US20140170668A1 - Monoclonal and oligoclonal anti-egfr antibodies for use in the treatment of tumors expressing predominantly high affinity egfr ligands or tumors expressing predominantly low affinity egfr ligands - Google Patents

Monoclonal and oligoclonal anti-egfr antibodies for use in the treatment of tumors expressing predominantly high affinity egfr ligands or tumors expressing predominantly low affinity egfr ligands Download PDF

Info

Publication number
US20140170668A1
US20140170668A1 US14/181,307 US201414181307A US2014170668A1 US 20140170668 A1 US20140170668 A1 US 20140170668A1 US 201414181307 A US201414181307 A US 201414181307A US 2014170668 A1 US2014170668 A1 US 2014170668A1
Authority
US
United States
Prior art keywords
egfr
kit
tumor
treatment
sample
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US14/181,307
Inventor
Raghida BUKHALID
Ulrik Nielsen
Shannon WERNER
Jeffrey David KEARNS
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Merrimack Pharmaceuticals Inc
Original Assignee
Merrimack Pharmaceuticals Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Merrimack Pharmaceuticals Inc filed Critical Merrimack Pharmaceuticals Inc
Priority to US14/181,307 priority Critical patent/US20140170668A1/en
Assigned to MERRIMACK PHARMACEUTICALS, INC reassignment MERRIMACK PHARMACEUTICALS, INC ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: Kearns, Jeffrey David, BUKHALID, RAGHIDA, NIELSEN, ULRIK, Werner, Shannon
Publication of US20140170668A1 publication Critical patent/US20140170668A1/en
Priority to US14/833,834 priority patent/US20160083800A1/en
Priority to US15/462,738 priority patent/US20170356049A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • C12Q1/6886Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material for cancer
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/3955Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against proteinaceous materials, e.g. enzymes, hormones, lymphokines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/39558Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against tumor tissues, cells, antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/08Drugs for disorders of the urinary system of the prostate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2863Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against receptors for growth factors, growth regulators
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/32Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against translation products of oncogenes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6844Nucleic acid amplification reactions
    • C12Q1/686Polymerase chain reaction [PCR]
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57484Immunoassay; Biospecific binding assay; Materials therefor for cancer involving compounds serving as markers for tumor, cancer, neoplasia, e.g. cellular determinants, receptors, heat shock/stress proteins, A-protein, oligosaccharides, metabolites
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57484Immunoassay; Biospecific binding assay; Materials therefor for cancer involving compounds serving as markers for tumor, cancer, neoplasia, e.g. cellular determinants, receptors, heat shock/stress proteins, A-protein, oligosaccharides, metabolites
    • G01N33/57496Immunoassay; Biospecific binding assay; Materials therefor for cancer involving compounds serving as markers for tumor, cancer, neoplasia, e.g. cellular determinants, receptors, heat shock/stress proteins, A-protein, oligosaccharides, metabolites involving intracellular compounds
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/74Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving hormones or other non-cytokine intercellular protein regulatory factors such as growth factors, including receptors to hormones and growth factors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • A61K2039/507Comprising a combination of two or more separate antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/71Receptors; Cell surface antigens; Cell surface determinants for growth factors; for growth regulators
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/21Immunoglobulins specific features characterized by taxonomic origin from primates, e.g. man
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/34Identification of a linear epitope shorter than 20 amino acid residues or of a conformational epitope defined by amino acid residues
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/55Fab or Fab'
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/565Complementarity determining region [CDR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/106Pharmacogenomics, i.e. genetic variability in individual responses to drugs and drug metabolism
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/475Assays involving growth factors
    • G01N2333/485Epidermal growth factor [EGF] (urogastrone)
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/475Assays involving growth factors
    • G01N2333/495Transforming growth factor [TGF]
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/52Predicting or monitoring the response to treatment, e.g. for selection of therapy based on assay results in personalised medicine; Prognosis

Definitions

  • the epidermal growth factor receptor is a cell surface transmembrane receptor of the HER/ErbB receptor family that transmits signals (including mitogenic signals that drive cell proliferation) to the interior of a cell when activated, typically by the binding of any of a number of extracellular ligands such as epidermal growth factor (EGF).
  • EGFR ligands vary in their affinity for EGFR and are categorized as either high- or low-affinity ligands. It is thought that the high- and low-affinity interactions between EGFR and its ligands activate different signaling pathways. This signal transmission occurs through a cascade of intracellular events beginning with protein phosphorylation mediated by receptor tyrosine kinase activity.
  • EGFR has proven a responsive target for anti-proliferative (e.g., anti-cancer) drugs, including “small molecule” tyrosine kinase inhibitor drugs (typically no larger than 700-900 AMU) that may be orally administered as well as monoclonal antibody based drugs that specifically bind to the extracellular domain of EGFR.
  • EGFR-targeted monoclonal antibodies are not always effective against EGFR-expressing tumors.
  • One approach taken with the aim of improving anti-EGFR antibody efficacy has been to develop mixtures of anti-EGFR monoclonal antibodies (i.e., oligoclonal antibodies) targeted to different sites (epitopes) on to the extracellular domain of EGFR.
  • Anti-EGFR antibodies such as those monoclonal and oligoclonal antibodies described in PCT Int. Pub. No. WO/2011/140254 and corresponding pending U.S. patent application Ser. No. 13/100,920, in pending U.S. provisional patent applications Nos. 61/504,633 and 61/558,945, and in U.S. Pat. No.
  • 7,887,805 (the “Oligoclonal Applications”), as well as oligoclonal mixtures of such antibodies in combination with other anti-EGFR antibodies, are useful for treatment of cancers, e.g., malignant (neoplastic) tumors.
  • cancers include but are not limited to, carcinoma, adenoma, blastoma, sarcoma, and lymphoma.
  • cancers include squamous cell cancer, small-cell lung cancer, non-small cell lung cancer, gastric cancer, pancreatic cancer, glial cell tumors such as glioblastoma and neurofibromatosis, cervical cancer, ovarian cancer, liver cancer, bladder cancer, hepatoma, breast cancer, colon cancer, melanoma, colorectal cancer, endometrial carcinoma, salivary gland carcinoma, kidney cancer, renal cancer, prostate cancer, vulvar cancer, thyroid cancer, hepatic carcinoma and various types of head and neck cancer.
  • a cancer treated or diagnosed using the methods disclosed herein is selected from melanoma, breast cancer, ovarian cancer, renal carcinoma, gastrointestinal/colon cancer, lung cancer (e.g., NSCLC), and prostate cancer.
  • theranostic methods predicting which tumors (e.g., malignant tumors) will be responsive to treatment with oligoclonal anti-EGFR antibodies, but not responsive to treatment with single monoclonal anti-EGFR antibodies, are provided. Also provided are kits useful for predicting responsiveness of tumors to anti-EGFR monoclonal and oligoclonal antibodies and methods of use thereof.
  • a preparation comprising only a single species of anti-EGFR monoclonal antibody (as opposed to preparations comprising mixtures of monoclonal antibodies) is provided for use in treating a patient having a tumor (e.g., a malignant tumor) that is predicted to have a favorable outcome (as described below) as a result of treatment #1, i.e., treatment with the preparation comprising only a single species of monoclonal antibody, and as a result of treatment #2, i.e., treatment with an oligoclonal anti-EGFR antibody preparation comprising a plurality of species of monoclonal anti-EGFR antibodies, one against each of at least two extracellular epitopes of EGFR, one of which antibody against at least two extracellular epitopes of EGFR being an antibody that inhibits ligand binding to EGFR.
  • a tumor e.g., a malignant tumor
  • treatment #2 i.e., treatment with an oligoclonal anti-EGFR antibody preparation comprising
  • a preparation comprising a plurality of species of monoclonal anti-EGFR antibodies is provided for use in treating a patient having a tumor that is predicted to have a favorable outcome as a result of treatment #2, but not as a result of treatment #1.
  • Identification of tumors predicted to respond to treatment #1 and treatment #2, or to treatment #2, but not to treatment #1 comprises obtaining a biopsy sample of the tumor and:
  • the at least two low affinity EGFR ligands is at least three low affinity ligands and the at least two high affinity EGFR ligands is three high affinity ligands. In another embodiment, the at least two low affinity EGFR ligands is four low affinity ligands and the at least two high affinity EGFR ligands is three high affinity ligands. In another embodiment, the at least two high affinity EGFR ligands is three high affinity ligands. In another embodiment, the at least two low affinity EGFR ligands is four low affinity ligands and the at least two high affinity EGFR ligands is at least two high affinity ligands.
  • a monoclonal antibody preparation comprising only a single species of monoclonal antibody is provided for use in the treatment of a patient predicted to have a favorable outcome from treatment with either of the monoclonal anti-EGFR antibody preparation or the oligoclonal anti-EGFR antibody preparation and the patient is subsequently treated with the monoclonal preparation. In another embodiment, this patient is subsequently treated with the oligoclonal anti-EGFR antibody preparation.
  • the patient is subsequently treated with the oligoclonal anti-EGFR antibody preparation.
  • the monoclonal anti-EGFR antibody preparation comprises a single monoclonal antibody that may be selected from, e.g., cetuximab, zalutumumab, nimotuzumab, matuzumab and panitumumab, all of which block ligand binding to EGFR, and therefore bind to the same or overlapping EGFRepitopes (Bin 1) and are not suitable for use together in oligoclonal antibody preparations.
  • Members of the plurality of anti-EGFR antibody species in an oligoclonal preparation separately and uniquely bind to two different extracellular epitopes of EGFR, may separately and uniquely bind to at least three extracellular epitopes of EGFR (in some cases no more than three).
  • Such a plurality may comprise two or three different species of monoclonal anti-EGFR antibodies, and in some embodiments no more than three different species. Mixtures comprising more than one antibody against any one epitope of EGFR are less preferred.
  • the oligoclonal preparations are duos, trios, or fourfold combinations of antibodies as disclosed in the Oligoclonal Applications.
  • the oligoclonal preparations provided herein comprise one or more of the anti-EGFR antibodies (e.g., of the above duos or trios or other oligoclonal combinations) that are not Bin 1 antibodies in combination with one of cetuximab, zalutumumab, nimotuzumab, matuzumab and panitumumab (which are all Bin 1 antibodies).
  • the anti-EGFR antibodies e.g., of the above duos or trios or other oligoclonal combinations
  • the oligoclonal preparations provided herein comprise one or more of the anti-EGFR antibodies (e.g., of the above duos or trios or other oligoclonal combinations) that are not Bin 1 antibodies in combination with one of cetuximab, zalutumumab, nimotuzumab, matuzumab and panitumumab (which are all Bin 1 antibodies).
  • the patient is subsequently treated with combination therapy comprising separate administration of at least two different monoclonal anti-EGFR antibodies.
  • the at least two different monoclonal anti-EGFR antibodies are selected from any of the anti-EGFR antibodies disclosed in the Oligoclonal Applications, as well as from cetuximab, zalutumumab, nimotuzumab, matuzumab and panitumumab, provided that one of the anti-EGFR antibodies is an antibody that inhibits ligand binding to EGFR.
  • the tumor is a tumor of the skin, central nervous system, head, neck, esophagus, stomach, colon, rectum, anus, liver, pancreas, bile duct, gallbladder, lung, breast, ovary, uterus, cervix, vagina, testis, germ cells, prostate, kidney, ureter, urinary bladder, adrenal, pituitary, thyroid, bone, muscle or connective tissue.
  • a method of treating a tumor in a patient by administration of a monoclonal anti-EGFR antibody preparation comprising a single species of monoclonal antibody comprising, prior to the administration, determining that the tumor does not have a level of high affinity ligands that is equal to or greater than the level of low affinity ligands in the tumor and not administering the monoclonal preparation if the tumor does have a level of high affinity ligands that is equal to or greater than the level of low affinity ligands in the tumor.
  • a method of treating a tumor in a patient by administration of an oligoclonal anti-EGFR antibody preparation comprising a plurality of species of monoclonal anti-EGFR antibodies, one against each of at least two extracellular epitopes of EGFR, one of the species of monoclonal anti-EGFR antibody inhibiting the binding of ligand to EGFR, is provided, the method comprising, prior to the administration, determining that the tumor has a level of high affinity ligands that is equal to or greater than the level of low affinity ligands in the tumor.
  • the oligoclonal anti-EGFR antibody preparation is MM-151.
  • kits for testing a tumor biopsy sample to determine levels of both high and low affinity EGFR ligands in the sample comprising;
  • the at least two pairs of high affinity EGFR ligand-specific polymerase chain reaction primers are specific to at least two of betacellulin, EGF, HB-EGF or TGF ⁇ and each of the at least two pairs of low affinity EGFR ligand-specific polymerase chain reaction primers are specific to at least two of amphiregulin, epigen, or epiregulin.
  • the at least two pairs of high affinity EGFR ligand-specific primers consist of all of betacellulin, EGF, HB-EGF and TGF ⁇ and the at least two pairs of high affinity EGFR ligand-specific primers consist of all of amphiregulin, epigen, and epiregulin.
  • the kit comprises at least one fluorescent reporter molecule suitable for use in a real-time RT-PCR assay.
  • the at least one RT-PCR reagent is one or more of an RNA-dependent DNA polymerase, a DNA-dependent DNA polymerase, a buffer, or a solution comprising at least micromolar concentrations of each of adenosine triphosphate (ATP), guanosine triphosphate (GTP), cytidine triphosphate (CTP), and thymidine triphosphate (TTP).
  • the one or more containers comprises either or both of at least one container that has an internal temperature of below 20° C. and above 0° C., and at least one container that has an internal temperature of below 0° C.
  • the contents of all, or at least one, of the at least one container has been prepared under cGMP conditions.
  • the oligoclonal anti-EGFR antibody preparation is a composition comprising a trio of anti-EGFR antibodies comprising a first antibody, a second antibody and a third antibody, wherein (i) the first antibody is, or competes for binding to EGFR with, or binds to the same epitope as, an antibody selected from the group consisting of ca, cb and cc; (ii) the second antibody is, or competes for binding to EGFR with, or binds to the same epitope as, an antibody selected from the group consisting of cd, ce and cf; and (iii) the third antibody is, or competes for binding to EGFR with, or binds to the same epitope as, an antibody selected from the group consisting of cg, ch, ci, cj and ck, wherein ca, cb, cd, ce, cf, cg, ch, ci, cj, and ck are each
  • FIGS. 1A-1C Phospho-EGF receptor and phospho-ERK signaling inhibition by single and pairwise combinations of Bin 1+Bin 2 or Bin 1+Bin 3 antibodies and comparisons with other known anti-EGFR antibodies such as cetuximab, nimotuzumab, and zalutumumab.
  • FIG. 1A shows inhibition of ERK activation by the Bin1/2 antibodies cb and cd.
  • FIG. 1B shows the inhibition of EGFR activation by the Bin1/2 antibodies and cd.
  • FIG. 1C shows the inhibition of ERK activation by the Bin1/3 antibodies cb and ch. Lines depict a five parameter logistic fit to the data from each combination.
  • FIGS. 2A-2G Inhibition of ligand-mediated tumor cell signaling in A431 cells preincubated with varying concentrations of anti-EGFR monoclonal antibodies cb (Bin1), cd (Bin2), cetuximab, zalutumumab, or nimotuzumab; as well as the oligoclonal combination of cb+cd; for 2 hrs. After incubation cells were stimulated with an EGFR ligand (8 nanomolar final concentration) for about 10 minutes.
  • FIG. 2A shows ELISA analysis of phospho-ERK (pERK) production (y-axis) as a function of antibody concentration (x-axis, in Log Molar concentration) after stimulation with the ligands amphiregulin ( FIG. 2A ), epigen ( FIG. 2B ), epiregulin ( FIG. 2C ), betacellulin ( FIG. 2D ), epidermal growth factor (EGF, FIG. 2E ), heparin-binding EGF-like growth factor (HB-EGF, FIG. 2F ), or transforming growth factor ⁇ (TGF- ⁇ , FIG. 2G ).
  • A431 cells incubated in the absence of anti-EGFR antibodies but with the ligand indicated in each graph (+Lig) or without ligand stimulation ( ⁇ Lig) were used as positive and negative controls, respectively.
  • FIGS. 3A-3G Inhibition of ligand-mediated tumor cell signaling in A431 cells preincubated with varying concentrations of anti-EGFR monoclonal antibodies cb (Bin1), ch (Bin3), cetuximab, zalutumumab, or nimotuzumab; as well as the oligoclonal combination of cb+ch; for 2 hrs. After pre-incubation with antibodies, cells were stimulated with an EGFR ligand (8 nanomolar final concentration) for 10 minutes.
  • FIG. 3A shows ELISA analysis of phospho-ERK (pERK) production (y-axis) as a function of antibody concentration (x-axis, in Log Molar concentration) after stimulation with the ligands amphiregulin ( FIG. 3A ), epigen ( FIG. 3B ), epiregulin ( FIG. 3C ), betacellulin ( FIG. 3D ), epidermal growth factor (EGF, FIG. 3E ), heparin-binding EGF-like growth factor (HB-EGF, FIG. 3F ), or transforming growth factor ⁇ (TGF- ⁇ , FIG. 3G ).
  • A431 cells incubated in the absence of anti-EGFR antibodies but with the ligand indicated in each graph (+Lig) or without ligand stimulation ( ⁇ Lig) were used as positive and negative controls, respectively.
  • FIGS. 4A-4L Inhibition of high affinity EGFR ligand-mediated tumor cell proliferation.
  • H322M cells FIGS. 4A-4D
  • H1975 cells FIGS. 4E-4H
  • LIM1215 cells FIGS. 4I-4L
  • FIGS. 4A-4D H322M cells
  • H1975 cells FIGS. 4E-4H
  • LIM1215 cells FIGS. 4I-4L
  • FIGS. 4I-4L were treated with varying concentrations of anti-EGFR monoclonal and oligoclonal antibodies in the presence of EGFR ligands.
  • Cells were treated with 200 ng/ml amphiregulin (AREG) ( FIGS. 4A , 4 E, and 4 I), 50 ng/ml EGF ( FIGS. 4B , 4 F, and 4 J), 50 ng/ml TGF ⁇ ( FIGS.
  • EGF amphiregulin
  • FIGS. 4D , 4 H, and 4 L 90 ng/ml HB-EGF
  • FIGS. 4D , 4 H, and 4 L 90 ng/ml HB-EGF
  • MM-151 open circles or cetuximab (CTX, solid squares; Bristol-Myers Squibb).
  • CX cetuximab
  • Cells treated with ligand (+Lig, upward arrow) or without ligand ( ⁇ Lig, downward arrow) in the absence of antibody treatment served as controls.
  • the y-axes represent cell viability as the fraction of the viability of the amphiregulin-treated control cells and the x-axes represent antibody concentration in Log (Molar).
  • FIGS. 5A-5L Effect of EGFR high affinity ligand titration on cell responsiveness to anti-EGFR inhibitors in vitro.
  • the non-small cell lung cancer (NSCLC) lines H322M ( FIGS. 5A-5D ), HCC827 ( FIGS. 5E-5H ), and H1975 ( FIGS. 5I-5L ) were tested.
  • Controls were growth in media with amphiregulin alone (+AREG, 200 ng/ml) or EGF alone as a control (+EGF, 20 ng/ml) or no added ligand ( ⁇ Lig).
  • Treatments were with varying concentrations (0.1-1 ⁇ M final concentration) of MM-151 or cetuximab (CTX) in the following conditions: amphiregulin alone (200 ng/ml, FIGS. 5A , 5 E, and 5 I); a 1000:1 amphiregulin:EGF ratio (0.2 ng/ml EGF, FIGS. 5B , 5 F, and 5 J); a 100:1 amphiregulin:EGF ratio (2 ng/ml EGF, FIGS. 5C , 5 G, and 5 K); and a 10:1 amphiregulin:EGF ratio (20 ng/ml EGF, FIGS. 5D , 5 H, and 5 L).
  • the y-axes represent cell viability as a fraction of the viability of the AREG-treated control cells, whereas and the x-axes represent antibody concentration in Log (Molar).
  • FIG. 6 Effect of EGFR ligand concentration on phopho-ERK cell signaling.
  • the epidermoid cancer cell line A431 was treated with media alone (“No Inhibitor”), MM-151 (100 nM) or cetuximab (100 nM) for 2 hrs, followed by the addition of various concentrations of EGF (0.16 ng/ml, 0.8 ng/ml, 4.0 ng/ml, 20 ng/ml, 100 ng/ml) or AREG (0.48 ng/ml, 2.4 ng/ml, 12 ng/ml, 60 ng/ml, 300 ng/ml), alone or in combination. Cells were incubated with the various EGF and AREG ligand combinations for 10 minutes, lysed, and levels of ERK phosphorylation measured by phospho-ERK ELISA.
  • EGFR human EGFR protein
  • HER1 human EGFR protein
  • 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 except for possible naturally occurring mutations that may be present in minor amounts. Such antibodies may be obtained, e.g. from hybridomas or by recombinant expression. Antigen binding fragments (including scFvs) of such immunoglobulins are also encompassed by the term “monoclonal antibody” as used herein. Monoclonal antibodies are highly specific, generally being directed against a single epitope on a single antigen site, e.g., on the extracellular domain of EGFR. Monoclonal antibodies include chimeric antibodies - whose variable regions derive from a first animal species (e.g., mouse) and whose constant regions derive from a second animal species (e.g., human), human antibodies and humanized antibodies.
  • first animal species e.g., mouse
  • a second animal species e.g
  • treat refers to therapeutic or preventative measures described herein.
  • the methods of “treatment” employ administration to a subject, an antibody or antibody pair or trio disclosed herein, for example, a subject having a disorder associated with EGFR dependent signaling or predisposed to having such a disease or disorder, in order to prevent, cure, delay, reduce the severity of, or ameliorate one or more symptoms of the disease or disorder or recurring disease or disorder, or in order to prolong the survival of a subject beyond that expected in the absence of such treatment.
  • anti-EGFR antibodies include cetuximab, panitumumab and nimotuzumab (which is not yet available in the US market).
  • Other pharmaceutical anti-EGFR antibodies include zalutumumab, and matuzumab, which are in development.
  • Still other anti-EGFR antibodies include those disclosed in the Oligoclonal Applications, e.g., the antibodies disclosed below.
  • P1X is a human IgG1 having a heavy chain variable region comprising SEQ ID NO: 1 and a light chain variable region comprising SEQ ID NO: 2;
  • P2X is a human IgG1 having a heavy chain variable region comprising SEQ ID NO: 3 and a light chain variable region comprising SEQ ID NO: 4;
  • P3X is a human IgG1 having a heavy chain variable region comprising SEQ ID NO: 5 and a light chain variable region comprising SEQ ID NO: 6.
  • MM-151 indicates a triple combination of P1X+P2X+P3X at a P1X:P2X:P3X molar ratio of 2:2:1.
  • patient includes human and other mammalian subjects that receive either prophylactic or therapeutic treatment.
  • sample refers to tissue, body fluid, or a cell (or a fraction of any of the foregoing) taken from a patient. Normally, the tissue or cell will be removed from the patient, but in vivo diagnosis is also contemplated.
  • a tissue sample can be taken from a surgically removed tumor and prepared for testing by conventional techniques.
  • lymphomas and leukemias lymphocytes, leukemic cells, or lymph tissues can be obtained (e.g., leukemic cells from blood) and appropriately prepared.
  • Other samples including urine, tears, serum, plasma, cerebrospinal fluid, feces, sputum, cell extracts etc. can also be useful for particular cancers.
  • a patient having a tumor predicted by the methods disclosed herein to have a favorable outcome following treatment with a monoclonal or oligoclonal anti-EGFR antibody, and who is then treated accordingly, may exhibit one of the following responses to therapy:
  • patients treated as disclosed herein may experience improvement in at least one sign of cancer.
  • the patient so treated exhibits pCR, CR, PR, or SD.
  • the patient so treated experiences tumor shrinkage and/or decrease in growth rate, i.e., suppression of tumor growth.
  • unwanted cell proliferation is reduced or inhibited.
  • one or more of the following can occur: the number of cancer cells can be reduced; tumor size can be reduced; cancer cell infiltration into peripheral organs can be inhibited, retarded, slowed, or stopped; tumor metastasis can be slowed or inhibited; tumor growth can be inhibited; recurrence of tumor can be prevented or delayed; one or more of the symptoms associated with cancer can be relieved to some extent.
  • such improvement is measured by a reduction in the quantity and/or size of measurable tumor lesions.
  • Measurable lesions are defined as those that can be accurately measured in at least one dimension (longest diameter is to be recorded) as ⁇ 10 mm by CT or MRI scan (e.g., CT scan slice thickness no greater than 5 mm), 10 mm caliper measurement by clinical exam or >20 mm by chest X-ray.
  • the size of non-target lesions can also be measured for improvement.
  • lesions can be measured on x-rays or CT or MRI images.
  • cytology or histology can be used to evaluate responsiveness to a therapy.
  • the cytological confirmation of the neoplastic origin of any effusion that appears or worsens during treatment when the measurable tumor has met criteria for response or stable disease can be considered to differentiate between response or stable disease (an effusion may be a side effect of the treatment) and progressive disease.
  • a beneficial response to therapy is indicated by at least one therapeutic effect selected from the group consisting of reduction in size of a tumor, reduction in number of metastatic lesions appearing over time, complete remission, partial remission, stable disease, increase in overall response rate, or a pathologic complete response.
  • compositions for use in the methods provided for herein are commercially available anti-EGFR compositions, e.g., of cetuximab, panitumumab and nimotuzumab, as well as the various pharmaceutical compositions provided in the Oligoclonal Applications.
  • oligoclonal anti-EGFR antibodies for the treatment of a disease associated with high-affinity EGFR ligand-driven signaling is also provided, as are methods of use of oligoclonal anti-EGFR antibodies for the treatment of tumor comprising protein or mRNA levels of at least two high-affinity EGFR ligands that are higher than levels in the tumor of at least two low-affinity EGFR ligands.
  • Cancers treated in accordance with the methods provided include melanoma, breast cancer, ovarian cancer, renal carcinoma, gastrointestinal cancer, gastro-esophageal junction cancer, colon cancer, lung cancer, pancreatic cancer, skin cancer, head and neck cancer glioblastoma, prostate cancer and other solid and/or metastatic tumors.
  • the monoclonal or oligoclonal antibody can be administered alone or with another therapeutic agent that acts in conjunction with or synergistically with the oligoclonal antibody to treat the disease associated with EGFR-mediated signaling.
  • kits for testing a tumor sample e.g., a tumor biopsy sample or a circulating tumor cell, to determine levels of both high and low affinity EGFR ligands in the sample, said kits being comprised by one or more containers comprising;
  • the kit may further contain instructions for use in determining how to treat a tumor in a patient following determination of levels of high and low affinity ligands in a sample of the tumor.
  • the kit may include an indication of the intended use of the contents of the kit (e.g., in the form of a label or other printed or recorded matter).
  • a cryopulverizer (COVARIS Inc.) is used for the pulverization of tumors. Tumors are stored in special bags (pre-weighed before the addition of the tumor) and placed in liquid nitrogen while handling them. For small tumors, 200 ⁇ L of Lysis buffer is first added to the bag containing the tumor, frozen in liquid nitrogen and then pulverized to improve the recovery of the tumor from the bag. Pulverized tumors are transferred to 2 mL EPPENDORF tubes and placed in liquid nitrogen until ready for further processing.
  • Tumors are lysed in Lysis buffer supplemented with protease and phosphatase inhibitors. Lysis Buffer is added to the tumor aliquots in a final concentration of about 62.5 mg/mL. Tumor samples are homogenized by vortexing for 30 sec and incubating on ice for about 30 min. The lysates are spun for about 10 min in Qiagen QIASHREDDER columns for further homogenization of the samples. Cleared lysates are aliquoted into fresh tubes for further processing.
  • A431 ATCC CRL-1555TM epidermoid carcinoma cells are seeded at a density of 35,000 cells/well or 17,500 cells per half well in 96 well tissue culture plates and grown in DMEM medium supplemented with antibiotics, 2 mM L-glutamine and 10% fetal bovine serum (FBS) for 24 hours at 37° C. and 5% carbon dioxide.
  • FBS fetal bovine serum
  • Cells are serum starved in 1% FBS medium with antibiotics and 2 mM L-glutamine for about 20 hours at 37° C. and 5% carbon dioxide. Cells are then treated as described below in each Example.
  • Lysates are either analyzed immediately by ELISA for phospho-ERK (a downstream effector of EGFR) or frozen at ' 1 80° C. until use.
  • 96-half well GREINER high binding plates (Cat. #675077; GREINER BIO-ONE, Monroe, N.C.) are coated with 50 ⁇ L of an EGFR antibody (4 ⁇ g/ml final concentration; EGFR Ab-11, Clone: 199.12, without BSA and azide, Fisher Scientific, cat #MS396P1ABX), and incubated overnight at room temperature. Next morning, plates are washed 3 times with 100 ⁇ l/well PBST (0.05% Tween-20) on a BIOTEK plate washer. Plates are subsequently blocked for about 1 hour at room temperature with 2% BSA in PBS.
  • the plates are washed 3 times with 100 ⁇ l/well PBST (0.05%Tween-20) on the BIOTEK plate washer.
  • Cell lysates 50 ⁇ l
  • standards pEGFR pY1068 ELISA kit, R&D Systems, cat #DYC3570
  • 50% Lysis buffer and 1%BSA-PBS per the manufacturer's recommendations
  • Plates are then washed 3 times with 100 ⁇ l/well in the BIOTEK plate washer with PBST (PBS with 0.05%Tween-20).
  • HRP horseradish peroxidase
  • a detection antibody pEGFR pY1068 ELISA kit, R&D Systems, cat #DYC3570 conjugated to horseradish peroxidase (HRP) diluted (as per manufacturer's instructions) in 2% BSA, PBS is added and incubated for about 2 hour at room temperature. The plate is washed 3 times with 100 ⁇ l/well in the BIOTEK plate washer with PBST (0.05%Tween-20). About 50 ⁇ L of SUPERSIGNAL PICO ELISA substrate is added and the plate is read using an Envision (Perkin Elmer) plate reader. The data are analyzed and duplicate samples are averaged and error bars are used to represent the standard deviation between the two replicates.
  • HRP horseradish peroxidase
  • the phospho-ERK ELISA is performed similarly to the phospho-EGFR ELISA with the following changes:
  • Human pERK ELISA DUOSET kit is purchased from R&D Systems (cat #DYC1018-5) and used as recommended by the manufacturer. The data are analyzed by subtracting background signal, regressing to a recombinant standard supplied by the manufacturer, and back-calculating the data (BCD) to correct for dilution factors. Duplicate samples are averaged and error bars are used when indicated to represent the standard deviation between two replicates.
  • A431 cells were treated with single antibodies or antibody pairs and their ability to inhibit EGFR-dependent signaling was compared to that each of cetuximab, nimotuzumab, and zalutumumab.
  • Cells were incubated with varying concentrations of anti-EGFR antibodies for 2 hrs, and then stimulated with an EGFR ligand for 10 minutes at 37° C. and 5% carbon dioxide.
  • the seven recombinant human EGFR ligands used individually were 100 ng/ml amphiregulin (“AREG,” R&D Systems, cat #262-AR/CF), 100 ng/ml betacellulin (R&D Systems, cat #261-CE-050/CF), EGF (PreproTech, cat #AF-100-15), 220 ng/ml epigen (epithelial mitogen homolog, PreproTech, cat #100-51), 150 ng/ml epiregulin (R&D Systems, cat #1195-EP/CF), 90 ng/ml HB-EGF (heparin-binding EGF-like growth factor, PreproTech, cat #100-47), and 50 ng/ml TGF ⁇ (transforming growth factor alpha, R&D Systems, cat #239-A).
  • FIGS. 1A-C ELISA measurements were performed as described above for pERK and pEGFR signaling and the results are shown in FIGS. 1A-C .
  • Single antibodies cb, cd, and ch, or pairs of cb and cd or cb and ch, (as described above in Example 1) were used to treat A431 cells at indicated total concentrations, and their ability to inhibit EGFR ligand-dependent signaling was compared to that of each single anti-EGFR antibodies cetuximab, nimotuzumab, and zalutumumab at the same concentrations. Cells were incubated with antibody for 2 hours followed by stimulation with EGFR ligand for 10 minutes.
  • amphiregulin 100 ng/ml
  • betacellulin 100 ng/ml
  • EGF 50 ng/ml
  • epigen 220 ng/ml
  • epiregulin 150 ng/ml
  • HB-EGF 90 ng/ml
  • TGF ⁇ 50 ng/ml
  • cb and cd were effective at inhibiting phospho-ERK signaling (i.e., inhibiting phosphorylation of ERK1 and ERK2) in response to the three ligands with low affinity for EGF receptor (amphiregulin, epigen, and epiregulin), but not in response to the four ligands with high affinity for EGF receptor (betacellulin, EGF, HB-EGF, and TGF ⁇ ). Only oligoclonal mixtures of Bin1/Bin2 antibodies cb and cd ( FIGS.
  • FIGS. 3A-G Bin1/Bin3 antibodies cb and ch
  • FIGS. 3A-G Bin1/Bin3 antibodies cb and ch
  • FIGS. 3A-G were effective at essentially completely inhibiting phospho-ERK signaling in response to all seven (both high- and low-affinity) EGFR ligands when compared to individual components of the mixtures, cb, cd, and ch and the other tested individual monoclonal antibodies, cetuximab, nimotuzumab, and zalutumumab.
  • Inhibition of tumor cell signaling in vitro is analyzed by the methods described above or minor variations thereof.
  • the epidermoid cancer cell line A431 was treated with media alone (“No Inhibitor”), MM-151 (100 nM) or cetuximab (100 nM) for 2 hrs, followed by the addition of various concentrations of EGF (0.16 ng/ml, 0.8 ng/ml, 4.0 ng/ml, 20 ng/ml, 100 ng/ml) or AREG (0.48 ng/ml, 2.4 ng/ml, 12 ng/ml, 60 ng/ml, 300 ng/ml), alone or in combination, as shown in FIG. 6 .
  • FIG. 6 shows MM-151 and cetuximab-mediated modulation of ERK signaling represented as fraction of the highest signal across all treatments.
  • Phospho-ERK signaling is inhibited by cetuximab in A431 cells under low-affinity EGFR ligand (AREG) stimulation, but become increasingly resistant to inhibition upon the addition of increasing amounts of the high-affinity EGFR ligand, EGF (middle panel), while inhibition of signaling by MM-151 is largely maintained under all conditions (lower panel).
  • H322M NCI, Frederick, Md. 21701
  • H1975 ATCC CRL-2868TM
  • LIM1215 Cell Bank Australia, NSW 2145
  • H322M and H1975 fetal calf serum
  • FCS fetal calf serum
  • RPMI-1640 medium supplemented with 25 mM HEPES, antibiotics, 2 mM L-glutamine, 10% FCS, 0.6 ⁇ g/ml insulin, 1 ⁇ g/ml hydrocortisone and 10 ⁇ M thioglycerol (LIM1215) for 24 hours at 37 degrees Celsius and 5% carbon dioxide.
  • RPMI-1640 with antibiotics, 2 mM L-glutamine, 1% FBS (for H322M and H1975) or RPMI-1640 with 25 mM HEPES, antibiotics, 2 mM L-glutamine, 1% FCS, 0.6 ⁇ g/ml Insulin, 1 ⁇ g/ml hydrocortisone and 10 ⁇ M thioglycerol (for LIM1215) supplemented with 200 ng/ml AREG, 50 ng/ml EGF, 50 ng/ml TGF ⁇ or 90 ng/ml HB-EGF in the presence of varying concentrations of MM-151 or cetuximab (Bristol-Myers Squibb).
  • CTG CellTiter-Glo® Luminescent Viable Cell Number Assay (Promega Corporation) according to manufacturer's instructions.
  • the CTG assay measures the number of viable cells in culture based upon quantitation of ATP present, which is an indicator of metabolically active cells.
  • Control treatments include cells treated with 1% FCS-containing medium (as detailed above) in the presence (“+Lig”) or absence (“ ⁇ Lig”) of the respective ligand treatment. Viable cell numbers are plotted in GraphPad Prism (GraphPad Software, La Jolla, Calif.) as a fraction of the respective ligand (“+Lig”) treatment control.
  • the non-small cell lung cancer (NSCLC) lines H322M and H1975 and colon cancer cell line LIM1215 were treated with varying concentrations of MM-151or cetuximab (0.1-1 ⁇ M final concentration). Potent inhibition of growth of H322M, H1975 and LIM1215 cells was obtained over a range of MM-151 concentrations in the presence of high affinity EGFR ligands (EGF, TGF ⁇ , HB-EGF), but not in the presence of cetuximab or in assay medium alone (1% FCS)— FIG. 4 (B-D, F-H, and J-L).
  • EGFR ligands EGF, TGF ⁇ , HB-EGF
  • FCS assay medium alone
  • non-small cell lung cancer (NSCLC) cell lines H322M, HCC827 and H1975 were treated with AREG alone (200 ng/ml) or with AREG plus increasing amounts of EGF (0.2, 2, 20 ng/ml) in the presence of varying concentrations of MM-151or cetuximab (0.1-1 ⁇ M final concentration).
  • NSCLC non-small cell lung cancer
  • the NSCLC cell lines respond to cetuximab under low-affinity EGFR ligand stimulation (AREG), but become increasingly unresponsive to treatment upon the addition of increasing amounts of the high-affinity EGFR ligand EGF, while sensitivity to MM-151 is largely maintained (see FIGS. 5A-5L ).
  • RAG low-affinity EGFR ligand stimulation
  • RT-qPCR real-time quantitative polymerase chain reaction
  • Total RNA is isolated from patient biopsy/tumor samples, e.g., by commercially available standard methods.
  • the method of total RNA isolation may be any method (including conventional methods) suitable for use with the type of patient biopsy sample being tested, e.g., fresh, fixed, frozen, formalin fixed paraffin embedded (FFPE), etc.
  • Total RNA is then converted to cDNA using the gene specific primers described below and Qiagen® OneStep RT-PCR reagents and protocol (Cat. #210210, Qiagen, Germantown, Md.).
  • the cDNA is then used for RT-qPCR using the following gene specific primers as TaqMan® probe sets obtained from Applied Biosystems (Carlsbad, Calif.) along with reagents and equipment from the same source, all as described below:

Abstract

Disclosed are pharmaceutical preparations for, and methods for determining, appropriate and effective treatment with therapeutic agents comprising a single species of anti-EGFR monoclonal antibody or therapeutic agents comprising a plurality of species of such antibodies, as well as kits useful for making such determinations.

Description

    RELATED APPLICATIONS
  • This application is a divisional of U.S. patent application Ser. No. 13/488,270, filed on Jun. 4, 2012, which claims the benefit of U.S. Provisional Application No. 61/493,252, filed on Jun. 3, 2011, U.S. Provisional Application No. 61/504,633, filed on Jul. 5, 2011, and U.S. Provisional Application No. 61/558,945, filed on Nov. 11, 2011, the entire contents of which are incorporated by reference herein.
  • BACKGROUND
  • The epidermal growth factor receptor (EGFR) is a cell surface transmembrane receptor of the HER/ErbB receptor family that transmits signals (including mitogenic signals that drive cell proliferation) to the interior of a cell when activated, typically by the binding of any of a number of extracellular ligands such as epidermal growth factor (EGF). EGFR ligands vary in their affinity for EGFR and are categorized as either high- or low-affinity ligands. It is thought that the high- and low-affinity interactions between EGFR and its ligands activate different signaling pathways. This signal transmission occurs through a cascade of intracellular events beginning with protein phosphorylation mediated by receptor tyrosine kinase activity. EGFR has proven a responsive target for anti-proliferative (e.g., anti-cancer) drugs, including “small molecule” tyrosine kinase inhibitor drugs (typically no larger than 700-900 AMU) that may be orally administered as well as monoclonal antibody based drugs that specifically bind to the extracellular domain of EGFR. EGFR-targeted monoclonal antibodies are not always effective against EGFR-expressing tumors. One approach taken with the aim of improving anti-EGFR antibody efficacy has been to develop mixtures of anti-EGFR monoclonal antibodies (i.e., oligoclonal antibodies) targeted to different sites (epitopes) on to the extracellular domain of EGFR. See, e.g., PCT Int. Pub. No. WO/2011/140254 and U.S. Pat. No. 7,887,805. These developments have created a need to enable the identification of cancer patients whose tumors have characteristics rendering them unresponsive to monoclonal anti-EGFR antibodies so that such patients may receive effective treatment via administration of oligoclonal anti-EGFR antibodies. The present disclosure answers this need and provides other benefits.
  • SUMMARY
  • Provided herein are theranostic methods for predicting responsiveness of tumor cells that express EGFR to therapeutic agents comprising anti-EGFR antibodies, and methods based on such predictions for treating patients having such tumors with such therapeutic agents. Anti-EGFR antibodies such as those monoclonal and oligoclonal antibodies described in PCT Int. Pub. No. WO/2011/140254 and corresponding pending U.S. patent application Ser. No. 13/100,920, in pending U.S. provisional patent applications Nos. 61/504,633 and 61/558,945, and in U.S. Pat. No. 7,887,805 (the “Oligoclonal Applications”), as well as oligoclonal mixtures of such antibodies in combination with other anti-EGFR antibodies, are useful for treatment of cancers, e.g., malignant (neoplastic) tumors. Examples of cancers include but are not limited to, carcinoma, adenoma, blastoma, sarcoma, and lymphoma. More particular examples of such cancers include squamous cell cancer, small-cell lung cancer, non-small cell lung cancer, gastric cancer, pancreatic cancer, glial cell tumors such as glioblastoma and neurofibromatosis, cervical cancer, ovarian cancer, liver cancer, bladder cancer, hepatoma, breast cancer, colon cancer, melanoma, colorectal cancer, endometrial carcinoma, salivary gland carcinoma, kidney cancer, renal cancer, prostate cancer, vulvar cancer, thyroid cancer, hepatic carcinoma and various types of head and neck cancer. In a particular embodiment, a cancer treated or diagnosed using the methods disclosed herein is selected from melanoma, breast cancer, ovarian cancer, renal carcinoma, gastrointestinal/colon cancer, lung cancer (e.g., NSCLC), and prostate cancer.
  • Accordingly, theranostic methods predicting which tumors (e.g., malignant tumors) will be responsive to treatment with oligoclonal anti-EGFR antibodies, but not responsive to treatment with single monoclonal anti-EGFR antibodies, are provided. Also provided are kits useful for predicting responsiveness of tumors to anti-EGFR monoclonal and oligoclonal antibodies and methods of use thereof.
  • In one aspect, a preparation comprising only a single species of anti-EGFR monoclonal antibody (as opposed to preparations comprising mixtures of monoclonal antibodies) is provided for use in treating a patient having a tumor (e.g., a malignant tumor) that is predicted to have a favorable outcome (as described below) as a result of treatment #1, i.e., treatment with the preparation comprising only a single species of monoclonal antibody, and as a result of treatment #2, i.e., treatment with an oligoclonal anti-EGFR antibody preparation comprising a plurality of species of monoclonal anti-EGFR antibodies, one against each of at least two extracellular epitopes of EGFR, one of which antibody against at least two extracellular epitopes of EGFR being an antibody that inhibits ligand binding to EGFR. In another aspect, a preparation comprising a plurality of species of monoclonal anti-EGFR antibodies is provided for use in treating a patient having a tumor that is predicted to have a favorable outcome as a result of treatment #2, but not as a result of treatment #1.
  • Identification of tumors predicted to respond to treatment #1 and treatment #2, or to treatment #2, but not to treatment #1, comprises obtaining a biopsy sample of the tumor and:
      • a) measuring levels in the sample of at least two (e.g., at least three or at least four) low affinity EGFR ligands, e.g., selected from amphiregulin, epigen, or epiregulin, which may, for each ligand independently, be measured as levels of ligand protein or as levels of the corresponding RNA species coding for the ligand protein, in the biopsy sample,
      • b) measuring levels in the sample of at least two (e.g., at least three) high affinity EGFR ligands, e.g., selected from betacellulin, EGF, HB-EGF or TGFα, which may, for each ligand independently, be measured as protein levels or as levels of the corresponding RNA species coding for them, in the biopsy sample,
        • in one embodiment, all of the levels measured in a) are protein levels and all of the levels measured in b) are protein levels; in another embodiment all of the levels measured in a) are mRNA levels and all of the levels measured in b) are mRNA levels.
      • and,
      • c) comparing the average level of each of the high affinity EGFR ligands, or of each corresponding RNA species, measured in a) to the average level of each of the low affinity EGFR ligands, or of each corresponding RNA species measured in b)
        where if the average level of low affinity EGFR ligands, or RNAs coding for them, measured in a) is greater than the average level of high affinity EGFR ligands, or RNAs coding for them, measured in b), the patient is predicted to have the favorable outcome as a result of treatment #1, as well as as a result of treatment #2, and if the average level of low affinity EGFR ligands, or RNAs coding for them, measured in a) is less than or equal to the average level of high affinity EGFR ligands, or RNAs coding for them, measured in b), the patient is predicted to have an unfavorable outcome from treatment #1, but is predicted to have a favorable outcome from treatment #2.
  • In one embodiment, the at least two low affinity EGFR ligands is at least three low affinity ligands and the at least two high affinity EGFR ligands is three high affinity ligands. In another embodiment, the at least two low affinity EGFR ligands is four low affinity ligands and the at least two high affinity EGFR ligands is three high affinity ligands. In another embodiment, the at least two high affinity EGFR ligands is three high affinity ligands. In another embodiment, the at least two low affinity EGFR ligands is four low affinity ligands and the at least two high affinity EGFR ligands is at least two high affinity ligands.
  • In one embodiment, a monoclonal antibody preparation comprising only a single species of monoclonal antibody is provided for use in the treatment of a patient predicted to have a favorable outcome from treatment with either of the monoclonal anti-EGFR antibody preparation or the oligoclonal anti-EGFR antibody preparation and the patient is subsequently treated with the monoclonal preparation. In another embodiment, this patient is subsequently treated with the oligoclonal anti-EGFR antibody preparation.
  • In yet another embodiment, if the patient is predicted to have an unfavorable outcome from treatment with the monoclonal anti-EGFR antibody preparation, the patient is subsequently treated with the oligoclonal anti-EGFR antibody preparation.
  • The monoclonal anti-EGFR antibody preparation comprises a single monoclonal antibody that may be selected from, e.g., cetuximab, zalutumumab, nimotuzumab, matuzumab and panitumumab, all of which block ligand binding to EGFR, and therefore bind to the same or overlapping EGFRepitopes (Bin 1) and are not suitable for use together in oligoclonal antibody preparations. Members of the plurality of anti-EGFR antibody species in an oligoclonal preparation separately and uniquely bind to two different extracellular epitopes of EGFR, may separately and uniquely bind to at least three extracellular epitopes of EGFR (in some cases no more than three). Such a plurality may comprise two or three different species of monoclonal anti-EGFR antibodies, and in some embodiments no more than three different species. Mixtures comprising more than one antibody against any one epitope of EGFR are less preferred. In certain embodiments the oligoclonal preparations are duos, trios, or fourfold combinations of antibodies as disclosed in the Oligoclonal Applications. In other embodiments, the oligoclonal preparations provided herein comprise one or more of the anti-EGFR antibodies (e.g., of the above duos or trios or other oligoclonal combinations) that are not Bin 1 antibodies in combination with one of cetuximab, zalutumumab, nimotuzumab, matuzumab and panitumumab (which are all Bin 1 antibodies).
  • In another embodiment, if the patient is predicted to have an unfavorable outcome from treatment with the monoclonal anti-EGFR antibody preparation, the patient is subsequently treated with combination therapy comprising separate administration of at least two different monoclonal anti-EGFR antibodies. In certain aspects, the at least two different monoclonal anti-EGFR antibodies are selected from any of the anti-EGFR antibodies disclosed in the Oligoclonal Applications, as well as from cetuximab, zalutumumab, nimotuzumab, matuzumab and panitumumab, provided that one of the anti-EGFR antibodies is an antibody that inhibits ligand binding to EGFR.
  • In another embodiment the tumor is a tumor of the skin, central nervous system, head, neck, esophagus, stomach, colon, rectum, anus, liver, pancreas, bile duct, gallbladder, lung, breast, ovary, uterus, cervix, vagina, testis, germ cells, prostate, kidney, ureter, urinary bladder, adrenal, pituitary, thyroid, bone, muscle or connective tissue.
  • In another aspect, a method of treating a tumor in a patient by administration of a monoclonal anti-EGFR antibody preparation comprising a single species of monoclonal antibody is provided, the method comprising, prior to the administration, determining that the tumor does not have a level of high affinity ligands that is equal to or greater than the level of low affinity ligands in the tumor and not administering the monoclonal preparation if the tumor does have a level of high affinity ligands that is equal to or greater than the level of low affinity ligands in the tumor.
  • In another aspect, a method of treating a tumor in a patient by administration of an oligoclonal anti-EGFR antibody preparation comprising a plurality of species of monoclonal anti-EGFR antibodies, one against each of at least two extracellular epitopes of EGFR, one of the species of monoclonal anti-EGFR antibody inhibiting the binding of ligand to EGFR, is provided, the method comprising, prior to the administration, determining that the tumor has a level of high affinity ligands that is equal to or greater than the level of low affinity ligands in the tumor. In one embodiment, the oligoclonal anti-EGFR antibody preparation is MM-151.
  • Also provided are kits for testing a tumor biopsy sample to determine levels of both high and low affinity EGFR ligands in the sample, said kits being comprised by one or more containers comprising;
      • a) at least two pairs of high affinity EGFR ligand-specific polymerase chain reaction (PCR) primers,
      • b) at least two pairs of low affinity EGFR ligand-specific PCR primers, and
      • c) at least one reverse transcription PCR (RT-PCR) reagent.
  • In one embodiment the at least two pairs of high affinity EGFR ligand-specific polymerase chain reaction primers are specific to at least two of betacellulin, EGF, HB-EGF or TGFα and each of the at least two pairs of low affinity EGFR ligand-specific polymerase chain reaction primers are specific to at least two of amphiregulin, epigen, or epiregulin. In another embodiment the at least two pairs of high affinity EGFR ligand-specific primers consist of all of betacellulin, EGF, HB-EGF and TGFα and the at least two pairs of high affinity EGFR ligand-specific primers consist of all of amphiregulin, epigen, and epiregulin. In another embodiment the kit comprises at least one fluorescent reporter molecule suitable for use in a real-time RT-PCR assay. In yet another embodiment the at least one RT-PCR reagent is one or more of an RNA-dependent DNA polymerase, a DNA-dependent DNA polymerase, a buffer, or a solution comprising at least micromolar concentrations of each of adenosine triphosphate (ATP), guanosine triphosphate (GTP), cytidine triphosphate (CTP), and thymidine triphosphate (TTP). In a further embodiment the one or more containers comprises either or both of at least one container that has an internal temperature of below 20° C. and above 0° C., and at least one container that has an internal temperature of below 0° C. Preferably the contents of all, or at least one, of the at least one container has been prepared under cGMP conditions.
  • In another aspect, a method is provided for treating a patient having a cancerous tumor, the method comprising determining, according to any of the methods described above, if the patient is predicted to have the favorable outcome as a result of treatment #1 and as a result of treatment #2 or if the patient is predicted to have the favorable outcome as a result of treatment #2 but not as a result of treatment #1, wherein if the patient is predicted to have a favorable outcome as a result of treatment #1 and as a result of treatment #2, the patient is treated with treatment #1 or treatment #2; and if the patient is predicted to have a favorable outcome as a result of treatment #2 but not as a result of treatment #1, the patient is treated with treatment 2 and not with treatment #1. Determining whether the patient will have a favorable outcome may be accomplished by use of any of the theranostic methods described above, which may be accomplished by use of a kit described above.
  • In certain aspects of the above methods, the oligoclonal anti-EGFR antibody preparation is a composition comprising a trio of anti-EGFR antibodies comprising a first antibody, a second antibody and a third antibody, wherein (i) the first antibody is, or competes for binding to EGFR with, or binds to the same epitope as, an antibody selected from the group consisting of ca, cb and cc; (ii) the second antibody is, or competes for binding to EGFR with, or binds to the same epitope as, an antibody selected from the group consisting of cd, ce and cf; and (iii) the third antibody is, or competes for binding to EGFR with, or binds to the same epitope as, an antibody selected from the group consisting of cg, ch, ci, cj and ck, wherein ca, cb, cd, ce, cf, cg, ch, ci, cj, and ck are each disclosed in PCT Int. Pub. No. WO/2011/140254 and corresponding pending U.S. patent application Ser. No. 13/100,920.
  • Other features and advantages of the invention will be apparent from the following detailed description, and from the claims.
  • BRIEF DESCRIPTION OF THE FIGURES
  • FIGS. 1A-1C: Phospho-EGF receptor and phospho-ERK signaling inhibition by single and pairwise combinations of Bin 1+Bin 2 or Bin 1+Bin 3 antibodies and comparisons with other known anti-EGFR antibodies such as cetuximab, nimotuzumab, and zalutumumab. FIG. 1A shows inhibition of ERK activation by the Bin1/2 antibodies cb and cd. FIG. 1B shows the inhibition of EGFR activation by the Bin1/2 antibodies and cd. FIG. 1C shows the inhibition of ERK activation by the Bin1/3 antibodies cb and ch. Lines depict a five parameter logistic fit to the data from each combination.
  • FIGS. 2A-2G: Inhibition of ligand-mediated tumor cell signaling in A431 cells preincubated with varying concentrations of anti-EGFR monoclonal antibodies cb (Bin1), cd (Bin2), cetuximab, zalutumumab, or nimotuzumab; as well as the oligoclonal combination of cb+cd; for 2 hrs. After incubation cells were stimulated with an EGFR ligand (8 nanomolar final concentration) for about 10 minutes. Figures show ELISA analysis of phospho-ERK (pERK) production (y-axis) as a function of antibody concentration (x-axis, in Log Molar concentration) after stimulation with the ligands amphiregulin (FIG. 2A), epigen (FIG. 2B), epiregulin (FIG. 2C), betacellulin (FIG. 2D), epidermal growth factor (EGF, FIG. 2E), heparin-binding EGF-like growth factor (HB-EGF, FIG. 2F), or transforming growth factor α (TGF-α, FIG. 2G). A431 cells incubated in the absence of anti-EGFR antibodies but with the ligand indicated in each graph (+Lig) or without ligand stimulation (−Lig) were used as positive and negative controls, respectively.
  • FIGS. 3A-3G: Inhibition of ligand-mediated tumor cell signaling in A431 cells preincubated with varying concentrations of anti-EGFR monoclonal antibodies cb (Bin1), ch (Bin3), cetuximab, zalutumumab, or nimotuzumab; as well as the oligoclonal combination of cb+ch; for 2 hrs. After pre-incubation with antibodies, cells were stimulated with an EGFR ligand (8 nanomolar final concentration) for 10 minutes. Figures show ELISA analysis of phospho-ERK (pERK) production (y-axis) as a function of antibody concentration (x-axis, in Log Molar concentration) after stimulation with the ligands amphiregulin (FIG. 3A), epigen (FIG. 3B), epiregulin (FIG. 3C), betacellulin (FIG. 3D), epidermal growth factor (EGF, FIG. 3E), heparin-binding EGF-like growth factor (HB-EGF, FIG. 3F), or transforming growth factor α (TGF-α, FIG. 3G). A431 cells incubated in the absence of anti-EGFR antibodies but with the ligand indicated in each graph (+Lig) or without ligand stimulation (−Lig) were used as positive and negative controls, respectively.
  • FIGS. 4A-4L: Inhibition of high affinity EGFR ligand-mediated tumor cell proliferation. H322M cells (FIGS. 4A-4D), H1975 cells (FIGS. 4E-4H), and LIM1215 cells (FIGS. 4I-4L) were treated with varying concentrations of anti-EGFR monoclonal and oligoclonal antibodies in the presence of EGFR ligands. Cells were treated with 200 ng/ml amphiregulin (AREG) (FIGS. 4A, 4E, and 4I), 50 ng/ml EGF (FIGS. 4B, 4F, and 4J), 50 ng/ml TGFα (FIGS. 4C, 4G, and 4K) or 90 ng/ml HB-EGF (FIGS. 4D, 4H, and 4L) in the presence of varying concentrations of MM-151 (open circles or cetuximab (CTX, solid squares; Bristol-Myers Squibb). Cells treated with ligand (+Lig, upward arrow) or without ligand (−Lig, downward arrow) in the absence of antibody treatment served as controls. The y-axes represent cell viability as the fraction of the viability of the amphiregulin-treated control cells and the x-axes represent antibody concentration in Log (Molar).
  • FIGS. 5A-5L: Effect of EGFR high affinity ligand titration on cell responsiveness to anti-EGFR inhibitors in vitro. The non-small cell lung cancer (NSCLC) lines H322M (FIGS. 5A-5D), HCC827 (FIGS. 5E-5H), and H1975 (FIGS. 5I-5L) were tested. Controls were growth in media with amphiregulin alone (+AREG, 200 ng/ml) or EGF alone as a control (+EGF, 20 ng/ml) or no added ligand (−Lig). Treatments were with varying concentrations (0.1-1 μM final concentration) of MM-151 or cetuximab (CTX) in the following conditions: amphiregulin alone (200 ng/ml, FIGS. 5A, 5E, and 5I); a 1000:1 amphiregulin:EGF ratio (0.2 ng/ml EGF, FIGS. 5B, 5F, and 5J); a 100:1 amphiregulin:EGF ratio (2 ng/ml EGF, FIGS. 5C, 5G, and 5K); and a 10:1 amphiregulin:EGF ratio (20 ng/ml EGF, FIGS. 5D, 5H, and 5L). The y-axes represent cell viability as a fraction of the viability of the AREG-treated control cells, whereas and the x-axes represent antibody concentration in Log (Molar).
  • FIG. 6: Effect of EGFR ligand concentration on phopho-ERK cell signaling. The epidermoid cancer cell line A431 was treated with media alone (“No Inhibitor”), MM-151 (100 nM) or cetuximab (100 nM) for 2 hrs, followed by the addition of various concentrations of EGF (0.16 ng/ml, 0.8 ng/ml, 4.0 ng/ml, 20 ng/ml, 100 ng/ml) or AREG (0.48 ng/ml, 2.4 ng/ml, 12 ng/ml, 60 ng/ml, 300 ng/ml), alone or in combination. Cells were incubated with the various EGF and AREG ligand combinations for 10 minutes, lysed, and levels of ERK phosphorylation measured by phospho-ERK ELISA.
  • DETAILED DESCRIPTION I. Definitions
  • The terms “EGFR,” and “EGF receptor” are used interchangeably herein to refer to human EGFR protein (also referred to as ErbB1 or HER1); see UniProtKB/Swiss-Prot entry P00533.
  • 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 except for possible naturally occurring mutations that may be present in minor amounts. Such antibodies may be obtained, e.g. from hybridomas or by recombinant expression. Antigen binding fragments (including scFvs) of such immunoglobulins are also encompassed by the term “monoclonal antibody” as used herein. Monoclonal antibodies are highly specific, generally being directed against a single epitope on a single antigen site, e.g., on the extracellular domain of EGFR. Monoclonal antibodies include chimeric antibodies - whose variable regions derive from a first animal species (e.g., mouse) and whose constant regions derive from a second animal species (e.g., human), human antibodies and humanized antibodies.
  • The terms “treat,” “treating,” and “treatment,” as used herein, refer to therapeutic or preventative measures described herein. The methods of “treatment” employ administration to a subject, an antibody or antibody pair or trio disclosed herein, for example, a subject having a disorder associated with EGFR dependent signaling or predisposed to having such a disease or disorder, in order to prevent, cure, delay, reduce the severity of, or ameliorate one or more symptoms of the disease or disorder or recurring disease or disorder, or in order to prolong the survival of a subject beyond that expected in the absence of such treatment.
  • Commercially available pharmaceutical anti-EGFR antibodies include cetuximab, panitumumab and nimotuzumab (which is not yet available in the US market). Other pharmaceutical anti-EGFR antibodies include zalutumumab, and matuzumab, which are in development. Still other anti-EGFR antibodies include those disclosed in the Oligoclonal Applications, e.g., the antibodies disclosed below.
  • P1X is a human IgG1 having a heavy chain variable region comprising SEQ ID NO: 1 and a light chain variable region comprising SEQ ID NO: 2;
  • P2X is a human IgG1 having a heavy chain variable region comprising SEQ ID NO: 3 and a light chain variable region comprising SEQ ID NO: 4; and
  • P3X is a human IgG1 having a heavy chain variable region comprising SEQ ID NO: 5 and a light chain variable region comprising SEQ ID NO: 6.
  • “MM-151” indicates a triple combination of P1X+P2X+P3X at a P1X:P2X:P3X molar ratio of 2:2:1.
  • TABLE 1
    Exemplary Antibodies
    P1X VH MGFGLSWLFLVAILKGVQC SEQ ID NO: 1
    QVQLVQSGAEVKKPGSSVKV
    SCKASGGTFSSYAISWVRQA
    PGQGLEWMGSIIPIFGTVNY
    AQKFQGRVTITADESTSTAY
    MELSSLRSEDTAVYYCARDP
    SVNLYWYFDLWGRGTLVTVSS
    P1X VL MGTPAQLLFLLLLWLPDTTG SEQ ID NO: 2
    DIQMTQSPSTLSASVGDRVT
    ITCRASQSISSWWAWYQQKP
    GKAPKLLIYDASSLESGVPS
    RFSGSGSGTEFTLTISSLQP
    DDFATYYCQQYHAHPTTFGG
    GTKVEIK
    P2X VH MGFGLSWLFLVAILKGVQC SEQ ID NO: 3
    QVQLVQSGAEVKKPGSSVKV
    SCKASGGTFGSYAISWVRQA
    PGQGLEWMGSIIPIFGAANP
    AQKSQGRVTITADESTSTAY
    MELSSLRSEDTAVYYCAKMG
    RGKVAFDIWGQGTMVTVSS
    P2X VL MGTPAQLLFLLLLWLPDTTG SEQ ID NO: 4
    DIVMTQSPDSLAVSLGERAT
    INCKSSQSVLYSPNNKNYLA
    WYQQKPGQPPKLLIYWASTR
    ESGVPDRFSGSGSGTDFTLT
    ISSLQAEDVAVYYCQQYYGS
    PITFGGGTKVEIK
    P3X VH MGFGLSWLFLVAILKGVQC SEQ ID NO: 5
    QVQLVQSGAEVKKPGASVKV
    SCKASGYAPTSYGINWVRQA
    PGQGLEWMGWISAYNGNTYY
    AQKLRGRVTMTTDTSTSTAY
    MELRSLRSDDTAVYYCARDL
    GGYGSGSVPFDPWGQGTLVTVSS
    P3X VL MGTPAQLLFLLLLWLPDTTG SEQ ID NO: 6
    EIVMTQSPATLSVSPGERAT
    LSCRASQSVSSNLAWYQQKP
    GQAPRLLIYGASTRATGIPA
    RFSGSGSGTEFTLTISSLQS
    EDFAVYYCQDYRTWPRRVFG
    GGTKVEIK
    zalutumumab  QVQLVESGGGVVQPGRSLRLS SEQ ID NO: 7
    VH CAASGFTFSTYGMHWVRQAPG
    KGLEWVAVIWDDGSYKYYGDS
    VKGRFTISRDNSKNTLYLQMN
    SLRAEDTAVYYCARDGITMVR
    GVMKDYFDYWGQGTLVTVSS
    zalutumumab  AIQLTQSPSSLSASVGDRVTI SEQ ID NO: 8
    VL TCRASQDISSALVWYQQKPGK
    APKLLIYDASSLESGVPSRFS
    GSESGTDFTLTISSLQPEDFA
    TYYCQQFNSYPLTFGGGTKVE
    IK
    Nimotuzumab  QVQLQQPGAELVKPGASVKLS SEQ ID NO: 9
    VH CKASGYTFTNYYIYWVKQRPG
    QGLEWIGAGINPTSGGSNFNE
    KFKTKATLTVDESSTTAYMQL
    SSLTSEDSAVYYCTRQGLWFD
    SDGRGFDFWGQGTTLTVSS
    Nimotuzumab  DVLMTQIPLSLPVSLGDQASISC SEQ ID NO: 10
    VL RSSQNIVHSNGNTYLDWYLQKP
    GQSPNLLIYKVSNRESGVPDRF
    RGSGSGTDFTLKISRVEAEDLGV
    YYCFQYSHVPWTFGGGTKLEIK
  • The term “patient” includes human and other mammalian subjects that receive either prophylactic or therapeutic treatment.
  • The term “sample” refers to tissue, body fluid, or a cell (or a fraction of any of the foregoing) taken from a patient. Normally, the tissue or cell will be removed from the patient, but in vivo diagnosis is also contemplated. In the case of a solid tumor, a tissue sample can be taken from a surgically removed tumor and prepared for testing by conventional techniques. In the case of lymphomas and leukemias, lymphocytes, leukemic cells, or lymph tissues can be obtained (e.g., leukemic cells from blood) and appropriately prepared. Other samples, including urine, tears, serum, plasma, cerebrospinal fluid, feces, sputum, cell extracts etc. can also be useful for particular cancers.
  • Various aspects of the disclosure are described in further detail in the following subsections.
  • II. Outcomes
  • A patient having a tumor predicted by the methods disclosed herein to have a favorable outcome following treatment with a monoclonal or oligoclonal anti-EGFR antibody, and who is then treated accordingly, may exhibit one of the following responses to therapy:
    • Pathologic complete response (pCR): absence of invasive cancer following primary systemic treatment.
    • Complete Response (CR): Disappearance of all target lesions.
    • Partial Response (PR): At least a 30% decrease in the sum of dimensions of target lesions, taking as reference the baseline sum diameters; or
    • Stable Disease (SD): Neither sufficient shrinkage to qualify for partial response, nor sufficient increase to qualify for progressive disease, taking as reference the smallest sum diameters while on study.
  • In exemplary outcomes, patients treated as disclosed herein may experience improvement in at least one sign of cancer.
  • In one embodiment the patient so treated exhibits pCR, CR, PR, or SD.
  • In another embodiment, the patient so treated experiences tumor shrinkage and/or decrease in growth rate, i.e., suppression of tumor growth. In another embodiment, unwanted cell proliferation is reduced or inhibited. In yet another embodiment, one or more of the following can occur: the number of cancer cells can be reduced; tumor size can be reduced; cancer cell infiltration into peripheral organs can be inhibited, retarded, slowed, or stopped; tumor metastasis can be slowed or inhibited; tumor growth can be inhibited; recurrence of tumor can be prevented or delayed; one or more of the symptoms associated with cancer can be relieved to some extent.
  • In other embodiments, such improvement is measured by a reduction in the quantity and/or size of measurable tumor lesions. Measurable lesions are defined as those that can be accurately measured in at least one dimension (longest diameter is to be recorded) as ≧10 mm by CT or MRI scan (e.g., CT scan slice thickness no greater than 5 mm), 10 mm caliper measurement by clinical exam or >20 mm by chest X-ray. The size of non-target lesions can also be measured for improvement. In one embodiment, lesions can be measured on x-rays or CT or MRI images.
  • In other embodiments, cytology or histology can be used to evaluate responsiveness to a therapy. The cytological confirmation of the neoplastic origin of any effusion that appears or worsens during treatment when the measurable tumor has met criteria for response or stable disease can be considered to differentiate between response or stable disease (an effusion may be a side effect of the treatment) and progressive disease.
  • In some embodiments, a beneficial response to therapy is indicated by at least one therapeutic effect selected from the group consisting of reduction in size of a tumor, reduction in number of metastatic lesions appearing over time, complete remission, partial remission, stable disease, increase in overall response rate, or a pathologic complete response.
  • III. Pharmaceutical Compositions
  • Pharmaceutical compositions for use in the methods provided for herein are commercially available anti-EGFR compositions, e.g., of cetuximab, panitumumab and nimotuzumab, as well as the various pharmaceutical compositions provided in the Oligoclonal Applications.
  • IV. Use of Oligoclonal Antibodies
  • Provided herein are methods of determining whether or not a monoclonal anti-EGFR antibody preparation comprising only a single species of anti-EGFR antibody should be used to treat a tumor. Use of oligoclonal anti-EGFR antibodies for the treatment of a disease associated with high-affinity EGFR ligand-driven signaling is also provided, as are methods of use of oligoclonal anti-EGFR antibodies for the treatment of tumor comprising protein or mRNA levels of at least two high-affinity EGFR ligands that are higher than levels in the tumor of at least two low-affinity EGFR ligands. Cancers treated in accordance with the methods provided include melanoma, breast cancer, ovarian cancer, renal carcinoma, gastrointestinal cancer, gastro-esophageal junction cancer, colon cancer, lung cancer, pancreatic cancer, skin cancer, head and neck cancer glioblastoma, prostate cancer and other solid and/or metastatic tumors.
  • The monoclonal or oligoclonal antibody can be administered alone or with another therapeutic agent that acts in conjunction with or synergistically with the oligoclonal antibody to treat the disease associated with EGFR-mediated signaling.
  • Also provided are kits for testing a tumor sample, e.g., a tumor biopsy sample or a circulating tumor cell, to determine levels of both high and low affinity EGFR ligands in the sample, said kits being comprised by one or more containers comprising;
      • a) at least two pairs of high affinity EGFR ligand-specific polymerase chain reaction (PCR) primers,
      • b) at least two pairs of low affinity EGFR ligand-specific PCR primers, and
      • c) at least one reverse transcription PCR (RT-PCR) reagent.
  • In another embodiment, the kit may further contain instructions for use in determining how to treat a tumor in a patient following determination of levels of high and low affinity ligands in a sample of the tumor. The kit may include an indication of the intended use of the contents of the kit (e.g., in the form of a label or other printed or recorded matter).
  • Other embodiments are described in the following non-limiting Examples.
  • The present invention is further illustrated by the following examples which should not be construed as further limiting. The contents of Sequence Listing, figures and all references, patents and published patent applications cited throughout this application are expressly incorporated herein by reference.
  • EXAMPLES Materials and Methods
  • Throughout the examples, the following materials and methods are used unless otherwise stated.
  • In general, unless otherwise indicated, conventional techniques of chemistry, molecular biology, recombinant DNA technology, immunology (especially, e.g., antibody technology), and standard techniques in polypeptide preparation are used. See, e.g., Sambrook, Fritsch and Maniatis, Molecular Cloning Cold Spring Harbor Laboratory Press (1989); Antibody Engineering Protocols (Methods in Molecular Biology), 510, Paul, S., Humana Pr (1996); Antibody Engineering: A Practical Approach (Practical Approach Series, 169), McCafferty, Ed., Irl Pr (1996); Antibodies: A Laboratory Manual, Harlow et al., C.S.H.L. Press, Pub. (1999); and Current Protocols in Molecular Biology, eds. Ausubel et al., John Wiley & Sons (1992).
  • Pulverization of Tumor Cells
  • A cryopulverizer (COVARIS Inc.) is used for the pulverization of tumors. Tumors are stored in special bags (pre-weighed before the addition of the tumor) and placed in liquid nitrogen while handling them. For small tumors, 200 μL of Lysis buffer is first added to the bag containing the tumor, frozen in liquid nitrogen and then pulverized to improve the recovery of the tumor from the bag. Pulverized tumors are transferred to 2 mL EPPENDORF tubes and placed in liquid nitrogen until ready for further processing.
  • Lysis of Tumor Cells
  • Tumors are lysed in Lysis buffer supplemented with protease and phosphatase inhibitors. Lysis Buffer is added to the tumor aliquots in a final concentration of about 62.5 mg/mL. Tumor samples are homogenized by vortexing for 30 sec and incubating on ice for about 30 min. The lysates are spun for about 10 min in Qiagen QIASHREDDER columns for further homogenization of the samples. Cleared lysates are aliquoted into fresh tubes for further processing.
  • Measurement of Inhibition of EGFR Ligand-Mediated Phosphorylation of ERK in Tumor Cells
  • Inhibition of ligand-mediated tumor cell signaling is investigated as follows: A431 (ATCC CRL-1555™) epidermoid carcinoma cells are seeded at a density of 35,000 cells/well or 17,500 cells per half well in 96 well tissue culture plates and grown in DMEM medium supplemented with antibiotics, 2 mM L-glutamine and 10% fetal bovine serum (FBS) for 24 hours at 37° C. and 5% carbon dioxide. Cells are serum starved in 1% FBS medium with antibiotics and 2 mM L-glutamine for about 20 hours at 37° C. and 5% carbon dioxide. Cells are then treated as described below in each Example. Cells are washed with ice-cold PBS and lysed in 50 μl ice-cold Lysis buffer (Mammalian Protein Extraction Lysis Reagent (M-PER, Pierce, Thermo Scientific product #78505) amended with 150 mM NaCl and protease inhibitor cocktail (Sigma, P714)) by incubating on ice for 30 minutes. Lysates are either analyzed immediately by ELISA for phospho-ERK (a downstream effector of EGFR) or frozen at '180° C. until use.
  • ELISA Assays
  • For the phospho-EGFR sandwich ELISA, 96-half well GREINER high binding plates (Cat. #675077; GREINER BIO-ONE, Monroe, N.C.) are coated with 50 μL of an EGFR antibody (4 μg/ml final concentration; EGFR Ab-11, Clone: 199.12, without BSA and azide, Fisher Scientific, cat #MS396P1ABX), and incubated overnight at room temperature. Next morning, plates are washed 3 times with 100 μl/well PBST (0.05% Tween-20) on a BIOTEK plate washer. Plates are subsequently blocked for about 1 hour at room temperature with 2% BSA in PBS. The plates are washed 3 times with 100 μl/well PBST (0.05%Tween-20) on the BIOTEK plate washer. Cell lysates (50 μl) or standards (pEGFR pY1068 ELISA kit, R&D Systems, cat #DYC3570) diluted in 50% Lysis buffer and 1%BSA-PBS (per the manufacturer's recommendations) are added to the plates in duplicates and incubated for 2 hrs at room temperature or overnight at 4° C. with shaking. Plates are then washed 3 times with 100 μl/well in the BIOTEK plate washer with PBST (PBS with 0.05%Tween-20). About 50 μl of a detection antibody (pEGFR pY1068 ELISA kit, R&D Systems, cat #DYC3570) conjugated to horseradish peroxidase (HRP) diluted (as per manufacturer's instructions) in 2% BSA, PBS is added and incubated for about 2 hour at room temperature. The plate is washed 3 times with 100 μl/well in the BIOTEK plate washer with PBST (0.05%Tween-20). About 50 μL of SUPERSIGNAL PICO ELISA substrate is added and the plate is read using an Envision (Perkin Elmer) plate reader. The data are analyzed and duplicate samples are averaged and error bars are used to represent the standard deviation between the two replicates.
  • The phospho-ERK ELISA is performed similarly to the phospho-EGFR ELISA with the following changes: Human pERK ELISA DUOSET kit is purchased from R&D Systems (cat #DYC1018-5) and used as recommended by the manufacturer. The data are analyzed by subtracting background signal, regressing to a recombinant standard supplied by the manufacturer, and back-calculating the data (BCD) to correct for dilution factors. Duplicate samples are averaged and error bars are used when indicated to represent the standard deviation between two replicates.
  • Example 1 Phospho-EGF Receptor and Phospho-ERK Signaling Inhibition by Single and Pairwise Combinations of Bin 1+Bin 2 or Bin 1+Bin 3 Antibodies and Comparisons with Each of Individual Monoclonal Antibodies Cetuximab, Nimotuzumab, and Zalutumumab
  • A431 cells were treated with single antibodies or antibody pairs and their ability to inhibit EGFR-dependent signaling was compared to that each of cetuximab, nimotuzumab, and zalutumumab. Cells were incubated with varying concentrations of anti-EGFR antibodies for 2 hrs, and then stimulated with an EGFR ligand for 10 minutes at 37° C. and 5% carbon dioxide. The seven recombinant human EGFR ligands used individually were 100 ng/ml amphiregulin (“AREG,” R&D Systems, cat #262-AR/CF), 100 ng/ml betacellulin (R&D Systems, cat #261-CE-050/CF), EGF (PreproTech, cat #AF-100-15), 220 ng/ml epigen (epithelial mitogen homolog, PreproTech, cat #100-51), 150 ng/ml epiregulin (R&D Systems, cat #1195-EP/CF), 90 ng/ml HB-EGF (heparin-binding EGF-like growth factor, PreproTech, cat #100-47), and 50 ng/ml TGFα (transforming growth factor alpha, R&D Systems, cat #239-A). ELISA measurements were performed as described above for pERK and pEGFR signaling and the results are shown in FIGS. 1A-C. Only mixtures of Bin1/Bin2 antibodies cb and cd (FIG. 1A) and Bin1/Bin3 antibodies cb and ch (FIG. 1C) were effective at completely inhibiting phospho-ERK signaling when compared to cetuximab, nimotuzumab, and zalutumumab, as well as to individual components cb, cd, and ch. All antibodies, including the mixtures, were effective at complete inhibition of Phospho-EGF receptor signaling, with the exception of nimotuzumab (FIG. 1B).
  • Example 2 Phospho-ERK Signaling Inhibition by Single and Pairwise Combinations of Bin 1, Bin 2, and Bin 3 Antibodies and Comparisons with Cetuximab, Nimotuzumab, and Zalutumumab
  • Single antibodies cb, cd, and ch, or pairs of cb and cd or cb and ch, (as described above in Example 1) were used to treat A431 cells at indicated total concentrations, and their ability to inhibit EGFR ligand-dependent signaling was compared to that of each single anti-EGFR antibodies cetuximab, nimotuzumab, and zalutumumab at the same concentrations. Cells were incubated with antibody for 2 hours followed by stimulation with EGFR ligand for 10 minutes. Seven EGFR ligands were used individually: amphiregulin (100 ng/ml), betacellulin (100 ng/ml), EGF (50 ng/ml), epigen (220 ng/ml), epiregulin (150 ng/ml), HB-EGF (90 ng/ml), and TGFα (50 ng/ml). Experiments were performed as described above and the results are shown in FIGS. 2A-G and 3A-G. Individually, cb and cd, as well as well cetuximab, nimotuzumab, and zalutumumab, were effective at inhibiting phospho-ERK signaling (i.e., inhibiting phosphorylation of ERK1 and ERK2) in response to the three ligands with low affinity for EGF receptor (amphiregulin, epigen, and epiregulin), but not in response to the four ligands with high affinity for EGF receptor (betacellulin, EGF, HB-EGF, and TGFα). Only oligoclonal mixtures of Bin1/Bin2 antibodies cb and cd (FIGS. 2A-G) and Bin1/Bin3 antibodies cb and ch (FIGS. 3A-G) were effective at essentially completely inhibiting phospho-ERK signaling in response to all seven (both high- and low-affinity) EGFR ligands when compared to individual components of the mixtures, cb, cd, and ch and the other tested individual monoclonal antibodies, cetuximab, nimotuzumab, and zalutumumab.
  • Example 3 Effect of EGFR Ligand Concentration on Phospho-ERK Cell Signaling
  • Inhibition of tumor cell signaling in vitro is analyzed by the methods described above or minor variations thereof. The epidermoid cancer cell line A431 was treated with media alone (“No Inhibitor”), MM-151 (100 nM) or cetuximab (100 nM) for 2 hrs, followed by the addition of various concentrations of EGF (0.16 ng/ml, 0.8 ng/ml, 4.0 ng/ml, 20 ng/ml, 100 ng/ml) or AREG (0.48 ng/ml, 2.4 ng/ml, 12 ng/ml, 60 ng/ml, 300 ng/ml), alone or in combination, as shown in FIG. 6. Cells were incubated with the various EGF and AREG ligand combinations for 10 minutes, lysed, and levels of ERK phosphorylation measured by phospho-ERK ELISA and analyzed as indicated in the methods. FIG. 6 shows MM-151 and cetuximab-mediated modulation of ERK signaling represented as fraction of the highest signal across all treatments. Phospho-ERK signaling is inhibited by cetuximab in A431 cells under low-affinity EGFR ligand (AREG) stimulation, but become increasingly resistant to inhibition upon the addition of increasing amounts of the high-affinity EGFR ligand, EGF (middle panel), while inhibition of signaling by MM-151 is largely maintained under all conditions (lower panel).
  • Example 4 Inhibition of Tumor Cell Proliferation in the Presence of High or Low Affinity EGFR Ligands Inhibition of Tumor Cell Proliferation In Vitro
  • Inhibition of cellular proliferation of cells expressing EGFR is examined in vitro as follows: H322M (NCI, Frederick, Md. 21701), H1975 (ATCC CRL-2868™), and LIM1215 (Cell Bank Australia, NSW 2145) cancer cells are separately seeded in 96 well tissue culture plates at 5,000 cells per well and grown in RPMI-1640 medium supplemented with antibiotics, 2 mM L-glutamine and 10% fetal calf serum (FCS) (H322M and H1975) or RPMI-1640 medium supplemented with 25 mM HEPES, antibiotics, 2 mM L-glutamine, 10% FCS, 0.6 μg/ml insulin, 1 μg/ml hydrocortisone and 10 μM thioglycerol (LIM1215) for 24 hours at 37 degrees Celsius and 5% carbon dioxide. Medium is then switched to RPMI-1640 with antibiotics, 2 mM L-glutamine, 1% FBS (for H322M and H1975) or RPMI-1640 with 25 mM HEPES, antibiotics, 2 mM L-glutamine, 1% FCS, 0.6 μg/ml Insulin, 1 μg/ml hydrocortisone and 10 μM thioglycerol (for LIM1215) supplemented with 200 ng/ml AREG, 50 ng/ml EGF, 50 ng/ml TGFα or 90 ng/ml HB-EGF in the presence of varying concentrations of MM-151 or cetuximab (Bristol-Myers Squibb). Cell viability is measured 72 hours post-treatment using the CellTiter-Glo® (CTG) Luminescent Viable Cell Number Assay (Promega Corporation) according to manufacturer's instructions. The CTG assay measures the number of viable cells in culture based upon quantitation of ATP present, which is an indicator of metabolically active cells. Control treatments include cells treated with 1% FCS-containing medium (as detailed above) in the presence (“+Lig”) or absence (“−Lig”) of the respective ligand treatment. Viable cell numbers are plotted in GraphPad Prism (GraphPad Software, La Jolla, Calif.) as a fraction of the respective ligand (“+Lig”) treatment control.
  • Results
  • The non-small cell lung cancer (NSCLC) lines H322M and H1975 and colon cancer cell line LIM1215 were treated with varying concentrations of MM-151or cetuximab (0.1-1 μM final concentration). Potent inhibition of growth of H322M, H1975 and LIM1215 cells was obtained over a range of MM-151 concentrations in the presence of high affinity EGFR ligands (EGF, TGFα, HB-EGF), but not in the presence of cetuximab or in assay medium alone (1% FCS)—FIG. 4 (B-D, F-H, and J-L). Potent inhibition of growth of H322M, H1975 and LIM1215 cells was obtained over a range of concentrations for both MM-151 and cetuximab, but not by assay medium alone (1% FCS) in the presence of the low affinity EGFR ligand amphiregulin (AREG)—FIG. 4 (A, E and I). These data demonstrate the ability of the MM-151 oligoclonal mixture to inhibit tumor cell proliferation in vitro in response to both high-affinity (EGF, TGFα, HB-EGF) and low-affinity (AREG) ligands, whereas cetuximab is only potently effective in cells treated with low-affinity (AREG) ligand.
  • Example 5 Effects of EGF Ligand Concentration on Cell Proliferation
  • Using methods essentially as described in the preceding Example, non-small cell lung cancer (NSCLC) cell lines H322M, HCC827 and H1975 were treated with AREG alone (200 ng/ml) or with AREG plus increasing amounts of EGF (0.2, 2, 20 ng/ml) in the presence of varying concentrations of MM-151or cetuximab (0.1-1 μM final concentration).
  • Results
  • The NSCLC cell lines respond to cetuximab under low-affinity EGFR ligand stimulation (AREG), but become increasingly unresponsive to treatment upon the addition of increasing amounts of the high-affinity EGFR ligand EGF, while sensitivity to MM-151 is largely maintained (see FIGS. 5A-5L).
  • Example 6 Assays and Kits Measurement of EGFR Family Ligand Expression Levels by RT-qPCR
  • Measurement of EGFR ligand expression in tumor biopsy samples by real-time quantitative polymerase chain reaction (RT-qPCR) of DNAs reverse transcribed from RNAs is carried out as follows:
  • Total RNA is isolated from patient biopsy/tumor samples, e.g., by commercially available standard methods. The method of total RNA isolation may be any method (including conventional methods) suitable for use with the type of patient biopsy sample being tested, e.g., fresh, fixed, frozen, formalin fixed paraffin embedded (FFPE), etc. Total RNA is then converted to cDNA using the gene specific primers described below and Qiagen® OneStep RT-PCR reagents and protocol (Cat. #210210, Qiagen, Germantown, Md.). The cDNA is then used for RT-qPCR using the following gene specific primers as TaqMan® probe sets obtained from Applied Biosystems (Carlsbad, Calif.) along with reagents and equipment from the same source, all as described below:
      • 1. TaqMan® Gene Expression Assay, Gene Name: betacellulin, Assay ID: Hs01101201_ml
      • 2. TaqMan® Gene Expression Assay, Gene Name: transforming growth factor, alpha, Assay ID: Hs00608187_ml
      • 3. TaqMan® Gene Expression Assay, Gene Name: heparin-binding EGF-like growth factor, Assay ID: Hs00181813_ml
      • 4. TaqMan® Gene Expression Assay, Gene Name: epiregulin, Assay ID: Hs00914313_ml
      • 5. TaqMan® Gene Expression Assay, Gene Name: amphiregulin, Assay ID: Hs00950669_ml
      • 6. TaqMan® Gene Expression Assay, Gene Name: epidermal growth factor, Assay ID: Hs01099999_ml.
      • 7. TaqMan® Gene Expression Assay, Gene Name: epithelial mitogen homolog (epigen), Assay ID Hs02385425_ml.
        5 μl of diluted cDNA is mixed with 10 μl of TaqMan® Fast Advanced Master Mix (Cat. #4444556), 2 μl of the above primer probe set and 3 μl of water in a MicroAmp® Fast Optical 96-Well Reaction Plate (Cat. #4366932). The plate is then placed in a Viia™ 7 RT-qPCR machine and a thermal cycling program completed as described in the manufacturers protocol. Data collection and analysis is carried out using the Viia™ 7-RUO-Software (Applied Biosystems).
  • Also see US Patent Publication Nos. 20030165952, 20040009489, 20050095634, 20050266420, 20070141587, 20070141588, 20070141589, 20080182255, 20090125247, 20090280490, 20100221754 and 20110086349, and U.S. Pat. Nos. 6,750,013, 6,808,888, 6,939,670, 6,964,850, 6,692,916, 7,081,340, 7,171,311, 7,526,387, 7,569,345, 7,622,251, 7,871,769, 7,838,224, 7,858,304, 7,930,104, and 8,071,286.
  • Equivalents
  • Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents of the specific embodiments described herein. Such equivalents are intended to be encompassed by the following claims. Any combination of the embodiments disclosed in the any plurality of the dependent claims is contemplated to be within the scope of the disclosure.

Claims (21)

What is claimed is:
1. A kit for testing a tumor sample to determine levels of both high and low affinity EGFR ligands in the sample, said kit comprising a container comprising; a) at least two pairs of high affinity EGFR ligand-specific polymerase chain reaction (PCR) primers, b) at least two pairs of low affinity EGFR ligand-specific PCR primers, and c) at least one reverse transcription PCR (RT-PCR) reagent.
2. The kit of claim 1, wherein the at least two pairs of high affinity EGFR ligand-specific polymerase chain reaction primers are specific to at least two of betacellulin, EGF, HB-EGF or TGF alpha and each of the at least two pairs of low affinity EGFR ligand-specific polymerase chain reaction primers are specific to at least two of amphiregulin, epigen, or epiregulin.
3. The kit of claim 2, wherein the at least two pairs of high affinity EGFR ligand-specific primers comprise specific primer pairs for betacellulin, EGF, HB-EGF and TGF alpha and the at least two pairs of high affinity EGFR ligand-specific primers comprise specific primer pairs for all of amphiregulin, epigen, and epiregulin.
4. The kit of claim 2, further comprising at least one fluorescent reporter molecule.
5. The kit of claim 1, wherein the at least one RT-PCR reagent is one or more of an RNA-dependent DNA polymerase, a DNA-dependent DNA polymerase, or a solution comprising at least a micromolar concentration of each of ATP, GTP, CTP and TTP.
6. The kit of claim 2, wherein the at least one RT-PCR reagent is one or more of an RNA-dependent DNA polymerase, a DNA-dependent DNA polymerase, or a solution comprising at least a micromolar concentration of each of ATP, GTP, CTP and TTP.
7. The kit of claim 5 wherein the primers, the reagents, or both, have been prepared under current Good Manufacturing Practice (cGMP) conditions.
8. The kit of claim 5, wherein container further comprises total RNA isolated from a tumor sample.
9. The kit of claim 8, wherein the tumor sample from which the RNA is isolated is a fresh, fixed, frozen, or formalin fixed paraffin embedded (FFPE) sample.
10. The kit of claim 9, wherein the tumor sample is a circulating tumor cell sample.
11. The kit of claim 9, wherein the tumor sample is a biopsy sample.
12. The kit of claim 9, wherein the tumor sample is from a tumor of the skin, central nervous system, head, neck, esophagus, stomach, colon, rectum, anus, liver, pancreas, bile duct, gallbladder, lung, breast, ovary, uterus, cervix, vagina, testis, germ cells, prostate, kidney, ureter, urinary bladder, adrenal, pituitary, thyroid, bone, muscle or connective tissue.
13. A kit for testing a tumor sample to determine levels of both high and low affinity EGFR ligands in the sample, said kit comprising one or more containers, one of said containers comprising; a) at least two pairs of high affinity EGFR ligand-specific polymerase chain reaction (PCR) primers, b) at least two pairs of low affinity EGFR ligand-specific PCR primers, c) at least one reverse transcription PCR (RT-PCR) reagent, and d) RNA isolated from a tumor sample.
14. The kit of claim 13, wherein the container comprising a), b), c), and d) is a 96 well plate.
15. The kit of claim 13, wherein the tumor sample from which the RNA is isolated is a fresh, fixed, frozen, or formalin fixed paraffin embedded (FFPE) sample.
16. The kit of claim 15, wherein the tumor sample is a circulating tumor cell sample.
17. The kit of claim 16, wherein the tumor sample is a biopsy sample.
18. The kit of claim 15, wherein the tumor sample was from a tumor of the skin, central nervous system, head, neck, esophagus, stomach, colon, rectum, anus, liver, pancreas, bile duct, gallbladder, lung, breast, ovary, uterus, cervix, vagina, testis, germ cells, prostate, kidney, ureter, urinary bladder, adrenal, pituitary, thyroid, bone, muscle or connective tissue.
19. The kit of claim 15, wherein the container comprising a), b), c), and d) is a 96 well plate
20. The kit of claim 18, wherein the container comprising a), b), c), and d) is a 96 well plate.
21. A monoclonal anti-EGFR antibody preparation comprising a single species of monoclonal antibody for treatment of a patient predicted by a method to have a favorable outcome from treatment with the monoclonal anti-EGFR antibody preparation by determining whether a patient having a tumor is predicted to have a favorable outcome, comprising reduction of growth of the tumor, as a result of treatment #1 with a monoclonal anti-EGFR antibody preparation comprising a single species of monoclonal antibody, and as a result of treatment #2 with an oligoclonal anti-EGFR antibody preparation comprising a plurality of species of monoclonal anti-EGFR antibodies, one against each of at least two extracellular epitopes of EGFR,
or,
whether the patient is predicted to have the favorable outcome as a result of treatment #2 but not as a result of treatment #1;
the method comprising:
obtaining a biopsy sample of the tumor and:
a) measuring levels of protein or mRNAs coding for at least two low affinity EGFR ligands selected from amphiregulin, epigen, or epiregulin in the biopsy sample,
b) measuring levels of protein or mRNAs coding for at least two high affinity EGFR ligands selected from betacellulin, EGF, HB-EGF or TGFα in the biopsy sample, and
c) comparing the average level of protein or mRNAs coding for each of the high affinity EGFR ligands measured in a) to the average level of protein or mRNAs coding for each of the low affinity EGFR ligands measured in b);
wherein,
if the average level of protein or mRNAs coding for low affinity EGFR ligands measured in a) is greater than the average level of protein or mRNAs coding for high affinity EGFR ligands measured in b), the patient is predicted to have the favorable outcome as a result of treatment #1 and the patient is also predicted to have the favorable outcome as a result of treatment #2, and
if the average level of protein or mRNAs coding for low affinity EGFR ligands measured in a) is less than or equal to the average level of protein or mRNAs coding for high affinity EGFR ligands measured in b), the patient is predicted to have an unfavorable outcome from treatment #1 and is predicted to have a favorable outcome from treatment #2.
US14/181,307 2011-06-03 2014-02-14 Monoclonal and oligoclonal anti-egfr antibodies for use in the treatment of tumors expressing predominantly high affinity egfr ligands or tumors expressing predominantly low affinity egfr ligands Abandoned US20140170668A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
US14/181,307 US20140170668A1 (en) 2011-06-03 2014-02-14 Monoclonal and oligoclonal anti-egfr antibodies for use in the treatment of tumors expressing predominantly high affinity egfr ligands or tumors expressing predominantly low affinity egfr ligands
US14/833,834 US20160083800A1 (en) 2011-06-03 2015-08-24 Monoclonal and oligoclonal anti-egfr antibodies for use in the treatment of tumors expressing predominantly high affinity egfr ligands or tumors expressing predominantly low affinity egfr ligands
US15/462,738 US20170356049A1 (en) 2011-06-03 2017-03-17 Monoclonal and oligoclonal anti-egfr antibodies for use in the treatment of tumors expressing predominantly high affinity egfr ligands or tumors expressing predominantly low affinity egfr ligands

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US201161493252P 2011-06-03 2011-06-03
US201161504633P 2011-07-05 2011-07-05
US201161558945P 2011-11-11 2011-11-11
US13/488,270 US8691231B2 (en) 2011-06-03 2012-06-04 Methods of treatment of tumors expressing predominantly high affinity EGFR ligands or tumors expressing predominantly low affinity EGFR ligands with monoclonal and oligoclonal anti-EGFR antibodies
US14/181,307 US20140170668A1 (en) 2011-06-03 2014-02-14 Monoclonal and oligoclonal anti-egfr antibodies for use in the treatment of tumors expressing predominantly high affinity egfr ligands or tumors expressing predominantly low affinity egfr ligands

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US13/488,270 Division US8691231B2 (en) 2011-06-03 2012-06-04 Methods of treatment of tumors expressing predominantly high affinity EGFR ligands or tumors expressing predominantly low affinity EGFR ligands with monoclonal and oligoclonal anti-EGFR antibodies

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US14/833,834 Continuation US20160083800A1 (en) 2011-06-03 2015-08-24 Monoclonal and oligoclonal anti-egfr antibodies for use in the treatment of tumors expressing predominantly high affinity egfr ligands or tumors expressing predominantly low affinity egfr ligands

Publications (1)

Publication Number Publication Date
US20140170668A1 true US20140170668A1 (en) 2014-06-19

Family

ID=46682898

Family Applications (9)

Application Number Title Priority Date Filing Date
US13/488,270 Expired - Fee Related US8691231B2 (en) 2011-06-03 2012-06-04 Methods of treatment of tumors expressing predominantly high affinity EGFR ligands or tumors expressing predominantly low affinity EGFR ligands with monoclonal and oligoclonal anti-EGFR antibodies
US14/147,331 Expired - Fee Related US9226964B2 (en) 2011-07-05 2014-01-03 Antibodies against epidermal growth factor receptor (EGFR) and uses thereof
US14/181,307 Abandoned US20140170668A1 (en) 2011-06-03 2014-02-14 Monoclonal and oligoclonal anti-egfr antibodies for use in the treatment of tumors expressing predominantly high affinity egfr ligands or tumors expressing predominantly low affinity egfr ligands
US14/266,387 Abandoned US20140234314A1 (en) 2011-07-05 2014-04-30 Antibodies against epidermal growth factor receptor (egfr) and uses thereof
US14/833,834 Abandoned US20160083800A1 (en) 2011-06-03 2015-08-24 Monoclonal and oligoclonal anti-egfr antibodies for use in the treatment of tumors expressing predominantly high affinity egfr ligands or tumors expressing predominantly low affinity egfr ligands
US14/847,297 Expired - Fee Related US10072299B2 (en) 2011-07-05 2015-09-08 Antibodies against epidermal growth factor receptor (EGFR) and uses thereof
US14/847,304 Expired - Fee Related US9885087B2 (en) 2011-07-05 2015-09-08 Antibodies against epidermal growth factor receptor (EGFR) and uses thereof
US14/847,291 Expired - Fee Related US9902999B2 (en) 2011-07-05 2015-09-08 Antibodies against epidermal growth factor receptor (EGFR) and uses thereof
US15/462,738 Abandoned US20170356049A1 (en) 2011-06-03 2017-03-17 Monoclonal and oligoclonal anti-egfr antibodies for use in the treatment of tumors expressing predominantly high affinity egfr ligands or tumors expressing predominantly low affinity egfr ligands

Family Applications Before (2)

Application Number Title Priority Date Filing Date
US13/488,270 Expired - Fee Related US8691231B2 (en) 2011-06-03 2012-06-04 Methods of treatment of tumors expressing predominantly high affinity EGFR ligands or tumors expressing predominantly low affinity EGFR ligands with monoclonal and oligoclonal anti-EGFR antibodies
US14/147,331 Expired - Fee Related US9226964B2 (en) 2011-07-05 2014-01-03 Antibodies against epidermal growth factor receptor (EGFR) and uses thereof

Family Applications After (6)

Application Number Title Priority Date Filing Date
US14/266,387 Abandoned US20140234314A1 (en) 2011-07-05 2014-04-30 Antibodies against epidermal growth factor receptor (egfr) and uses thereof
US14/833,834 Abandoned US20160083800A1 (en) 2011-06-03 2015-08-24 Monoclonal and oligoclonal anti-egfr antibodies for use in the treatment of tumors expressing predominantly high affinity egfr ligands or tumors expressing predominantly low affinity egfr ligands
US14/847,297 Expired - Fee Related US10072299B2 (en) 2011-07-05 2015-09-08 Antibodies against epidermal growth factor receptor (EGFR) and uses thereof
US14/847,304 Expired - Fee Related US9885087B2 (en) 2011-07-05 2015-09-08 Antibodies against epidermal growth factor receptor (EGFR) and uses thereof
US14/847,291 Expired - Fee Related US9902999B2 (en) 2011-07-05 2015-09-08 Antibodies against epidermal growth factor receptor (EGFR) and uses thereof
US15/462,738 Abandoned US20170356049A1 (en) 2011-06-03 2017-03-17 Monoclonal and oligoclonal anti-egfr antibodies for use in the treatment of tumors expressing predominantly high affinity egfr ligands or tumors expressing predominantly low affinity egfr ligands

Country Status (13)

Country Link
US (9) US8691231B2 (en)
EP (2) EP3090759A1 (en)
JP (2) JP6249945B2 (en)
KR (1) KR20140048230A (en)
CN (1) CN103781494B (en)
AU (1) AU2012279132B2 (en)
BR (1) BR112014000158A8 (en)
CA (1) CA2840088A1 (en)
HK (1) HK1197071A1 (en)
IL (1) IL229951B (en)
MX (1) MX350861B (en)
TW (1) TWI583700B (en)
WO (1) WO2013006547A2 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9044460B2 (en) 2010-05-04 2015-06-02 Merrimack Pharmaceuticals, Inc. Antibodies against epidermal growth factor receptor (EGFR) and uses thereof
US9226964B2 (en) 2011-07-05 2016-01-05 Merrimack Pharmaceuticals, Inc. Antibodies against epidermal growth factor receptor (EGFR) and uses thereof
US9657108B2 (en) 2014-05-14 2017-05-23 Merrimack Pharmaceuticals, Inc. Dosage and administration of anti-EGFR therapeutics

Families Citing this family (32)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9944707B2 (en) 2012-05-17 2018-04-17 Sorrento Therapeutics, Inc. Antibodies that bind epidermal growth factor receptor (EGFR)
US9717724B2 (en) 2012-06-13 2017-08-01 Ipsen Biopharm Ltd. Methods for treating pancreatic cancer using combination therapies
AU2013202947B2 (en) 2012-06-13 2016-06-02 Ipsen Biopharm Ltd. Methods for treating pancreatic cancer using combination therapies comprising liposomal irinotecan
EP2879712B1 (en) 2012-07-31 2018-04-25 Crown Bioscience, Inc. (Taicang) Histological markers for identifying non-small cell lung carcinoma patients for treatment with an anti-egfr drug
KR101854003B1 (en) 2013-07-02 2018-06-14 경희대학교 산학협력단 Video including multi layers encoding and decoding method
EP2955522A1 (en) * 2014-06-13 2015-12-16 Fundació Privada Institut d'Investigació Oncològica de Vall Hebron A method for monitoring the treatment of patients with tumors expressing EGFR
JP2018516870A (en) * 2015-04-24 2018-06-28 メリマック ファーマシューティカルズ インコーポレーティッド Method for treating a patient having a mutation in the extracellular domain of epidermal growth factor receptor (EGFR) using a combination of three fully human monoclonal anti-EGFR antibodies
US11318131B2 (en) 2015-05-18 2022-05-03 Ipsen Biopharm Ltd. Nanoliposomal irinotecan for use in treating small cell lung cancer
SI3337467T1 (en) 2015-08-20 2021-03-31 Ipsen Biopharm Ltd. Ash Road Combination therapy using liposomal irinotecan and a parp inhibitor for cancer treatment
EP3791876A1 (en) 2015-08-21 2021-03-17 Ipsen Biopharm Ltd. Methods for treating metastatic pancreatic cancer using combination therapies comprising liposomal irinotecan and oxaliplatin
WO2017035482A1 (en) * 2015-08-27 2017-03-02 Merrimack Pharmaceuticals, Inc Combination therapies for treatment of heregulin positive cancers
CN108366965B (en) 2015-10-16 2021-10-01 易普森生物制药有限公司 Stable camptothecin pharmaceutical compositions
WO2017070456A1 (en) * 2015-10-22 2017-04-27 Merrimack Pharmaceuticals, Inc. Methods of treating cancer by administering a bispecific antibody antagonist of igf-ir and erbb3 and a combination of anti-egfr antibodies
WO2017070475A1 (en) * 2015-10-22 2017-04-27 Merrimack Pharmaceuticals, Inc. Methods of treating cancer by administering a mek inhibitor and a combination of anti-egfr antibodies
WO2017070356A1 (en) * 2015-10-23 2017-04-27 Merrimack Pharmaceuticals, Inc. DOSAGE AND ADMINISTRATION OF ANTI-c-MET, ANTI-EpCAM BISPECIFIC ANTIBODIES, USES THEREOF AND MENTHODS OF TREATMENT THEREWITH
US20180271998A1 (en) 2015-12-04 2018-09-27 Merrimack Pharmaceuticals, Inc. Disulfide-stabilized fabs
CN108883176A (en) * 2016-03-28 2018-11-23 东丽株式会社 The treatment of cancer and/or prophylactic compositions
US20170336392A1 (en) * 2016-05-02 2017-11-23 Emmanuel A. Theodorakis Imaging methods to assess the efficacy of anticancer drugs in vitro cancer using spontaneously- forming spheroids
WO2018006785A1 (en) * 2016-07-05 2018-01-11 江苏恒瑞医药股份有限公司 Egfr antibody-drug conjugate and pharmaceutical use thereof
MX2019004783A (en) 2016-11-02 2019-08-12 Ipsen Biopharm Ltd Treating gastric cancer using combination therapies comprising liposomal irinotecan, oxaliplatin, 5-fluoruracil (and leucovorin).
US11124575B2 (en) * 2017-02-22 2021-09-21 University Of Saskatchewan EGFR-binding agents and uses thereof
WO2018169953A1 (en) * 2017-03-17 2018-09-20 R.P. Scherer Technologies, Llc Antibody conjugates and methods of making and using the same
EP3645563A4 (en) * 2017-06-27 2021-03-17 Neuracle Science Co., Ltd Anti-fam19a5 antibodies and uses thereof
CN109402063B (en) * 2017-08-18 2020-11-24 河北省科学院生物研究所 Porcine hemoglobin-resistant hybridoma cell strain, monoclonal antibody and application thereof
US20200347140A1 (en) 2017-08-30 2020-11-05 Symphogen A/S Compositions and methods for treating cancer with anti-egfr antibodies
CU24558B1 (en) * 2017-11-28 2021-12-08 Ct Inmunologia Molecular MONOCLONAL ANTIBODIES RECOGNIZING THE EPIDERMAL GROWTH FACTOR RECEPTOR AND ITS DERIVED FRAGMENTS
AU2019235900A1 (en) * 2018-03-14 2020-08-13 Dana-Farber Cancer Institute, Inc. Engineered cells, T cell immune modulating antibodies and methods for using the same
CA3097999A1 (en) 2018-04-24 2019-10-31 Neuracle Science Co., Ltd. Use of anti-family with sequence similarity 19, member a5 antibodies for the treatment of neuropathic pain
TW202039581A (en) 2018-12-28 2020-11-01 日商協和麒麟股份有限公司 Bispecific antibody binding to TfR
CN110812479A (en) * 2019-11-18 2020-02-21 青海晨菲制药有限公司 Gallic acid and EGFR target antibody composition and application thereof in lung cancer
US20230027739A1 (en) 2021-05-10 2023-01-26 Entrada Therapeutics, Inc. Antigen-binding and antigen degradation constructs
WO2024046396A1 (en) * 2022-08-31 2024-03-07 洪明奇 Anti-meegfr antibody, antigen-binding fragment thereof, and use thereof

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090298701A1 (en) * 2008-05-14 2009-12-03 Baker Joffre B Predictors of patient response to treatment with egf receptor inhibitors
WO2010019952A2 (en) * 2008-08-15 2010-02-18 Merrimack Pharmaceuticals, Inc. Methods, systems and products for predicting response of tumor cells to a therapeutic agent and treating a patient according to the predicted response
US8008003B2 (en) * 2002-11-15 2011-08-30 Genomic Health, Inc. Gene expression profiling of EGFR positive cancer
WO2011132182A1 (en) * 2010-04-18 2011-10-27 Yeda Research And Development Co. Ltd. MOLECULES AND METHODS OF USING SAME FOR TREATING ErbB/ErbB LIGANDS ASSOCIATED DISEASES

Family Cites Families (87)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4475196A (en) 1981-03-06 1984-10-02 Zor Clair G Instrument for locating faults in aircraft passenger reading light and attendant call control system
US4447233A (en) 1981-04-10 1984-05-08 Parker-Hannifin Corporation Medication infusion pump
US4439196A (en) 1982-03-18 1984-03-27 Merck & Co., Inc. Osmotic drug delivery system
US4522811A (en) 1982-07-08 1985-06-11 Syntex (U.S.A.) Inc. Serial injection of muramyldipeptides and liposomes enhances the anti-infective activity of muramyldipeptides
US4447224A (en) 1982-09-20 1984-05-08 Infusaid Corporation Variable flow implantable infusion apparatus
US4487603A (en) 1982-11-26 1984-12-11 Cordis Corporation Implantable microinfusion pump system
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US4486194A (en) 1983-06-08 1984-12-04 James Ferrara Therapeutic device for administering medicaments through the skin
US4596556A (en) 1985-03-25 1986-06-24 Bioject, Inc. Hypodermic injection apparatus
US5374548A (en) 1986-05-02 1994-12-20 Genentech, Inc. Methods and compositions for the attachment of proteins to liposomes using a glycophospholipid anchor
MX9203291A (en) 1985-06-26 1992-08-01 Liposome Co Inc LIPOSOMAS COUPLING METHOD.
US4881175A (en) 1986-09-02 1989-11-14 Genex Corporation Computer based system and method for determining and displaying possible chemical structures for converting double- or multiple-chain polypeptides to single-chain polypeptides
US4946778A (en) 1987-09-21 1990-08-07 Genex Corporation Single polypeptide chain binding molecules
US5260203A (en) 1986-09-02 1993-11-09 Enzon, Inc. Single polypeptide chain binding molecules
US5013653A (en) 1987-03-20 1991-05-07 Creative Biomolecules, Inc. Product and process for introduction of a hinge region into a fusion protein to facilitate cleavage
US5132405A (en) 1987-05-21 1992-07-21 Creative Biomolecules, Inc. Biosynthetic antibody binding sites
US5258498A (en) 1987-05-21 1993-11-02 Creative Biomolecules, Inc. Polypeptide linkers for production of biosynthetic proteins
US5091513A (en) 1987-05-21 1992-02-25 Creative Biomolecules, Inc. Biosynthetic antibody binding sites
DE3856559T2 (en) 1987-05-21 2004-04-29 Micromet Ag Multifunctional proteins with predetermined objectives
US4790824A (en) 1987-06-19 1988-12-13 Bioject, Inc. Non-invasive hypodermic injection device
US4941880A (en) 1987-06-19 1990-07-17 Bioject, Inc. Pre-filled ampule and non-invasive hypodermic injection device assembly
US5223409A (en) 1988-09-02 1993-06-29 Protein Engineering Corp. Directed evolution of novel binding proteins
GB8823869D0 (en) 1988-10-12 1988-11-16 Medical Res Council Production of antibodies
US5312335A (en) 1989-11-09 1994-05-17 Bioject Inc. Needleless hypodermic injection device
US5064413A (en) 1989-11-09 1991-11-12 Bioject, Inc. Needleless hypodermic injection device
US6075181A (en) 1990-01-12 2000-06-13 Abgenix, Inc. Human antibodies derived from immunized xenomice
EP1690935A3 (en) 1990-01-12 2008-07-30 Abgenix, Inc. Generation of xenogeneic antibodies
US6673986B1 (en) 1990-01-12 2004-01-06 Abgenix, Inc. Generation of xenogeneic antibodies
US6150584A (en) 1990-01-12 2000-11-21 Abgenix, Inc. Human antibodies derived from immunized xenomice
US5427908A (en) 1990-05-01 1995-06-27 Affymax Technologies N.V. Recombinant library screening methods
US6172197B1 (en) 1991-07-10 2001-01-09 Medical Research Council Methods for producing members of specific binding pairs
GB9015198D0 (en) 1990-07-10 1990-08-29 Brien Caroline J O Binding substance
US5877397A (en) 1990-08-29 1999-03-02 Genpharm International Inc. Transgenic non-human animals capable of producing heterologous antibodies of various isotypes
US6300129B1 (en) 1990-08-29 2001-10-09 Genpharm International Transgenic non-human animals for producing heterologous antibodies
ES2246502T3 (en) 1990-08-29 2006-02-16 Genpharm International, Inc. TRANSGENIC NON-HUMAN ANIMALS ABLE TO PRODUCE HETEROLOGICAL ANTIBODIES.
US6255458B1 (en) 1990-08-29 2001-07-03 Genpharm International High affinity human antibodies and human antibodies against digoxin
US5770429A (en) 1990-08-29 1998-06-23 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
US5789650A (en) 1990-08-29 1998-08-04 Genpharm International, Inc. Transgenic non-human animals for producing heterologous antibodies
US5661016A (en) 1990-08-29 1997-08-26 Genpharm International Inc. Transgenic non-human animals capable of producing heterologous antibodies of various isotypes
US5545806A (en) 1990-08-29 1996-08-13 Genpharm International, Inc. Ransgenic non-human animals for producing heterologous antibodies
US5625126A (en) 1990-08-29 1997-04-29 Genpharm International, Inc. Transgenic non-human animals for producing heterologous antibodies
US5874299A (en) 1990-08-29 1999-02-23 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
US5814318A (en) 1990-08-29 1998-09-29 Genpharm International Inc. Transgenic non-human animals for producing heterologous antibodies
US5633425A (en) 1990-08-29 1997-05-27 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
ATE275198T1 (en) 1991-12-02 2004-09-15 Medical Res Council PRODUCTION OF ANTIBODIES ON PHAGE SURFACES BASED ON ANTIBODIES SEGMENT LIBRARIES.
JPH07503132A (en) 1991-12-17 1995-04-06 ジェンファーム インターナショナル,インコーポレイティド Transgenic non-human animals capable of producing xenoantibodies
US5383851A (en) 1992-07-24 1995-01-24 Bioject Inc. Needleless hypodermic injection device
ES2091684T3 (en) 1992-11-13 1996-11-01 Idec Pharma Corp THERAPEUTIC APPLICATION OF CHEMICAL AND RADIO-MARKED ANTIBODIES AGAINST THE RESTRICTED DIFFERENTIATION ANTIGEN OF HUMAN B-LYMPHOCYTES FOR THE TREATMENT OF B-CELL LYMPHOMA.
AU6819494A (en) 1993-04-26 1994-11-21 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
US6214388B1 (en) 1994-11-09 2001-04-10 The Regents Of The University Of California Immunoliposomes that optimize internalization into target cells
AU6113396A (en) 1995-06-14 1997-01-15 Regents Of The University Of California, The Novel high affinity human antibodies to tumor antigens
US6056973A (en) 1996-10-11 2000-05-02 Sequus Pharmaceuticals, Inc. Therapeutic liposome composition and method of preparation
TW520297B (en) 1996-10-11 2003-02-11 Sequus Pharm Inc Fusogenic liposome composition and method
US6224903B1 (en) 1996-10-11 2001-05-01 Sequus Pharmaceuticals, Inc. Polymer-lipid conjugate for fusion of target membranes
US6210707B1 (en) 1996-11-12 2001-04-03 The Regents Of The University Of California Methods of forming protein-linked lipidic microparticles, and compositions thereof
IT1303776B1 (en) 1998-11-19 2001-02-23 S I S S A Scuola Internaz Supe PROCESS FOR THE PREPARATION OF GENOTECHE, OF POLYPEPTIDES USING THESE GENOTECHE AND THE OCTOPUS OBTAINED.
EP1212422B1 (en) 1999-08-24 2007-02-21 Medarex, Inc. Human ctla-4 antibodies and their uses
LT2857516T (en) 2000-04-11 2017-09-11 Genentech, Inc. Multivalent antibodies and uses therefor
IL155977A0 (en) 2000-11-30 2003-12-23 Medarex Inc Transgenic transchromosomal rodents for making human antibodies
KR100983997B1 (en) 2001-01-09 2010-09-28 메르크 파텐트 게엠베하 Combination therapy using receptor tyrosine kinase inhibitors and angiogenesis inhibitors
WO2004000102A2 (en) * 2002-06-19 2003-12-31 Abgenix, Inc. Method for predicting response to epidermal growth factor receptor-directed therapy
KR20050057631A (en) 2002-10-10 2005-06-16 메르크 파텐트 게엠베하 Bispecific anti-erb-b antibodies and their use in tumor therapy
AU2004232928A1 (en) 2003-04-22 2004-11-04 Ibc Pharmaceuticals Polyvalent protein complex
DK1687338T3 (en) 2003-11-07 2011-02-07 Ablynx Nv Camelidae single-domain antibodies VHH targeting epidermal growth factor receptor and uses thereof
EP3173073A1 (en) 2004-05-03 2017-05-31 Merrimack Pharmaceuticals, Inc. Liposomes for drug delivery
EP1778270A4 (en) 2004-05-05 2009-09-02 Merrimack Pharmaceuticals Inc Bispecific binding agents for modulating biological activity
AU2005327973A1 (en) 2005-02-23 2006-08-31 Merrimack Pharmaceuticals, Inc. Bispecific binding agents for modulating biological activity
US7498142B2 (en) 2006-01-31 2009-03-03 Yeda Research And Development Co., Ltd. Methods of identifying combinations of antibodies with an improved anti-tumor activity and compositions and methods using the antibodies
EP2089543B1 (en) 2006-11-15 2020-07-01 BioFire Diagnostics, LLC High density self-contained biological analysis
TW200846362A (en) 2007-02-09 2008-12-01 Symphogen As A polyclonal antibody product
PL2132229T3 (en) 2007-03-01 2016-12-30 Recombinant anti-epidermal growth factor receptor antibody compositions
EP2190878A1 (en) 2007-09-06 2010-06-02 Genmab A/S Novel methods and antibodies for treating cancer
EP3753947A1 (en) 2007-09-14 2020-12-23 Adimab, LLC Rationally designed, synthetic antibody libraries and uses therefor
US8159931B2 (en) 2007-12-11 2012-04-17 Broadcom Corporation Orthogonal pilot code construction
JO2913B1 (en) 2008-02-20 2015-09-15 امجين إنك, Antibodies directed to angiopoietin-1 and angiopoietin-2 and uses thereof
DE102010014680A1 (en) 2009-11-18 2011-08-18 Evonik Degussa GmbH, 45128 Cells, nucleic acids, enzymes and their use, as well as methods for producing sophorolipids
JP2013533211A (en) 2010-05-04 2013-08-22 メリマック ファーマシューティカルズ インコーポレーティッド Antibodies against epidermal growth factor receptor (EGFR) and uses thereof
WO2011140151A1 (en) 2010-05-04 2011-11-10 Dyax Corp. Antibodies against epidermal growth factor receptor (egfr)
US20150231238A1 (en) 2011-03-15 2015-08-20 Merrimack Pharmaceuticals, Inc. Overcoming resistance to erbb pathway inhibitors
US8691231B2 (en) 2011-06-03 2014-04-08 Merrimack Pharmaceuticals, Inc. Methods of treatment of tumors expressing predominantly high affinity EGFR ligands or tumors expressing predominantly low affinity EGFR ligands with monoclonal and oligoclonal anti-EGFR antibodies
US20170314079A1 (en) 2011-07-05 2017-11-02 Merrimack Pharmaceuticals, Inc. Antibodies against epidermal growth factor receptor (egfr) and uses thereof
EP2554551A1 (en) 2011-08-03 2013-02-06 Fundacio Institut mar d'Investigacions Médiques (IMIM) Mutations in the epidermal growth factor receptor gene
US9273143B2 (en) 2011-09-30 2016-03-01 Regeneron Pharmaceuticals, Inc. Methods and compositions comprising a combination of an anti-ErbB3 antibody and an anti-EGFR antibody
AU2013201584A1 (en) 2012-03-12 2013-09-26 Merrimack Pharmaceuticals, Inc. Methods for treating pancreatic cancer using combination therapies comprising an anti-ErbB3 antibody
AU2013202947B2 (en) 2012-06-13 2016-06-02 Ipsen Biopharm Ltd. Methods for treating pancreatic cancer using combination therapies comprising liposomal irinotecan
MA39599A (en) 2014-05-14 2016-10-05 Merrimack Pharmaceuticals Inc Dosage and administration anti-egfr therapeutics
JP2018516870A (en) 2015-04-24 2018-06-28 メリマック ファーマシューティカルズ インコーポレーティッド Method for treating a patient having a mutation in the extracellular domain of epidermal growth factor receptor (EGFR) using a combination of three fully human monoclonal anti-EGFR antibodies

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8008003B2 (en) * 2002-11-15 2011-08-30 Genomic Health, Inc. Gene expression profiling of EGFR positive cancer
US20090298701A1 (en) * 2008-05-14 2009-12-03 Baker Joffre B Predictors of patient response to treatment with egf receptor inhibitors
WO2010019952A2 (en) * 2008-08-15 2010-02-18 Merrimack Pharmaceuticals, Inc. Methods, systems and products for predicting response of tumor cells to a therapeutic agent and treating a patient according to the predicted response
WO2011132182A1 (en) * 2010-04-18 2011-10-27 Yeda Research And Development Co. Ltd. MOLECULES AND METHODS OF USING SAME FOR TREATING ErbB/ErbB LIGANDS ASSOCIATED DISEASES

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
Qiagen product guide 2004. *
RT2 ProfilerTM PCR Array system manual-from SA Biociences, Version 5.01, 9/16/2010. *

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9044460B2 (en) 2010-05-04 2015-06-02 Merrimack Pharmaceuticals, Inc. Antibodies against epidermal growth factor receptor (EGFR) and uses thereof
US9226964B2 (en) 2011-07-05 2016-01-05 Merrimack Pharmaceuticals, Inc. Antibodies against epidermal growth factor receptor (EGFR) and uses thereof
US9885087B2 (en) 2011-07-05 2018-02-06 Merrimack Pharmaceuticals, Inc. Antibodies against epidermal growth factor receptor (EGFR) and uses thereof
US9902999B2 (en) 2011-07-05 2018-02-27 Merrimack Pharmaceuticals, Inc. Antibodies against epidermal growth factor receptor (EGFR) and uses thereof
US10072299B2 (en) 2011-07-05 2018-09-11 Merrimack Pharmaceuticals, Inc. Antibodies against epidermal growth factor receptor (EGFR) and uses thereof
US9657108B2 (en) 2014-05-14 2017-05-23 Merrimack Pharmaceuticals, Inc. Dosage and administration of anti-EGFR therapeutics
US9920131B2 (en) 2014-05-14 2018-03-20 Merrimack Pharmaceuticals, Inc. Dosage and administration of anti-EGFR therapeutics

Also Published As

Publication number Publication date
US9902999B2 (en) 2018-02-27
CN103781494B (en) 2016-05-18
US9226964B2 (en) 2016-01-05
CN103781494A (en) 2014-05-07
MX2014000270A (en) 2014-07-09
WO2013006547A2 (en) 2013-01-10
US9885087B2 (en) 2018-02-06
JP2014522850A (en) 2014-09-08
WO2013006547A9 (en) 2013-02-21
US20120308576A1 (en) 2012-12-06
BR112014000158A8 (en) 2018-03-06
US20150376284A1 (en) 2015-12-31
TW201313739A (en) 2013-04-01
JP6249945B2 (en) 2017-12-20
AU2012279132B2 (en) 2014-12-18
HK1197071A1 (en) 2015-01-02
EP3090759A1 (en) 2016-11-09
US20150368347A1 (en) 2015-12-24
CA2840088A1 (en) 2013-01-10
WO2013006547A3 (en) 2013-04-18
MX350861B (en) 2017-09-25
KR20140048230A (en) 2014-04-23
US20140127207A1 (en) 2014-05-08
EP2729172A2 (en) 2014-05-14
IL229951B (en) 2018-10-31
BR112014000158A2 (en) 2017-02-07
US20160083800A1 (en) 2016-03-24
US10072299B2 (en) 2018-09-11
US20150368346A1 (en) 2015-12-24
US20140234314A1 (en) 2014-08-21
AU2012279132A1 (en) 2013-03-14
JP2018070622A (en) 2018-05-10
TWI583700B (en) 2017-05-21
EP2729172B1 (en) 2018-09-05
US20170356049A1 (en) 2017-12-14
US8691231B2 (en) 2014-04-08

Similar Documents

Publication Publication Date Title
US8691231B2 (en) Methods of treatment of tumors expressing predominantly high affinity EGFR ligands or tumors expressing predominantly low affinity EGFR ligands with monoclonal and oligoclonal anti-EGFR antibodies
RU2710735C2 (en) Compositions and methods of treating and diagnosing cancer-resistant cancer
Villella et al. HER‐2/neu overexpression in uterine papillary serous cancers and its possible therapeutic implications
US20110217309A1 (en) Biological markers predictive of anti-cancer response to insulin-like growth factor-1 receptor kinase inhibitors
EP1861715A2 (en) Biological markers predictive of anti-cancer response to epidermal growth factor receptor kinase inhibitors
Mahadevan et al. Novel receptor tyrosine kinase targeted combination therapies for imatinib-resistant gastrointestinal stromal tumors (GIST)
US8834873B2 (en) Method for detecting and controlling cancer
Guerard et al. Nuclear translocation of IGF1R by intracellular amphiregulin contributes to the resistance of lung tumour cells to EGFR-TKI
JP6675300B2 (en) Use of EGFR biomarkers for the treatment of gastric cancer with anti-EGFR drugs
Peeters et al. Panitumumab in combination with cytotoxic chemotherapy for the treatment of metastatic colorectal carcinoma
US20170234880A1 (en) Diagnosis of cancer
Kolb et al. Expression and differential signaling of heregulins in pancreatic cancer cells
TW201601753A (en) Protein biomarker and uses thereof
US20110275644A1 (en) Biological markers predictive of anti-cancer response to insulin-like growth factor-1 receptor kinase inhibitors
JP2017535548A (en) Prediction of VEGF antagonist response
US10184006B2 (en) Biomarkers for predicting outcomes of cancer therapy with ErbB3 inhibitors
EP2530462A1 (en) Methods and kits for predicting tumor responses to antibodies against epidermal growth factor receptor (EGFR) and uses thereof
US20170306415A1 (en) Predicting tumor responses to antibodies against hepatocyte growth factor (hgf) and/or its cognate receptor, c-met
Flaherty et al. Her-2 targeted therapy: beyond breast cancer and trastuzumab
Nutzinger et al. Management of HER2 alterations in non-small cell lung cancer–The past, present, and future
US20240042020A1 (en) Anti-garp-tgf-beta1/pd-1 combination therapy
Khan Prognostic significance, predictive value, and targeting of the HER-family members in pancreatic cancer
Bhosle Modulation of DNA strand break induction and repair by tyrosine kinase inhibitors targeted against EGFR and HER2
Joyce Varughese et al. Cervical carcinomas overexpress human trophoblast cell-surface marker (Trop-2) and are highly sensitive to immunotherapy with hRS7, a humanized monoclonal anti-Trop-2 antibody

Legal Events

Date Code Title Description
AS Assignment

Owner name: MERRIMACK PHARMACEUTICALS, INC, MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:BUKHALID, RAGHIDA;NIELSEN, ULRIK;WERNER, SHANNON;AND OTHERS;SIGNING DATES FROM 20120612 TO 20120613;REEL/FRAME:032872/0240

STCB Information on status: application discontinuation

Free format text: EXPRESSLY ABANDONED -- DURING EXAMINATION