, e.g. a Chinese Hamster Ovary (CHO) cell or lymphoid cell (e.g., Y0, NSO, Sp20 cell).
  • a method of making an anti-IGF-lR antibody comprises culturing a host cell comprising a nucleic acid encoding the antibody, as provided above, under conditions suitable for expression of the antibody, and optionally recovering the antibody from the host cell (or host cell culture medium).
  • nucleic acid encoding an antibody is isolated and inserted into one or more vectors for further cloning and/or expression in a host cell.
  • nucleic acid may be readily isolated and sequenced using conventional procedures (e.g., by using oligonucleotide probes that are capable of binding specifically to genes encoding the heavy and light chains of the antibody).
  • Suitable host cells for cloning or expression of antibody-encoding vectors include prokaryotic or eukaryotic cells described herein.
  • antibodies may be produced in bacteria, in particular when glycosylation and Fc effector function are not needed.
  • U.S. Patent Nos. 5,648,237, 5,789,199, and 5,840,523. See also Charlton, K.A., Methods in Molecular Biology, Vol. 248 (B.K.C. Lo, ed., Humana Press, Totowa, NJ), (2004) pp. 245-254, describing expression of antibody fragments in E. coli.
  • the antibody may be isolated from the bacterial cell paste in a soluble fraction and can be further purified.
  • eukaryotic microbes such as filamentous fungi or yeast are suitable cloning or expression hosts for antibody-encoding vectors, including fungi and yeast strains whose glycosylation pathways have been "humanized,” resulting in the production of an antibody with a partially or fully human glycosylation pattern. See Gerngross, T.U., Nat. Biotech. 22 (2004) 1409-1414, and Li, H., et al, Nat. Biotech. 24 (2006) 210-215.
  • Suitable host cells for the expression of glycosylated antibody are also derived from multicellular organisms (invertebrates and vertebrates). Examples of invertebrate cells include plant and insect cells. Numerous baculoviral strains have been identified which may be used in conjunction with insect cells, particularly for transfection of Spodoptera frugiperda cells.
  • Plant cell cultures can also be utilized as hosts. See, e.g., US Patent Nos. 5,959,177, 6,040,498, 6,420,548, 7,125,978, and 6,417,429 (describing PLANTIBODIESTM technology for producing antibodies in transgenic plants).
  • Vertebrate cells may also be used as hosts.
  • mammalian cell lines that are adapted to grow in suspension may be useful.
  • Other examples of useful mammalian host cell lines are monkey kidney CV1 line transformed by SV40 (COS-7); human embryonic kidney line (293 or 293 cells as described, e.g., in Graham, F.L., et al, J. Gen Virol. 36 (1977) 59-74); baby hamster kidney cells (BHK); mouse Sertoli cells (TM4 cells as described, e.g., in Mather, J.P., Biol. Reprod.
  • monkey kidney cells (CV1); African green monkey kidney cells (VERO-76); human cervical carcinoma cells (HELA); canine kidney cells (MDCK; buffalo rat liver cells (BRL 3A); human lung cells (W138); human liver cells (Hep G2); mouse mammary tumor (MMT 060562); TRI cells, as described, e.g., in Mather, J.P., et al, Annals N.Y. Acad. Sci. 383 (1982) 44-68; MRC 5 cells; and FS4 cells.
  • Other useful mammalian host cell lines include Chinese hamster ovary (CHO) cells, including DHFR- CHO cells (Urlaub et al, Proc. Natl. Acad. Sci.
  • the invention also provides immunoconjugates comprising an anti-IGF-lR antibody herein conjugated to one or more cytotoxic agents, such as chemotherapeutic agents or drugs, growth inhibitory agents, toxins (e.g., protein toxins, enzymatically active toxins of bacterial, fungal, plant, or animal origin, or fragments thereof), or radioactive isotopes.
  • cytotoxic agents such as chemotherapeutic agents or drugs, growth inhibitory agents, toxins (e.g., protein toxins, enzymatically active toxins of bacterial, fungal, plant, or animal origin, or fragments thereof), or radioactive isotopes.
  • an immunoconjugate is an antibody-drug conjugate (ADC) in which an antibody is conjugated to one or more drugs, including but not limited to a maytansinoid (see U.S. Patent Nos. 5,208,020, 5,416,064 and European Patent EP 0 425 235 Bl); an auristatin such as monomethylauristatin drug moieties DE and DF (MMAE and MMAF) (see U.S. Patent Nos. 5,635,483 and 5,780,588, and 7,498,298); a dolastatin; a calicheamicin or derivative thereof (see U.S. Patent Nos.
  • ADC antibody-drug conjugate
  • drugs including but not limited to a maytansinoid (see U.S. Patent Nos. 5,208,020, 5,416,064 and European Patent EP 0 425 235 Bl); an auristatin such as monomethylauristatin drug moieties DE and DF (MMAE and MMAF) (
  • an immunoconjugate comprises an antibody as described herein conjugated to an enzymatically active toxin or fragment thereof, including but not limited to 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, PAPII, and PAP-S), momordica charantia inhibitor, curcin, crotin, sapaonaria officinalis inhibitor, gelonin, mitogellin, restrictocin, phenomycin, enomycin, and the tricothecenes.
  • an enzymatically active toxin or fragment thereof including but not limited to diphtheria A chain, nonbinding active fragments of diphtheria toxin, exotoxin A chain (
  • an immunoconjugate comprises an antibody as described herein conjugated to a radioactive atom to form a radioconjugate.
  • a variety of radioactive isotopes are available for the production of radioconjugates. Examples include At 211 , I 131 , I 125 , Y 90 , Re 186 , Re 188 , Sm 153 , Bi 212 , P 32 , Pb 212 and radioactive isotopes of Lu.
  • the radioconjugate When used for detection, it may comprise a radioactive atom for scintigraphic studies, for example tc99m or 1123, or a spin label for nuclear magnetic resonance (NMR) imaging (also known as magnetic resonance imaging, mri), such as iodine- 123 again, iodine-131, indium-I l l, fluorine- 19, carbon- 13, nitrogen- 15, oxygen- 17, gadolinium, manganese or iron.
  • NMR nuclear magnetic resonance
  • Conjugates of an antibody and cytotoxic agent may be made using a variety of bifunctional protein coupling agents such as N-succinimidyl-3-(2-pyridyldithio) propionate (SPDP), succinimidyl-4-(N-maleimidomethyl) cyclohexane-1- carboxylate (SMCC), iminothiolane (IT), bifunctional derivatives of imidoesters (such as dimethyl adipimidate HC1), active esters (such as disuccinimidyl suberate), aldehydes (such as glutaraldehyde), bis-azido compounds (such as bis (p-azidobenzoyl) hexanediamine), bis-diazonium derivatives (such as bis- (p-diazoniumbenzoyl)-ethylenediamine), diisocyanates (such as toluene 2,6- diisocyanate), and bis-active fluorine compounds (such as l
  • a ricin immunotoxin can be prepared as described in Vitella, E.S., et al, Science 238 (1987) 1098-1104.
  • Carbon- 14-labeled 1- isothiocyanatobenzyl-3-methyldiethylene triaminepentaacetic acid is an exemplary chelating agent for conjugation of radionucleotide to the antibody. See WO 94/11026.
  • the linker may be a "cleavable linker" facilitating release of a cytotoxic drug in the cell.
  • an acid-labile linker for example, an acid-labile linker, peptidase-sensitive linker, photolabile linker, dimethyl linker or disulfide-containing linker (Chari, R.V., et al, Cancer Res. 52 (1992) 127-131; U.S. Patent No. 5,208,020) may be used.
  • the immunuoconjugates or ADCs herein expressly contemplate, but are not limited to such conjugates prepared with cross-linker reagents including, but not limited to, BMPS, EMCS, GMBS, HBVS, LC-SMCC, MBS, MPBH, SBAP, SIA, SIAB, SMCC, SMPB, SMPH, sulfo-EMCS, sulfo-GMBS, sulfo-KMUS, sulfo-MBS, sulfo-SIAB, sulfo-SMCC, and sulfo-SMPB, and SVSB (succinimidyl- (4-vinylsulfone)benzoate) which are commercially available (e.g., from Pierce Biotechnology, Inc., Rockford, IL., U.S.A).
  • cross-linker reagents including, but not limited to, BMPS, EMCS, GMBS, HBVS, LC-
  • any of the anti-IGF-lR antibodies provided herein is useful for detecting the presence of IGF-1R in a biological sample.
  • the term "detecting" as used herein encompasses quantitative or qualitative detection.
  • a biological sample comprises a cell or tissue, such as tumor tissue.
  • an anti-IGF-lR antibody for use in a method of diagnosis or detection is provided.
  • a method of detecting the presence of IGF-1R in a biological sample comprises contacting the biological sample with an anti-IGF-lR antibody as described herein under conditions permissive for binding of the anti-IGF-lR antibody to IGF-1R, and detecting whether a complex is formed beTween® the anti-IGF-lR antibody and IGF-1R.
  • Such method may be an in vitro or in vivo method.
  • an anti-IGF-lR antibody is used to select subjects eligible for therapy with an anti-IGF-lR antibody, e.g. where IGF-1R is a biomarker for selection of patients.
  • Exemplary disorders that may be diagnosed using an antibody of the invention is tumor tissue.
  • labeled anti-IGF-lR antibodies include, but are not limited to, labels or moieties that are detected directly (such as fluorescent, chromophoric, electron-dense, chemiluminescent, and radioactive labels), as well as moieties, such as enzymes or ligands, that are detected indirectly, e.g., through an enzymatic reaction or molecular interaction.
  • labels or moieties that are detected directly such as fluorescent, chromophoric, electron-dense, chemiluminescent, and radioactive labels
  • moieties such as enzymes or ligands
  • 32 14 125 3 131 include, but are not limited to, the radioisotopes P, C, I, H, and I, fluorophores such as rare earth chelates or fluorescein and its derivatives, rhodamine and its derivatives, dansyl, umbelliferone, luceriferases, e.g., firefly luciferase and bacterial luciferase (U.S. Patent No.
  • luciferin 2,3-dihydrophthalazinediones
  • horseradish peroxidase HRP
  • alkaline phosphatase alkaline phosphatase
  • ⁇ -galactosidase glucoamylase
  • lysozyme saccharide oxidases, e.g., glucose oxidase, galactose oxidase, and glucose-6-phosphate dehydrogenase
  • heterocyclic oxidases such as uricase and xanthine oxidase, coupled with an enzyme that employs hydrogen peroxide to oxidize a dye precursor such as HRP, lactoperoxidase, or microperoxidase, biotin/avidin, spin labels, bacteriophage labels, stable free radicals, and the like.
  • compositions of an anti-IGF-lR antibody as described herein are prepared by mixing such antibody having the desired degree of purity with one or more optional pharmaceutically acceptable carriers (Osol, A., et al., (eds.) Remington's Pharmaceutical Sciences, 16th edition, Mack Publishing Company, Pennsylvania (1980)), in the form of lyophilized formulations or aqueous solutions.
  • Osol A., et al., (eds.) Remington's Pharmaceutical Sciences, 16th edition, Mack Publishing Company, Pennsylvania (1980)
  • Pharmaceutically acceptable carriers are generally nontoxic to recipients at the dosages and concentrations employed, and include, but are not limited to: buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyldimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride; benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); low molecular weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, histidine, arg
  • sHASEGP soluble neutral-active hyaluronidase glycoproteins
  • rHuPH20 HYLENEX ® , Baxter International, Inc.
  • Certain exemplary sHASEGPs and methods of use, including rHuPH20, are described in US Patent Publication Nos. 2005/0260186 and 2006/0104968.
  • a sHASEGP is combined with one or more additional glycosaminoglycanases such as chondroitinases.
  • Exemplary lyophilized antibody formulations are described in US Patent No. 6,267,958.
  • Aqueous antibody formulations include those described in US Patent No. 6,171,586 and WO 2006/044908, the latter formulations including a histidine- acetate buffer.
  • the formulation herein may also contain more than one active ingredients as necessary for the particular indication being treated, preferably those with complementary activities that do not adversely affect each other.
  • active ingredients are suitably present in combination in amounts that are effective for the purpose intended.
  • Active ingredients may be entrapped in microcapsules prepared, for example, by coacervation techniques or by interfacial polymerization, for example, hydroxymethylcellulose or gelatin-microcapsules and poly-(methylmethacylate) microcapsules, respectively, in colloidal drug delivery systems (for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules) or in macroemulsions.
  • colloidal drug delivery systems for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules
  • Sustained-release preparations may be prepared. Suitable examples of sustained- release preparations include semipermeable matrices of solid hydrophobic polymers containing the antibody, which matrices are in the form of shaped articles, e.g. films, or microcapsules.
  • the formulations to be used for in vivo administration are generally sterile. Sterility may be readily accomplished, e.g., by filtration through sterile filtration membranes.
  • anti-IGF-lR antibodies Any of the anti-IGF-lR antibodies provided herein may be used in therapeutic methods.
  • an anti-IGF-lR antibody for use as a medicament is provided.
  • an anti-IGF-lR antibody for use in treating tumor disease is provided.
  • an anti-IGF-lR antibody for use in a method of treatment is provided.
  • the invention provides an anti-IGF- 1R antibody for use in a method of treating an individual having tumor disease comprising administering to the individual an effective amount of the anti-IGF-lR antibody.
  • the method further comprises administering to the individual an effective amount of at least one additional therapeutic agent, e.g., as described below.
  • the invention provides an anti-IGF-lR antibody for use in inhibiting cell proliferation.
  • the invention provides an anti-IGF-lR antibody for use in a method inhibiting cell proliferationin an individual comprising administering to the individual an effective of the anti-IGF-lR antibody.
  • An "individual" according to any of the above embodiments is preferably a human.
  • the invention provides for the use of an anti-IGF-lR antibody in the manufacture or preparation of a medicament.
  • the medicament is for treatment of tumor disease.
  • the medicament is for use in a method of treating tumor disease comprising administering to an individual having tumor disease an effective amount of the medicament.
  • the method further comprises administering to the individual an effective amount of at least one additional therapeutic agent, e.g., as described below.
  • the medicament is for inhibiting cell proliferation.
  • the medicament is for use in a method of inhibiting cell proliferation in an individual comprising administering to the individual an amount effective of themedicament.
  • An "individual" according to any of the above embodiments may be a human.
  • the invention provides a method for treating a tumor disease.
  • the method comprises administering to an individual having such tumor disease an effective amount of an anti-IGF-lR antibody.
  • the method further comprises administering to the individual an effective amount of at least one additional therapeutic agent, as described below.
  • An "individual" according to any of the above embodiments may be a human.
  • the invention provides a method for inhibiting cell proliferation in an individual.
  • the method comprises administering to the individual an effective amount of an anti-IGF-lR antibody.
  • an "individual" is a human.
  • the invention provides pharmaceutical formulations comprising any of the anti-IGF-lR antibodies provided herein, e.g., for use in any of the above therapeutic methods.
  • a pharmaceutical formulation comprises any of the anti-IGF-lR antibodies provided herein and a pharmaceutically acceptable carrier.
  • a pharmaceutical formulation comprises any of the anti-IGF-lR antibodies provided herein and at least one additional therapeutic agent, e.g., as described below.
  • Antibodies of the invention can be used either alone or in combination with other agents in a therapy.
  • an antibody of the invention may be co-administered with at least one additional therapeutic agent.
  • an additional therapeutic agent is gemcitabine.
  • Such combination therapies noted above encompass combined administration (where two or more therapeutic agents are included in the same or separate formulations), and separate administration, in which case, administration of the antibody of the invention can occur prior to, simultaneously, and/or following, administration of the additional therapeutic agent and/or adjuvant.
  • Antibodies of the invention can also be used in combination with radiation therapy.
  • An antibody of the invention can be administered by any suitable means, including parenteral, intrapulmonary, and intranasal, and, if desired for local treatment, intralesional administration.
  • Parenteral infusions include intramuscular, intravenous, intraarterial, intraperitoneal, or subcutaneous administration. Dosing can be by any suitable route, e.g. by injections, such as intravenous or subcutaneous injections, depending in part on whether the administration is brief or chronic.
  • Various dosing schedules including but not limited to single or multiple administrations over various time -points, bolus administration, and pulse infusion are contemplated herein.
  • Antibodies of the invention would be formulated, dosed, and administered in a fashion consistent with good medical practice. Factors for consideration in this context include the particular disorder being treated, the particular mammal being treated, the clinical condition of the individual patient, the cause of the disorder, the site of delivery of the agent, the method of administration, the scheduling of administration, and other factors known to medical practitioners.
  • the antibody need not be, but is optionally formulated with one or more agents currently used to prevent or treat the disorder in question. The effective amount of such other agents depends on the amount of antibody present in the formulation, the type of disorder or treatment, and other factors discussed above. These are generally used in the same dosages and with administration routes as described herein, or about from 1 to 99% of the dosages described herein, or in any dosage and by any route that is empirically/clinically determined to be appropriate.
  • an antibody of the invention when used alone or in combination with one or more other additional therapeutic agents, will depend on the type of disease to be treated, the type of antibody, the severity and course of the disease, whether the antibody is administered for preventive or therapeutic purposes, previous therapy, the patient's clinical history and response to the antibody, and the discretion of the attending physician.
  • the antibody is suitably administered to the patient at one time or over a series of treatments.
  • about 1 ⁇ g/kg to 15 mg/kg (e.g. O.lmg/kg-lOmg/kg) of antibody can be an initial candidate dosage for administration to the patient, whether, for example, by one or more separate administrations, or by continuous infusion.
  • One typical daily dosage might range from about 1 ⁇ g/kg to 100 mg/kg or more, depending on the factors mentioned above. For repeated administrations over several days or longer, depending on the condition, the treatment would generally be sustained until a desired suppression of disease symptoms occurs.
  • One exemplary dosage of the antibody would be in the range from about 0.05 mg/kg to about 10 mg/kg.
  • one or more doses of about 0.5 mg/kg, 2.0 mg/kg, 4.0 mg/kg or 10 mg/kg (or any combination thereof) may be administered to the patient.
  • Such doses may be administered intermittently, e.g. every week or every three weeks (e.g. such that the patient receives from about two to about twenty, or e.g. about six doses of the antibody).
  • An initial higher loading dose, followed by one or more lower doses may be administered.
  • An exemplary dosing regimen comprises administering about 4 to 10 mg/kg. However, other dosage regimens may be useful. The progress of this therapy is easily monitored by conventional techniques and assays.
  • an article of manufacture containing materials useful for the treatment, prevention and/or diagnosis of the disorders described above comprises a container and a label or package insert on or associated with the container.
  • Suitable containers include, for example, bottles, vials, syringes, IV solution bags, etc.
  • the containers may be formed from a variety of materials such as glass or plastic.
  • the container holds a composition which is by itself or combined with another composition effective for treating, preventing and/or diagnosing the condition and may have a sterile access port (for example the container may be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle).
  • At least one active agent in the composition is an antibody of the invention.
  • the label or package insert indicates that the composition is used for treating the condition of choice.
  • the article of manufacture may comprise (a) a first container with a composition contained therein, wherein the composition comprises an antibody of the invention; and (b) a second container with a composition contained therein, wherein the composition comprises a further cytotoxic or otherwise therapeutic agent.
  • the article of manufacture in this embodiment of the invention may further comprise a package insert indicating that the compositions can be used to treat a particular condition.
  • the article of manufacture may further comprise a second (or third) container comprising a pharmaceutically- acceptable buffer, such as bacteriostatic water for injection (BWFI), phosphate- buffered saline, Ringer's solution and dextrose solution. It may further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, needles, and syringes.
  • BWFI bacteriostatic water for injection
  • phosphate- buffered saline such as bacterio
  • any of the above articles of manufacture may include an immunoconjugate of the invention in place of or in addition to an anti-IGF-lR antibody.
  • Single amino acid substitutions in the VH CDRH3 region of mAb IR18 (9 positions: ELGRRYFDL) were generated based on a plasmid encoding the wild- type immunoglobulin heavy chain of mAb IR18. To this end, individual DNA fragments encoding the altered CDRH3 regions for the respective single amino acid substitutions were generated synthetically and cloned into the corresponding position of the plasmid.
  • Plasmid-DNA was prepared from all clones and co-transfected into HEK293 cells together with a plasmid encoding the wild-type light chain of mAb IR18.
  • lxl 0 6 HEK293 cells were co-transfected with plasmids encoding i) a mutant heavy chain and ii) the wild-type light chain gene.
  • Transfection was performed in 24-well plates in a culture volume of 1 ml using the 293-fection reagent. After seven days of culture the supernatants were harvested, frozen, and stored at -80°C.
  • the antibodies were purified using Protein G sepharose affinity chromatography, and the antibody concentrations were determined using an ELISA assay specific for human immunoglobulin. Subsequently, all supernatants containing human immunoglobulin were analysed for their binding affinity to the human IGFl-R using the Biacore technology.
  • the kinetic constants of mAb IR18 and its CDRH3 variants binding IGFl-R were measured via BiaCore.
  • the antibodies were captured to a CI -Chip via an amine-coupled capture antibody (goat-anti-human IgG).
  • Ligand-loading of the capture antibody was ca. 200 RU (-1.3 fmol/mm 2 ).
  • the concentration of the mAbs was 1 nM in phosphate-buffered saline (PBS) containing 0.05% Tween® 20 and 0.1%) (w/v) bovine serum albumin (BSA). Capturing was performed for 40 seconds.
  • PBS phosphate-buffered saline
  • BSA bovine serum albumin
  • Samples were diluted in PBS containing 0.05% Tween® 20 and 0.1% BSA (w/v), running buffer was PBS containing 0.05% Tween® 20.
  • a classical concentration series was measured (120 seconds association, 10 minutes dissociation, 6 concentrations). The flow rate was 30 ⁇ / ⁇ .
  • the chip was regenerated using 10 mM glycine pH 1.5 (2 minutes injection time) after which the intact, free capture antibody was available again.
  • Human tumor cells (HT29, NCI H322M, 0.5 to 1 x 10 5 /ml) are plated in RPMI 1640 medium (PAA, Cat. No. El 5-039) supplemented with 2 mM L-Glutamin, lx non-essential amino acids (Gibco, Cat. No. 11140-035), 1 mM sodium pyruvate (Gibco, Cat. No. 11360-039) and 10% heat inactivated FCS (PAA, Cat. No. A15- 771).
  • Six bottles in the T175 format are inoculated with 20 ml cells in the respective medium for each experiment and cultivated for two days at 37°C and 5% C0 2 to obtain confluent cell monolayers.
  • Cells are reisolated by centrifugation for 10 minutes at 1000 rpm (Heraeus sepatech, Omnifuge 2.0 RS) and resuspended in 50 ml of binding buffer (120 mM NaCl, 5 mM KC1, 1.2 mM MgS0 4 , 1 mM EDTA, 10 mM D(+)glucose, 15 mM NaAc, 100 mM Hepes pH 7.6, 1%) BSA). Cells are counted, reisolated by centrifugation and adjusted with binding buffer to 1 x 10 6 cells/ml.
  • binding buffer 120 mM NaCl, 5 mM KC1, 1.2 mM MgS0 4 , 1 mM EDTA, 10 mM D(+)glucose, 15 mM NaAc, 100 mM Hepes pH 7.6, 1%) BSA.
  • 1125-labeled IGF-I and IGF-II peptides are diluted in binding buffer to a final activity of 4 x 10 5 counts/(minute x ml).
  • 75 ⁇ of antibody at the specified concentrations together with 25 ⁇ of prediluted 1125-labeled IGF-I or IGF-II peptide is added to 200 ⁇ of cell suspension and incubated for 3,5 h at 4°C. Cells are reisolated by centrifugation for 5 minutes at 2000 rpm (Eppendorf, 5415C) and supernatant removed.
  • IGF-IR IGF-I receptor
  • Human tumor cells (HT29, NCI H322M, 5 x 10 4 /ml) are plated in RPMI 1640 medium (PAA, Cat. No. E15-039) supplemented with 2 mM L-Glutamin, lx nonessential aminoacids (Gibco, Cat. No. 11140-035), 1 mM sodium pyruvate (Gibco, Cat. No. 11360-039) and 0.5% heat inactivated FCS (PAA, Cat. No. A15-771).
  • IC50 values 12 well plates are inoculated with 1 ml cells in the respective medium for each experiment and cultivated for two days at 37°C and 5% C0 2 .
  • the cells are detached using a cell scraper (Corning, Cat. No. 3010) and well contents transferred to Eppendorf reaction tubes. Cell fragments are removed by centrifugation for 10 minutes at 13000 rpm and 4°C and half of the supernatant is added to 2x Laemmli sample buffer in a 1 : 1 (v/v) ratio.
  • IGF-IR immunoprecipitation of IGF-IR
  • the remaining supernatant of cell lysates underwent a clearifying spin (10 minutes at 13000 rpm and 4°C) right before 1 ⁇ of an polyclonal antibody against IGF-IRB (C-20, Santa Cruz Biotechnologies) or a murine monoclonal antibody (IgGl) which recognizes an epitope within amino acids 440-586 of the extracellular domain (a-chain) of the human IGF Type 1 Receptor is added (mAb 24-55, GroPep).
  • IGF-IRB C-20, Santa Cruz Biotechnologies
  • IgGl murine monoclonal antibody
  • a phosphotyrosine specific antibody (Upstate, clone 4G10, Cat. No. 05-321) is used to determine phosphorylation status of immunopurified IGF-IR.
  • an antibody with specificity for phosphorylated Ser473 (Cell Signalling, Cat. No. 9271) is applied.
  • IGF-I receptor IGF-IR
  • Human tumor cells (HT29, 5 x 10 4 cells/ml) in RPMI 1640 medium (PAA, Cat. No. El 5-039) supplemented with 2 mM L-Glutamin, lx non-essential aminoacids (Gibco, Cat. No. 11140-035), 1 mM sodium pyruvate (Gibco, Cat. No. 11360-039) and 10% heat inactivated FCS (PAA, Cat. No. A15-771). For each incubation period one 12 well plate is inoculated with 1 ml cells in the respective medium for each experiment and cultivated for 24 hours at 37°C and 5% C0 2 .
  • PAA Human tumor cells
  • medium is carefully removed and replaced by different concentrations of antibody diluted in the respective medium.
  • medium is replaced by either medium without antibody or medium with a control antibody (AB-1, Oncogene, Cat. No. GR11).
  • AB-1 Oncogene, Cat. No. GR11
  • Cells are incubated at 37°C and 5% C0 2 and individual plates are taken out for further processing after 15 minutes, 24 hours and 48 hours.
  • the medium is carefully removed by aspiration and 100 ⁇ of cold lysis buffer is added per well (50mM Hepes pH 7.2, 150 mM NaCl, lmM EGTA, 10% glycerol, 1% TritonTM -XI 00, lOOmM NaF, 10 mM Na 4 P 2 0 7 , CompleteTM protease inhibitor).
  • the cells are detached using a cell scraper (Corning, Cat. No. 3010) and well contents transferred to Eppendorf reaction tubes. Cell fragments are removed by centrifugation for 10 minutes at 13000 rpm and 4°C and the supernatant is added to 2x Laemmli sample buffer in a 1 : 1 (v/v) ratio. Cellular proteins are separated by SDS-PAGE and transferred to a nitrocellulose membrane (PROTRANTM BA 85, Schleicher&Schuell, Cat. No. 10 401196) by semi-dry western-blotting.
  • IGF-IR An antibody specific for IGF-IR (C-20, Santa Cruz Biotechnologies, Cat. No. sc- 713) is used to determine protein levels of IGF-IR.
  • 3T3 cells (1 x 10 5 /ml) expressing recombinantly high numbers (>10 5 ) human IR are plated in MEM Dulbecco medium (DMEM) with high glucose (PAA, Cat. No. El 5-009) supplemented with 2mM L-Glutamin (Gibco, Cat. No. 25030-024) and 10% heat inactivated FCS (PAA, Cat. No. A15-771).
  • DMEM MEM Dulbecco medium
  • PAA high glucose
  • 2mM L-Glutamin Gibco, Cat. No. 25030-02
  • 10% heat inactivated FCS (PAA, Cat. No. A15-771).
  • Six bottles in the T175 format are inoculated with 20 ml cells in the respective medium for each experiment and cultivated for two days at 37°C and 5% C02 to obtain confluent cell monolayers.
  • lx Trypsin/EDTA (Gibco, Cat. No. 25300-054) per T175 flask are added and detachment of cells monitored with a microscope. The cells are collected and medium with 10% FCS as described before is added to a total volume of 50 ml. Cells are reisolated by centrifugation for 10 minutes at 1000 rpm and resuspended in 50 ml of binding buffer (120 mM NaCl, 5 mM KC1, 1.2 mM MgS0 4 , 1 mM EDTA, 10 mM D(+)glucose, 15 mM NaAc, 100 mM Hepes pH 7.6, 1% BSA). Cells are counted, reisolated by centrifugation and adjusted with binding buffer to 1 x 10 6 cells/ml.
  • binding buffer 120 mM NaCl, 5 mM KC1, 1.2 mM MgS0 4 , 1 mM EDTA, 10
  • 1125-labeled insulin peptide (Amersham, Cat. No. IM166, -2000 Ci/mmol), solubilized in 0.1% CH3COOH, are diluted in binding buffer to a final activity of 4xl0 5 counts/(minute*ml).
  • 75 ⁇ of antibody together with 25 ⁇ of prediluted 1125-labeled insulin peptide is added to 200 ⁇ of cell suspension (final antibody concentration 200 nM) and incubated for 3,5 h at 4°C. Cells are reisolated by centrifugation for 5 minutes at 2000 rpm and supernatant is removed. After washing two times in 1 ml binding buffer, cells are resuspended in 1 ml binding buffer and transferred to scintillation tubes. The amount of radioactive peptide bound to the cell surface receptors is measured on a scintillation counter.
  • phosphorylation of IGF-IR is determined in the absence of IGF-I ligand but in the presence of the antibody of the invention and a reference antibody ( IR3, Oncogene, Germany). This is performed by a western-blotting analysis with phosphorylation-state specific antibodies.
  • 3T3 cells (ATCC CRL 1658) transfected with IGF-IR (5xl0 4 cells/ml, Pietrzkowski, Z., et al, Cell Growth Differ. 3 (1992) 199-205) are plated in MEM Dulbecco medium (DMEM) with high glucose (PAA, CatNo.
  • the medium is carefully removed and replaced by different concentrations of antibody diluted in the respective medium.
  • Cells are incubated for 15 minutes under the conditions mentioned above.
  • the medium is carefully removed by aspiration and 100 ⁇ of cold lysis buffer is added per well (50mM Hepes pH 7.2, 150 mM NaCl, ImM EGTA, 10% glycerol, 1% Triton-XlOO, lOOmM NaF, 10 mM Na 4 P 2 0 7 , CompleteTM protease inhibitor).
  • the cells are detached using a cell scraper (Corning, CatNo. 3010) and well contents transferred to Eppendorf reaction tubes.
  • Cell fragments are removed by centrifugation for 10 minutes at 13000 rpm and 4°C (Eppendorf centrifuge 5415R) and half of the supernatant is added to 2x Laemmli sample buffer in a 1 : 1 (v/v) ratio.
  • the remaining supernatant of cell lysates underwent a clearifying spin (10 minutes at 13000 rpm and 4°C) right before 1 ⁇ of an antibody against IGF-IR is added (C-20, Santa Cruz Biotechnologies, CatNo. sc-713 or mAb 24-55, GroPep, CatNo. MAD1).
  • Protein G SepharoseTM beads (Amersham Biosciences, CatNo. 17-0618-01) are added followed by another incubation step of 1 hour at 4°C.
  • the beads with bound antibody-protein-complexes are isolated by centrifugation (1 minute at 2000 rpm and 4°C) and washed three times with wash buffer (lysis buffer with only 0.1 %> Triton-XlOO). After boiling the beads in Laemmli sample buffer, cellular proteins are separated by SDS-PAGE and transferred to a nitrocellulose membrane (PROTRAN BA 85, Schleicher&Schuell, CatNo. 10 401196) by semi-dry western- blotting.
  • a phosphotyrosin specific antibody (Upstate, clone 4G10, CatNo. 05-321, recognizing tyrosine -phosphorylated proteins) is used to determine phosphorylation status of immunopurified IGF-IR.
  • phosphorylated Akt/PKB an antibody against Aktl with specificity for phosphorylated Ser473 (Cell Signalling, CatNo. 9271) is applied.
  • Tumors are induced in nude mice and treated once with different concentrations of the antibody of the invention. 24 hours after treatment the tumors are extracted and homogenized under liquid nitrogen.
  • Cold lysis buffer is added (50mM Hepes pH 7.2, 150 mM NaCl, lmM EGTA, 10% glycerol, 1% Triton-XlOO, lOOmM NaF, 1 mM Na 3 V0 4 , 10 mM Na 4 P 2 0 7 , CompleteTM protease inhibitor, lmM PMSF) in a buffer- volume to tumor- weight ratio of 3 : 1 and thoroughly mixed with the thawing tumor homogenate.
  • insoluble fragments are removed by centrifugation for 10 minutes at 13000 rpm and 4°C (Eppendorf centrifuge 5415R).
  • the protein concentration of the samples is determined with the Micro BCATM Reagents (Pierce) and lysis buffer is added to adjust equal concentrations. Part of the supernatant is added to 2x Laemmli sample buffer in a 1 : 1 (v/v) ratio.
  • Cellular proteins are separated by SDS-PAGE and transferred to a nitrocellulose membrane (PROTRAN BA 85, Schleicher&Schuell, CatNo. 10 401196) by semi-dry western-blotting.
  • An IGF-IR specific antibody (C-20, Santa Cruz Biotechnologies, CatNo. sc-713) is used to to detect IGF-IR.
  • tumor volume [mg] (length x (width)2).
  • Antibody is administered intraperitoneally (i.p.) at 10ml/ kg. Treatment is started with doubled doses of the antibody administered in doubled volumes. Tumors are induced in nude mice as described above. After tumors had grown to an average volume of 160 mg, mice are treated intraperitoneally six times once a week with 6, 0.6 and 0.06 mg/ kg of antibody as consecutive doses starting with 12, 1.2 and 0.12 mg/ kg as loading dose given once on the first day of treatment.
  • antibody according to the invention is tested in combination with gemcitabine in the same model. Tumors are induced as described above and treatment is initiated when tumors had established and grown to 170mm3 average in all groups.
  • Antibody is administered once a week i.p. at 6 and 0.6 mg/kg and in combination with 62 mg/ kg of gemcitabine at 0.6 mg.
  • Gemcitabine is administered one cycle i.e. every third day for four times in total. Again, treatment is started by administering doubled doses of the antibody.

Abstract

The invention provides anti-IGF-1R antibodies and methods of using the same.

Description

Antibodies against insulin-like growth factor I receptor and uses thereof Field of the invention
The present invention relates to antibodies against human insulin-like growth factor I receptor (IGF-IR), methods for their production, pharmaceutical compositions containing said antibodies, and uses thereof.
Background of the invention
Insulin-like growth factor I receptor (IGF-IR, EC 2.7.10.1, CD 221 antigen) belongs to the family of transmembrane protein tyrosine kinases (LeRoith, D., et al, Endocrin. Rev. 16 (1995) 143-163; and Adams, T.E., et al, Cell. Mol. Life Sci. 57 (2000) 1050-1093). IGF IR binds IGF I with high affinity and initiates the physiological response to this ligand in vivo. IGF IR also binds to IGF II, however with slightly lower affinity. The IGF-1 system, including IGF-IR, plays an important role during proliferation of (normal and neoplastic) cells. IGF-IR is found on normal human tissues e. g. from placenta, prostate, bladder, kidney, duodenum, small bowel, gallbladder, common bile duct, intrahepatic bile duct, bronchi, tonsil, thymus, breast, sebaceous gl, salivary gland, uterine cervix, and salpinx. IGF IR overexpression promotes the neoplastic transformation of cells and there exists evidence that IGF IR is involved in malignant transformation of cells and is therefore a useful target for the development of therapeutic agents for the treatment of cancer (Adams, T.E., et al, Cell. Mol. Life Sci. 57 (2000) 1050-1093).
Antibodies against IGF IR are well-known in the state of the art and investigated for their antitumor effects in vitro and in vivo (Benini, S., et al, Clin. Cancer Res. 7 (2001) 1790-1797; Scotlandi, K., et al, Cancer Gene Ther. 9 (2002) 296-307; Scotlandi, K., et al., Int. J. Cancer 101 (2002) 11-16; Brunetti, A., et al., Biochem. Biophys. Res. Commun. 165 (1989) 212-218; Prigent, S.A., et al, J. Biol. Chem. 265 (1990) 9970-9977; Li, S.L., et al, Cancer Immunol. Immunother. 49 (2000) 243-252; Pessino, A., et al, Biochem. Biophys. Res. Commun. 162 (1989) 1236- 1243; Surinya, K.H., et al, J. Biol. Chem. 277 (2002) 16718-16725; Soos, M.A., et al, J. Biol. Chem., 267 (1992) 12955-12963; Soos, M.A., et al, Proc. Natl. Acad. Sci. USA 86 (1989) 5217-5221; O'Brien, R.M., et al, EMBO J. 6 (1987) 4003- 4010; Taylor, R., et al, Biochem. J. 242 (1987) 123-129; Soos, M.A., et al, Biochem. J. 235 (1986) 199-208; Li, S.L., et al, Biochem. Biophys. Res. Commun. 196 (1993) 92-98; Delafontaine, P., et al, J. Mol. Cell. Cardiol. 26 (1994) 1659- 1673; Kull, F.C., et al. J. Biol. Chem. 258 (1983) 6561-6566; Morgan, D.O., and Roth, R.A., Biochemistry 25 (1986) 1364-1371; Forsayeth, J.R., et al, Proc. Natl. Acad. Sci. USA 84 (1987) 3448-3451; Schaefer, E.M., et al, J. Biol. Chem. 265 (1990) 13248-13253; Gustafson, T.A., and Rutter, W.J., J. Biol. Chem. 265 (1990) 18663-18667; Hoyne, P.A., et al, FEBS Lett. 469 (2000) 57-60; Tulloch, P.A., et al, J. Struct. Biol. 125 (1999) 11-18; Rohlik, Q.T., et al, Biochem. Biophys. Res. Comm. 149 (1987) 276-281; and Adams, T.E., et al, Cell. Mol. Life Sci. 57 (2000) 1050-1093; Dricu, A., et al, Glycobiology 9 (1999) 571-579; Kanter-Lewensohn, L., et al, Melanoma Res. 8 (1998) 389-397; Li, S.L., et al, Cancer Immunol. Immunother. 49 (2000) 243-252). Antibodies against IGF-IR are also described in a lot of further publications, e.g., Arteaga, C.L., et al, Breast Cancer Res. Treatment 22 (1992) 101-106; and Hailey, J., et al, Mol. Cancer Ther. 1 (2002) 1349-1353.
In particular, the monoclonal antibody against IGF IR called aIR3 is widely used in the investigation of studying IGF IR mediated processes and IGF I mediated diseases such as cancer. Alpha IR 3 was described by Kull, F.C., et al, J. Biol. Chem. 258 (1983) 6561-6566. In the meantime, about a hundred publications have been published dealing with the investigation and therapeutic use of aIR3 in regard to its antitumor effect, alone and together with cytostatic agents such as doxorubicin and vincristine. aIR3 is a murine monoclonal antibody which is known to inhibit IGF I binding to IGF receptor but not IGF II binding to IGF-IR. aIR3 stimulates at high concentrations tumor cell proliferation and IGF-IR phosphorylation (Bergmann, U., et al, Cancer Res. 55 (1995) 2007-2011; Kato, FL, et al, J. Biol. Chem. 268 (1993) 2655-2661). There exist other antibodies (e.g., 1H7, Li, S.L., et al, Cancer Immunol. Immunother. 49 (2000) 243-252) which inhibit IGF II binding to IGF IR more potently than IGF I binding. A summary of the state of the art of antibodies and their properties and characteristics is described by Adams, T.E., et al, Cell. Mol. Life Sci. 57 (2000) 1050-1093.
Most of the antibodies described in the state of the art are of mouse origin. Such antibodies are, as is well known in the state of the art, not useful for the therapy of human patients without further alterations like chimerization or humanization. Based on these drawbacks, human antibodies are clearly preferred as therapeutic agents in the treatment of human patients. Human antibodies are well-known in the state of the art (van Dijk, M.A., and van de Winkel, J.G., Curr. Opin. Chem. Biol. 5 (2001) 368-374). Based on such technology, human antibodies against a great variety of targets can be produced. Examples of human antibodies against IGF-IR are described in WO 2002/053596, WO 2004/071529, WO 2005/016967, WO 2006/008639, US 2005/0249730, US 2005/0084906, WO 2005/058967, WO 2006/013472, US 2003/0165502, WO 2005/082415, WO 2005/016970, WO 2003/106621, WO 2004/083248, WO 2003/100008, WO 2004/087756, WO 2005/005635 and WO 2005/094376.
WO 2004/087756 describes antibodies binding to IGF-IR and inhibiting the binding of IGF-I and IGF-II to IGF-IR characterized in being of human IgGl isotype, and showing a ratio of inhibition of the binding of IGF-I to IGF-IR to the inhibition of binding of IGF-II to IGF-IR of 1 :3 to 3: 1, and induces cell death of 20% or more cells of a preparation of IGF-IR expressing cells after 24 hours at a concentration of said antibody of 100 nM by ADCC.
WO 2005/005635 describes an anti-IGF-lR antibody characterized in comprising a CDRH3 region of SEQ ID NO: 1, a CDRH2 region of SEQ ID NO:2 and a CDRH1 region of SEQ ID NO: 3 and in that the light chain variable domain a CDRL3 region of SEQ ID NO: 4, a CDRL2 region of SEQ ID NO:5 and a CDRLl region of SEQ ID NO: 6 and an antibody characterized in that the heavy chain variable domain comprises SEQ ID NO:7 and the light chain variable domain comprises SEQ ID NO:8.
There is still a need for antibodies against IGF IR with convincing benefits for patients in need of antitumor therapy. The relevant benefit for the patient is, in simple terms, reduction in tumor growth and a significant prolongation of time to progression caused by the treatment with the antitumorigenic agent.
Summary of the invention
The invention comprises an antibody specifically binding to human IGF-IR, characterized in comprising as heavy chain variable domain CDRH3 region a CDRH3 region of SEQ ID NO: 1 with a mutation selected from the group consisting of E1Q, L2E, L2F, L2H, L2M, L2T, G3K, G3L, G3Q, R4H, R4K, Y6F, Y6H, Y6W, D8A, D8G, D8Q, D8S, L9G, L9I, L9N, L9P, L9Q, L9R, L9T, or L9V.
The antibodies according to the invention differ from the antibodies described in WO 2005/005635 by mutations in the heavy chain CDRH3 region.
Preferably the antibody according to the invention is characterized in that the heavy chain variable domain comprises a CDRH3 region of SEQ ID NO: 1 with a mutation selected from the group consisting of E1Q, L2E, L2F, L2H, L2M, L2T, G3K, G3L, G3Q, R4H, R4K, Y6F, Y6H, Y6W, D8A, D8G, D8Q, D8S, L9G, L9I, L9N, L9P, L9Q, L9R, L9T, or L9V, a CDRH2 region of SEQ ID NO:2 and a CDRH1 region of SEQ ID NO:3.
Preferably the antibody according to the invention is characterized in that the heavy chain variable domain comprises a CDRH3 region of SEQ ID NO: 1 with a mutation selected from the group consisting of EIQ, L2E, L2F, L2H, L2M, L2T, G3K, G3L, G3Q, R4H, R4K, Y6F, Y6H, Y6W, D8A, D8G, D8Q, D8S, L9G, L9I, L9N, L9P, L9Q, L9R, L9T, or L9V, a CDRH2 region of SEQ ID NO:2 and a CDRH1 region of SEQ ID NO: 3 and the light chain variable domain comprises a CDRL3 region of SEQ ID NO: 4, a CDRL2 region of SEQ ID NO:5 and a CDRL1 region of SEQ ID NO:6.
Preferably the antibody according to the invention is characterized in that the heavy chain variable domain comprises SEQ ID NO: 7 comprising a mutation selected from the group consisting of EIQ, L2E, L2F, L2H, L2M, L2T, G3K, G3L, G3Q, R4H, R4K, Y6F, Y6H, Y6W, D8A, D8G, D8Q, D8S, L9G, L9I, L9N, L9P, L9Q, L9R, L9T, or L9V.
Preferably the antibody according to the invention is characterized in that the heavy chain variable domain comprises SEQ ID NO: 7 comprising a double mutation selected from the group consisting of E99Q EIQ, L100E L2E, LI OOF L2F, L100H L2H, L100M L2M, L100T L2T, G101K G3K, G101L G3L, G101Q G3Q, R102H R4H, R102K R4K, Y104F Y6F, Y104H Y6H, Y104W Y6W, D106A D8A, D106G D8G, D106Q D8Q, D106S D8S, L107G L9G, L107I L9I, L107N L9N, L107P L9P, L107Q L9Q, L107R L9R, L107T L9T, or L107V L9V.
Preferably the antibody according to the invention is characterized in that the heavy chain variable domain comprises SEQ ID NO: 7 comprising a mutation selected from the group consisting of EIQ, L2E, L2F, L2H, L2M, L2T, G3K, G3L, G3Q, R4H, R4K, Y6F, Y6H, Y6W, D8A, D8G, D8Q, D8S, L9G, L9I, L9N, L9P, L9Q, L9R, L9T, or L9V and the light chain variable domain comprises SEQ ID NO:8.
Preferably the antibody according to the invention is characterized in that the heavy chain variable domain comprises SEQ ID NO: 7 comprising a double mutation selected from the group consisting of E99Q EIQ, L100E L2E, LI OOF L2F, L100H L2H, L100M L2M, L100T L2T, G101K G3K, G101L G3L, G101Q G3Q, R102H R4H, R102K R4K, Y104F Y6F, Y104H Y6H, Y104W Y6W, D106A D8A, D106G D8G, D106Q D8Q, D106S D8S, L107G L9G, L107I L9I, L107N L9N, L107P L9P, L107Q L9Q, L107R L9R, L107T L9T, or LI 07V L9V and the light chain variable domain comprises SEQ ID NO:8.
Heavy chain variable domain SEQ ID NO: 7 and light chain variable domain SEQ ID NO:8 are preferably of human lambda isotype.
Preferably the antibody according to the invention is characterized by the above mentioned amino acid sequences or amino acid sequence fragments and properties. The antibody according to the invention preferably comprises a Fc part derived from human origin. Preferably the antibody according to the invention is of human IgGl or IgG4 isotype. Preferably the antibody is of human IgGl class. Preferably the antibody is humanized or is a human antibody.
Preferably the antibody is specifically binding to IGF-IR and inhibiting the binding of IGF-I and IGF -II to IGF-IR. In addition the antibody according to the invention is preferably of IgGl isotype, partially fucosylated, and/or is not activating the IGF-IR signaling even in IGF-IR overexpressing cells at a 200-fold concentration or even 20.000-fold concentration of its IC50 value.
Preferably, at a concentration of 5 nM the antibodies according to the invention completely inhibit IGF-I mediated signal transduction of IGF-IR in tumor cells.
The antibody is preferably a monoclonal antibody and, in addition, a chimeric antibody (human constant chain), a humanized antibody and especially preferably a human antibody.
The antibody preferably binds to IGF-IR human (EC 2.7.1.10.1, SwissProt P08069) in competition to antibody 18, which is described in WO 2005/005635 and comprises the heavy chain variable domain of SEQ ID NO: 7 and the light chain variable domain of SEQ ID NO:8.
The antibody is preferably further characterized by an affinity of 10"8 M (KD) or less, preferably of about 10"9 to 10"13 M.
The antibody shows preferably no detectable concentration dependent inhibition of insulin binding to the insulin receptor.
The antibody is preferably of IgGl type. The antibody according to the invention considerably prolongates the time to progression in relevant xenograft tumor models in comparison with vehicle treated animals and reduces tumor growth. The antibody inhibits the binding of IGF I and IGF II to IGF IR in vitro and in vivo, preferably in about an equal manner for IGF-I and IGF-II.
The invention further provides methods for the recombinant production of such antibodies.
The invention further provides a nucleic acid encoding a heavy chain variable domain characterized in that the heavy chain variable domain comprises SEQ ID NO:7 with a mutation selected from the group consisting of EIQ, L2E, L2F, L2H, L2M, L2T, G3K, G3L, G3Q, R4H, R4K, Y6F, Y6H, Y6W, D8A, D8G, D8Q, D8S, L9G, L9I, L9N, L9P, L9Q, L9R, L9T, or L9V.
The invention further provides a nucleic acid encoding a heavy chain variable domain characterized in that the heavy chain variable domain comprises SEQ ID NO:7 with a double mutation selected from the group consisting of E99Q EIQ, L100E L2E, LI OOF L2F, L100H L2H, L100M L2M, L100T L2T, G101K G3K, G101L G3L, G101Q G3Q, R102H R4H, R102K R4K, Y104F Y6F, Y104H Y6H, Y104W Y6W, D106A D8A, D106G D8G, D106Q D8Q, D106S D8S, L107G L9G, LI 071 L9I, L107N L9N, L107P L9P, L107Q L9Q, L107R L9R, L107T L9T, or LI 07V L9V.
The invention further provides a nucleic acid encoding a heavy chain variable domain characterized in that the heavy chain variable domain comprises SEQ ID NO:7 with a mutation selected from the group consisting of EIQ, L2E, L2F, L2H, L2M, L2T, G3K, G3L, G3Q, R4H, R4K, Y6F, Y6H, Y6W, D8A, D8G, D8Q, D8S, L9G, L9I, L9N, L9P, L9Q, L9R, L9T, or L9V and the light chain variable domain comprises SEQ ID NO:8.
The invention further provides a nucleic acid encoding a heavy chain variable domain characterized in that the heavy chain variable domain comprises SEQ ID NO:7 with a double mutation selected from the group consisting of E99Q EIQ, L100E L2E, LI OOF L2F, L100H L2H, L100M L2M, L100T L2T, G101K G3K, G101L G3L, G101Q G3Q, R102H R4H, R102K R4K, Y104F Y6F, Y104H Y6H, Y104W Y6W, D106A D8A, D106G D8G, D106Q D8Q, D106S D8S, L107G L9G, LI 071 L9I, L107N L9N, L107P L9P, L107Q L9Q, L107R L9R, L107T L9T, or LI 07V L9V and the light chain variable domain comprises SEQ ID NO:8. The invention further comprises the use of an antibody according to the invention, for the treatment of diseases, preferably of tumor diseases.
The invention further comprises an immunoconjugate comprising an antibody according to the invention and a cytotoxic agent.
The invention further comprises a pharmaceutical formulation comprising an antibody according to the invention and a pharmaceutically acceptable carrier.
The invention further comprises an antibody according to the invention for use as a medicament.
The invention further comprises an antibody according to the invention for use in treating tumor diseases.
The invention further comprises an antibody according to the invention for use in inhibiting the binding of human IGF I and human IGF-II to human IGF-IR.
The invention further comprises an antibody according to the invention for use in the manufacture of a medicament.
The invention further comprises an antibody according to the invention for use in the manufacture of a medicament, wherein the medicament is for treatment of a tumor disease.
The invention further comprises the use of an according to the invention in the manufacture of a medicament for the treatment of a tumor disease.
The invention further comprises a method of treating an individual having a tumor disease comprising administering to the individual an effective amount of the antibody according to the invention
The invention further comprises a method for inhibiting the binding of human IGF I and human IGF-II to human IGF-IR in an individual comprising administering to the individual an effective amount of the antibody according to the invention.
The invention further comprises the use of an antibody according to the invention for the manufacture of a medicament for the treatment of diseases, preferably of tumor diseases. The invention further comprises a method for the manufacture of a medicament for the treatment of diseases, preferably of tumor diseases (especially breast cancer, primary glioma, pancreatic cancers, bladder papilloma, colon adenocarcinoma, melanoma, medulloblastoma, pediatric tumors, fibrosarcoma), characterized in comprising an antibody according to the invention.
Antibodies according to the invention show benefits for patients in need of antitumor therapy and provide reduction of tumor growth and a significant prolongation of the time to progression. The antibodies according to the invention have new and inventive properties causing a benefit for a patient suffering from a disease associated with an IGF deregulation, especially a tumor disease. The antibodies according to the invention are characterized by the above mentioned properties.
The invention further provides methods for treating cancer, preferably breast cancer, pancreatic cancer, prostate cancer, bladder cancer, malignal melanoma, Ewing's sarcoma, Neuroblastoma, Osteosarcoma, Rhabdomyosarcoma, and/or NSCLC, comprising administering to a patient diagnosed as having cancer (and therefore being in need of an antitumor therapy) an antibody against IGF IR according to the invention. The antibody may be administered alone, in a pharmaceutical composition, or alternatively in combination with other inhibitors of cancer related signaling pathways such as EGFR, Her2/neu or estrogen receptor or in combination with a cytotoxic treatment such as radiotherapy or a cytotoxic agent or a prodrug thereof. The antibody is administered in a pharmaceutically effective amount.
The invention further comprises the use of an antibody according to the invention for cancer treatment, preferably breast cancer, pancreatic cancer and/or NSCLC, and for the manufacture of a pharmaceutical composition according to the invention. In addition, the invention comprises a method for the manufacture of a pharmaceutical composition according to the invention.
The invention further comprises an antibody according to the invention for cancer treatment, preferably breast cancer, pancreatic cancer, prostate cancer, bladder cancer, malignal melanoma, Ewing's sarcoma, Neuroblastoma, Osteosarcoma, Rhabdomyosarcoma and/or NSCLC. The invention further comprises a pharmaceutical composition containing an antibody according to the invention, optionally together with a buffer and/or an adjuvant useful for the formulation of antibodies for pharmaceutical purposes.
The invention further comprises a pharmaceutical composition comprising an antibody according to the invention.
The invention further provides pharmaceutical compositions comprising such antibodies in a pharmaceutically acceptable carrier. In one embodiment, the pharmaceutical composition may be included in an article of manufacture or kit. The invention further provides the use of an antibody according to the invention for the manufacture of a pharmaceutical composition for the treatment of cancer. The antibody is used in a pharmaceutically effective amount.
The invention further comprises the use of an antibody according to the invention for the manufacture of a pharmaceutical composition for the treatment of cancer, preferably breast cancer, pancreatic cancer and/or NSCLC. The antibody is used in a pharmaceutically effective amount.
The invention further comprises a method for the production of a recombinant human antibody according to the invention, characterized by expressing a nucleic acid encoding an antibody binding to IGF-IR in a CHO host cell and recovering said antibody from said cell.
The invention further comprises a process for the production of an antibody against IGF-IR according to the invention, comprising the steps of transforming a host cell, preferably a CHO cell.
Detailed description of the embodiments of the invention I. Definitions
An "acceptor human framework" for the purposes herein is a framework comprising the amino acid sequence of a light chain variable domain (VL) framework or a heavy chain variable domain (VH) framework derived from a human immunoglobulin framework or a human consensus framework, as defined below. An acceptor human framework "derived from" a human immunoglobulin framework or a human consensus framework may comprise the same amino acid sequence thereof, or it may contain amino acid sequence changes. In some embodiments, the number of amino acid changes are 10 or less, 9 or less, 8 or less, 7 or less, 6 or less, 5 or less, 4 or less, 3 or less, or 2 or less. In some embodiments, the VL acceptor human framework is identical in sequence to the VL human immunoglobulin framework sequence or human consensus framework sequence.
"Affinity" refers to the strength of the sum total of noncovalent interactions between a single binding site of a molecule (e.g., an antibody) and its binding partner (e.g., an antigen). Unless indicated otherwise, as used herein, "binding affinity" refers to intrinsic binding affinity which reflects a 1 : 1 interaction between members of a binding pair (e.g., antibody and antigen). The affinity of a molecule X for its partner Y can generally be represented by the dissociation constant (Kd). Affinity can be measured by common methods known in the art, including those described herein. Specific illustrative and exemplary embodiments for measuring binding affinity are described in the following.
The terms "anti-IGF-lR antibody" and "an antibody that binds to IGF-1R" refer to an antibody that is capable of binding IGF-1R with sufficient affinity such that the antibody is useful as a diagnostic and/or therapeutic agent in targeting IGF-1R. In one embodiment, the extent of binding of an anti-IGF-lR antibody to an unrelated, non-IGF-lR protein is less than about 10% of the binding of the antibody to IGF-1R as measured, by surface Plasmon resonance. In certain embodiments, an antibody that binds to IGF-1R has a dissociation constant (Kd) of 10"8 M or less, e.g. from 10"8 M to 10"13 M, e.g., from 10"9 M to 10"13 M).
The term "antibody" herein is used in the broadest sense and encompasses various antibody structures, including but not limited to monoclonal antibodies, polyclonal antibodies, multispecific antibodies (e.g., bispecific antibodies), and antibody fragments so long as they exhibit the desired antigen-binding activity.
An "antibody fragment" refers to a molecule other than an intact antibody that comprises a portion of an intact antibody that binds the antigen to which the intact antibody binds. Examples of antibody fragments include but are not limited to Fv, Fab, Fab', Fab'-SH, F(ab')2; diabodies; linear antibodies; single-chain antibody molecules (e.g. scFv); and multispecific antibodies formed from antibody fragments.
The term "chimeric" antibody refers to an antibody in which a portion of the heavy and/or light chain is derived from a particular source or species, while the remainder of the heavy and/or light chain is derived from a different source or species. The "class" of an antibody refers to the type of constant domain or constant region possessed by its heavy chain. There are five major classes of antibodies: IgA, IgD, IgE, IgG, and IgM, and several of these may be further divided into subclasses (isotypes), e.g., IgGi, IgG2, IgG3, IgG4, IgAls and IgA2. The heavy chain constant domains that correspond to the different classes of immunoglobulins are called a, δ, ε, γ, and μ, respectively.
The term "cytotoxic agent" as used herein refers to a substance that inhibits or prevents a cellular function and/or causes cell death or destruction. Cytotoxic
21 1 131 125 agents include, but are not limited to, radioactive isotopes (e.g., At , 1 , 1 , Y90, Re186, Re188, Sm153, Bi212, P32, Pb212 and radioactive isotopes of Lu); chemotherapeutic agents or drugs (e.g., methotrexate, adriamicin, vinca alkaloids (vincristine, vinblastine, etoposide), doxorubicin, melphalan, mitomycin C, chlorambucil, daunorubicin or other intercalating agents); growth inhibitory agents; enzymes and fragments thereof such as nucleolytic enzymes; antibiotics; toxins such as small molecule toxins or enzymatically active toxins of bacterial, fungal, plant or animal origin, including fragments and/or variants thereof; and the various antitumor or anticancer agents disclosed below.
"Effector functions" refer to those biological activities attributable to the Fc region of an antibody, which vary with the antibody isotype. Examples of antibody effector functions include: Clq binding and complement dependent cytotoxicity (CDC); Fc receptor binding; antibody-dependent cell-mediated cytotoxicity (ADCC); phagocytosis; down regulation of cell surface receptors (e.g. B cell receptor); and B cell activation.
An "effective amount" of an agent, e.g., a pharmaceutical formulation, refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired therapeutic or prophylactic result.
The term "Fc region" herein is used to define a C-terminal region of an immunoglobulin heavy chain that contains at least a portion of the constant region. The term includes native sequence Fc regions and variant Fc regions. In one embodiment, a human IgG heavy chain Fc region extends from Cys226, or from Pro230, to the carboxyl-terminus of the heavy chain. However, the C-terminal lysine (Lys447) of the Fc region may or may not be present. Unless otherwise specified herein, numbering of amino acid residues in the Fc region or constant region is according to the EU numbering system, also called the EU index, as described in Kabat et al, Sequences of Proteins of Immunological Interest, 5th ed. Public Health Service, NIH Publication 91-3242, National Institutes of Health, Bethesda, MD (1991).
"Framework" or "FR" refers to variable domain residues other than hypervariable region (HVR) residues. The FR of a variable domain generally consists of four FR domains: FR1, FR2, FR3, and FR4. Accordingly, the CDR and FR sequences generally appear in the following sequence in VH (or VL): FR1-CDR1(L1)-FR2- CDR2(L2)-FR3-CDR3(L3)-FR4.
The terms "full length antibody," "intact antibody," and "whole antibody" are used herein interchangeably to refer to an antibody having a structure substantially similar to a native antibody structure or having heavy chains that contain an Fc region as defined herein.
The terms "host cell," "host cell line," and "host cell culture" are used interchangeably and refer to cells into which exogenous nucleic acid has been introduced, including the progeny of such cells. Host cells include "transformants" and "transformed cells," which include the primary transformed cell and progeny derived therefrom without regard to the number of passages. Progeny may not be completely identical in nucleic acid content to a parent cell, but may contain mutations. Mutant progeny that have the same function or biological activity as screened or selected for in the originally transformed cell are included herein.
A "human antibody" is one which possesses an amino acid sequence which corresponds to that of an antibody produced by a human or a human cell or derived from a non-human source that utilizes human antibody repertoires or other human antibody-encoding sequences. This definition of a human antibody specifically excludes a humanized antibody comprising non-human antigen-binding residues.
A "human consensus framework" is a framework which represents the most commonly occurring amino acid residues in a selection of human immunoglobulin VL or VH framework sequences. Generally, the selection of human immunoglobulin VL or VH sequences is from a subgroup of variable domain sequences. Generally, the subgroup of sequences is a subgroup as in Kabat et al, Sequences of Proteins of Immunological Interest, 5th ed., Public Health Service, NIH Publication 91-3242, National Institutes of Health, Bethesda, MD (1991). In one embodiment, for the VL, the subgroup is subgroup kappa I as in Kabat et al, supra. In one embodiment, for the VH, the subgroup is subgroup III as in Kabat et al., supra.
A "humanized" antibody refers to a chimeric antibody comprising amino acid residues from non-human CDRs and amino acid residues from human FRs. In certain embodiments, a humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the CDRs correspond to those of a non-human antibody, and all or substantially all of the FRs correspond to those of a human antibody. A humanized antibody optionally may comprise at least a portion of an antibody constant region derived from a human antibody. A "humanized form" of an antibody, e.g., a non-human antibody, refers to an antibody that has undergone humanization.
The term "complementarity determining regions" or "CDR," as used herein, refers to each of the regions of an antibody variable domain which are hypervariable in sequence and/or form structurally defined loops ("hypervariable loops). (Kabat et al, Sequences of Proteins of Immunological Interest, 5th ed. Public Health Service, NIH Publication 91-3242, National Institutes of Health, Bethesda, MD (1991)). With the exception of CDR1 in VH, CDRs generally comprise the amino acid residues that form the hypervariable loops. Unless otherwise indicated, CDR residues and other residues in the variable domain (e.g., FR residues) are numbered herein according to Kabat et al., supra.
An "immunoconjugate" is an antibody conjugated to one or more heterologous molecule(s), including but not limited to a cytotoxic agent.
An "individual" or "subject" is a mammal. Mammals include, but are not limited to, domesticated animals (e.g., cows, sheep, cats, dogs, and horses), primates (e.g., humans and non-human primates such as monkeys), rabbits, and rodents (e.g., mice and rats). In certain embodiments, the individual or subject is a human.
Inhibiting binding of IGF I and II to IGF-1R can be determined using a IGF-1R expressing cell line (HT29) and radioactive labeled IGF-I or IGF-II. The amount of radioactive peptide bound to the cell surface receptors is measured. An antibody according to the invention should inhibit the binding like antibody 18 described in WO 2005/005635 with an IC50 value of 0.5 nM or lower.
An "isolated" antibody is one which has been separated from a component of its natural environment. In some embodiments, an antibody is purified to greater than 95% or 99% purity as determined by, for example, electrophoretic (e.g., SDS- PAGE, isoelectric focusing (IEF), capillary electrophoresis) or chromatographic (e.g., ion exchange or reverse phase HPLC). For review of methods for assessment of antibody purity, see, e.g., Flatman, S., et al, J. Chromatogr. B 848 (2007) 79-87.
An "isolated" nucleic acid refers to a nucleic acid molecule that has been separated from a component of its natural environment. An isolated nucleic acid includes a nucleic acid molecule contained in cells that ordinarily contain the nucleic acid molecule, but the nucleic acid molecule is present extrachromosomally or at a chromosomal location that is different from its natural chromosomal location.
"Isolated nucleic acid encoding an anti-IGF-lR antibody" refers to one or more nucleic acid molecules encoding antibody heavy and light chains (or fragments thereof), including such nucleic acid molecule(s) in a single vector or separate vectors, and such nucleic acid molecule(s) present at one or more locations in a host cell.
The term "monoclonal antibody" as used herein refers to an antibody obtained from a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical and/or bind the same epitope, except for possible variant antibodies, e.g., containing naturally occurring mutations or arising during production of a monoclonal antibody preparation, such variants generally being present in minor amounts. In contrast to polyclonal antibody preparations, which typically include different antibodies directed against different determinants (epitopes), each monoclonal antibody of a monoclonal antibody preparation is directed against a single determinant on an antigen. Thus, the modifier "monoclonal" indicates the character of the antibody as being obtained from a substantially homogeneous population of antibodies, and is not to be construed as requiring production of the antibody by any particular method. For example, the monoclonal antibodies to be used in accordance with the present invention may be made by a variety of techniques, including but not limited to the hybridoma method, recombinant DNA methods, phage-display methods, and methods utilizing transgenic animals containing all or part of the human immunoglobulin loci, such methods and other exemplary methods for making monoclonal antibodies being described herein.
The term "mutation" as used herein refers to one or more amino acid substitutions in a CDR and/or variable region of an antibody according to the invention. The terms EIQ, L2E, L2F, L2H, L2M, L2T, G3K, G3L, G3Q, R4H, R4K, Y6F, Y6H, Y6W, D8A, D8G, D8Q, D8S, L9G, L9I, L9N, L9P, L9Q, L9R, L9T, or L9V relate to substitutions in the heavy chain CDRH3 (SEQ ID NO: l). The numbering 1 to 9 relates therefore to amino acids 1 to 9 of SEQ ID NO: l . EIQ means therefore that the first amino acid of SEQ ID NO: l, which is E (Glu), is replaced by amino acid Q (Gin). Terms with higher numbers than 9, like E99Q, L100E, L100F, L100H, L100M, L100T, G101K, G101L, G101Q, R102H, R102K, Y104F, Y104H, Y104W, D106A, D106G, D106Q, D106S, L107G, L107I, L107N, L107P, L107Q, L107R, L107T, or LI 07V relate to the heavy chain variable domain comprises SEQ ID NO:7. E99Q means therefore that amino acid 99 of SEQ ID NO:7, which is E (Glu), is replaced by amino acid Q (Gin). A double mutation E99Q, EIQ describes therefore an antibody according to the invention wherein E99 of SEQ ID NO:7 is replaced by Q and El of SEQ ID NO: l is replaced by Q.
A "naked antibody" refers to an antibody that is not conjugated to a heterologous moiety (e.g., a cytotoxic moiety) or radiolabel. The naked antibody may be present in a pharmaceutical formulation.
"Native antibodies" refer to naturally occurring immunoglobulin molecules with varying structures. For example, native IgG antibodies are heterotetrameric glycoproteins of about 150,000 daltons, composed of two identical light chains and two identical heavy chains that are disulfide-bonded. From N- to C-terminus, each heavy chain has a variable region (VH), also called a variable heavy domain or a heavy chain variable domain, followed by three constant domains (CHI, CH2, and CH3). Similarly, from N- to C-terminus, each light chain has a variable region (VL), also called a variable light domain or a light chain variable domain, followed by a constant light (CL) domain. The light chain of an antibody may be assigned to one of two types, called kappa (κ) and lambda (λ), based on the amino acid sequence of its constant domain.
The term "package insert" is used to refer to instructions customarily included in commercial packages of therapeutic products, that contain information about the indications, usage, dosage, administration, combination therapy, contraindications and/or warnings concerning the use of such therapeutic products.
"Percent (%) amino acid sequence identity" with respect to a reference polypeptide sequence is defined as the percentage of amino acid residues in a candidate sequence that are identical with the amino acid residues in the reference polypeptide sequence, after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent sequence identity, and not considering any conservative substitutions as part of the sequence identity. Alignment for purposes of determining percent amino acid sequence identity can be achieved in various ways that are within the skill in the art, for instance, using publicly available computer software such as BLAST, BLAST-2, ALIGN or Megalign (DNASTAR) software. Those skilled in the art can determine appropriate parameters for aligning sequences, including any algorithms needed to achieve maximal alignment over the full length of the sequences being compared. For purposes herein, however, % amino acid sequence identity values are generated using the sequence comparison computer program ALIGN-2. The ALIGN -2 sequence comparison computer program was authored by Genentech, Inc., and the source code has been filed with user documentation in the U.S. Copyright Office, Washington D.C., 20559, where it is registered under U.S. Copyright Registration No. TXU510087. The ALIGN-2 program is publicly available from Genentech, Inc., South San Francisco, California, or may be compiled from the source code. The ALIGN-2 program should be compiled for use on a UNIX operating system, including digital UNIX V4.0D. All sequence comparison parameters are set by the ALIGN-2 program and do not vary.
In situations where ALIGN-2 is employed for amino acid sequence comparisons, the % amino acid sequence identity of a given amino acid sequence A to, with, or against a given amino acid sequence B (which can alternatively be phrased as a given amino acid sequence A that has or comprises a certain % amino acid sequence identity to, with, or against a given amino acid sequence B) is calculated as follows:
100 times the fraction X/Y where X is the number of amino acid residues scored as identical matches by the sequence alignment program ALIGN-2 in that program's alignment of A and B, and where Y is the total number of amino acid residues in B. It will be appreciated that where the length of amino acid sequence A is not equal to the length of amino acid sequence B, the % amino acid sequence identity of A to B will not equal the % amino acid sequence identity of B to A. Unless specifically stated otherwise, all % amino acid sequence identity values used herein are obtained as described in the immediately preceding paragraph using the ALIGN-2 computer program. The term "pharmaceutical formulation" refers to a preparation which is in such form as to permit the biological activity of an active ingredient contained therein to be effective, and which contains no additional components which are unacceptably toxic to a subject to which the formulation would be administered.
A "pharmaceutically acceptable carrier" refers to an ingredient in a pharmaceutical formulation, other than an active ingredient, which is nontoxic to a subject., A pharmaceutically acceptable carrier includes, but is not limited to, a buffer, excipient, stabilizer, or preservative.
The term "IGF-IR," as used herein, refers to human insulin- like growth factor I receptor (IGF-IR, EC 2.7.10.1, CD 221 antigen) which belongs to the family of transmembrane protein tyrosine kinases (LeRoith, D., et al, Endocrin. Rev. 16 (1995) 143-163; and Adams, T.E., et al, Cell. Mol. Life Sci. 57 (2000) 1050-1093. The IGF-I receptor consists of an alpha subunit (a 130-135 kD protein that is involved in ligand binding) and a beta subunit (a 95-kD transmembrane protein with kinase activity).
As used herein, "treatment" (and grammatical variations thereof such as "treat" or "treating") refers to clinical intervention in an attempt to alter the natural course of the individual being treated, and can be performed either for prophylaxis or during the course of clinical pathology. Desirable effects of treatment include, but are not limited to, preventing occurrence or recurrence of disease, alleviation of symptoms, diminishment of any direct or indirect pathological consequences of the disease, preventing metastasis, decreasing the rate of disease progression, amelioration or palliation of the disease state, and remission or improved prognosis. In some embodiments, antibodies of the invention are used to delay development of a disease or to slow the progression of a disease.
The term "variable region" or "variable domain" refers to the domain of an antibody heavy or light chain that is involved in binding the antibody to antigen. The variable domains of the heavy chain and light chain (VH and VL, respectively) of a native antibody generally have similar structures, with each domain comprising four conserved framework regions (FRs) and three hypervariable regions (HVRs). (See, e.g., Kindt et al, Kuby Immunology, 6th ed., W.H. Freeman and Co., New York, (2007), page 91). A single VH or VL domain may be sufficient to confer antigen-binding specificity. Furthermore, antibodies that bind a particular antigen may be isolated using a VH or VL domain from an antibody that binds the antigen to screen a library of complementary VL or VH domains, respectively. See, e.g., Portolano, S., et al, J. Immunol. 150 (1993) 880-887; Clackson, T., et al, Nature 352 (1991) 624-628.
The term "vector," as used herein, refers to a nucleic acid molecule capable of propagating another nucleic acid to which it is linked. The term includes the vector as a self-replicating nucleic acid structure as well as the vector incorporated into the genome of a host cell into which it has been introduced. Certain vectors are capable of directing the expression of nucleic acids to which they are operatively linked. Such vectors are referred to herein as "expression vectors."
II. Compositions and methods
In one aspect, the invention is based on antibodies that bind to IGF-1R. Antibodies of the invention are useful, e.g., for the diagnosis or treatment of tumor diseases.
A. Exemplary Anti-IGF-IR Antibodies
In a further aspect of the invention, an anti-IGF-lR antibody according to any of the above embodiments is a monoclonal antibody, including a chimeric, humanized or human antibody. In one embodiment, an anti-IGF-lR antibody is an antibody fragment, e.g., a Fv, Fab, Fab', scFv, diabody, or F(ab')2 fragment. In another embodiment, the antibody is a full length antibody, e.g., an intact IgGl antibody or other antibody class or isotype as defined herein.
In a further aspect, an anti-IGF-lR antibody according to any of the above embodiments may incorporate any of the features, singly or in combination, as described in the sections below:
1. Antibody Affinity
In certain embodiments, an antibody provided herein has a dissociation constant (Kd) of 10"8 M or less, e.g. from 10"8 M to 10"13 M, e.g., from 10"9 M to 10"13 M).
According to another embodiment, Kd is measured using surface plasmon resonance assays using a BIACORE®-2000 or a BIACORE ®-3000 (BIAcore, Inc., Piscataway, NJ) at 25°C with immobilized antigen CM5 chips at ~10 response units (RU). Briefly, carboxymethylated dextran biosensor chips (CM5, BIACORE, Inc.) are activated with N-ethyl-N'- (3-dimethylaminopropyl)-carbodiimide hydrochloride (EDC) and N-hydroxysuccinimide (NHS) according to the supplier's instructions. Antigen is diluted with 10 mM sodium acetate, pH 4.8, to 5 μg/ml (-0.2 μΜ) before injection at a flow rate of 5 μΐ/minute to achieve approximately 10 response units (RU) of coupled protein. Following the injection of antigen, 1 M ethanolamine is injected to block unreacted groups. For kinetics measurements, two-fold serial dilutions of Fab (0.78 nM to 500 nM) are injected in PBS with 0.05% polysorbate 20 (TWEEN®-20™) surfactant (PBST) at 25°C at a flow rate of approximately 25 μΐ/min. Association rates (kon) and dissociation rates (k0ff) are calculated using a simple one-to-one Langmuir binding model (BIACORE Evaluation Software version 3.2) by simultaneously fitting the association and dissociation sensorgrams. The equilibrium dissociation constant (Kd) is calculated as the ratio koff kon See, e.g., Chen, Y., et al, J. Mol. Biol. 293 (1999) 865-881. If the on-rate exceeds 10^ M~l s~l by the surface plasmon resonance assay above, then the on-rate can be determined by using a fluorescent quenching technique that measures the increase or decrease in fluorescence emission intensity (excitation = 295 nm; emission = 340 nm, 16 nm band-pass) at 25°C of a 20 nM anti-antigen antibody (Fab form) in PBS, pH 7.2, in the presence of increasing concentrations of antigen as measured in a spectrometer, such as a stop-flow equipped spectrophometer (Aviv Instruments) or a 8000-series SLM-AMINCO ™ spectrophotometer (ThermoSpectronic) with a stirred cuvette.
2. Antibody Fragments
In certain embodiments, an antibody provided herein is an antibody fragment. Antibody fragments include, but are not limited to, Fab, Fab', Fab'-SH, F(ab')2, Fv, and scFv fragments, and other fragments described below. For a review of certain antibody fragments, see Hudson, P.J., et al, Nat. Med. 9 (2003) 129-134. For a review of scFv fragments, see, e.g., Plueckthun, A., in The Pharmacology of Monoclonal Antibodies, vol. 113, Rosenburg and Moore eds., (Springer-Verlag, New York), (1994) pp. 269-315; see also WO 93/16185; and U.S. Patent Nos. 5,571,894 and 5,587,458. For discussion of Fab and F(ab')2 fragments comprising salvage receptor binding epitope residues and having increased in vivo half-life, see U.S. Patent No. 5,869,046.
Diabodies are antibody fragments with two antigen-binding sites that may be bivalent or bispecific. See, for example, EP 404,097; WO 1993/01161; Hudson, P.J., et al, Nat. Med. 9 (2003) 129-134; and Holliger, P., et al, Proc. Natl. Acad. Sci. USA 90 (1993) 6444-6448. Triabodies and tetrabodies are also described in Hudson, P.J., et al, Nat. Med. 9 (2003) 129-134. Single-domain antibodies are antibody fragments comprising all or a portion of the heavy chain variable domain or all or a portion of the light chain variable domain of an antibody. In certain embodiments, a single-domain antibody is a human single-domain antibody (Domantis, Inc., Waltham, MA; see, e.g., U.S. Patent No. 6,248,516 Bl).
Antibody fragments can be made by various techniques, including but not limited to proteolytic digestion of an intact antibody as well as production by recombinant host cells (e.g. E. coli or phage), as described herein.
3. Multispecific Antibodies
In certain embodiments, an antibody provided herein is a multispecific antibody, e.g. a bispecific antibody. Multispecific antibodies are monoclonal antibodies that have binding specificities for at least two different sites. In certain embodiments, one of the binding specificities is for IGF-1R and the other is for any other antigen. In certain embodiments, bispecific antibodies may bind to two different epitopes of IGF-1R. Bispecific antibodies may also be used to localize cytotoxic agents to cells which express IGF-1R. Bispecific antibodies can be prepared as full length antibodies or antibody fragments.
Techniques for making multispecific antibodies include, but are not limited to, recombinant co-expression of two immunoglobulin heavy chain-light chain pairs having different specificities (see Milstein, C, and Cuello, A.C., Nature 305 (1983) 537-540), WO 93/08829, and Traunecker, A., et al, EMBO J. 10 (1991) 3655- 3659), and "knob-in-hole" engineering (see, e.g., U.S. Patent No. 5,731,168). Multi-specific antibodies may also be made by engineering electrostatic steering effects for making antibody Fc-heterodimeric molecules (WO 2009/089004A1); cross-linking two or more antibodies or fragments (see, e.g., US Patent No. 4,676,980, and Brennan, M., et al, Science 229 (1985) 81-83); using leucine zippers to produce bi-specific antibodies (see, e.g., Kostelny, S.A., et al, J. Immunol. 148 (1992) 1547-1553); using "diabody" technology for making bispecific antibody fragments (see, e.g., Holliger, P., et al, Proc. Natl. Acad. Sci. USA 90 (1993) 6444-6448); and using single-chain Fv (sFv) dimers (see, e.g. Gruber, M., et al, J. Immunol. 152 (1994) 5368-5374); and preparing trispecific antibodies as described, e.g., in Tutt, A., et al, J. Immunol. 147 (1991) 60-69. Engineered antibodies with three or more functional antigen binding sites, including "Octopus antibodies," are also included herein (see, e.g. US 2006/0025576A1).
The antibody or fragment herein also includes a "Dual Acting FAb" or "DAF" comprising an antigen binding site that binds to IGF-1R as well as another, different antigen (see, US 2008/0069820, for example).
4. Antibody Variants a) Glycosylation variants
In certain embodiments, an antibody provided herein is altered to increase or decrease the extent to which the antibody is glycosylated. Addition or deletion of glycosylation sites to an antibody may be conveniently accomplished by altering the amino acid sequence such that one or more glycosylation sites is created or removed.
Where the antibody comprises an Fc region, the carbohydrate attached thereto may be altered. Native antibodies produced by mammalian cells typically comprise a branched, biantennary oligosaccharide that is generally attached by an N-linkage to Asn297 of the CH2 domain of the Fc region. See, e.g., Wright, A., et al, TIBTECH 15 (1997) 26-32. The oligosaccharide may include various carbohydrates, e.g., mannose, N-acetyl glucosamine (GlcNAc), galactose, and sialic acid, as well as a fucose attached to a GlcNAc in the "stem" of the biantennary oligosaccharide structure. In some embodiments, modifications of the oligosaccharide in an antibody of the invention may be made in order to create antibody variants with certain improved properties.
In one embodiment, antibody variants are provided having a carbohydrate structure that lacks fucose attached (directly or indirectly) to an Fc region. For example, the amount of fucose in such antibody may be from 1% to 80%, from 1% to 65%, from 5%> to 65%o or from 20%> to 40%>. The amount of fucose is determined by calculating the average amount of fucose within the sugar chain at Asn297, relative to the sum of all glycostructures attached to Asn 297 (e. g. complex, hybrid and high mannose structures) as measured by MALDI-TOF mass spectrometry, as described in WO 2008/077546, for example. Asn297 refers to the asparagine residue located at about position 297 in the Fc region (Eu numbering of Fc region residues); however, Asn297 may also be located about ± 3 amino acids upstream or downstream of position 297, i.e., beTween® positions 294 and 300, due to minor sequence variations in antibodies. Such fucosylation variants may have improved ADCC function. See, e.g., US Patent Publication Nos. US 2003/0157108 (Presta, L.); US 2004/0093621 (Kyowa Hakko Kogyo Co., Ltd). Examples of publications related to "defucosylated" or "fucose-deficient" antibody variants include: US 2003/0157108; WO 2000/61739; WO 2001/29246; US 2003/0115614; US 2002/0164328; US 2004/0093621; US 2004/0132140; US 2004/0110704; US 2004/0110282; US 2004/0109865; WO 2003/085119; WO 2003/084570; WO 2005/035586; WO 2005/035778; WO2005/053742; WO2002/031140; Okazaki, A., et al, J. Mol. Biol. 336 (2004) 1239-1249; Yamane-Ohnuki, N., et al, Biotech. Bioeng. 87 (2004) 614-622. Examples of cell lines capable of producing defucosylated antibodies include Led 3 CHO cells deficient in protein fucosylation (Ripka, J. et al, Arch. Biochem. Biophys. 249 (1986) 533-545; US Pat Appl No. US 2003/0157108 Al, Presta, L; and WO 2004/056312 Al, Adams et al, especially at Example 11), and knockout cell lines, such as alpha- 1,6- fucosyltransferase gene, FUT8, knockout CHO cells (see, e.g., Yamane-Ohnuki, N., et al, Biotech. Bioeng. 87 (2004) 614-622; Kanda, Y. et al, Biotechnol. Bioeng. 94 (2006) 680-688; and WO 2003/085107).
Antibodies variants are further provided with bisected oligosaccharides, e.g., in which a biantennary oligosaccharide attached to the Fc region of the antibody is bisected by GlcNAc. Such antibody variants may have reduced fucosylation and/or improved ADCC function. Examples of such antibody variants are described, e.g., in WO 2003/011878 (Jean-Mairet, et al); US Patent No. 6,602,684 (Umana, et al); and US 2005/0123546 (Umana, et al.). Antibody variants with at least one galactose residue in the oligosaccharide attached to the Fc region are also provided. Such antibody variants may have improved CDC function. Such antibody variants are described, e.g., in WO 1997/30087 (Patel, et al); WO 1998/58964 (Raju, S.); and WO 1999/22764 (Raju, S.). b) Fc region variants
In certain embodiments, one or more amino acid modifications may be introduced into the Fc region of an antibody provided herein, thereby generating an Fc region variant. The Fc region variant may comprise a human Fc region sequence {e.g., a human IgGl, IgG2, IgG3 or IgG4 Fc region) comprising an amino acid modification {e.g. a substitution) at one or more amino acid positions. In certain embodiments, the invention contemplates an antibody variant that possesses some but not all effector functions, which make it a desirable candidate for applications in which the half life of the antibody in vivo is important yet certain effector functions (such as complement and ADCC) are unnecessary or deleterious. In vitro and/or in vivo cytotoxicity assays can be conducted to confirm the reduction/depletion of CDC and/or ADCC activities. For example, Fc receptor (FcR) binding assays can be conducted to ensure that the antibody lacks FcyR binding (hence likely lacking ADCC activity), but retains FcRn binding ability. The primary cells for mediating ADCC, NK cells, express Fc(RIII only, whereas monocytes express Fc(RI, Fc(RII and Fc(RIII. FcR expression on hematopoietic cells is summarized in Table 3 on page 464 of Ravetch, J.V., and Kinet, Annu. Rev. Immunol. 9 (1991) 457-492. Non-limiting examples of in vitro assays to assess ADCC activity of a molecule of interest is described in U.S. Patent No. 5,500,362 (see, e.g. Hellstrom, I., et al, Proc. Nat'l Acad. Sci. USA 83 (1986) 7059-7063) and Hellstrom, I., et al, Proc. Nat'l Acad. Sci. USA 82 (1985) 1499- 1502; US 5,821,337 (see Bruggemann, M., et al, J. Exp. Med. 166 (1987) 1351- 1361). Alternatively, non-radioactive assays methods may be employed (see, for example, ACTI™ non-radioactive cytotoxicity assay for flow cytometry (CellTechnology, Inc. Mountain View, CA; and CytoTox 96® non-radioactive cytotoxicity assay (Promega, Madison, WI). Useful effector cells for such assays include peripheral blood mononuclear cells (PBMC) and Natural Killer (NK) cells. Alternatively, or additionally, ADCC activity of the molecule of interest may be assessed in vivo, e.g., in a animal model such as that disclosed in Clynes, R., et al, Proc. Natl. Acad. Sci. USA 95 (1998) 652-656. Clq binding assays may also be carried out to confirm that the antibody is unable to bind Clq and hence lacks CDC activity. See, e.g., Clq and C3c binding ELISA in WO 2006/029879 and WO 2005/100402. To assess complement activation, a CDC assay may be performed (see, for example, Gazzano-Santoro, H., et al., J. Immunol. Methods 202 (1997) 163ff; Cragg, M.S., et al, Blood 101 (2003) 1045-1052; and Cragg, M.S. and Glenni, M.J., Blood 103 (2004) 2738-2743). FcRn binding and in vivo clearance/half life determinations can also be performed using methods known in the art (see, e.g., Petkova, S.B., et al, Int'l. Immunol. 18 (2006) 1759-1769).
Antibodies with reduced effector function include those with substitution of one or more of Fc region residues 238, 265, 269, 270, 297, 327 and 329 (U.S. Patent No. 6,737,056). Such Fc mutants include Fc mutants with substitutions at two or more of amino acid positions 265, 269, 270, 297 and 327, including the so-called "DANA" Fc mutant with substitution of residues 265 and 297 to alanine (US Patent No. 7,332,581).
Certain antibody variants with improved or diminished binding to FcRs are described. (See, e.g., U.S. Patent No. 6,737,056; WO 2004/056312, and Shields, R.L., et al, J. Biol. Chem. 9 (2001) 6591-6604).
In certain embodiments, an antibody variant comprises an Fc region with one or more amino acid substitutions which improve ADCC, e.g., substitutions at positions 298, 333, and/or 334 of the Fc region (EU numbering of residues).
In some embodiments, alterations are made in the Fc region that result in altered (i.e., either improved or diminished) Clq binding and/or Complement Dependent Cytotoxicity (CDC), e.g., as described in US Patent No. 6,194,551, WO 99/51642, and Idusogie, E.E., et al, J. Immunol. 164 (2000) 4178-4184.
Antibodies with increased half lives and improved binding to the neonatal Fc receptor (FcRn), which is responsible for the transfer of maternal IgGs to the fetus (Guyer, R.L., et al, J. Immunol. 117 (1976) 587-593, and Kim, J.K., et al, European Journalof Immunology 24 (1994) 2429-2434), are described in US 2005/0014934A1 (Hinton et al.). Those antibodies comprise an Fc region with one or more substitutions therein which improve binding of the Fc region to FcRn. Such Fc variants include those with substitutions at one or more of Fc region residues: 238, 256, 265, 272, 286, 303, 305, 307, 311, 312, 317, 340, 356, 360, 362, 376, 378, 380, 382, 413, 424 or 434, e.g., substitution of Fc region residue 434 (US Patent No. 7,371,826).
See also Duncan, A.R., and Winter, G., Nature 332 (1988) 738-740; U.S. Patent No. 5,648,260; U.S. Patent No. 5,624,821; and WO 94/29351 concerning other examples of Fc region variants. c) Cysteine engineered antibody variants
In certain embodiments, it may be desirable to create cysteine engineered antibodies, e.g., "thioMAbs," in which one or more residues of an antibody are substituted with cysteine residues. In particular embodiments, the substituted residues occur at accessible sites of the antibody. By substituting those residues with cysteine, reactive thiol groups are thereby positioned at accessible sites of the antibody and may be used to conjugate the antibody to other moieties, such as drug moieties or linker-drug moieties, to create an immunoconjugate, as described further herein. In certain embodiments, any one or more of the following residues may be substituted with cysteine: V205 (Kabat numbering) of the light chain; Al 18 (EU numbering) of the heavy chain; and S400 (EU numbering) of the heavy chain Fc region. Cysteine engineered antibodies may be generated as described, e.g., in U.S. Patent No. 7,521,541. d) Antibody Derivatives
In certain embodiments, an antibody provided herein may be further modified to contain additional nonproteinaceous moieties that are known in the art and readily available. The moieties suitable for derivatization of the antibody include but are not limited to water soluble polymers. Non-limiting examples of water soluble polymers include, but are not limited to, polyethylene glycol (PEG), copolymers of ethylene glycol/propylene glycol, carboxymethylcellulose, dextran, polyvinyl alcohol, polyvinyl pyrrolidone, poly-1, 3-dioxolane, poly-l,3,6-trioxane, ethylene/maleic anhydride copolymer, polyaminoacids (either homopolymers or random copolymers), and dextran or poly(n-vinyl pyrrolidone)poly ethylene glycol, propropylene glycol homopolymers, prolypropylene oxide/ethylene oxide co-polymers, polyoxyethylated polyols (e.g., glycerol), polyvinyl alcohol, and mixtures thereof. Polyethylene glycol propionaldehyde may have advantages in manufacturing due to its stability in water. The polymer may be of any molecular weight, and may be branched or unbranched. The number of polymers attached to the antibody may vary, and if more than one polymer are attached, they can be the same or different molecules. In general, the number and/or type of polymers used for derivatization can be determined based on considerations including, but not limited to, the particular properties or functions of the antibody to be improved, whether the antibody derivative will be used in a therapy under defined conditions, etc.
In another embodiment, conjugates of an antibody and nonproteinaceous moiety that may be selectively heated by exposure to radiation are provided. In one embodiment, the nonproteinaceous moiety is a carbon nanotube (Kam, N.W., et al, Proc. Natl. Acad. Sci. USA 102 (2005) 11600-11605). The radiation may be of any wavelength, and includes, but is not limited to, wavelengths that do not harm ordinary cells, but which heat the nonproteinaceous moiety to a temperature at which cells proximal to the antibody-nonproteinaceous moiety are killed. B. Recombinant Methods and Compositions
Antibodies may be produced using recombinant methods and compositions, e.g., as described in U.S. Patent No. 4,816,567. In one embodiment, isolated nucleic acid encoding an anti-IGF-lR antibody described herein is provided. Such nucleic acid may encode an amino acid sequence comprising the VL and/or an amino acid sequence comprising the VH of the antibody (e.g., the light and/or heavy chains of the antibody). In a further embodiment, one or more vectors (e.g., expression vectors) comprising such nucleic acid are provided. In a further embodiment, a host cell comprising such nucleic acid is provided. In one such embodiment, a host cell comprises (e.g., has been transformed with): (1) a vector comprising a nucleic acid that encodes an amino acid sequence comprising the VL of the antibody and an amino acid sequence comprising the VH of the antibody, or (2) a first vector comprising a nucleic acid that encodes an amino acid sequence comprising the VL of the antibody and a second vector comprising a nucleic acid that encodes an amino acid sequence comprising the VH of the antibody. In one embodiment, the host cell is eukaryotic, e.g. a Chinese Hamster Ovary (CHO) cell or lymphoid cell (e.g., Y0, NSO, Sp20 cell). In one embodiment, a method of making an anti-IGF-lR antibody is provided, wherein the method comprises culturing a host cell comprising a nucleic acid encoding the antibody, as provided above, under conditions suitable for expression of the antibody, and optionally recovering the antibody from the host cell (or host cell culture medium).
For recombinant production of an anti-IGF-lR antibody, nucleic acid encoding an antibody, e.g., as described above, is isolated and inserted into one or more vectors for further cloning and/or expression in a host cell. Such nucleic acid may be readily isolated and sequenced using conventional procedures (e.g., by using oligonucleotide probes that are capable of binding specifically to genes encoding the heavy and light chains of the antibody).
Suitable host cells for cloning or expression of antibody-encoding vectors include prokaryotic or eukaryotic cells described herein. For example, antibodies may be produced in bacteria, in particular when glycosylation and Fc effector function are not needed. For expression of antibody fragments and polypeptides in bacteria, see, e.g., U.S. Patent Nos. 5,648,237, 5,789,199, and 5,840,523. (See also Charlton, K.A., Methods in Molecular Biology, Vol. 248 (B.K.C. Lo, ed., Humana Press, Totowa, NJ), (2004) pp. 245-254, describing expression of antibody fragments in E. coli.) After expression, the antibody may be isolated from the bacterial cell paste in a soluble fraction and can be further purified.
In addition to prokaryotes, eukaryotic microbes such as filamentous fungi or yeast are suitable cloning or expression hosts for antibody-encoding vectors, including fungi and yeast strains whose glycosylation pathways have been "humanized," resulting in the production of an antibody with a partially or fully human glycosylation pattern. See Gerngross, T.U., Nat. Biotech. 22 (2004) 1409-1414, and Li, H., et al, Nat. Biotech. 24 (2006) 210-215.
Suitable host cells for the expression of glycosylated antibody are also derived from multicellular organisms (invertebrates and vertebrates). Examples of invertebrate cells include plant and insect cells. Numerous baculoviral strains have been identified which may be used in conjunction with insect cells, particularly for transfection of Spodoptera frugiperda cells.
Plant cell cultures can also be utilized as hosts. See, e.g., US Patent Nos. 5,959,177, 6,040,498, 6,420,548, 7,125,978, and 6,417,429 (describing PLANTIBODIES™ technology for producing antibodies in transgenic plants).
Vertebrate cells may also be used as hosts. For example, mammalian cell lines that are adapted to grow in suspension may be useful. Other examples of useful mammalian host cell lines are monkey kidney CV1 line transformed by SV40 (COS-7); human embryonic kidney line (293 or 293 cells as described, e.g., in Graham, F.L., et al, J. Gen Virol. 36 (1977) 59-74); baby hamster kidney cells (BHK); mouse Sertoli cells (TM4 cells as described, e.g., in Mather, J.P., Biol. Reprod. 23 (1980) 243-252); monkey kidney cells (CV1); African green monkey kidney cells (VERO-76); human cervical carcinoma cells (HELA); canine kidney cells (MDCK; buffalo rat liver cells (BRL 3A); human lung cells (W138); human liver cells (Hep G2); mouse mammary tumor (MMT 060562); TRI cells, as described, e.g., in Mather, J.P., et al, Annals N.Y. Acad. Sci. 383 (1982) 44-68; MRC 5 cells; and FS4 cells. Other useful mammalian host cell lines include Chinese hamster ovary (CHO) cells, including DHFR- CHO cells (Urlaub et al, Proc. Natl. Acad. Sci. USA 77 (1980) 4216-4220); and myeloma cell lines such as Y0, NSO and Sp2/0. For a review of certain mammalian host cell lines suitable for antibody production, see, e.g., Yazaki, P. J., and Wu, A.M., Methods in Molecular Biology, Antibody Engineering: Methods and Protocols, Vol. 248 (B.K.C. Lo, (ed.), Humana Press, Totowa, NJ), (2004) pp. 255-268. C. Immunoconjugates
The invention also provides immunoconjugates comprising an anti-IGF-lR antibody herein conjugated to one or more cytotoxic agents, such as chemotherapeutic agents or drugs, growth inhibitory agents, toxins (e.g., protein toxins, enzymatically active toxins of bacterial, fungal, plant, or animal origin, or fragments thereof), or radioactive isotopes.
In one embodiment, an immunoconjugate is an antibody-drug conjugate (ADC) in which an antibody is conjugated to one or more drugs, including but not limited to a maytansinoid (see U.S. Patent Nos. 5,208,020, 5,416,064 and European Patent EP 0 425 235 Bl); an auristatin such as monomethylauristatin drug moieties DE and DF (MMAE and MMAF) (see U.S. Patent Nos. 5,635,483 and 5,780,588, and 7,498,298); a dolastatin; a calicheamicin or derivative thereof (see U.S. Patent Nos. 5,712,374, 5,714,586, 5,739,116, 5,767,285, 5,770,701, 5,770,710, 5,773,001, and 5,877,296; Hinman, L.M., et al, Cancer Res. 53 (1993) 3336-3342; and Lode, H.N., et al, Cancer Res. 58 (1998) 2925-2928); an anthracycline such as daunomycin or doxorubicin (see Kratz, F., et al, Current Med. Chem. 13 (2006) 477-523; Jeffrey, S.C., et al, Bioorganic & Med. Chem. Letters 16 (2006) 358- 362; Torgov, M.Y., et al, Bioconj. Chem. 16 (2005) 717-721; Nagy, A., et al, Proc. Natl. Acad. Sci. USA 97 (2000) 829-834; Dubowchik, G.M., et al, Bioorg. & Med. Chem. Letters 12 (2002) 1529-1532; King, H.D., et al, J. Med. Chem. 45 (2002) 4336-4343; and U.S. Patent No. 6,630,579); methotrexate; vindesine; a taxane such as docetaxel, paclitaxel, larotaxel, tesetaxel, and ortataxel; a trichothecene; and CC1065.
In another embodiment, an immunoconjugate comprises an antibody as described herein conjugated to an enzymatically active toxin or fragment thereof, including but not limited to 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, PAPII, and PAP-S), momordica charantia inhibitor, curcin, crotin, sapaonaria officinalis inhibitor, gelonin, mitogellin, restrictocin, phenomycin, enomycin, and the tricothecenes.
In another embodiment, an immunoconjugate comprises an antibody as described herein conjugated to a radioactive atom to form a radioconjugate. A variety of radioactive isotopes are available for the production of radioconjugates. Examples include At211, I131, I125, Y90, Re186, Re188, Sm153, Bi212, P32, Pb212 and radioactive isotopes of Lu. When the radioconjugate is used for detection, it may comprise a radioactive atom for scintigraphic studies, for example tc99m or 1123, or a spin label for nuclear magnetic resonance (NMR) imaging (also known as magnetic resonance imaging, mri), such as iodine- 123 again, iodine-131, indium-I l l, fluorine- 19, carbon- 13, nitrogen- 15, oxygen- 17, gadolinium, manganese or iron.
Conjugates of an antibody and cytotoxic agent may be made using a variety of bifunctional protein coupling agents such as N-succinimidyl-3-(2-pyridyldithio) propionate (SPDP), succinimidyl-4-(N-maleimidomethyl) cyclohexane-1- carboxylate (SMCC), iminothiolane (IT), bifunctional derivatives of imidoesters (such as dimethyl adipimidate HC1), active esters (such as disuccinimidyl suberate), aldehydes (such as glutaraldehyde), bis-azido compounds (such as bis (p-azidobenzoyl) hexanediamine), bis-diazonium derivatives (such as bis- (p-diazoniumbenzoyl)-ethylenediamine), diisocyanates (such as toluene 2,6- diisocyanate), and bis-active fluorine compounds (such as l,5-difluoro-2,4- dinitrobenzene). For example, a ricin immunotoxin can be prepared as described in Vitella, E.S., et al, Science 238 (1987) 1098-1104. Carbon- 14-labeled 1- isothiocyanatobenzyl-3-methyldiethylene triaminepentaacetic acid (MX-DTPA) is an exemplary chelating agent for conjugation of radionucleotide to the antibody. See WO 94/11026. The linker may be a "cleavable linker" facilitating release of a cytotoxic drug in the cell. For example, an acid-labile linker, peptidase-sensitive linker, photolabile linker, dimethyl linker or disulfide-containing linker (Chari, R.V., et al, Cancer Res. 52 (1992) 127-131; U.S. Patent No. 5,208,020) may be used.
The immunuoconjugates or ADCs herein expressly contemplate, but are not limited to such conjugates prepared with cross-linker reagents including, but not limited to, BMPS, EMCS, GMBS, HBVS, LC-SMCC, MBS, MPBH, SBAP, SIA, SIAB, SMCC, SMPB, SMPH, sulfo-EMCS, sulfo-GMBS, sulfo-KMUS, sulfo-MBS, sulfo-SIAB, sulfo-SMCC, and sulfo-SMPB, and SVSB (succinimidyl- (4-vinylsulfone)benzoate) which are commercially available (e.g., from Pierce Biotechnology, Inc., Rockford, IL., U.S.A).
D. Methods and Compositions for Diagnostics and Detection
In certain embodiments, any of the anti-IGF-lR antibodies provided herein is useful for detecting the presence of IGF-1R in a biological sample. The term "detecting" as used herein encompasses quantitative or qualitative detection. In certain embodiments, a biological sample comprises a cell or tissue, such as tumor tissue.
In one embodiment, an anti-IGF-lR antibody for use in a method of diagnosis or detection is provided. In a further aspect, a method of detecting the presence of IGF-1R in a biological sample is provided. In certain embodiments, the method comprises contacting the biological sample with an anti-IGF-lR antibody as described herein under conditions permissive for binding of the anti-IGF-lR antibody to IGF-1R, and detecting whether a complex is formed beTween® the anti-IGF-lR antibody and IGF-1R. Such method may be an in vitro or in vivo method. In one embodiment, an anti-IGF-lR antibody is used to select subjects eligible for therapy with an anti-IGF-lR antibody, e.g. where IGF-1R is a biomarker for selection of patients.
Exemplary disorders that may be diagnosed using an antibody of the invention is tumor tissue.
In certain embodiments, labeled anti-IGF-lR antibodies are provided. Labels include, but are not limited to, labels or moieties that are detected directly (such as fluorescent, chromophoric, electron-dense, chemiluminescent, and radioactive labels), as well as moieties, such as enzymes or ligands, that are detected indirectly, e.g., through an enzymatic reaction or molecular interaction. Exemplary labels
32 14 125 3 131 include, but are not limited to, the radioisotopes P, C, I, H, and I, fluorophores such as rare earth chelates or fluorescein and its derivatives, rhodamine and its derivatives, dansyl, umbelliferone, luceriferases, e.g., firefly luciferase and bacterial luciferase (U.S. Patent No. 4,737,456), luciferin, 2,3-dihydrophthalazinediones, horseradish peroxidase (HRP), alkaline phosphatase, β-galactosidase, glucoamylase, lysozyme, saccharide oxidases, e.g., glucose oxidase, galactose oxidase, and glucose-6-phosphate dehydrogenase, heterocyclic oxidases such as uricase and xanthine oxidase, coupled with an enzyme that employs hydrogen peroxide to oxidize a dye precursor such as HRP, lactoperoxidase, or microperoxidase, biotin/avidin, spin labels, bacteriophage labels, stable free radicals, and the like.
E. Pharmaceutical Formulations
Pharmaceutical formulations of an anti-IGF-lR antibody as described herein are prepared by mixing such antibody having the desired degree of purity with one or more optional pharmaceutically acceptable carriers (Osol, A., et al., (eds.) Remington's Pharmaceutical Sciences, 16th edition, Mack Publishing Company, Pennsylvania (1980)), in the form of lyophilized formulations or aqueous solutions. Pharmaceutically acceptable carriers are generally nontoxic to recipients at the dosages and concentrations employed, and include, but are not limited to: buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyldimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride; benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); low molecular weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, histidine, arginine, or lysine; monosaccharides, disaccharides, and other carbohydrates including glucose, mannose, or dextrins; chelating agents such as EDTA; sugars such as sucrose, mannitol, trehalose or sorbitol; salt-forming counter-ions such as sodium; metal complexes (e.g. Zn-protein complexes); and/or non-ionic surfactants such as polyethylene glycol (PEG). Exemplary pharmaceutically acceptable carriers herein further include insterstitial drug dispersion agents such as soluble neutral-active hyaluronidase glycoproteins (sHASEGP), for example, human soluble PH-20 hyaluronidase glycoproteins, such as rHuPH20 (HYLENEX®, Baxter International, Inc.). Certain exemplary sHASEGPs and methods of use, including rHuPH20, are described in US Patent Publication Nos. 2005/0260186 and 2006/0104968. In one aspect, a sHASEGP is combined with one or more additional glycosaminoglycanases such as chondroitinases.
Exemplary lyophilized antibody formulations are described in US Patent No. 6,267,958. Aqueous antibody formulations include those described in US Patent No. 6,171,586 and WO 2006/044908, the latter formulations including a histidine- acetate buffer.
The formulation herein may also contain more than one active ingredients as necessary for the particular indication being treated, preferably those with complementary activities that do not adversely affect each other. For example, it may be desirable to further provide gemcitabine or gemcitabine in combination with erlotinib. Such active ingredients are suitably present in combination in amounts that are effective for the purpose intended. Active ingredients may be entrapped in microcapsules prepared, for example, by coacervation techniques or by interfacial polymerization, for example, hydroxymethylcellulose or gelatin-microcapsules and poly-(methylmethacylate) microcapsules, respectively, in colloidal drug delivery systems (for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules) or in macroemulsions. Such techniques are disclosed in Osol, A., et al., (eds.) Remington's Pharmaceutical Sciences 16th edition, Mack Publishing Company, Pennsylvania (1980).
Sustained-release preparations may be prepared. Suitable examples of sustained- release preparations include semipermeable matrices of solid hydrophobic polymers containing the antibody, which matrices are in the form of shaped articles, e.g. films, or microcapsules.
The formulations to be used for in vivo administration are generally sterile. Sterility may be readily accomplished, e.g., by filtration through sterile filtration membranes.
F. Therapeutic Methods and Compositions
Any of the anti-IGF-lR antibodies provided herein may be used in therapeutic methods.
In one aspect, an anti-IGF-lR antibody for use as a medicament is provided. In further aspects, an anti-IGF-lR antibody for use in treating tumor disease is provided. In certain embodiments, an anti-IGF-lR antibody for use in a method of treatment is provided. In certain embodiments, the invention provides an anti-IGF- 1R antibody for use in a method of treating an individual having tumor disease comprising administering to the individual an effective amount of the anti-IGF-lR antibody. In one such embodiment, the method further comprises administering to the individual an effective amount of at least one additional therapeutic agent, e.g., as described below. In further embodiments, the invention provides an anti-IGF-lR antibody for use in inhibiting cell proliferation. In certain embodiments, the invention provides an anti-IGF-lR antibody for use in a method inhibiting cell proliferationin an individual comprising administering to the individual an effective of the anti-IGF-lR antibody. An "individual" according to any of the above embodiments is preferably a human. In a further aspect, the invention provides for the use of an anti-IGF-lR antibody in the manufacture or preparation of a medicament. In one embodiment, the medicament is for treatment of tumor disease. In a further embodiment, the medicament is for use in a method of treating tumor disease comprising administering to an individual having tumor disease an effective amount of the medicament. In one such embodiment, the method further comprises administering to the individual an effective amount of at least one additional therapeutic agent, e.g., as described below. In a further embodiment, the medicament is for inhibiting cell proliferation. In a further embodiment, the medicament is for use in a method of inhibiting cell proliferation in an individual comprising administering to the individual an amount effective of themedicament. An "individual" according to any of the above embodiments may be a human.
In a further aspect, the invention provides a method for treating a tumor disease. In one embodiment, the method comprises administering to an individual having such tumor disease an effective amount of an anti-IGF-lR antibody. In one such embodiment, the method further comprises administering to the individual an effective amount of at least one additional therapeutic agent, as described below. An "individual" according to any of the above embodiments may be a human.
In a further aspect, the invention provides a method for inhibiting cell proliferation in an individual. In one embodiment, the method comprises administering to the individual an effective amount of an anti-IGF-lR antibody. In one embodiment, an "individual" is a human.
In a further aspect, the invention provides pharmaceutical formulations comprising any of the anti-IGF-lR antibodies provided herein, e.g., for use in any of the above therapeutic methods. In one embodiment, a pharmaceutical formulation comprises any of the anti-IGF-lR antibodies provided herein and a pharmaceutically acceptable carrier. In another embodiment, a pharmaceutical formulation comprises any of the anti-IGF-lR antibodies provided herein and at least one additional therapeutic agent, e.g., as described below.
Antibodies of the invention can be used either alone or in combination with other agents in a therapy. For instance, an antibody of the invention may be co-administered with at least one additional therapeutic agent. In certain embodiments, an additional therapeutic agent is gemcitabine. Such combination therapies noted above encompass combined administration (where two or more therapeutic agents are included in the same or separate formulations), and separate administration, in which case, administration of the antibody of the invention can occur prior to, simultaneously, and/or following, administration of the additional therapeutic agent and/or adjuvant. Antibodies of the invention can also be used in combination with radiation therapy.
An antibody of the invention (and any additional therapeutic agent) can be administered by any suitable means, including parenteral, intrapulmonary, and intranasal, and, if desired for local treatment, intralesional administration. Parenteral infusions include intramuscular, intravenous, intraarterial, intraperitoneal, or subcutaneous administration. Dosing can be by any suitable route, e.g. by injections, such as intravenous or subcutaneous injections, depending in part on whether the administration is brief or chronic. Various dosing schedules including but not limited to single or multiple administrations over various time -points, bolus administration, and pulse infusion are contemplated herein.
Antibodies of the invention would be formulated, dosed, and administered in a fashion consistent with good medical practice. Factors for consideration in this context include the particular disorder being treated, the particular mammal being treated, the clinical condition of the individual patient, the cause of the disorder, the site of delivery of the agent, the method of administration, the scheduling of administration, and other factors known to medical practitioners. The antibody need not be, but is optionally formulated with one or more agents currently used to prevent or treat the disorder in question. The effective amount of such other agents depends on the amount of antibody present in the formulation, the type of disorder or treatment, and other factors discussed above. These are generally used in the same dosages and with administration routes as described herein, or about from 1 to 99% of the dosages described herein, or in any dosage and by any route that is empirically/clinically determined to be appropriate.
For the prevention or treatment of disease, the appropriate dosage of an antibody of the invention (when used alone or in combination with one or more other additional therapeutic agents) will depend on the type of disease to be treated, the type of antibody, the severity and course of the disease, whether the antibody is administered for preventive or therapeutic purposes, previous therapy, the patient's clinical history and response to the antibody, and the discretion of the attending physician. The antibody is suitably administered to the patient at one time or over a series of treatments. Depending on the type and severity of the disease, about 1 μg/kg to 15 mg/kg (e.g. O.lmg/kg-lOmg/kg) of antibody can be an initial candidate dosage for administration to the patient, whether, for example, by one or more separate administrations, or by continuous infusion. One typical daily dosage might range from about 1 μg/kg to 100 mg/kg or more, depending on the factors mentioned above. For repeated administrations over several days or longer, depending on the condition, the treatment would generally be sustained until a desired suppression of disease symptoms occurs. One exemplary dosage of the antibody would be in the range from about 0.05 mg/kg to about 10 mg/kg. Thus, one or more doses of about 0.5 mg/kg, 2.0 mg/kg, 4.0 mg/kg or 10 mg/kg (or any combination thereof) may be administered to the patient. Such doses may be administered intermittently, e.g. every week or every three weeks (e.g. such that the patient receives from about two to about twenty, or e.g. about six doses of the antibody). An initial higher loading dose, followed by one or more lower doses may be administered. An exemplary dosing regimen comprises administering about 4 to 10 mg/kg. However, other dosage regimens may be useful. The progress of this therapy is easily monitored by conventional techniques and assays.
It is understood that any of the above formulations or therapeutic methods may be carried out using an immunoconjugate of the invention in place of or in addition to an anti-IGF-lR antibody.
G. Articles of Manufacture
In another aspect of the invention, an article of manufacture containing materials useful for the treatment, prevention and/or diagnosis of the disorders described above is provided. The article of manufacture comprises a container and a label or package insert on or associated with the container. Suitable containers include, for example, bottles, vials, syringes, IV solution bags, etc. The containers may be formed from a variety of materials such as glass or plastic. The container holds a composition which is by itself or combined with another composition effective for treating, preventing and/or diagnosing the condition and may have a sterile access port (for example the container may be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle). At least one active agent in the composition is an antibody of the invention. The label or package insert indicates that the composition is used for treating the condition of choice. Moreover, the article of manufacture may comprise (a) a first container with a composition contained therein, wherein the composition comprises an antibody of the invention; and (b) a second container with a composition contained therein, wherein the composition comprises a further cytotoxic or otherwise therapeutic agent. The article of manufacture in this embodiment of the invention may further comprise a package insert indicating that the compositions can be used to treat a particular condition. Alternatively, or additionally, the article of manufacture may further comprise a second (or third) container comprising a pharmaceutically- acceptable buffer, such as bacteriostatic water for injection (BWFI), phosphate- buffered saline, Ringer's solution and dextrose solution. It may further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, needles, and syringes.
It is understood that any of the above articles of manufacture may include an immunoconjugate of the invention in place of or in addition to an anti-IGF-lR antibody.
III. EXAMPLES
The following are examples of methods and compositions of the invention. It is understood that various other embodiments may be practiced, given the general description provided above.
Although the foregoing invention has been described in some detail by way of illustration and example for purposes of clarity of understanding, the descriptions and examples should not be construed as limiting the scope of the invention. The disclosures of all patent and scientific literature cited herein are expressly incorporated in their entirety by reference.
Example 1
Antibody generation
Single amino acid substitutions in the VH CDRH3 region of mAb IR18 (9 positions: ELGRRYFDL) were generated based on a plasmid encoding the wild- type immunoglobulin heavy chain of mAb IR18. To this end, individual DNA fragments encoding the altered CDRH3 regions for the respective single amino acid substitutions were generated synthetically and cloned into the corresponding position of the plasmid.
Plasmid-DNA was prepared from all clones and co-transfected into HEK293 cells together with a plasmid encoding the wild-type light chain of mAb IR18. In brief, lxl 06 HEK293 cells were co-transfected with plasmids encoding i) a mutant heavy chain and ii) the wild-type light chain gene. Transfection was performed in 24-well plates in a culture volume of 1 ml using the 293-fection reagent. After seven days of culture the supernatants were harvested, frozen, and stored at -80°C. Later, the antibodies were purified using Protein G sepharose affinity chromatography, and the antibody concentrations were determined using an ELISA assay specific for human immunoglobulin. Subsequently, all supernatants containing human immunoglobulin were analysed for their binding affinity to the human IGFl-R using the Biacore technology.
Example 2
Antibody Affinity
The kinetic constants of mAb IR18 and its CDRH3 variants binding IGFl-R were measured via BiaCore. To this end, the antibodies were captured to a CI -Chip via an amine-coupled capture antibody (goat-anti-human IgG). Ligand-loading of the capture antibody was ca. 200 RU (-1.3 fmol/mm2). The concentration of the mAbs was 1 nM in phosphate-buffered saline (PBS) containing 0.05% Tween® 20 and 0.1%) (w/v) bovine serum albumin (BSA). Capturing was performed for 40 seconds.
Human IGFl-R ECD (R&D Systems Cat. No. 391-GR) was used at concentrations of c = 400 nM, 200 nM, 100 nM, 25 nM, 6.25 nM, and 1.5625 nM. Samples were diluted in PBS containing 0.05% Tween® 20 and 0.1% BSA (w/v), running buffer was PBS containing 0.05% Tween® 20. A classical concentration series was measured (120 seconds association, 10 minutes dissociation, 6 concentrations). The flow rate was 30 μΕ/ηιίη.
After each cycle the chip was regenerated using 10 mM glycine pH 1.5 (2 minutes injection time) after which the intact, free capture antibody was available again.
The use of a CI -chip and the low capture level of the antibodies which were to be tested (5-10 RU) made sure that the binding of the dimeric IGFl-R molecules was monovalent. This way the distance between the neighbouring antibodies was sufficiently large to make sure that each dimeric IGFl-R molecule was able to interact with only one antibody. In such a setting the kj-rate is significantly higher than in experiments with higher antibody density causing bivalent binding of the receptor. Results are shown in table 1
Table 1:
Figure imgf000039_0001
Example 3
Inhibition of IGF-I and IGF-II binding to tumor cells expressing IGF-IR
In order to determine the ability of the antibody of the invention to block binding of the ligands IGF-I and IGF-II to the IGF-I receptor (IGF-IR), competition experiments with radioactively labeled ligand peptides are performed.
Human tumor cells (HT29, NCI H322M, 0.5 to 1 x 105/ml) are plated in RPMI 1640 medium (PAA, Cat. No. El 5-039) supplemented with 2 mM L-Glutamin, lx non-essential amino acids (Gibco, Cat. No. 11140-035), 1 mM sodium pyruvate (Gibco, Cat. No. 11360-039) and 10% heat inactivated FCS (PAA, Cat. No. A15- 771). Six bottles in the T175 format are inoculated with 20 ml cells in the respective medium for each experiment and cultivated for two days at 37°C and 5% C02 to obtain confluent cell monolayers.
To collect individual cells, 2 ml of lx Trypsin/EDTA (Gibco, Cat. No. 25300-054) per T175 flask are added and detachment of cells monitored with a Zeiss Axiovert25 microscope. The cells are collected and medium with 10% FCS as described before is added to a total volume of 50 ml. Cells are reisolated by centrifugation for 10 minutes at 1000 rpm (Heraeus sepatech, Omnifuge 2.0 RS) and resuspended in 50 ml of binding buffer (120 mM NaCl, 5 mM KC1, 1.2 mM MgS04, 1 mM EDTA, 10 mM D(+)glucose, 15 mM NaAc, 100 mM Hepes pH 7.6, 1%) BSA). Cells are counted, reisolated by centrifugation and adjusted with binding buffer to 1 x 106 cells/ml.
1125-labeled IGF-I and IGF-II peptides (Amersham, -2000 Ci/mmol, Cat. No. IM172 and IM238), solubilized in 0.1% CH3COOH, are diluted in binding buffer to a final activity of 4 x 105 counts/(minute x ml). 75 μΐ of antibody at the specified concentrations together with 25 μΐ of prediluted 1125-labeled IGF-I or IGF-II peptide is added to 200 μΐ of cell suspension and incubated for 3,5 h at 4°C. Cells are reisolated by centrifugation for 5 minutes at 2000 rpm (Eppendorf, 5415C) and supernatant removed. After washing two times in 1 ml binding buffer, cells are resuspended in 1 ml binding buffer and transferred to scintillation tubes. The amount of radioactive peptide bound to the cell surface receptors is measured on a scintillation counter.
Example 4
Inhibition of IGF-I mediated phosphorylation of IGF-IR and Akt/PKB
In order to determine the ability of the antibody of the invention to inhibit activation and phosphorylation of the IGF-I receptor (IGF-IR), competition experiments are performed with IGF-I peptide and subsequent Western blotting analysis with antibodies specific for phosphorylated tyrosine.
Human tumor cells (HT29, NCI H322M, 5 x 104/ml) are plated in RPMI 1640 medium (PAA, Cat. No. E15-039) supplemented with 2 mM L-Glutamin, lx nonessential aminoacids (Gibco, Cat. No. 11140-035), 1 mM sodium pyruvate (Gibco, Cat. No. 11360-039) and 0.5% heat inactivated FCS (PAA, Cat. No. A15-771). For determination of IC50 values, 12 well plates are inoculated with 1 ml cells in the respective medium for each experiment and cultivated for two days at 37°C and 5% C02.
After 48 hours of cultivation with low serum medium, the medium is carefully removed and replaced by different concentrations of antibody diluted in the respective medium. After 5 minutes incubation at 37°C and 5% C02 IGF-I peptide is added at a final concentration of 2 nM and cells are again incubated for 10 minutes under the conditions mentioned above. The medium is carefully removed by aspiration and 100 μΐ of cold lysis buffer is added per well (50mM Hepes pH 7.2, 150 mM NaCl, lmM EGTA, 10% glycerol, 1% Triton™ -XI 00, lOOmM NaF, 10 mM Na4P207, Complete™ protease inhibitor). The cells are detached using a cell scraper (Corning, Cat. No. 3010) and well contents transferred to Eppendorf reaction tubes. Cell fragments are removed by centrifugation for 10 minutes at 13000 rpm and 4°C and half of the supernatant is added to 2x Laemmli sample buffer in a 1 : 1 (v/v) ratio. For immunoprecipitation of IGF-IR, the remaining supernatant of cell lysates underwent a clearifying spin (10 minutes at 13000 rpm and 4°C) right before 1 μΐ of an polyclonal antibody against IGF-IRB (C-20, Santa Cruz Biotechnologies) or a murine monoclonal antibody (IgGl) which recognizes an epitope within amino acids 440-586 of the extracellular domain (a-chain) of the human IGF Type 1 Receptor is added (mAb 24-55, GroPep). After 2 hours incubation at 4°C in a rotating Eppendorf reaction tube, 25 μΐ Protein G Sepharose™ beads (Amersham Biosciences, Cat. No. 17-0618-01) are added followed by another incubation step of 1 hour at 4°C. The beads with bound antibody-protein-complexes are isolated by centrifugation (1 minute at 2000 rpm and 4°C) and ished three times with wash buffer (lysis buffer with only 0.1% Triton™ -XI 00). After boiling the beads in Laemmli sample buffer, cellular proteins are separated by SDS-PAGE and transferred to a nitrocellulose membrane (PROTRAN™ BA 85, Schleicher&Schuell) by semi-dry Western blotting.
A phosphotyrosine specific antibody (Upstate, clone 4G10, Cat. No. 05-321) is used to determine phosphorylation status of immunopurified IGF-IR. For the detection of phosphorylated Akt/PKB an antibody with specificity for phosphorylated Ser473 (Cell Signalling, Cat. No. 9271) is applied. Example 5
Induction of antibody mediated downregulation of IGF-IR in-vitro
In order to detect effects of the antibody of the invention on the amount of IGF-I receptor (IGF-IR) in tumor cells, time-course experiments and subsequent western- blotting analysis with IGF-IR specific antibodies are performed.
Human tumor cells (HT29, 5 x 104 cells/ml) in RPMI 1640 medium (PAA, Cat. No. El 5-039) supplemented with 2 mM L-Glutamin, lx non-essential aminoacids (Gibco, Cat. No. 11140-035), 1 mM sodium pyruvate (Gibco, Cat. No. 11360-039) and 10% heat inactivated FCS (PAA, Cat. No. A15-771). For each incubation period one 12 well plate is inoculated with 1 ml cells in the respective medium for each experiment and cultivated for 24 hours at 37°C and 5% C02.
The medium is carefully removed and replaced by different concentrations of antibody diluted in the respective medium. In two control wells, medium is replaced by either medium without antibody or medium with a control antibody (AB-1, Oncogene, Cat. No. GR11). Cells are incubated at 37°C and 5% C02 and individual plates are taken out for further processing after 15 minutes, 24 hours and 48 hours.
The medium is carefully removed by aspiration and 100 μΐ of cold lysis buffer is added per well (50mM Hepes pH 7.2, 150 mM NaCl, lmM EGTA, 10% glycerol, 1% Triton™ -XI 00, lOOmM NaF, 10 mM Na4P207, Complete™ protease inhibitor). The cells are detached using a cell scraper (Corning, Cat. No. 3010) and well contents transferred to Eppendorf reaction tubes. Cell fragments are removed by centrifugation for 10 minutes at 13000 rpm and 4°C and the supernatant is added to 2x Laemmli sample buffer in a 1 : 1 (v/v) ratio. Cellular proteins are separated by SDS-PAGE and transferred to a nitrocellulose membrane (PROTRAN™ BA 85, Schleicher&Schuell, Cat. No. 10 401196) by semi-dry western-blotting.
An antibody specific for IGF-IR (C-20, Santa Cruz Biotechnologies, Cat. No. sc- 713) is used to determine protein levels of IGF-IR.
Downregulation of IGF-IR induced by the antibody of the invention after less than 24 hours after addition of the antibody is observed. Example 6
Inhibition of insulin binding to 3T3-cells expressing human insulin receptor
In order to determine whether the antibody of the invention also blocks binding of insulin to the insulin receptor (IR), competition experiments are performed with a radioactive ly labeled ligand peptide.
3T3 cells (1 x 105/ml) expressing recombinantly high numbers (>105) human IR are plated in MEM Dulbecco medium (DMEM) with high glucose (PAA, Cat. No. El 5-009) supplemented with 2mM L-Glutamin (Gibco, Cat. No. 25030-024) and 10% heat inactivated FCS (PAA, Cat. No. A15-771). Six bottles in the T175 format are inoculated with 20 ml cells in the respective medium for each experiment and cultivated for two days at 37°C and 5% C02 to obtain confluent cell monolayers.
To collect individual cells, 2 ml of lx Trypsin/EDTA (Gibco, Cat. No. 25300-054) per T175 flask are added and detachment of cells monitored with a microscope. The cells are collected and medium with 10% FCS as described before is added to a total volume of 50 ml. Cells are reisolated by centrifugation for 10 minutes at 1000 rpm and resuspended in 50 ml of binding buffer (120 mM NaCl, 5 mM KC1, 1.2 mM MgS04, 1 mM EDTA, 10 mM D(+)glucose, 15 mM NaAc, 100 mM Hepes pH 7.6, 1% BSA). Cells are counted, reisolated by centrifugation and adjusted with binding buffer to 1 x 106 cells/ml.
1125-labeled insulin peptide (Amersham, Cat. No. IM166, -2000 Ci/mmol), solubilized in 0.1% CH3COOH, are diluted in binding buffer to a final activity of 4xl05 counts/(minute*ml). 75 μΐ of antibody together with 25 μΐ of prediluted 1125-labeled insulin peptide is added to 200 μΐ of cell suspension (final antibody concentration 200 nM) and incubated for 3,5 h at 4°C. Cells are reisolated by centrifugation for 5 minutes at 2000 rpm and supernatant is removed. After washing two times in 1 ml binding buffer, cells are resuspended in 1 ml binding buffer and transferred to scintillation tubes. The amount of radioactive peptide bound to the cell surface receptors is measured on a scintillation counter.
Example 7
No stimulation of IGF-IR and Akt/PKB phosphorylation
In order to exclude IGF-IR stimulating activities of the antibody of the invention, phosphorylation of IGF-IR is determined in the absence of IGF-I ligand but in the presence of the antibody of the invention and a reference antibody ( IR3, Oncogene, Germany). This is performed by a western-blotting analysis with phosphorylation-state specific antibodies. 3T3 cells (ATCC CRL 1658) transfected with IGF-IR (5xl04cells/ml, Pietrzkowski, Z., et al, Cell Growth Differ. 3 (1992) 199-205) are plated in MEM Dulbecco medium (DMEM) with high glucose (PAA, CatNo. El 5-009) supplemented with 2mM L-Glutamin (Gibco, CatNo. 25030-024) and 0.5% heat inactivated FCS (PAA, CatNo. Al 5-771) or human tumor cells (HT29, NCI H322M, 5xl04/ml) in RPMI 1640 medium (PAA, CatNo. E15-039) supplemented with 2 mM L-Glutamin, lx non-essential aminoacids (Gibco, CatNo. 11140-035), 1 mM sodium pyruvate (Gibco, CatNo. 11360-039) and 0.5% heat inactivated FCS (PAA, CatNo. A15-771). For determination of IC50 values, 12 well plates are inoculated with 1 ml cells in the respective medium for each experiment and cultivated for two days at 37°C and 5%> C02.
After 48 hours of cultivation with low serum medium, the medium is carefully removed and replaced by different concentrations of antibody diluted in the respective medium. Cells are incubated for 15 minutes under the conditions mentioned above. The medium is carefully removed by aspiration and 100 μΐ of cold lysis buffer is added per well (50mM Hepes pH 7.2, 150 mM NaCl, ImM EGTA, 10% glycerol, 1% Triton-XlOO, lOOmM NaF, 10 mM Na4P207, Complete™ protease inhibitor). The cells are detached using a cell scraper (Corning, CatNo. 3010) and well contents transferred to Eppendorf reaction tubes. Cell fragments are removed by centrifugation for 10 minutes at 13000 rpm and 4°C (Eppendorf centrifuge 5415R) and half of the supernatant is added to 2x Laemmli sample buffer in a 1 : 1 (v/v) ratio. For immunoprecipitation of IGF-IR, the remaining supernatant of cell lysates underwent a clearifying spin (10 minutes at 13000 rpm and 4°C) right before 1 μΐ of an antibody against IGF-IR is added (C-20, Santa Cruz Biotechnologies, CatNo. sc-713 or mAb 24-55, GroPep, CatNo. MAD1). After 2 hours incubation at 4°C in a rotating Eppendorf reaction tube, 25 μΐ Protein G Sepharose™ beads (Amersham Biosciences, CatNo. 17-0618-01) are added followed by another incubation step of 1 hour at 4°C. The beads with bound antibody-protein-complexes are isolated by centrifugation (1 minute at 2000 rpm and 4°C) and washed three times with wash buffer (lysis buffer with only 0.1 %> Triton-XlOO). After boiling the beads in Laemmli sample buffer, cellular proteins are separated by SDS-PAGE and transferred to a nitrocellulose membrane (PROTRAN BA 85, Schleicher&Schuell, CatNo. 10 401196) by semi-dry western- blotting. A phosphotyrosin specific antibody (Upstate, clone 4G10, CatNo. 05-321, recognizing tyrosine -phosphorylated proteins) is used to determine phosphorylation status of immunopurified IGF-IR. For the detection of phosphorylated Akt/PKB an antibody against Aktl with specificity for phosphorylated Ser473 (Cell Signalling, CatNo. 9271) is applied.
Example 8
Induction of receptor down-regulation in H322M xenograft models
Tumors are induced in nude mice and treated once with different concentrations of the antibody of the invention. 24 hours after treatment the tumors are extracted and homogenized under liquid nitrogen. Cold lysis buffer is added (50mM Hepes pH 7.2, 150 mM NaCl, lmM EGTA, 10% glycerol, 1% Triton-XlOO, lOOmM NaF, 1 mM Na3V04, 10 mM Na4P207, Complete™ protease inhibitor, lmM PMSF) in a buffer- volume to tumor- weight ratio of 3 : 1 and thoroughly mixed with the thawing tumor homogenate. After solubilizing the tissue for 15 minutes on ice, insoluble fragments are removed by centrifugation for 10 minutes at 13000 rpm and 4°C (Eppendorf centrifuge 5415R). The protein concentration of the samples is determined with the Micro BCA™ Reagents (Pierce) and lysis buffer is added to adjust equal concentrations. Part of the supernatant is added to 2x Laemmli sample buffer in a 1 : 1 (v/v) ratio. Cellular proteins are separated by SDS-PAGE and transferred to a nitrocellulose membrane (PROTRAN BA 85, Schleicher&Schuell, CatNo. 10 401196) by semi-dry western-blotting. An IGF-IR specific antibody (C-20, Santa Cruz Biotechnologies, CatNo. sc-713) is used to to detect IGF-IR.
Example 9
Growth inhibition of H322M tumors
The effects of an antibody according to the invention in vivo is investigated by inducing tumors in athymic nude mice according to established methods. Human H322M NSCLC cells are coinjected together with Matrigel subcutaneously into 6- 7 week-old athymic nu mice (nu/nu). For that purpose, 5 x 106 H322M cells are concentrated in ΙΟΟμΙ culture medium and mixed with 100 μΐ Matrigel. 200μ1 of this mixture are injected into the right flanks of the mice. Tumor volume is calculated by measuring tumor diameters with Vernier calipers twice a week according to the formula first published by Geran et al., ("Protocols for screening chemical agents and natural products against animal tumors and other biological systems", Cancer Chemother. Rep. 11.301, (1972)) where tumor volume [mg] = (length x (width)2). Antibody is administered intraperitoneally (i.p.) at 10ml/ kg. Treatment is started with doubled doses of the antibody administered in doubled volumes. Tumors are induced in nude mice as described above. After tumors had grown to an average volume of 160 mg, mice are treated intraperitoneally six times once a week with 6, 0.6 and 0.06 mg/ kg of antibody as consecutive doses starting with 12, 1.2 and 0.12 mg/ kg as loading dose given once on the first day of treatment.
In addition antibody according to the invention is tested in combination with gemcitabine in the same model. Tumors are induced as described above and treatment is initiated when tumors had established and grown to 170mm3 average in all groups. Antibody is administered once a week i.p. at 6 and 0.6 mg/kg and in combination with 62 mg/ kg of gemcitabine at 0.6 mg. Gemcitabine is administered one cycle i.e. every third day for four times in total. Again, treatment is started by administering doubled doses of the antibody.

Claims

Patent Claims
1. An isolated antibody specifically binding to human IGF-1R, characterized in comprising as heavy chain variable domain CDRH3 region a CDRH3 region of SEQ ID NO: 1 with a mutation selected from the group consisting of E1Q, L2E, L2F, L2H, L2M, L2T, G3K, G3L, G3Q, R4H, R4K, Y6F, Y6H, Y6W, D8A, D8G, D8Q, D8S, L9G, L9I, L9N, L9P, L9Q, L9R, L9T, or L9V.
2. The antibody of claim 1, characterized in comprising a CDRH2 region of SEQ ID NO:2 and a CDRH1 region of SEQ ID NO:3.
3. The antibody of claim 1 or 2, characterized in comprising a light chain variable domain comprising a CDRL3 region of SEQ ID NO: 4, a CDRL2 region of SEQ ID NO:5 and a CDRLl region of SEQ ID NO:6.
4. The antibody of any one of claim 1 to 3, characterized in comprising a heavy chain variable domain of SEQ ID NO: 7 including a heavy chain variable domain CDRH3 region of SEQ ID NO: 1 with a double mutation selected from the group consisting of E99Q E1Q, L100E L2E, L100F L2F, L100H L2H, L100M L2M, L100T L2T, G101K G3K, G101L G3L, G101Q G3Q, R102H R4H, R102K R4K, Y104F Y6F, Y104H Y6H, Y104W Y6W, D106A D8A, D106G D8G,D106Q D8Q, D106S D8S, L107G L9G, L107I L9I, L107N L9N, L107P L9P, L107Q L9Q, L107R L9R, L107T L9T, or LI 07V L9V.
5. The antibody of claim 1, characterized in comprising a heavy chain variable domain of SEQ ID NO: 7 with a mutation selected from the group consisting of E1Q, L2E, L2F, L2H, L2M, L2T, G3K, G3L, G3Q, R4H, R4K, Y6F, Y6H, Y6W, D8A, D8G, D8Q, D8S, L9G, L9I, L9N, L9P, L9Q, L9R, L9T, or L9V and a light chain variable domain of SEQ ID NO:8.
6. The antibody of claim 4, characterized in comprising the light chain variable domain of SEQ ID NO:8.
7. Isolated nucleic acid encoding the antibody of claim 1 to 6.
8. A host cell comprising the nucleic acid of claim 7.
9. A method of producing an antibody comprising culturing the host cell of claim 8.
10. An immunoconjugate comprising the antibody of any one of claim 1 to 6 and a cytotoxic agent.
11. A pharmaceutical formulation comprising the antibody of any one of claim 1 to 6 and a pharmaceutically acceptable carrier.
12. The antibody of any one of claim 1 to 6 for use as a medicament.
13. The antibody of any one of claim 1 to 6 for use in treating tumor diseases.
14. The antibody of any one of claim 1 to 6 for use in inhibiting the binding of human IGF I and human IGF-II to human IGF-IR.
15. Use of the antibody of any one of claim 1 to 6 in the manufacture of a medicament.
16. Use of the antibody of any one of claim 1 to 6 in the manufacture of a medicament for the treatment of a tumor disease.
17. A method of treating an individual having a tumor disease comprising administering to the individual an effective amount of the antibody of any one of claims 1 to 6.
18. A method inhibiting the binding of human IGF I and human IGF-II to human IGF-IR in an individual comprising administering to the individual an effective amount of the antibody of any one of claim 1 to 6.
PCT/EP2011/060376 2010-06-22 2011-06-21 Antibodies against insulin-like growth factor i receptor and uses thereof WO2011161119A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP10166860.6 2010-06-22
EP10166860 2010-06-22

Publications (1)

Publication Number Publication Date
WO2011161119A1 true WO2011161119A1 (en) 2011-12-29

Family

ID=42562726

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2011/060376 WO2011161119A1 (en) 2010-06-22 2011-06-21 Antibodies against insulin-like growth factor i receptor and uses thereof

Country Status (1)

Country Link
WO (1) WO2011161119A1 (en)

Citations (93)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4676980A (en) 1985-09-23 1987-06-30 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Target specific cross-linked heteroantibodies
US4737456A (en) 1985-05-09 1988-04-12 Syntex (U.S.A.) Inc. Reducing interference in ligand-receptor binding assays
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
EP0404097A2 (en) 1989-06-22 1990-12-27 BEHRINGWERKE Aktiengesellschaft Bispecific and oligospecific, mono- and oligovalent receptors, production and applications thereof
WO1993001161A1 (en) 1991-07-11 1993-01-21 Pfizer Limited Process for preparing sertraline intermediates
US5208020A (en) 1989-10-25 1993-05-04 Immunogen Inc. Cytotoxic agents comprising maytansinoids and their therapeutic use
WO1993008829A1 (en) 1991-11-04 1993-05-13 The Regents Of The University Of California Compositions that mediate killing of hiv-infected cells
WO1993016185A2 (en) 1992-02-06 1993-08-19 Creative Biomolecules, Inc. Biosynthetic binding protein for cancer marker
WO1994011026A2 (en) 1992-11-13 1994-05-26 Idec Pharmaceuticals Corporation Therapeutic application of chimeric and radiolabeled antibodies to human b lymphocyte restricted differentiation antigen for treatment of b cell lymphoma
WO1994029351A2 (en) 1993-06-16 1994-12-22 Celltech Limited Antibodies
US5500362A (en) 1987-01-08 1996-03-19 Xoma Corporation Chimeric antibody with specificity to human B cell surface antigen
EP0425235B1 (en) 1989-10-25 1996-09-25 Immunogen Inc Cytotoxic agents comprising maytansinoids and their therapeutic use
US5571894A (en) 1991-02-05 1996-11-05 Ciba-Geigy Corporation Recombinant antibodies specific for a growth factor receptor
US5587458A (en) 1991-10-07 1996-12-24 Aronex Pharmaceuticals, Inc. Anti-erbB-2 antibodies, combinations thereof, and therapeutic and diagnostic uses thereof
US5624821A (en) 1987-03-18 1997-04-29 Scotgen Biopharmaceuticals Incorporated Antibodies with altered effector functions
US5635483A (en) 1992-12-03 1997-06-03 Arizona Board Of Regents Acting On Behalf Of Arizona State University Tumor inhibiting tetrapeptide bearing modified phenethyl amides
US5648237A (en) 1991-09-19 1997-07-15 Genentech, Inc. Expression of functional antibody fragments
WO1997030087A1 (en) 1996-02-16 1997-08-21 Glaxo Group Limited Preparation of glycosylated antibodies
US5712374A (en) 1995-06-07 1998-01-27 American Cyanamid Company Method for the preparation of substantiallly monomeric calicheamicin derivative/carrier conjugates
US5714586A (en) 1995-06-07 1998-02-03 American Cyanamid Company Methods for the preparation of monomeric calicheamicin derivative/carrier conjugates
US5731168A (en) 1995-03-01 1998-03-24 Genentech, Inc. Method for making heteromultimeric polypeptides
US5739116A (en) 1994-06-03 1998-04-14 American Cyanamid Company Enediyne derivatives useful for the synthesis of conjugates of methyltrithio antitumor agents
US5770710A (en) 1987-10-30 1998-06-23 American Cyanamid Company Antitumor and antibacterial substituted disulfide derivatives prepared from compounds possessing a methlytrithio group
US5770701A (en) 1987-10-30 1998-06-23 American Cyanamid Company Process for preparing targeted forms of methyltrithio antitumor agents
US5780588A (en) 1993-01-26 1998-07-14 Arizona Board Of Regents Elucidation and synthesis of selected pentapeptides
US5789199A (en) 1994-11-03 1998-08-04 Genentech, Inc. Process for bacterial production of polypeptides
US5821337A (en) 1991-06-14 1998-10-13 Genentech, Inc. Immunoglobulin variants
US5840523A (en) 1995-03-01 1998-11-24 Genetech, Inc. Methods and compositions for secretion of heterologous polypeptides
WO1998058964A1 (en) 1997-06-24 1998-12-30 Genentech, Inc. Methods and compositions for galactosylated glycoproteins
US5869046A (en) 1995-04-14 1999-02-09 Genentech, Inc. Altered polypeptides with increased half-life
WO1999022764A1 (en) 1997-10-31 1999-05-14 Genentech, Inc. Methods and compositions comprising glycoprotein glycoforms
US5959177A (en) 1989-10-27 1999-09-28 The Scripps Research Institute Transgenic plants expressing assembled secretory antibodies
WO1999051642A1 (en) 1998-04-02 1999-10-14 Genentech, Inc. Antibody variants and fragments thereof
US6040498A (en) 1998-08-11 2000-03-21 North Caroline State University Genetically engineered duckweed
WO2000061739A1 (en) 1999-04-09 2000-10-19 Kyowa Hakko Kogyo Co., Ltd. Method for controlling the activity of immunologically functional molecule
US6171586B1 (en) 1997-06-13 2001-01-09 Genentech, Inc. Antibody formulation
US6194551B1 (en) 1998-04-02 2001-02-27 Genentech, Inc. Polypeptide variants
WO2001029246A1 (en) 1999-10-19 2001-04-26 Kyowa Hakko Kogyo Co., Ltd. Process for producing polypeptide
US6248516B1 (en) 1988-11-11 2001-06-19 Medical Research Council Single domain ligands, receptors comprising said ligands methods for their production, and use of said ligands and receptors
US6267958B1 (en) 1995-07-27 2001-07-31 Genentech, Inc. Protein formulation
WO2002031140A1 (en) 2000-10-06 2002-04-18 Kyowa Hakko Kogyo Co., Ltd. Cells producing antibody compositions
WO2002053596A2 (en) 2001-01-05 2002-07-11 Pfizer Inc. Antibodies to insulin-like growth factor i receptor
US6420548B1 (en) 1999-10-04 2002-07-16 Medicago Inc. Method for regulating transcription of foreign genes
US20020164328A1 (en) 2000-10-06 2002-11-07 Toyohide Shinkawa Process for purifying antibody
WO2003011878A2 (en) 2001-08-03 2003-02-13 Glycart Biotechnology Ag Antibody glycosylation variants having increased antibody-dependent cellular cytotoxicity
US20030115614A1 (en) 2000-10-06 2003-06-19 Yutaka Kanda Antibody composition-producing cell
US6602684B1 (en) 1998-04-20 2003-08-05 Glycart Biotechnology Ag Glycosylation engineering of antibodies for improving antibody-dependent cellular cytotoxicity
US20030157108A1 (en) 2001-10-25 2003-08-21 Genentech, Inc. Glycoprotein compositions
US20030165502A1 (en) 2000-06-13 2003-09-04 City Of Hope Single-chain antibodies against human insulin-like growth factor I receptor: expression, purification, and effect on tumor growth
US6630579B2 (en) 1999-12-29 2003-10-07 Immunogen Inc. Cytotoxic agents comprising modified doxorubicins and daunorubicins and their therapeutic use
WO2003084570A1 (en) 2002-04-09 2003-10-16 Kyowa Hakko Kogyo Co., Ltd. DRUG CONTAINING ANTIBODY COMPOSITION APPROPRIATE FOR PATIENT SUFFERING FROM FcϜRIIIa POLYMORPHISM
WO2003085107A1 (en) 2002-04-09 2003-10-16 Kyowa Hakko Kogyo Co., Ltd. Cells with modified genome
WO2003085119A1 (en) 2002-04-09 2003-10-16 Kyowa Hakko Kogyo Co., Ltd. METHOD OF ENHANCING ACTIVITY OF ANTIBODY COMPOSITION OF BINDING TO FcϜ RECEPTOR IIIa
WO2003100008A2 (en) 2002-05-24 2003-12-04 Schering Corporation Neutralizing human anti-igfr antibody
WO2003106621A2 (en) 2002-06-14 2003-12-24 Immunogen, Inc. Anti-igf-i receptor antibody
US20040093621A1 (en) 2001-12-25 2004-05-13 Kyowa Hakko Kogyo Co., Ltd Antibody composition which specifically binds to CD20
US6737056B1 (en) 1999-01-15 2004-05-18 Genentech, Inc. Polypeptide variants with altered effector function
US20040110282A1 (en) 2002-04-09 2004-06-10 Kyowa Hakko Kogyo Co., Ltd. Cells in which activity of the protein involved in transportation of GDP-fucose is reduced or lost
US20040109865A1 (en) 2002-04-09 2004-06-10 Kyowa Hakko Kogyo Co., Ltd. Antibody composition-containing medicament
WO2004056312A2 (en) 2002-12-16 2004-07-08 Genentech, Inc. Immunoglobulin variants and uses thereof
US20040132140A1 (en) 2002-04-09 2004-07-08 Kyowa Hakko Kogyo Co., Ltd. Production process for antibody composition
WO2004071529A2 (en) 2003-02-13 2004-08-26 Pfizer Products Inc. Uses of anti-insulin-like growth factor i receptor antibodies
WO2004083248A1 (en) 2003-03-14 2004-09-30 Pharmacia Corporation Antibodies to igf-i receptor for the treatment of cancers
WO2004087756A2 (en) 2003-04-02 2004-10-14 F. Hoffmann-La Roche Ag Antibodies against insulin-like growth factor i receptor and uses thereof
US20050014934A1 (en) 2002-10-15 2005-01-20 Hinton Paul R. Alteration of FcRn binding affinities or serum half-lives of antibodies by mutagenesis
WO2005005635A2 (en) 2003-07-10 2005-01-20 F. Hoffmann-La Roche Ag Antibodies against insulin-like growth factor i receptor and uses thereof
WO2005016970A2 (en) 2003-05-01 2005-02-24 Imclone Systems Incorporated Fully human antibodies directed against the human insulin-like growth factor-1 receptor
WO2005016967A2 (en) 2003-08-13 2005-02-24 Pfizer Products Inc. Modified human igf-1r antibodies
US20050084906A1 (en) 2002-01-18 2005-04-21 Liliane Goetsch Novel anti-IGF-IR antibodies and uses thereof
WO2005035778A1 (en) 2003-10-09 2005-04-21 Kyowa Hakko Kogyo Co., Ltd. PROCESS FOR PRODUCING ANTIBODY COMPOSITION BY USING RNA INHIBITING THE FUNCTION OF α1,6-FUCOSYLTRANSFERASE
WO2005035586A1 (en) 2003-10-08 2005-04-21 Kyowa Hakko Kogyo Co., Ltd. Fused protein composition
US20050123546A1 (en) 2003-11-05 2005-06-09 Glycart Biotechnology Ag Antigen binding molecules with increased Fc receptor binding affinity and effector function
WO2005053742A1 (en) 2003-12-04 2005-06-16 Kyowa Hakko Kogyo Co., Ltd. Medicine containing antibody composition
WO2005058967A2 (en) 2003-12-16 2005-06-30 Pierre Fabre Medicament Novel anti-insulin/igf-i hybrid receptor or anti-insulin/igf-i hybrid receptor and igf-ir antibodies and uses thereof
WO2005082415A2 (en) 2004-02-25 2005-09-09 Dana Farber Cancer Institute, Inc. Inhibitors of insulin-like growth factor receptor-1 for inhibiting tumor cell growth
WO2005094376A2 (en) 2004-03-31 2005-10-13 Bristol-Myers Squibb Company Synergistic methods and compositions for treating cancer
WO2005100402A1 (en) 2004-04-13 2005-10-27 F.Hoffmann-La Roche Ag Anti-p-selectin antibodies
US20050249730A1 (en) 2002-01-18 2005-11-10 Pierre Fabre Medicament Novel anti-IGF-IR and/or anti-insulin/IGF-I hybrid receptors antibodies and uses thereof
US20050260186A1 (en) 2003-03-05 2005-11-24 Halozyme, Inc. Soluble glycosaminoglycanases and methods of preparing and using soluble glycosaminoglycanases
WO2006008639A1 (en) 2004-07-16 2006-01-26 Pfizer Products Inc. Combination treatment for non-hematologic malignancies using an anti-igf-1r antibody
US20060025576A1 (en) 2000-04-11 2006-02-02 Genentech, Inc. Multivalent antibodies and uses therefor
WO2006013472A2 (en) 2004-07-29 2006-02-09 Pierre Fabre Medicament Novel anti-igf-ir antibodies and uses thereof
WO2006029879A2 (en) 2004-09-17 2006-03-23 F.Hoffmann-La Roche Ag Anti-ox40l antibodies
WO2006044908A2 (en) 2004-10-20 2006-04-27 Genentech, Inc. Antibody formulation in histidine-acetate buffer
US20060104968A1 (en) 2003-03-05 2006-05-18 Halozyme, Inc. Soluble glycosaminoglycanases and methods of preparing and using soluble glycosaminogly ycanases
US7125978B1 (en) 1999-10-04 2006-10-24 Medicago Inc. Promoter for regulating expression of foreign genes
WO2007126876A2 (en) * 2006-03-28 2007-11-08 Biogen Idec Ma Inc. Anti-igf-ir antibodies and uses thereof
US20080069820A1 (en) 2006-08-30 2008-03-20 Genentech, Inc. Multispecific antibodies
US7371826B2 (en) 1999-01-15 2008-05-13 Genentech, Inc. Polypeptide variants with altered effector function
WO2008077546A1 (en) 2006-12-22 2008-07-03 F. Hoffmann-La Roche Ag Antibodies against insulin-like growth factor i receptor and uses thereof
US7498298B2 (en) 2003-11-06 2009-03-03 Seattle Genetics, Inc. Monomethylvaline compounds capable of conjugation to ligands
US7521541B2 (en) 2004-09-23 2009-04-21 Genetech Inc. Cysteine engineered antibodies and conjugates
WO2009089004A1 (en) 2008-01-07 2009-07-16 Amgen Inc. Method for making antibody fc-heterodimeric molecules using electrostatic steering effects

Patent Citations (101)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US4737456A (en) 1985-05-09 1988-04-12 Syntex (U.S.A.) Inc. Reducing interference in ligand-receptor binding assays
US4676980A (en) 1985-09-23 1987-06-30 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Target specific cross-linked heteroantibodies
US5500362A (en) 1987-01-08 1996-03-19 Xoma Corporation Chimeric antibody with specificity to human B cell surface antigen
US5624821A (en) 1987-03-18 1997-04-29 Scotgen Biopharmaceuticals Incorporated Antibodies with altered effector functions
US5648260A (en) 1987-03-18 1997-07-15 Scotgen Biopharmaceuticals Incorporated DNA encoding antibodies with altered effector functions
US5770701A (en) 1987-10-30 1998-06-23 American Cyanamid Company Process for preparing targeted forms of methyltrithio antitumor agents
US5770710A (en) 1987-10-30 1998-06-23 American Cyanamid Company Antitumor and antibacterial substituted disulfide derivatives prepared from compounds possessing a methlytrithio group
US6248516B1 (en) 1988-11-11 2001-06-19 Medical Research Council Single domain ligands, receptors comprising said ligands methods for their production, and use of said ligands and receptors
EP0404097A2 (en) 1989-06-22 1990-12-27 BEHRINGWERKE Aktiengesellschaft Bispecific and oligospecific, mono- and oligovalent receptors, production and applications thereof
US5416064A (en) 1989-10-25 1995-05-16 Immunogen, Inc. Cytotoxic agents comprising maytansinoids and their therapeutic use
EP0425235B1 (en) 1989-10-25 1996-09-25 Immunogen Inc Cytotoxic agents comprising maytansinoids and their therapeutic use
US5208020A (en) 1989-10-25 1993-05-04 Immunogen Inc. Cytotoxic agents comprising maytansinoids and their therapeutic use
US5959177A (en) 1989-10-27 1999-09-28 The Scripps Research Institute Transgenic plants expressing assembled secretory antibodies
US6417429B1 (en) 1989-10-27 2002-07-09 The Scripps Research Institute Transgenic plants expressing assembled secretory antibodies
US5571894A (en) 1991-02-05 1996-11-05 Ciba-Geigy Corporation Recombinant antibodies specific for a growth factor receptor
US5821337A (en) 1991-06-14 1998-10-13 Genentech, Inc. Immunoglobulin variants
WO1993001161A1 (en) 1991-07-11 1993-01-21 Pfizer Limited Process for preparing sertraline intermediates
US5648237A (en) 1991-09-19 1997-07-15 Genentech, Inc. Expression of functional antibody fragments
US5587458A (en) 1991-10-07 1996-12-24 Aronex Pharmaceuticals, Inc. Anti-erbB-2 antibodies, combinations thereof, and therapeutic and diagnostic uses thereof
WO1993008829A1 (en) 1991-11-04 1993-05-13 The Regents Of The University Of California Compositions that mediate killing of hiv-infected cells
WO1993016185A2 (en) 1992-02-06 1993-08-19 Creative Biomolecules, Inc. Biosynthetic binding protein for cancer marker
WO1994011026A2 (en) 1992-11-13 1994-05-26 Idec Pharmaceuticals Corporation Therapeutic application of chimeric and radiolabeled antibodies to human b lymphocyte restricted differentiation antigen for treatment of b cell lymphoma
US5635483A (en) 1992-12-03 1997-06-03 Arizona Board Of Regents Acting On Behalf Of Arizona State University Tumor inhibiting tetrapeptide bearing modified phenethyl amides
US5780588A (en) 1993-01-26 1998-07-14 Arizona Board Of Regents Elucidation and synthesis of selected pentapeptides
WO1994029351A2 (en) 1993-06-16 1994-12-22 Celltech Limited Antibodies
US5877296A (en) 1994-06-03 1999-03-02 American Cyanamid Company Process for preparing conjugates of methyltrithio antitumor agents
US5739116A (en) 1994-06-03 1998-04-14 American Cyanamid Company Enediyne derivatives useful for the synthesis of conjugates of methyltrithio antitumor agents
US5773001A (en) 1994-06-03 1998-06-30 American Cyanamid Company Conjugates of methyltrithio antitumor agents and intermediates for their synthesis
US5767285A (en) 1994-06-03 1998-06-16 American Cyanamid Company Linkers useful for the synthesis of conjugates of methyltrithio antitumor agents
US5789199A (en) 1994-11-03 1998-08-04 Genentech, Inc. Process for bacterial production of polypeptides
US5731168A (en) 1995-03-01 1998-03-24 Genentech, Inc. Method for making heteromultimeric polypeptides
US5840523A (en) 1995-03-01 1998-11-24 Genetech, Inc. Methods and compositions for secretion of heterologous polypeptides
US5869046A (en) 1995-04-14 1999-02-09 Genentech, Inc. Altered polypeptides with increased half-life
US5712374A (en) 1995-06-07 1998-01-27 American Cyanamid Company Method for the preparation of substantiallly monomeric calicheamicin derivative/carrier conjugates
US5714586A (en) 1995-06-07 1998-02-03 American Cyanamid Company Methods for the preparation of monomeric calicheamicin derivative/carrier conjugates
US6267958B1 (en) 1995-07-27 2001-07-31 Genentech, Inc. Protein formulation
WO1997030087A1 (en) 1996-02-16 1997-08-21 Glaxo Group Limited Preparation of glycosylated antibodies
US6171586B1 (en) 1997-06-13 2001-01-09 Genentech, Inc. Antibody formulation
WO1998058964A1 (en) 1997-06-24 1998-12-30 Genentech, Inc. Methods and compositions for galactosylated glycoproteins
WO1999022764A1 (en) 1997-10-31 1999-05-14 Genentech, Inc. Methods and compositions comprising glycoprotein glycoforms
US6194551B1 (en) 1998-04-02 2001-02-27 Genentech, Inc. Polypeptide variants
WO1999051642A1 (en) 1998-04-02 1999-10-14 Genentech, Inc. Antibody variants and fragments thereof
US6602684B1 (en) 1998-04-20 2003-08-05 Glycart Biotechnology Ag Glycosylation engineering of antibodies for improving antibody-dependent cellular cytotoxicity
US6040498A (en) 1998-08-11 2000-03-21 North Caroline State University Genetically engineered duckweed
US7371826B2 (en) 1999-01-15 2008-05-13 Genentech, Inc. Polypeptide variants with altered effector function
US7332581B2 (en) 1999-01-15 2008-02-19 Genentech, Inc. Polypeptide variants with altered effector function
US6737056B1 (en) 1999-01-15 2004-05-18 Genentech, Inc. Polypeptide variants with altered effector function
WO2000061739A1 (en) 1999-04-09 2000-10-19 Kyowa Hakko Kogyo Co., Ltd. Method for controlling the activity of immunologically functional molecule
US6420548B1 (en) 1999-10-04 2002-07-16 Medicago Inc. Method for regulating transcription of foreign genes
US7125978B1 (en) 1999-10-04 2006-10-24 Medicago Inc. Promoter for regulating expression of foreign genes
WO2001029246A1 (en) 1999-10-19 2001-04-26 Kyowa Hakko Kogyo Co., Ltd. Process for producing polypeptide
US6630579B2 (en) 1999-12-29 2003-10-07 Immunogen Inc. Cytotoxic agents comprising modified doxorubicins and daunorubicins and their therapeutic use
US20060025576A1 (en) 2000-04-11 2006-02-02 Genentech, Inc. Multivalent antibodies and uses therefor
US20030165502A1 (en) 2000-06-13 2003-09-04 City Of Hope Single-chain antibodies against human insulin-like growth factor I receptor: expression, purification, and effect on tumor growth
US20030115614A1 (en) 2000-10-06 2003-06-19 Yutaka Kanda Antibody composition-producing cell
WO2002031140A1 (en) 2000-10-06 2002-04-18 Kyowa Hakko Kogyo Co., Ltd. Cells producing antibody compositions
US20020164328A1 (en) 2000-10-06 2002-11-07 Toyohide Shinkawa Process for purifying antibody
WO2002053596A2 (en) 2001-01-05 2002-07-11 Pfizer Inc. Antibodies to insulin-like growth factor i receptor
WO2003011878A2 (en) 2001-08-03 2003-02-13 Glycart Biotechnology Ag Antibody glycosylation variants having increased antibody-dependent cellular cytotoxicity
US20030157108A1 (en) 2001-10-25 2003-08-21 Genentech, Inc. Glycoprotein compositions
US20040093621A1 (en) 2001-12-25 2004-05-13 Kyowa Hakko Kogyo Co., Ltd Antibody composition which specifically binds to CD20
US20050249730A1 (en) 2002-01-18 2005-11-10 Pierre Fabre Medicament Novel anti-IGF-IR and/or anti-insulin/IGF-I hybrid receptors antibodies and uses thereof
US20050084906A1 (en) 2002-01-18 2005-04-21 Liliane Goetsch Novel anti-IGF-IR antibodies and uses thereof
US20040110704A1 (en) 2002-04-09 2004-06-10 Kyowa Hakko Kogyo Co., Ltd. Cells of which genome is modified
US20040109865A1 (en) 2002-04-09 2004-06-10 Kyowa Hakko Kogyo Co., Ltd. Antibody composition-containing medicament
WO2003084570A1 (en) 2002-04-09 2003-10-16 Kyowa Hakko Kogyo Co., Ltd. DRUG CONTAINING ANTIBODY COMPOSITION APPROPRIATE FOR PATIENT SUFFERING FROM FcϜRIIIa POLYMORPHISM
US20040132140A1 (en) 2002-04-09 2004-07-08 Kyowa Hakko Kogyo Co., Ltd. Production process for antibody composition
US20040110282A1 (en) 2002-04-09 2004-06-10 Kyowa Hakko Kogyo Co., Ltd. Cells in which activity of the protein involved in transportation of GDP-fucose is reduced or lost
WO2003085119A1 (en) 2002-04-09 2003-10-16 Kyowa Hakko Kogyo Co., Ltd. METHOD OF ENHANCING ACTIVITY OF ANTIBODY COMPOSITION OF BINDING TO FcϜ RECEPTOR IIIa
WO2003085107A1 (en) 2002-04-09 2003-10-16 Kyowa Hakko Kogyo Co., Ltd. Cells with modified genome
WO2003100008A2 (en) 2002-05-24 2003-12-04 Schering Corporation Neutralizing human anti-igfr antibody
WO2003106621A2 (en) 2002-06-14 2003-12-24 Immunogen, Inc. Anti-igf-i receptor antibody
US20050014934A1 (en) 2002-10-15 2005-01-20 Hinton Paul R. Alteration of FcRn binding affinities or serum half-lives of antibodies by mutagenesis
WO2004056312A2 (en) 2002-12-16 2004-07-08 Genentech, Inc. Immunoglobulin variants and uses thereof
WO2004071529A2 (en) 2003-02-13 2004-08-26 Pfizer Products Inc. Uses of anti-insulin-like growth factor i receptor antibodies
US20060104968A1 (en) 2003-03-05 2006-05-18 Halozyme, Inc. Soluble glycosaminoglycanases and methods of preparing and using soluble glycosaminogly ycanases
US20050260186A1 (en) 2003-03-05 2005-11-24 Halozyme, Inc. Soluble glycosaminoglycanases and methods of preparing and using soluble glycosaminoglycanases
WO2004083248A1 (en) 2003-03-14 2004-09-30 Pharmacia Corporation Antibodies to igf-i receptor for the treatment of cancers
WO2004087756A2 (en) 2003-04-02 2004-10-14 F. Hoffmann-La Roche Ag Antibodies against insulin-like growth factor i receptor and uses thereof
WO2005016970A2 (en) 2003-05-01 2005-02-24 Imclone Systems Incorporated Fully human antibodies directed against the human insulin-like growth factor-1 receptor
WO2005005635A2 (en) 2003-07-10 2005-01-20 F. Hoffmann-La Roche Ag Antibodies against insulin-like growth factor i receptor and uses thereof
WO2005016967A2 (en) 2003-08-13 2005-02-24 Pfizer Products Inc. Modified human igf-1r antibodies
WO2005035586A1 (en) 2003-10-08 2005-04-21 Kyowa Hakko Kogyo Co., Ltd. Fused protein composition
WO2005035778A1 (en) 2003-10-09 2005-04-21 Kyowa Hakko Kogyo Co., Ltd. PROCESS FOR PRODUCING ANTIBODY COMPOSITION BY USING RNA INHIBITING THE FUNCTION OF α1,6-FUCOSYLTRANSFERASE
US20050123546A1 (en) 2003-11-05 2005-06-09 Glycart Biotechnology Ag Antigen binding molecules with increased Fc receptor binding affinity and effector function
US7498298B2 (en) 2003-11-06 2009-03-03 Seattle Genetics, Inc. Monomethylvaline compounds capable of conjugation to ligands
WO2005053742A1 (en) 2003-12-04 2005-06-16 Kyowa Hakko Kogyo Co., Ltd. Medicine containing antibody composition
WO2005058967A2 (en) 2003-12-16 2005-06-30 Pierre Fabre Medicament Novel anti-insulin/igf-i hybrid receptor or anti-insulin/igf-i hybrid receptor and igf-ir antibodies and uses thereof
WO2005082415A2 (en) 2004-02-25 2005-09-09 Dana Farber Cancer Institute, Inc. Inhibitors of insulin-like growth factor receptor-1 for inhibiting tumor cell growth
WO2005094376A2 (en) 2004-03-31 2005-10-13 Bristol-Myers Squibb Company Synergistic methods and compositions for treating cancer
WO2005100402A1 (en) 2004-04-13 2005-10-27 F.Hoffmann-La Roche Ag Anti-p-selectin antibodies
WO2006008639A1 (en) 2004-07-16 2006-01-26 Pfizer Products Inc. Combination treatment for non-hematologic malignancies using an anti-igf-1r antibody
WO2006013472A2 (en) 2004-07-29 2006-02-09 Pierre Fabre Medicament Novel anti-igf-ir antibodies and uses thereof
WO2006029879A2 (en) 2004-09-17 2006-03-23 F.Hoffmann-La Roche Ag Anti-ox40l antibodies
US7521541B2 (en) 2004-09-23 2009-04-21 Genetech Inc. Cysteine engineered antibodies and conjugates
WO2006044908A2 (en) 2004-10-20 2006-04-27 Genentech, Inc. Antibody formulation in histidine-acetate buffer
WO2007126876A2 (en) * 2006-03-28 2007-11-08 Biogen Idec Ma Inc. Anti-igf-ir antibodies and uses thereof
US20080069820A1 (en) 2006-08-30 2008-03-20 Genentech, Inc. Multispecific antibodies
WO2008077546A1 (en) 2006-12-22 2008-07-03 F. Hoffmann-La Roche Ag Antibodies against insulin-like growth factor i receptor and uses thereof
WO2009089004A1 (en) 2008-01-07 2009-07-16 Amgen Inc. Method for making antibody fc-heterodimeric molecules using electrostatic steering effects

Non-Patent Citations (90)

* Cited by examiner, † Cited by third party
Title
"Remington's Pharmaceutical Sciences", 1980, MACK PUBLISHING COMPANY
ADAMS, T.E. ET AL., CELL. MOL. LIFE SCI., vol. 57, 2000, pages 1050 - 1093
ARTEAGA, C.L. ET AL., BREAST CANCER RES. TREATMENT, vol. 22, 1992, pages 101 - 106
BENINI, S. ET AL., CLIN. CANCER RES., vol. 7, 2001, pages 1790 - 1797
BERGMANN, U. ET AL., CANCER RES., vol. 55, 1995, pages 2007 - 2011
BRENNAN, M. ET AL., SCIENCE, vol. 229, 1985, pages 81 - 83
BRUGGEMANN, M. ET AL., J. EXP. MED., vol. 166, 1987, pages 1351 - 1361
BRUNETTI, A. ET AL., BIOCHEM. BIOPHYS. RES. COMMUN., vol. 165, 1989, pages 212 - 218
CHARI, R.V. ET AL., CANCER RES., vol. 52, 1992, pages 127 - 131
CHARLTON, K.A.: "Methods in Molecular Biology", vol. 248, 2004, HUMANA PRESS, pages: 245 - 254
CHEN, Y. ET AL., J. MOL. BIOL., vol. 293, 1999, pages 865 - 881
CLACKSON, T. ET AL., NATURE, vol. 352, 1991, pages 624 - 628
CLYNES, R. ET AL., PROC. NATL. ACAD. SCI. USA, vol. 95, 1998, pages 652 - 656
CRAGG, M.S. ET AL., BLOOD, vol. 101, 2003, pages 1045 - 1052
CRAGG, M.S., GLENNI, M.J, BLOOD, vol. 103, 2004, pages 2738 - 2743
DELAFONTAINE, P. ET AL., J. MOL. CELL. CARDIOL., vol. 26, 1994, pages 1659 - 1673
DRICU, A. ET AL., GLYCOBIOLOGY, vol. 9, 1999, pages 571 - 579
DUBOWCHIK, G.M. ET AL., BIOORG. & MED. CHEM. LETTERS, vol. 12, 2002, pages 1529 - 1532
DUNCAN, A.R., WINTER, G., NATURE, vol. 332, 1988, pages 738 - 740
FLATMAN, S. ET AL., J. CHROMATOGR. B, vol. 848, 2007, pages 79 - 87
FORSAYETH, J.R. ET AL., PROC. NATL. ACAD. SCI. USA, vol. 84, 1987, pages 3448 - 3451
GAZZANO-SANTORO, H. ET AL., J. IMMUNOL. METHODS, vol. 202, 1997
GERAN ET AL.: "Protocols for screening chemical agents and natural products against animal tumors and other biological systems", CANCER CHEMOTHER. REP., vol. 11, 1972, pages 301
GERNGROSS, T.U., NAT. BIOTECH., vol. 22, 2004, pages 1409 - 1414
GRAHAM, F.L. ET AL., J. GEN VIROL., vol. 36, 1977, pages 59 - 74
GRUBER, M. ET AL., J. IMMUNOL., vol. 152, 1994, pages 5368 - 5374
GUSTAFSON, T.A., RUTTER, W.J., J. BIOL. CHEM., vol. 265, 1990, pages 18663 - 18667
GUYER, R.L. ET AL., J. IMMUNOL., vol. 117, 1976, pages 587 - 593
HAILEY, J. ET AL., MOL. CANCER THER., vol. 1, 2002, pages 1349 - 1353
HELLSTROM, I. ET AL., PROC. NAT'L ACAD. SCI. USA, vol. 82, 1985, pages 1499 - 1502
HELLSTROM, I. ET AL., PROC. NAT'L ACAD. SCI. USA, vol. 83, 1986, pages 7059 - 7063
HINMAN, L.M. ET AL., CANCER RES., vol. 53, 1993, pages 3336 - 3342
HOLLIGER, P. ET AL., PROC. NATL. ACAD. SCI. USA, vol. 90, 1993, pages 6444 - 6448
HOYNE, P.A. ET AL., FEBS LETT., vol. 469, 2000, pages 57 - 60
HUDSON, P.J. ET AL., NAT. MED., vol. 9, 2003, pages 129 - 134
IDUSOGIE, E.E. ET AL., J. IMMUNOL., vol. 164, 2000, pages 4178 - 4184
JEFFREY, S.C. ET AL., BIOORGANIC & MED. CHEM. LETTERS, vol. 16, 2006, pages 358 - 362
KABAT ET AL.: "Sequences of Proteins of Immunological Interest", 1991, NIH PUBLICATION 91-3242
KABAT ET AL.: "Sequences of Proteins of Immunological Interest", 1991, NIH PUBLICATION 91-3242, NATIONAL INSTITUTES OF HEALTH
KAM, N.W. ET AL., PROC. NATL. ACAD. SCI. USA, vol. 102, 2005, pages 11600 - 11605
KANDA, Y. ET AL., BIOTECHNOL. BIOENG., vol. 94, 2006, pages 680 - 688
KANTER-LEWENSOHN, L. ET AL., MELANOMA RES., vol. 8, 1998, pages 389 - 397
KATO, H. ET AL., J. BIOL. CHEM., vol. 268, 1993, pages 2655 - 2661
KIM, J.K. ET AL., EUROPEAN JOURNALOF IMMUNOLOGY, vol. 24, 1994, pages 2429 - 2434
KINDT ET AL.: "Kuby Immunology", 2007, W.H. FREEMAN AND CO., pages: 91
KING, H.D. ET AL., J. MED. CHEM., vol. 45, 2002, pages 4336 - 4343
KOSTELNY, S.A. ET AL., J. IMMUNOL., vol. 148, 1992, pages 1547 - 1553
KRATZ, F. ET AL., CURRENT MED. CHEM., vol. 13, 2006, pages 477 - 523
KULL, F.C. ET AL., J. BIOL. CHEM., vol. 258, 1983, pages 6561 - 6566
LEROITH, D. ET AL., ENDOCRIN. REV., vol. 16, 1995, pages 143 - 163
LI, H. ET AL., NAT. BIOTECH., vol. 24, 2006, pages 210 - 215
LI, S.L. ET AL., BIOCHEM. BIOPHYS. RES. COMMUN., vol. 196, 1993, pages 92 - 98
LI, S.L. ET AL., CANCER IMMUNOL. IMMUNOTHER., vol. 49, 2000, pages 243 - 252
LODE, H.N. ET AL., CANCER RES., vol. 58, 1998, pages 2925 - 2928
MATHER, J.P. ET AL., ANNALS N.Y. ACAD. SCI., vol. 383, 1982, pages 44 - 68
MATHER, J.P., BIOL. REPROD., vol. 23, 1980, pages 243 - 252
MILSTEIN, C., CUELLO, A.C., NATURE, vol. 305, 1983, pages 537 - 540
MORGAN, D.O., ROTH, R.A., BIOCHEMISTRY, vol. 25, 1986, pages 1364 - 1371
NAGY, A. ET AL., PROC. NATL. ACAD. SCI. USA, vol. 97, 2000, pages 829 - 834
O'BRIEN, R.M. ET AL., EMBO J., vol. 6, 1987, pages 4003 - 4010
OKAZAKI, A. ET AL., J. MOL. BIOL., vol. 336, 2004, pages 1239 - 1249
PESSINO, A. ET AL., BIOCHEM. BIOPHYS. RES. COMMUN., vol. 162, 1989, pages 1236 - 1243
PETKOVA, S.B. ET AL., INT'L. IMMUNOL., vol. 18, 2006, pages 1759 - 1769
PIETRZKOWSKI, Z. ET AL., CELL GROWTH DIFFER., vol. 3, 1992, pages 199 - 205
PLUECKTHUN, A.: "The Pharmacology of Monoclonal Antibodies", vol. 113, 1994, SPRINGER-VERLAG, pages: 269 - 315
PORTOLANO, S. ET AL., J. IMMUNOL., vol. 150, 1993, pages 880 - 887
PRIGENT, S.A. ET AL., J. BIOL. CHEM., vol. 265, 1990, pages 9970 - 9977
RAVETCH, J.V., KINET, ANNU., REV. IMMUNOL., vol. 9, 1991, pages 457 - 492
RIPKA, J. ET AL., ARCH. BIOCHEM. BIOPHYS., vol. 249, 1986, pages 533 - 545
ROHLIK, Q.T. ET AL., BIOCHEM. BIOPHYS. RES. COMM., vol. 149, 1987, pages 276 - 281
SCHAEFER, E.M. ET AL., J. BIOL. CHEM., vol. 265, 1990, pages 13248 - 13253
SCOTLANDI, K. ET AL., CANCER GENE THER., vol. 9, 2002, pages 296 - 307
SCOTLANDI, K. ET AL., INT. J. CANCER, vol. 101, 2002, pages 11 - 16
SHIELDS, R.L. ET AL., J. BIOL. CHEM., vol. 9, 2001, pages 6591 - 6604
SOOS, M.A. ET AL., BIOCHEM. J., vol. 235, 1986, pages 199 - 208
SOOS, M.A. ET AL., J. BIOL. CHEM., vol. 267, 1992, pages 12955 - 12963
SOOS, M.A. ET AL., PROC. NATL. ACAD. SCI. USA, vol. 86, 1989, pages 5217 - 5221
SURINYA, K.H. ET AL., J. BIOL. CHEM., vol. 277, 2002, pages 16718 - 16725
TAYLOR, R. ET AL., BIOCHEM. J., vol. 242, 1987, pages 123 - 129
TORGOV, M.Y. ET AL., BIOCONJ. CHEM., vol. 16, 2005, pages 717 - 721
TRAUNECKER, A. ET AL., EMBO J., vol. 10, 1991, pages 3655 - 3659
TULLOCH, P.A. ET AL., J. STRUCT. BIOL., vol. 125, 1999, pages 11 - 18
TUTT, A. ET AL., J. IMMUNOL., vol. 147, 1991, pages 60 - 69
URLAUB ET AL., PROC. NATL. ACAD. SCI. USA, vol. 77, 1980, pages 4216 - 4220
VAN DIJK, M.A., VAN DE WINKEL, J.G., CURR. OPIN. CHEM. BIOL., vol. 5, 2001, pages 368 - 374
VITELLA, E.S. ET AL., SCIENCE, vol. 238, 1987, pages 1098 - 1104
WARK K L ET AL: "Latest technologies for the enhancement of antibody affinity", ADVANCED DRUG DELIVERY REVIEWS, ELSEVIER BV, AMSTERDAM, NL, vol. 58, no. 5-6, 7 August 2006 (2006-08-07), pages 657 - 670, XP024892147, ISSN: 0169-409X, [retrieved on 20060807], DOI: 10.1016/J.ADDR.2006.01.025 *
WRIGHT, A. ET AL., TIBTECH, vol. 15, 1997, pages 26 - 32
YAMANE-OHNUKI, N. ET AL., BIOTECH. BIOENG., vol. 87, 2004, pages 614 - 622
YAZAKI, P.J., WU, A.M.: "Methods in Molecular Biology, Antibody Engineering: Methods and Protocols", vol. 248, 2004, HUMANA PRESS, pages: 255 - 268

Similar Documents

Publication Publication Date Title
US11192950B2 (en) Humanized and affinity matured antibodies to FcRH5 and methods of use
EP4089115A1 (en) Anti-tigit antibodies and usage method
AU2019204128B2 (en) Antibodies and methods of use
US9732148B2 (en) Anti-α-synuclein antibodies and methods of use
EP3296321B1 (en) Anti-mesothelin antibodies and immunoconjugates
JP2022137087A (en) Anti-her2 antibody and method for using the same
IL228740A (en) Anti-fgfr1 agonistic antibody and uses thereof
WO2011159980A1 (en) Anti-axl antibodies and methods of use
WO2012010582A1 (en) Anti-cxcr5 antibodies and methods of use
WO2020153467A1 (en) Novel cancer antigens and antibodies of said antigens
WO2011161119A1 (en) Antibodies against insulin-like growth factor i receptor and uses thereof
EP4155321A1 (en) Anti-ddr2 antibodies and uses thereof
US20230194533A1 (en) Monoclonal Antibodies Against Carcinoembryonic Antigens, and Their Uses
NZ737942B2 (en) Humanized and affinity matured antibodies to fcrh5 and methods of use

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 11726455

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 11726455

Country of ref document: EP

Kind code of ref document: A1