WO2015031626A1 - Dosage de cmet soluble - Google Patents

Dosage de cmet soluble Download PDF

Info

Publication number
WO2015031626A1
WO2015031626A1 PCT/US2014/053181 US2014053181W WO2015031626A1 WO 2015031626 A1 WO2015031626 A1 WO 2015031626A1 US 2014053181 W US2014053181 W US 2014053181W WO 2015031626 A1 WO2015031626 A1 WO 2015031626A1
Authority
WO
WIPO (PCT)
Prior art keywords
seq
amino acid
acid sequence
scmet
concentration
Prior art date
Application number
PCT/US2014/053181
Other languages
English (en)
Inventor
Mark Gerald Anderson
Gerard J. Davis
Philip M. HEMKEN
Nicolette M. JEANBLANC
Jieyi Wang
Peter James Ansell
Original Assignee
Abbvie Inc.
Abbott Laboratories
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 Abbvie Inc., Abbott Laboratories filed Critical Abbvie Inc.
Publication of WO2015031626A1 publication Critical patent/WO2015031626A1/fr

Links

Classifications

    • 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
    • G01N33/5748Immunoassay; Biospecific binding assay; Materials therefor for cancer involving oncogenic proteins
    • 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
    • G01N2333/705Assays involving receptors, cell surface antigens or cell surface determinants
    • G01N2333/71Assays involving receptors, cell surface antigens or cell surface determinants for growth factors; for growth regulators

Definitions

  • the present invention relates to methods and immunoassay platforms for detecting soluble cMet and for determining a prognosis, diagnosis or risk identification of disease in a patient using anti-cMet antibodies.
  • the anti-cMet antibodies may be used to assess the efficacy of a therapeutic/prophylactic treatment of a subject suffering from disease.
  • cMet is an oncogenic receptor tyrosine kinase, with hepatocyte growth factor (HGF) being its only known ligand.
  • HGF hepatocyte growth factor
  • the primary single chain precursor protein is post-translationally cleaved to produce the alpha (50 kDa) and beta (145 kDa) subunits, which are linked together by a disulfide bridge to form the mature receptor.
  • Ligand dependent or independent dimerization is needed for cMet signaling. Both cMet and HGF are required for normal mammalian
  • cMet is normally expressed by cells of epithelial origin, while expression of HGF is restricted to cells of mesenchymal origin. Upon HGF stimulation, cMet induces several biological responses that collectively give rise to a program known as invasive growth. cMet activation triggers mitogenesis and morphogenesis. cMet is normally expressed only in stem cells and progenitor cells, which allows these cells to grow invasively in order to generate new tissues in an embryo or regenerate damaged tissues in an adult. cMet is involved in liver regeneration and wound healing during adulthood.
  • cMet is deregulated and plays a role in tumor progression, angiogenesis, invasive growth, metastasis and resistance to therapies.
  • Aberrantly active cMet triggers tumor growth (i.e., transformation/mitogenesis), formation of new blood vessels that supply the tumor with nutrients, functions anti-apoptotically, and invasion/metastasis (i.e., spread of cancer to other organs).
  • cMet may be a good biomarker for cancer.
  • current cMet assays are limited by the range of detecting cMet and its sensitivity to background.
  • cMet there is a need in the art to use cMet to determine, monitor, and treat cancer related diseases.
  • the present invention is directed to a method for determining scMet concentration in a test sample, the method comprising contacting the test sample with at least one capture antibody, wherein the capture antibody binds to an epitope on scMet or a fragment of scMet to form a capture antibody-scMet antigen complex; contacting the capture antibody-scMet antigen complex with at least one detection antibody comprising a detectable label, wherein the detection antibody binds to an epitope on scMet that is not bound by the capture antibody and forms a capture antibody-scMet antigen-detection antibody complex; and determining the scMet concentration in the test sample based on the signal generated by the detectable label in the capture antibody-scMet antigen-detection antibody complex.
  • the at least one capture antibody comprises a variable heavy domain comprising the amino acid sequence of SEQ ID NO: l; a variable light domain comprising the amino acid sequence of SEQ ID NO:5; a variable heavy domain comprising the amino acid sequence of SEQ ID NO: l and a variable light domain comprising the amino acid sequence of SEQ ID NO:5; a variable heavy chain comprising a complementarity determining region (CDR)1 comprising the amino acid sequence of SEQ ID NO:2, a CDR2 comprising the amino acid sequence of SEQ ID NO:3, and a CDR3 comprising the amino acid sequence of SEQ ID NO:4; a variable light chain comprising a CDRl comprising the amino acid sequence of SEQ ID NO:6, a CDR2 comprising the amino acid sequence of SEQ ID NO:7, and a CDR3 comprising the amino acid sequence of SEQ ID NO:8; or a variable heavy chain comprising a CDRl comprising the amino acid sequence of SEQ ID NO:2, a CDR2 comprising the
  • the at least one detection antibody comprises: a variable heavy domain comprising the amino acid sequence of SEQ ID NO:9; a variable light domain comprising the amino acid sequence of SEQ ID NO: 13; a variable heavy domain comprising the amino acid sequence of SEQ ID NO: 9 and a variable light domain comprising the amino acid sequence of SEQ ID NO: 13; a variable heavy chain comprising a CDR1 comprising the amino acid sequence of SEQ ID NO: 10, a CDR2 comprising the amino acid sequence of SEQ ID NO: 11 , and a CDR3 comprising the amino acid sequence of SEQ ID NO: 12; a variable light chain comprising a CDR1 comprising the amino acid sequence of SEQ ID NO: 14, a CDR2 comprising the amino acid sequence of SEQ ID NO: 15, and a CDR3 comprising the amino acid sequence of SEQ ID NO: 16; or a variable heavy chain comprising a CDR1 comprising the amino acid sequence of SEQ ID NO: 10, a CDR2 comprising the amino acid sequence of SEQ ID NO
  • the method may further comprise comparing the signal generated by the detectable label as a direct or indirect indication of the scMet concentration in the test sample to a signal generated as a direct or indirect indication of the scMet concentration in a control or calibrator.
  • the scMet concentration in the test sample may be used to determine or assess whether a subject has or is at risk of developing disease.
  • the disease may be liver disease or cancer.
  • An increased scMet concentration as compared to the scMet concentration in a control or calibrator indicates that the subject may have liver fibrosis or liver cancer.
  • An increased scMet concentration as compared to the scMet concentration in a control or calibrator indicates that the subject may have cancer.
  • the cancer may be gastric cancer, breast cancer, prostate cancer, liver cancer, or colon cancer.
  • the at least one capture antibody is an antibody produced by a hybridoma cell line deposited at the CNCM, Institut Pasteur, Paris, on March 14, 2007 under the number 1-3724 and the at least one detection antibody is an antibody produced by a hybridoma cell line deposited at the CNCM, Institut Pasteur, Paris, on March 12, 2008 under the number 1-3949.
  • the at least one detection antibody may be an F(ab') 2 fragment.
  • the present invention is also directed to a method of diagnosing and treating liver disease in a subject, the method comprising obtaining a biological sample comprising blood from the subject; determining the scMet concentration in the biological sample from the subject using the method for determining scMet concentration in a test sample described above; comparing the scMet concentration in the biological sample with the scMet concentration in a normal control or calibrator; diagnosing the subject as having liver disease if the scMet concentration in the biological sample is greater than the scMet concentration in the normal control or calibrator; and administering a liver disease treatment regimen to the subject diagnosed as having liver disease.
  • the biological sample of a subject is selected from a tissue sample, bodily fluid, whole blood, plasma, serum, urine, bronchoalveolar lavage fluid, and a cell culture suspension or fraction thereof.
  • the liver disease may be liver fibrosis or liver cancer.
  • the present invention is also directed to a method of diagnosing and treating cancer in a subject, the method comprising obtaining a biological sample comprising blood from the subject; determining the scMet concentration in the biological sample from the subject using the method for determining scMet concentration in a test sample described above; comparing the scMet concentration in the sample with the scMet concentration in a normal control or calibrator; diagnosing the subject as having cancer if the scMet concentration in the biological sample is greater than the scMet concentration in the normal control or calibrator; and administering a cancer treatment regimen to the subject diagnosed as having cancer.
  • the biological sample of a subject is selected from a tissue sample, bodily fluid, whole blood, plasma, serum, urine, bronchoalveolar lavage fluid, and a cell culture suspension or fraction thereof.
  • the cancer may be gastric cancer, breast cancer, prostate cancer, liver cancer, or colon cancer.
  • the present invention is also directed to a method for determining if a subject is responding to the administration of one or more pharmaceutical compositions, the method comprising: measuring the scMet concentration in a sample from the subject using the method for determining scMet concentration in a test sample as described above; comparing the scMet concentration in the sample with the scMet concentration in a normal control or calibrator, wherein an altered scMet concentration indicates that the subject is not responding to the administration of one or more pharmaceutical compositions; and adjusting the treatment of the subject if the subject is not responding to the administration of one or more pharmaceutical compositions.
  • the present invention is also directed to a method of diagnosing and treating liver disease in a subject, the method comprising contacting the test sample with at least one capture antibody, wherein the capture antibody binds to an epitope on scMet or a fragment of scMet to form a capture antibody-scMet antigen complex; contacting the capture antibody-scMet antigen complex with at least one detection antibody comprising a detectable label, wherein the detection antibody binds to an epitope on scMet that is not bound by the capture antibody and forms a capture antibody-scMet antigen-detection antibody complex; determining the scMet
  • the concentration in the test sample based on the signal generated by the detectable label in the capture antibody-scMet antigen-detection antibody complex formed in (b), whereupon the scMet concentration in the test sample is determined; comparing the scMet concentration in the sample with the scMet concentration in a normal control or calibrator; diagnosing the subject as having liver disease if the scMet concentration in the biological sample is greater than the scMet concentration in the normal control or calibrator; and administering a liver disease treatment regimen to the subject diagnosed as having liver disease.
  • the at least one capture antibody comprises a variable heavy domain comprising the amino acid sequence of SEQ ID NO: l; a variable light domain comprising the amino acid sequence of SEQ ID NO:5; a variable heavy domain comprising the amino acid sequence of SEQ ID NO: l and a variable light domain comprising the amino acid sequence of SEQ ID NO:5; a variable heavy chain comprising a complementarity determining region (CDR)1 comprising the amino acid sequence of SEQ ID NO:2, a CDR2 comprising the amino acid sequence of SEQ ID NO:3, and a CDR3 comprising the amino acid sequence of SEQ ID NO:4; a variable light chain comprising a CDR1 comprising the amino acid sequence of SEQ ID NO:6, a CDR2 comprising the amino acid sequence of SEQ ID NO:7, and a CDR3 comprising the amino acid sequence of SEQ ID NO:8; or a variable heavy chain comprising a CDR1 comprising the amino acid sequence of SEQ ID NO:2, a CDR2 comprising the
  • the at least one detection antibody comprises a variable heavy domain comprising the amino acid sequence of SEQ ID NO:9; a variable light domain comprising the amino acid sequence of SEQ ID NO: 13; a variable heavy domain comprising the amino acid sequence of SEQ ID NO: 9 and a variable light domain comprising the amino acid sequence of SEQ ID NO: 13; a variable heavy chain comprising a CDR1 comprising the amino acid sequence of SEQ ID NO: 10, a CDR2 comprising the amino acid sequence of SEQ ID NO: 11 , and a CDR3 comprising the amino acid sequence of SEQ ID NO: 12; a variable light chain comprising a CDR1 comprising the amino acid sequence of SEQ ID NO: 14, a CDR2 comprising the amino acid sequence of SEQ ID NO: 15, and a CDR3 comprising the amino acid sequence of SEQ ID NO: 16; or a variable heavy chain comprising a CDR1 comprising the amino acid sequence of SEQ ID NO: 10, a CDR2 comprising the amino acid sequence of SEQ ID NO:
  • the present invention is also directed to a method of diagnosing and treating cancer in a subject, the method comprising contacting the test sample with at least one capture antibody, wherein the capture antibody binds to an epitope on scMet or a fragment of scMet to form a capture antibody-scMet antigen complex; contacting the capture antibody-scMet antigen complex with at least one detection antibody comprising a detectable label, wherein the detection antibody binds to an epitope on scMet that is not bound by the capture antibody and forms a capture antibody-scMet antigen-detection antibody complex; determining the scMet
  • the concentration in the test sample based on the signal generated by the detectable label in the capture antibody-scMet antigen-detection antibody complex formed in (b), whereupon the scMet concentration in the test sample is determined; comparing the scMet concentration in the sample with the scMet concentration in a normal control or calibrator; diagnosing the subject as having cancer if the scMet concentration in the biological sample is greater than the scMet concentration in the normal control or calibrator; and administering a cancer treatment regimen to the subject diagnosed as having cancer.
  • the at least one capture antibody comprises a variable heavy domain comprising the amino acid sequence of SEQ ID NO: 1 ; a variable light domain comprising the amino acid sequence of SEQ ID NO:5; a variable heavy domain comprising the amino acid sequence of SEQ ID NO: 1 and a variable light domain comprising the amino acid sequence of SEQ ID NO:5; a variable heavy chain comprising a complementarity determining region (CDR)l comprising the amino acid sequence of SEQ ID NO:2, a CDR2 comprising the amino acid sequence of SEQ ID NO:3, and a CDR3 comprising the amino acid sequence of SEQ ID NO:4; a variable light chain comprising a CDR1 comprising the amino acid sequence of SEQ ID NO:6, a CDR2 comprising the amino acid sequence of SEQ ID NO:7, and a CDR3 comprising the amino acid sequence of SEQ ID NO: 8; or a variable heavy chain comprising a CDR1 comprising the amino acid sequence of SEQ ID NO:2, a CDR2 comprising the amino
  • the at least one detection antibody comprises a variable heavy domain comprising the amino acid sequence of SEQ ID NO:9; a variable light domain comprising the amino acid sequence of SEQ ID NO: 13; a variable heavy domain comprising the amino acid sequence of SEQ ID NO: 9 and a variable light domain comprising the amino acid sequence of SEQ ID NO: 13; a variable heavy chain comprising a CDR1 comprising the amino acid sequence of SEQ ID NO: 10, a CDR2 comprising the amino acid sequence of SEQ ID NO : 11 , and a CDR3 comprising the amino acid sequence of SEQ ID NO : 12; a variable light chain comprising a CDR1 comprising the amino acid sequence of SEQ ID NO: 14, a CDR2 comprising the amino acid sequence of SEQ ID NO: 15, and a CDR3 comprising the amino acid sequence of SEQ ID NO: 16; or a variable heavy chain comprising a CDR1 comprising the amino acid sequence of SEQ ID NO: 10, a CDR2 comprising the amino acid sequence of SEQ ID
  • the present invention is also directed to a kit for assaying a test sample for scMet.
  • the kit comprises at least one capture antibody, wherein the capture antibody binds to an epitope on scMet or a fragment of scMet, at least one detection antibody, wherein the detection antibody binds to an epitope on scMet that is not bound by the capture antibody, and instructions for assaying the test sample for scMet.
  • the at least one capture antibody comprises a variable heavy domain comprising the amino acid sequence of SEQ ID NO: 1 ; a variable light domain comprising the amino acid sequence of SEQ ID NO:5; a variable heavy domain comprising the amino acid sequence of SEQ ID NO: 1 and a variable light domain comprising the amino acid sequence of SEQ ID NO:5; a variable heavy chain comprising a complementarity determining region (CDR)1 comprising the amino acid sequence of SEQ ID NO:2, a CDR2 comprising the amino acid sequence of SEQ ID NO:3, and a CDR3 comprising the amino acid sequence of SEQ ID NO:4; a variable light chain comprising a CDR1 comprising the amino acid sequence of SEQ ID NO:6, a CDR2 comprising the amino acid sequence of SEQ ID NO:7, and a CDR3 comprising the amino acid sequence of SEQ ID NO: 8; or a variable heavy chain comprising a CDR1 comprising the amino acid sequence of SEQ ID NO:2, a CDR2 comprising the amino
  • the at least one detection antibody comprises a variable heavy domain comprising the amino acid sequence of SEQ ID NO:9; a variable light domain comprising the amino acid sequence of SEQ ID NO: 13; a variable heavy domain comprising the amino acid sequence of SEQ ID NO: 9 and a variable light domain comprising the amino acid sequence of SEQ ID NO: 13; a variable heavy chain comprising a CDR1 comprising the amino acid sequence of SEQ ID NO: 10, a CDR2 comprising the amino acid sequence of SEQ ID NO : 11 , and a CDR3 comprising the amino acid sequence of SEQ ID NO : 12; a variable light chain comprising a CDR1 comprising the amino acid sequence of SEQ ID NO: 14, a CDR2 comprising the amino acid sequence of SEQ ID NO: 15, and a CDR3 comprising the amino acid sequence of SEQ ID NO: 16; or a variable heavy chain comprising a CDR1 comprising the amino acid sequence of SEQ ID NO: 10, a CDR2 comprising the amino acid sequence of SEQ ID
  • the at least one detection antibody may be an F(ab') 2 fragment.
  • the at least one detection antibody may be optionally detectably labeled.
  • the kit may further comprise a reference standard indicating a scMet concentration in a control or calibrator.
  • the reference standard may indicate scMet concentration levels of 200 ng/mL (High), 50 ng/mL (Medium) and 12.5 ng/mL (Low).
  • FIG. 1 shows a schematic diagram of the ARCHITECT® immunometric assay principle.
  • FIG. 2 shows a representative sandwich array
  • FIG. 3 shows the binding curve of mAb 11E1 and cMet-A488.
  • FIG. 4 shows a graph of the ARCHITECT® scMet assays using Dynal uP.
  • FIG. 5 shows the complex testing of the ARCHITECT® scMet assay.
  • FIG. 6 shows a standard curve of scMet assay using 11E1 uP and 224D10 F(ab') 2 fragment conjugate where the circle on the graph represents a range of current samples tested.
  • FIGS. 7A-7B show the sample stability at room temperature at 4 and 24 hrs.
  • FIG. 7A shows the endogenous sample stability at room temperature at 4 and 24 hrs.
  • FIG. 7B shows the spiked sample stability at room temperature at 4 and 24 hrs.
  • FIGS. 8A-8B show the sample stability at 2-8°C at 24 and 72 hrs.
  • FIG. 8A shows the endogenous sample stability at 2-8°C at 24 and 72 hrs.
  • FIG. 8B shows the spiked sample stability at 2-8°C at 24 and 72 hrs.
  • FIGS. 9A-9B show the sample stability at -80°C at 24 and 72 hrs and day 12 and day 12 with 3 freeze/thaw cycles.
  • FIG. 9A shows the endogenous sample stability at -80°C at 24 and 72 hrs and day 12 and day 12 with 3 freeze/thaw cycles.
  • FIG. 9B shows the spiked sample stability at -80°C at 24 and 72 hrs and day 12 and day 12 with 3 freeze/thaw cycles.
  • FIG. 10 shows the levels of scMet in human patients with various diseases as measured by the ARCHITECT® scMet assays using the 11E1 antibody and 224D10 F(ab') 2 fragment antibody.
  • FIG. 11 shows a receiver operating characteristic (ROC) curve for prostate cancer vs normal healthy subjects.
  • FIG. 12 shows an ROC curve for breast cancer vs normal healthy subjects.
  • FIG. 13 shows an ROC curve for gastric cancer vs normal healthy subjects.
  • the present invention relates to an immunoassay for analyzing the levels of soluble cMet (scMet) to identify, diagnose and treat a disease in subjects in need thereof.
  • cMet is an oncogenic receptor tyrosine kinase involved in mitogenesis and morphogenesis.
  • cMet is found to be deregulated in many types of cancer, including liver, gastric, breast, prostate, and colon.
  • cMet is frequently overexpressed in cancer tissues, however, the activity of the cMet receptor may also play a role in cancer development alone or in combination with overexpressed cMet.
  • Table 1 shows the percentage of various types of cancers that have MET gene amplification (i.e., increased copy number of the MET gene), overexpression of cMet mRNA and/or overexpression of HGF mRNA.
  • the present invention immunoassay differs from previous scMet immunoassays by using a unique combination of antibodies to detect scMet in a sample over a wider range of concentrations, thus providing a more versatile and sensitive assay.
  • the present invention immunoassay detects scMet in a range that is about 235 times greater than the range of commercially available assays.
  • the increased detectable range of concentration of the disclosed immunoassay provides a more accurate and sensitive assay for diagnosing and distinguishing liver disease and cancer in a patient.
  • the present invention allows for more diseases to be more accurately diagnosed compared to scMet immunoassays that measure scMet levels using a different combination of antibodies.
  • the disclosed immunoassay may be used to detect increased or decreased scMet concentrations in a sample compared to a control or calibrator sample and thus be used to identify in a patient various liver diseases or cancers.
  • the wider range of detection of the disclosed immunoassay allows the determination and distinguishment of various diseases, which may vary in scMet concentrations by tens of nanogram amounts.
  • the use of the scMet immunoassay may provide accurate diagnosing and subsequent treatment of patients with liver disease or cancer.
  • each intervening number there between with the same degree of precision is explicitly contemplated.
  • the numbers 7 and 8 are contemplated in addition to 6 and 9, and for the range 6.0-7.0, the number 6.0, 6.1, 6.2, 6.3, 6.4, 6.5, 6.6, 6.7, 6.8, 6.9, and 7.0 are explicitly contemplated.
  • Antibody and “antibodies” as used herein refers to monoclonal antibodies, multispecific antibodies, human antibodies, humanized antibodies (fully or partially humanized), animal antibodies such as, but not limited to, a bird (for example, a duck or a goose), a shark, a whale, and a mammal, including a non-primate (for example, a cow, a pig, a camel, a llama, a horse, a goat, a rabbit, a sheep, a hamster, a guinea pig, a cat, a dog, a rat, a mouse, etc.) or a non-human primate (for example, a monkey, a chimpanzee, etc.), recombinant antibodies, chimeric antibodies, single-chain Fvs (“scFv”), single chain antibodies, single domain antibodies, Fab fragments, F(ab') fragments, F(ab')2 fragments, disulfide
  • antibodies include immunoglobulin molecules and immunologically active fragments of immunoglobulin molecules, namely, molecules that contain an analyte-binding site.
  • Immunoglobulin molecules can be of any type (for example, IgG, IgE, IgM, IgD, IgA, and IgY), class (for example, IgGl, IgG2, IgG3, IgG4, IgAl, and IgA2), or subclass.
  • an antibody against an analyte is frequently referred to herein as being either an "anti-analyte antibody” or merely an “analyte antibody” (e.g., an anti-scMet antibody or a scMet antibody).
  • Antibody fragment refers to a portion of an intact antibody comprising the antigen-binding site or variable region. The portion does not include the constant heavy chain domains (i.e. CH2, CH3, or CH4, depending on the antibody isotype) of the Fc region of the intact antibody.
  • antibody fragments include, but are not limited to, Fab fragments, Fab' fragments, Fab'-SH fragments, F(ab')2 fragments, Fd fragments, Fv fragments, diabodies, single-chain Fv (scFv) molecules, single-chain polypeptides containing only one light chain variable domain, single-chain polypeptides containing the three CDRs of the light-chain variable domain, single-chain polypeptides containing only one heavy chain variable region, and single-chain polypeptides containing the three CDRs of the heavy chain variable region.
  • AUC area under curve
  • AUC under a ROC curve is a measure of accuracy.
  • An area of 1 represents a perfect test, whereas an area of 0.5 represents an insignificant test.
  • a preferred AUC may be at least approximately 0.700, at least approximately 0.750, at least approximately 0.800, at least approximately 0.850, at least approximately 0.900, at least approximately 0.910, at least approximately 0.920 ⁇ at least approximately 0.930, at least approximately 0.940, at least approximately 0.950, at least approximately 0.960, at least approximately 0.970, at least approximately 0.980, at least approximately 0.990, or at least approximately 0.995.
  • Breast cancer refers to cancer that starts in the tissues of the breast.
  • the two main types of breast cancer are ductal carcinoma, which starts in the tubes (ducts) that move milk from the breast to the nipple, and lobular carcinoma, which starts in the parts of the breast, called lobules, that produce milk.
  • Breast cancer may also start in other areas of the breast.
  • Breast cancer may be invasive or noninvasive (in situ).
  • Cancer refers to the uncontrolled and unregulated growth of abnormal cells in the body. Cancerous cells are also called malignant cells. Cancer may invade nearby parts of the body and may also spread to more distant parts of the body through the lymphatic system or bloodstream. Cancers include Adrenocortical Carcinoma, Anal Cancer, Bladder Cancer, Brain Tumor, Breast Cancer, Carcinoid Tumor, Gastrointestinal, Carcinoma of
  • Lymphoma Central Nervous System (Primary) Lymphoma, Cutaneous T-Cell Lymphoma, Hodgkin's Disease Lymphoma, Non-Hodgkin's Disease Lymphoma, Malignant Mesothelioma, Melanoma, Merkel Cell Carcinoma, Metasatic Squamous Neck Cancer with Occult Primary, Multiple Myeloma and Other Plasma Cell Neoplasms, Mycosis Fungoides, Myelodysplasia Syndrome, Myeloproliferative Disorders, Nasopharyngeal Cancer, euroblastoma, Oral Cancer, Oropharyngeal Cancer, Osteosarcoma, Ovarian Epithelial Cancer, Ovarian Germ Cell Tumor, Pancreatic Cancer, Exocrine, Pancreatic Cancer, Islet Cell Carcinoma, Paranasal Sinus and Nasal Cavity Cancer, Parathyroid Cancer, Penile Cancer, Pituitary Cancer, Plasma Cell Neoplasm, Prostate Cancer, Rhabdom
  • cMet also known as MET or MNNG HOS Transforming gene
  • HGFR hepatocyte growth factor receptor
  • cMet proto-oncogene has a total length of 125,982 bp and is located in the 7q31 locus of chromosome 7.
  • the primary single chain precursor protein is post-translationally cleaved to produce the alpha (50 kDa) and beta (145 kDa) subunits, which are linked together by a disulfide bridge to form the mature receptor.
  • the ligand for cMet is hepatocyte growth factor (HGF).
  • scMet refers to soluble cMet.
  • scMet and cMet refer to scMet and cMet in a sample regardless of whether or not the scMet or cMet is bound to HGF.
  • the binding of HGF to proto-oncogenic cMet activate a tyrosine kinase signaling cascade that stimulates mitogenesis, cell motility, and matrix invasion.
  • cMet plays a role in angiogenesis, tumorogenesis, and tissue regeneration.
  • cMet overexpression is typically found in most cancers, such as gastric cancer, prostate cancer, breast cancer, liver cancer, and colon cancer. cMet activity may be more important in gastric cancer.
  • Colon cancer refers to cancer that starts in the large intestine (colon) or the rectum (end of the colon) and is also known as colorectal cancer. Almost all colon cancers start in glands in the lining of the colon and rectum. Nearly all colon cancers begin as noncancerous (benign) polyps that slowly develop into cancer.
  • F(ab') 2 fragment refers to antibodies generated by pepsin digestion of whole IgG antibodies to remove most of the Fc region while leaving intact some of the hinge region.
  • F(ab') 2 fragments have two antigen-binding F(ab) portions linked together by disulfide bonds, and therefore are divalent with a molecular weight of about 110 kDa.
  • Divalent antibody fragments are smaller than whole IgG molecules and enable a better penetration into tissue thus facilitating better antigen recognition in immunohistochemistry.
  • the use of F(ab') 2 fragments also avoids unspecific binding to Fc receptor on live cells or to Protein A/G.
  • F(ab') 2 fragments can both bind and precipitate antigens.
  • Gastric Cancer refers to cancer originating in the stomach. Gastric cancer may be caused by infection by Helicobacter pylori, autoimmune atrophic gastritis, or intestinal metaplasia. Various genetic factors are associated with increased risk levels of gastric cancer.
  • HGF refers to hepatocyte growth factor, which is a paracrine cellular growth, motility, and morphogenic factor. HGF is a pleotropic growth factor variously designated as scatter factor, hematopoietin A and mammary growth factor. HGF has a central role in angiogenesis, tumorogenesis, and tissue regeneration as it stimulates mitogenesis, cell motility, and matrix invasion. HGF is secreted by mesenchymal cells and acts as a
  • HGF multifunctional cytokine on cells of mainly epithelial origin.
  • HGF targets and acts primarily upon epithelial cells and endothelial cells but also haemopoietic progenitor cells.
  • HGF plays a role in embryonic organ development, adult organ regeneration, and wound healing.
  • HGF regulates cell growth, cell motility, and morphogenesis by activating a tyrosine kinase signaling cascade after binding to the proto-oncogenic cMet receptor.
  • HGF may interact with copper.
  • Label and “detectable label” as used herein refer to a moiety attached to an antibody or an analyte to render the reaction between the antibody and the analyte detectable, and the antibody or analyte so labeled is referred to as “detectably labeled.”
  • a label can produce a signal that is detectable by visual or instrumental means.
  • Various labels include signal-producing substances, such as chromogens, fluorescent compounds, chemiluminescent compounds, radioactive compounds, and the like.
  • Representative examples of labels include moieties that produce light, e.g., acridinium compounds, and moieties that produce fluorescence, e.g., fluorescein. Other labels are described herein.
  • the moiety itself, may not be detectable but may become detectable upon reaction with yet another moiety. Use of the term “detectably labeled” is intended to encompass such labeling.
  • the detectable label can be a radioactive label (such as 3 H, 14 C, 32 P, 33 P, 35 S, 90 Y, 99 Tc, m In, 125 1, 131 I,
  • an enzymatic label such as horseradish peroxidase, alkaline
  • chemiluminescent label such as acridinium esters, thioesters, or sulfonamides; luminol, isoluminol, phenanthridinium esters, and the like
  • fluorescent label such as fluorescein (e.g., 5 -fluorescein, 6-carboxyfiuorescein, 3'6- carboxyfiuorescein, 5(6)-carboxyfiuorescein, 6-hexachloro-fluorescein, 6-tetrachloro fluorescein, fluorescein isothiocyanate, and the like)
  • fluorescein e.g., 5 -fluorescein, 6-carboxyfiuorescein, 3'6- carboxyfiuorescein, 5(6)-carboxyfiuorescein, 6-hexachloro-fluorescein, 6-tetrachloro fluorescein, fluorescein isothiocyanate, and the like
  • An acridinium compound can be used as a detectable label in a homogeneous chemiluminescent assay (see, e.g., Adamczyk et al, Bioorg. Med. Chem. Lett. 16: 1324-1328 (2006); Adamczyk et al, Bioorg. Med. Chem. Lett. 4: 2313-2317 (2004); Adamczyk et al, Biorg. Med. Chem. Lett. 14: 3917-3921 (2004); and Adamczyk et al, Org. Lett. 5: 3779- 3782 (2003)).
  • the acridinium compound is an acridinium-9-carboxamide.
  • Methods for preparing acridinium 9-carboxamides are described in Mattingly, J. Biolumin. Chemilumin. 6: 107-114 (1991); Adamczyk et al, J. Org. Chem. 63: 5636-5639 (1998); Adamczyk et al, Tetrahedron 55: 10899-10914 (1999); Adamczyk et al, Org. Lett. 1 : 779-781 (1999); Adamczyk et al., Bioconjugate Chem.
  • an acridinium compound is an acridinium-9-carboxylate aryl ester.
  • An example of an acridinium-9-carboxylate aryl ester of formula II is 10-methyl-9- (phenoxycarbonyl)acridinium fluorosulfonate (available from Cayman Chemical, Ann Arbor, MI).
  • Methods for preparing acridinium 9-carboxylate aryl esters are described in McCapra et al., Photochem. Photobiol. 4: 1111-21 (1965); Razavi et al, Luminescence 15: 245-249 (2000); Razavi et al, Luminescence 15: 239-244 (2000); and U.S. Patent No.
  • acridinium-9-carboxylate aryl esters are efficient chemiluminescent indicators for hydrogen peroxide produced in the oxidation of an analyte by at least one oxidase in terms of the intensity of the signal and/or the rapidity of the signal.
  • the course of the chemiluminescent emission for the acridinium-9-carboxylate aryl ester is completed rapidly, i.e., in under 1 second, while the acridinium-9-carboxamide chemiluminescent emission extends over 2 seconds.
  • Acridinium-9- carboxylate aryl ester however, loses its chemiluminescent properties in the presence of protein. Therefore, its use requires the absence of protein during signal generation and detection.
  • the amount of protein removed or separated from the test sample can be about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, or about 95%.
  • acridinium-9-carboxylate aryl ester and its use are set forth in U.S. Pat. App. No. 11/697,835, filed April 9, 2007.
  • Acridinium-9-carboxylate aryl esters can be dissolved in any suitable solvent, such as degassed anhydrous N,N-dimethylformamide (DMF) or aqueous sodium cholate.
  • Liver cancer refers to cancer that originates in the liver. Liver cancer includes hepatocellular carcinoma and fibrolamellar carcinoma. In most cases, the cause of liver cancer is usually scarring of the liver (i.e., cirrhosis).
  • Liver cirrhosis refers to the consequence of chronic liver disease characterized by replacement of liver tissue by fibrosis, scar tissue, and regenerative nodules (lumps that occur because of a process in which damaged tissue is regenerated, leading to loss of liver function.
  • the architectural organization of the functional units of the liver become so disrupted that blood flow through the liver and liver function become disrupted.
  • Cirrhosis is most commonly caused by alcoholism, hepatitis B and C, and fatty liver disease, but has many other possible causes. Some cases are idiopathic (i.e., of unknown cause). Once cirrhosis has developed, the serious complications of liver disease may occur including portal hypertension, liver failure, and liver cancer.
  • Cirrhosis may be caused by alcohol abuse, autoimmune diseases of the liver, Hepatitis B or C virus infection, inflammation of the liver that is long-term (chronic), and iron overload in the body
  • hemochromatosis Patients with hepatitis B or C are at risk for liver cancer, even if they have not developed cirrhosis.
  • Liver disease refers to damage to or disease of the liver. Symptoms of liver dysfunction include both physical signs and a variety of symptoms related to digestive problems, blood sugar problems, immune disorders, abnormal absorption of fats, and metabolism problems. Liver disease includes liver fibrosis, liver cirrhosis, and liver cancer. All chronic liver diseases can lead to liver fibrosis. Chronic liver disease may be caused by chronic viral hepatitis B and alcoholic liver disease.
  • Liver fibrosis refers to an excessive accumulation of extracellular matrix proteins including collagen that occurs in most types of chronic liver diseases. Liver fibrosis is the scarring process that represents the liver's response to injury or illness. Liver fibrosis may be cause by infections due hepatitis B and C, parasites, excessive alcohol use and exposure to toxic chemicals, including pharmaceutical drugs and blocked bile ducts. Advanced liver fibrosis results in cirrhosis, liver failure, and portal hypertension and often requires liver transplantation.
  • Monoclonal antibody 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. Monoclonal antibodies are highly specific, being directed against a single antigen. Furthermore, in contrast to polyclonal antibody preparations that typically include different antibodies directed against different determinants (epitopes), each monoclonal antibody is directed against a single determinant on the antigen.
  • the monoclonal antibodies herein specifically include "chimeric" antibodies in which a portion of the heavy and/or light chain is identical with or homologous to corresponding sequences in antibodies derived from a particular species or belonging to a particular antibody class or subclass, while the remainder of the chain(s) is identical with or homologous to corresponding sequences in antibodies derived from another species or belonging to another antibody class or subclass, as well as fragments of such antibodies, so long as they exhibit the desired biological.
  • Prostate cancer refers to cancer that starts in the prostate gland.
  • Predetermined cutoff and predetermined level refer to an assay cutoff value that is used to assess diagnostic, prognostic, or therapeutic efficacy results by comparing the assay results against the predetermined cutoff/level, where the predetermined cutoff/level already has been linked or associated with various clinical parameters (e.g., presence of disease, stage of disease, severity of disease, progression, non-progression, or improvement of disease, etc.).
  • the disclosure provides exemplary predetermined levels.
  • cutoff values may vary depending on the nature of the immunoassay (e.g., antibodies employed, reaction conditions, sample purity, etc.).
  • Pretreatment reagent e.g., lysis, precipitation and/or solubilization reagent, as used in a diagnostic assay as described herein is one that lyses any cells and/or solubilizes any analyte that is/are present in a test sample. Pretreatment is not necessary for all samples, as described further herein. Among other things, solubilizing the analyte (i.e., HGF, fragments of HGF, variants of HGF or any combinations thereof) entails release of the analyte from any endogenous binding proteins present in the sample.
  • a pretreatment reagent may be homogeneous (not requiring a separation step) or heterogeneous (requiring a separation step).
  • the pretreatment reagent optionally can comprise: (a) one or more solvents and salt, (b) one or more solvents, salt and detergent, (c) detergent, (d) detergent and salt, or (e) any reagent or combination of reagents appropriate for cell lysis and/or solubilization of analyte.
  • Quadrature reagents in the context of immunoassays and kits described herein, include, but are not limited to, calibrators, controls, and sensitivity panels.
  • a "calibrator” or “standard” typically is used (e.g., one or more, such as a plurality) in order to establish calibration (standard) curves for interpolation of the concentration of an analyte, such as an antibody or an analyte.
  • a single calibrator which is near a predetermined positive/negative cutoff, can be used.
  • Risk assessment refers to the evaluation of factors including biomarkers, to predict the risk of occurrence of future events including disease onset or disease progression, so that treatment decisions regarding the subject may be made on a more informed basis.
  • sample may be used interchangeable and may be a sample of blood, tissue, urine, serum, plasma, amniotic fluid, cerebrospinal fluid, placental cells or tissue, endothelial cells, leukocytes, or monocytes.
  • the sample can be used directly as obtained from a patient or can be pre-treated, such as by filtration, distillation, extraction, concentration, centrifugation, inactivation of interfering components, addition of reagents, and the like, to modify the character of the sample in some manner as discussed herein or otherwise as is known in the art.
  • any cell type, tissue, or bodily fluid may be utilized to obtain a sample.
  • Such cell types, tissues, and fluid may include sections of tissues such as biopsy and autopsy samples, frozen sections taken for histologic purposes, blood (such as whole blood), plasma, serum, sputum, stool, tears, mucus, saliva, bronchoalveolar lavage (BAL) fluid, hair, skin, red blood cells, platelets, interstitial fluid, ocular lens fluid, cerebral spinal fluid, sweat, nasal fluid, synovial fluid, menses, amniotic fluid, semen, etc.
  • Cell types and tissues may also include lymph fluid, ascetic fluid, gynecological fluid, urine, peritoneal fluid, cerebrospinal fluid, a fluid collected by vaginal rinsing, or a fluid collected by vaginal flushing.
  • a tissue or cell type may be provided by removing a sample of cells from an animal, but can also be accomplished by using previously isolated cells (e.g., isolated by another person, at another time, and/or for another purpose).
  • Archival tissues such as those having treatment or outcome history, may also be used. Protein or nucleotide isolation and/or purification may not be necessary.
  • Series of calibrating compositions refers to a plurality of compositions comprising a known concentration of scMet, wherein each of the compositions differs from the other compositions in the series by the concentration of scMet.
  • Solid phase refers to any material that is insoluble, or can be made insoluble by a subsequent reaction.
  • the solid phase can be chosen for its intrinsic ability to attract and immobilize a capture agent.
  • the solid phase can have affixed thereto a linking agent that has the ability to attract and immobilize the capture agent.
  • the linking agent can include a charged substance that is oppositely charged with respect to the capture agent itself or to a charged substance conjugated to the capture agent.
  • the linking agent can be any binding partner (preferably specific) that is immobilized on (attached to) the solid phase and that has the ability to immobilize the capture agent through a binding reaction.
  • the linking agent enables the indirect binding of the capture agent to a solid phase material before the performance of the assay or during the performance of the assay.
  • the solid phase can be plastic, derivatized plastic, magnetic or non-magnetic metal, glass or silicon, including, for example, a test tube, microtiter well, sheet, bead, microparticle, chip, and other
  • a mammal e.g., cow, pig, camel, llama, horse, goat, rabbit, sheep, hamsters, guinea pig, cat, dog, rat, and mouse
  • a non-human primate for example, a monkey, such as a cynomolgous or rhesus monkey, chimpanzee, etc.
  • a human primate for example, a monkey, such as a cynomolgous or
  • the subject may be a human or a non-human.
  • the subject or patient may be undergoing other forms of treatment.
  • Treatment are each used interchangeably herein to describe reversing, alleviating, or inhibiting the progress of a disease, or one or more symptoms of such disease, to which such term applies.
  • the term also refers to preventing a disease, and includes preventing the onset of a disease, or preventing the symptoms associated with a disease.
  • a treatment may be either performed in an acute or chronic way.
  • the term also refers to reducing the severity of a disease or symptoms associated with such disease prior to affliction with the disease.
  • Such prevention or reduction of the severity of a disease prior to affliction refers to administration of an antibody or pharmaceutical composition of the present invention to a subject that is not at the time of administration afflicted with the disease.
  • Preventing also refers to preventing the recurrence of a disease or of one or more symptoms associated with such disease.
  • Treatment and “therapeutically,” refer to the act of treating, as “treating” is defined above.
  • the present invention is directed to method of detecting and measuring scMet in a sample from a subject using antibodies that bind to different scMet epitopes.
  • the method includes (a) obtaining a biological sample from a subject, (b) contacting the biological sample with a capture antibody, which binds to an epitope on scMet or scMet fragment to form a capture antibody-scMet antigen complex, (c) contacting the capture antibody-scMet antigen complex with a detection antibody which includes a detectable label and binds to an epitope on scMet or scMet fragment that is not bound by the capture antibody, to form a capture antibody-scMet antigen-detection antibody, and (d) determining the presence, amount or concentration of scMet or scMet fragment in the biological sample based on the signal generated by the detectable label in the capture antibody-scMet antigen-detection antibody complex.
  • the present invention is further directed to a method for diagnosing a disease in a subject based on the scMet levels in a sample from the subject.
  • the method includes the steps of (a) obtaining a biological sample from a subject, (b) determining the level of scMet in the biological sample, (c) comparing the level of scMet in the biological sample to a reference level of scMet, (d) identifying the subject as having a disease if the level of scMet in the biological sample is greater than the reference level of scMet, and (e) administering a treatment regiment to the subject identified as having disease.
  • Ranges of scMet detection have at least 5%, 10%, 25%, 50%, 75%, 100%, 110%, 120%, 130%, 140%, 150%, 160%, 170%, 180%, 190%, 200%, 210%, 220%, 230%, 240%, 250%, 260%, 270%, 280%, 290%, 300%, 400%, or 500% improved range size compared to other
  • scMet, and/or peptides or fragments thereof may be analyzed using an immunoassay.
  • the presence or amount of scMet or scMet fragment can be determined using antibodies and detecting specific binding to scMet or scMet fragment.
  • the antibody, or antibody fragment thereof may specifically bind to scMet or scMet fragment.
  • one or more of the antibodies can be used in combination with one or more of the antibodies.
  • the presence or amount of scMet or scMet fragment present in a body sample may be readily determined using an immunoassay, such as sandwich immunoassay (e.g., monoclonal- polyclonal sandwich immunoassays, including radioisotope detection (radioimmunoassay (RIA)) and enzyme detection (enzyme immunoassay (EIA) or enzyme-linked immunosorbent assay (ELISA) (e.g., Quantikine ELISA assays, R&D Systems, Minneapolis, MN)).
  • sandwich immunoassay e.g., monoclonal- polyclonal sandwich immunoassays, including radioisotope detection (radioimmunoassay (RIA)) and enzyme detection (enzyme immunoassay (EIA) or enzyme-linked immunosorbent assay (ELISA) (e.g., Quantikine ELISA assays, R&D Systems, Minneapolis, MN)).
  • sandwich immunoassay
  • chemiluminescent microparticle immunoassay in particular one employing the ARCHITECT® automated analyzer (Abbott Laboratories, Abbott Park, IL), is an example of a preferred immunoassay.
  • Other methods include, for example, mass spectrometry and
  • the use of immobilized antibodies or antibody fragments thereof may be incorporated into the immunoassay.
  • the antibodies may be immobilized onto a variety of supports, such as magnetic or chromatographic matrix particles, the surface of an assay plate (such as microtiter wells), pieces of a solid substrate material, and the like.
  • An assay strip can be prepared by coating the antibody or plurality of antibodies in an array on a solid support. This strip can then be dipped into the test biological sample and processed quickly through washes and detection steps to generate a measurable signal, such as a colored spot.
  • a heterogeneous format may be used. For example, after the test sample is obtained from a subject, a first mixture is prepared. The mixture contains the test sample being assessed for scMet or scMet fragment and a first specific binding partner, wherein the first specific binding partner and any scMet contained in the test sample form a first specific binding partner- scMet antigen complex.
  • the first specific binding partner may be an anti-scMet antibody that binds to an epitope having an amino acid sequence comprising at least three contiguous (3) amino acids of SEQ ID NO: 33 ("cMet (25-932)-HIEGRMD-6His"; amino acid 25-932 of cMet with a HIEGRMD linker (SEQ ID NO:36) and 6-histidine tag (SEQ ID NO:37)).
  • SEQ ID NO: 33 amino acid sequence comprising at least three contiguous (3) amino acids of SEQ ID NO: 33
  • cMet (25-932)-HIEGRMD-6His amino acid 25-932 of cMet with a HIEGRMD linker
  • SEQ ID NO:37 6-histidine tag
  • the solid phase used in the immunoassay can be any solid phase known in the art, such as, but not limited to, a magnetic particle, a bead, a test tube, a microtiter plate, a cuvette, a membrane, a scaffolding molecule, a film, a filter paper, a disc, and a chip.
  • any unbound scMet is removed from the complex using any technique known in the art.
  • the unbound scMet can be removed by washing.
  • the first specific binding partner is present in excess of any scMet present in the test sample, such that all scMet that is present in the test sample is bound by the first specific binding partner.
  • a second specific binding partner is added to the mixture to form a first specific binding partner-scMet antigen-second specific binding partner complex.
  • the second specific binding partner may be an anti-scMet antibody that binds to an epitope having an amino acid sequence comprising at least three contiguous (3) amino acids of SEQ ID NO: 33 ("cMet (25-932)-HIEGRMD-6His").
  • the second specific binding partner is labeled with or contains a detectable label as described above.
  • immobilized antibodies or antibody fragments thereof may be incorporated into the immunoassay.
  • the antibodies may be immobilized onto a variety of supports, such as magnetic or chromatographic matrix particles, the surface of an assay plate (such as microtiter wells), pieces of a solid substrate material, and the like.
  • An assay strip can be prepared by coating the antibody or plurality of antibodies in an array on a solid support. This strip can then be dipped into the test biological sample and processed quickly through washes and detection steps to generate a measurable signal, such as a colored spot.
  • the Sandwich ELISA measures the amount of antigen between two layers of antibodies (i.e., at least one capture antibody) and a detection antibody (i.e. at least one detection antibody).
  • the capture antibody and the detection antibody bind to different epitopes on the antigen, e.g., scMet. Desirably, binding of the capture antibody to an epitope does not interfere with binding of the detection antibody to an epitope.
  • Either monoclonal or polyclonal antibodies may be used as the capture and detection antibodies in the sandwich ELISA.
  • At least two antibodies are employed to separate and quantify scMet or scMet fragment in a test sample. More specifically, the at least two antibodies bind to certain epitopes of scMet or a scMet fragment forming an immune complex which is referred to as a "sandwich".
  • One or more antibodies can be used to capture the scMet or scMet fragment in the test sample (these antibodies are frequently referred to as a “capture” antibody or “capture” antibodies) and one or more antibodies is used to bind a detectable (namely, quantifiable) label to the sandwich (these antibodies are frequently referred to as the "detection" antibody or
  • “detection” antibodies In a sandwich assay, the binding of an antibody to its epitope desirably is not diminished by the binding of any other antibody in the assay to its respective epitope.
  • Antibodies are selected so that the one or more first antibodies brought into contact with a test sample suspected of containing scMet or scMet fragment do not bind to all or part of an epitope recognized by the second or subsequent antibodies, thereby interfering with the ability of the one or more second detection antibodies to bind to the scMet or scMet fragment.
  • the antibodies may be used as a first antibody in said immunoassay.
  • the antibody immunospecifically binds to epitopes on scMet.
  • said immunoassay may comprise a second antibody that immunospecifically binds to epitopes that are not recognized or bound by the first antibody.
  • a test sample suspected of containing scMet or scMet fragment can be contacted with at least one first capture antibody (or antibodies) and at least one second detection antibodies either simultaneously or sequentially.
  • a test sample suspected of containing scMet or scMet fragment is first brought into contact with the at least one first capture antibody that specifically binds to a particular epitope under conditions which allow the formation of a first antibody-scMet antigen complex. If more than one capture antibody is used, a first multiple capture antibody-scMet antigen complex is formed.
  • the antibodies, preferably, the at least one capture antibody are used in molar excess amounts of the maximum amount of scMet or scMet fragment expected in the test sample. For example, from about 5 ⁇ g/ml to about 1 mg/ml of antibody per ml of microparticle coating buffer may be used.
  • the at least one first capture antibody can be bound to a solid support which facilitates the separation the first antibody-scMet antigen complex from the test sample.
  • a solid support known in the art can be used, including but not limited to, solid supports made out of polymeric materials in the forms of wells, tubes, or beads.
  • the antibody (or antibodies) can be bound to the solid support by adsorption, by covalent bonding using a chemical coupling agent or by other means known in the art, provided that such binding does not interfere with the ability of the antibody to bind scMet or scMet fragment.
  • the solid support can be derivatized to allow reactivity with various functional groups on the antibody.
  • test sample suspected of containing scMet or scMet fragment is brought into contact with the at least one first capture antibody
  • the test sample is incubated in order to allow for the formation of a first capture antibody (or multiple antibody)-scMet antigen complex.
  • the incubation can be carried out at a pH of from about 4.5 to about 10.0, at a temperature of from about 2°C to about 45°C, and for a period from at least about one (1) minute to about eighteen (18) hours, from about 2-6 minutes, or from about 3-4 minutes.
  • the complex is then contacted with at least one second detection antibody (under conditions that allow for the formation of a first/multiple antibody-scMet antigen-second antibody complex). If the first antibody-scMet antigen complex is contacted with more than one detection antibody, then a first/multiple capture antibody-scMet antigen-multiple antibody detection complex is formed. As with first antibody, when the at least second (and subsequent) antibody is brought into contact with the first antibody-scMet antigen complex, a period of incubation under conditions similar to those described above is required for the formation of the first/multiple antibody-scMet antigen-second/multiple antibody complex.
  • At least one second antibody contains a detectable label.
  • the detectable label can be bound to the at least one second antibody prior to, simultaneously with or after the formation of the first/multiple antibody-scMet antigen-second/multiple antibody complex. Any detectable label known in the art can be used.
  • Chemiluminescent assays can be performed in accordance with the methods described in Adamczyk et al., Anal. Chim. Acta 579(1): 61-67 (2006). While any suitable assay format can be used, a microplate chemiluminometer (Mithras LB-940, Berthold Technologies U.S.A., LLC, Oak Ridge, TN) enables the assay of multiple samples of small volumes rapidly.
  • the chemiluminometer can be equipped with multiple reagent injectors using 96-well black polystyrene microplates (Costar #3792). Each sample can be added into a separate well, followed by the simultaneous/sequential addition of other reagents as determined by the type of assay employed. Desirably, the formation of pseudobases in neutral or basic solutions employing an acridinium aryl ester is avoided, such as by acidification. The chemiluminescent response is then recorded well-by-well. In this regard, the time for recording the
  • chemiluminescent response will depend, in part, on the delay between the addition of the reagents and the particular acridinium employed.
  • the order in which the test sample and the specific binding partner(s) are added to form the mixture for chemiluminescent assay is not critical. If the first specific binding partner is detectably labeled with an acridinium compound, detectably labeled first specific binding partner-scMet antigen complexes form. Alternatively, if a second specific binding partner is used and the second specific binding partner is detectably labeled with an acridinium compound, detectably labeled first specific binding partner-scMet antigen-second specific binding partner complexes form. Any unbound specific binding partner, whether labeled or unlabeled, can be removed from the mixture using any technique known in the art, such as washing.
  • Hydrogen peroxide can be generated in situ in the mixture or provided or supplied to the mixture before, simultaneously with, or after the addition of an above-described acridinium compound. Hydrogen peroxide can be generated in situ in a number of ways such as would be apparent to one skilled in the art.
  • a source of hydrogen peroxide can be simply added to the mixture.
  • the source of the hydrogen peroxide can be one or more buffers or other solutions that are known to contain hydrogen peroxide.
  • a solution of hydrogen peroxide can simply be added.
  • a detectable signal namely, a chemiluminescent signal, indicative of the presence of scMet or scMet fragment is generated.
  • the basic solution contains at least one base and has a pH greater than or equal to 10, preferably, greater than or equal to 12.
  • Examples of basic solutions include, but are not limited to, sodium hydroxide, potassium hydroxide, calcium hydroxide, ammonium hydroxide, magnesium hydroxide, sodium carbonate, sodium
  • the amount of basic solution added to the sample depends on the concentration of the basic solution. Based on the concentration of the basic solution used, one skilled in the art can easily determine the amount of basic solution to add to the sample.
  • the chemiluminescent signal that is generated can be detected using routine techniques known to those skilled in the art. Based on the intensity of the signal generated, the amount of scMet or scMet fragment in the sample can be quantified. Specifically, the amount of scMet in the sample is proportional to the intensity of the signal generated. The amount of scMet present can be quantified by comparing the amount of light generated to a standard curve for scMet or by comparison to a reference standard. The standard curve can be generated using serial dilutions or solutions of known concentrations of scMet by mass spectroscopy, gravimetric methods, and other techniques known in the art.
  • the conjugate diluent pH should be about 6.0 +/- 0.2
  • the microparticle coating buffer should be maintained at room temperature (i.e., at about 17 to about 27 ° C)
  • the microparticle coating buffer pH should be about 6.5 +/- 0.2
  • the microparticle diluent pH should be about 7.8 +/- 0.2.
  • Solids preferably are less than about 0.2%, such as less than about 0.15%, less than about 0.14%, less than about 0.13%, less than about 0.12%, or less than about 0.11%, such as about 0.10%.
  • the anti-cMet antibodies used in the methods described above may be a chimeric anti- cMet or humanized anti-cMet antibody.
  • the antibody may inhibit binding of human hepatocyte growth factor.
  • both the humanized antibody and chimeric antibody are monovalent.
  • both the humanized antibody and chimeric antibody comprise a single Fab region linked to an Fc region.
  • the antibody may comprise the F(ab') 2 fragment.
  • IC50 values are determined across an antibody concentration range from about 0.01 nM to around 1000 nM, about 0.1 nM to around 1000 nM, about 1.0 nM to around 1000 nM, about 10.0 nM to around 1000 nM, about 20.0 nM to around 1000 nM, about 50.0 nM to around 1000 nM, about 100.0 nM to around 1000 nM, about 200.0 nM to around 1000 nM, 0.01 nM to around 900 nM, about 0.1 nM to around 900 nM, about 1.0 nM to around 900 nM, about 10.0 nM to around 900 nM, about 20.0 nM to around 900 nM, about 50.0 nM to around 900 nM, about 100.0 nM to around 900 nM, about 200.0 nM to around 900 nM, 0.01 nM to around 800 nM, about 0.1 nM to around 800
  • the anti-cMet antibody may be an 1 1E1 monoclonal antibody, a 224D10 monoclonal antibody, or an antibody fragment thereof. (i) 1 IE 1 antibody
  • 11E1 or “ml 1E1” refers to a monoclonal antibody produced by hybridoma cell line deposited on March 14, 2007 under the number 1-3724 at the Collection Nationale de Cultures de Microorganismes (CNCM, National Collection of Microorganism Cultures) (Institut Pasteur, Paris, France) as described in WO 2009/007427, which is herein incorporated by reference in its entirety.
  • 11E1 binds to an epitope on cMet that is different from the epitope that 224D10 F(ab') 2 binds.
  • 11E1 has a heavy chain amino acid sequence of SEQ ID NO: 1, which is encoded by a nucleotide sequence of SEQ ID NO: 17, and a light chain amino acid sequence of SEQ ID NO: 5, which is encoded by a nucleotide sequence of SEQ ID NO: 21.
  • 11E1 includes CDR-Hl (SEQ ID NO: 2), CDR-H2 (SEQ ID NO: 3), and CDR-H3 (SEQ ID NO: 4) and CDR-L1 (SEQ ID NO: 6), CDR-L2 (SEQ ID NO: 7), and CDR-L3 (SEQ ID NO: 8), which are encoded by nucleotide sequences of SEQ ID NOs: 18-20 and 22-24, respectively.
  • 224D 10 or "m224D 10” refers to a monoclonal antibody produced by hybridoma cell line deposited on March 12, 2008 under the number 1-3949 at the Collection Nationale de Cultures de Microorganismes (CNCM, National Collection of Microorganism Cultures) (Institut Pasteur, Paris, France) as described in US 2012/0149031.
  • 224D10 binds to an epitope on cMet that is different from the epitope that 11E1 binds.
  • 224D10 has a heavy chain amino acid sequence of SEQ ID NO: 9, which is encoded by a nucleotide sequence of SEQ ID NO: 25, and a light chain amino acid sequence of SEQ ID NO: 13, which is encoded by a nucleotide sequence of SEQ ID NO: 29.
  • 224D10 includes CDR-Hl (SEQ ID NO: 10), CDR-H2 (SEQ ID NO: 11), and CDR-H3 (SEQ ID NO: 12) and CDR-L1 (SEQ ID NO: 14), CDR-L2 (SEQ ID NO: 15), and CDR-L3 (SEQ ID NO: 16), which are encoded by nucleotide sequences of SEQ ID NOs: 26-28 and 30-32, respectively.
  • the present invention is directed to a method for determining the presence, amount, or concentration of scMet or scMet fragment in a test sample using an anti-cMet antibody, such as the 11E1 antibody and 224D10 antibody, or antibody fragments thereof.
  • an anti-cMet antibody such as the 11E1 antibody and 224D10 antibody, or antibody fragments thereof.
  • the method includes the steps of (a) contacting the test sample with a capture antibody, which binds to an epitope on scMet or scMet fragment, so as to form a capture antibody-scMet antigen complex; (b) contacting the capture antibody-scMet antigen complex with at least one detection antibody, which comprises a detectable label and binds to an epitope on scMet or scMet fragment that is not bound by the capture antibody, to form a capture antibody-scMet antigen-detection antibody complex; and (c) determining the presence, amount or concentration of scMet or scMet fragment in the test sample based on the signal generated by the detectable label in the capture antibody- scMet antigen-detection antibody complex whereupon the present, amount, or concentration of scMet or scMet fragment in the test sample is determined.
  • the capture antibody includes the 11E1 antibody or a domain or region of: a variable heavy domain comprising the amino acid sequence of SEQ ID NO: 1 ; a variable light domain comprising the amino acid sequence of SEQ ID NO:5; a variable heavy domain comprising the amino acid sequence of SEQ ID NO: l and a variable light domain comprising the amino acid sequence of SEQ ID NO:5; a variable heavy chain comprising a complementarity determining region (CDR)1 comprising the amino acid sequence of SEQ ID NO:2, a CDR2 comprising the amino acid sequence of SEQ ID NO:3, and a CDR3 comprising the amino acid sequence of SEQ ID NO:4; a variable light chain comprising a CDR1 comprising the amino acid sequence of SEQ ID NO:6, a CDR2 comprising the amino acid sequence of SEQ ID NO:7, and a CDR3 comprising the amino acid sequence of SEQ ID NO:8; or a variable heavy chain comprising a CDR1 comprising the amino acid sequence of SEQ ID NO
  • the detection antibody includes a 224D10 antibody or a domain or region of: a variable heavy domain comprising the amino acid sequence of SEQ ID NO:9; a variable light domain comprising the amino acid sequence of SEQ ID NO: 13; a variable heavy domain comprising the amino acid sequence of SEQ ID NO:9 and a variable light domain comprising the amino acid sequence of SEQ ID NO: 13; a variable heavy chain comprising a CDR1 comprising the amino acid sequence of SEQ ID NO: 10, a CDR2 comprising the amino acid sequence of SEQ ID NO: 11 , and a CDR3 comprising the amino acid sequence of SEQ ID NO: 12; a variable light chain comprising a CDR1 comprising the amino acid sequence of SEQ ID NO: 14, a CDR2 comprising the amino acid sequence of SEQ ID NO: 15, and a CDR3 comprising the amino acid sequence of SEQ ID NO: 16; or a variable heavy chain comprising a CDR1 comprising the amino acid sequence of SEQ ID NO: 10, a CDR2
  • control sample may be analyzed concurrently with the sample from the subject as described above.
  • the results obtained from the subject sample can be compared to the results obtained from the control sample.
  • Standard curves may be provided, with which assay results for the biological sample may be compared.
  • Such standard curves present levels of marker as a function of assay units, i.e. fluorescent signal intensity, if a fluorescent label is used.
  • standard curves can be provided for control levels of the scMet in normal healthy tissue, as well as for "at- risk" levels of the scMet in tissue taken from donors, who may have one or more of the characteristics set forth above.
  • a method for determining the presence, amount, or concentration of scMet or scMet fragment in a test sample comprises assaying the test sample for scMet by an immunoassay, for example, employing at least one capture antibody that binds to an epitope on scMet or a fragment of scMet and at least one detection antibody that binds to an epitope on scMet which is different from the epitope for the capture antibody and optionally includes a detectable label, and comprising comparing a signal generated by the detectable label as a direct or indirect indication of the presence, amount or concentration of scMet in the test sample to a signal generated as a direct or indirect indication of the presence, amount or concentration of scMet in a calibrator.
  • the calibrator is optionally, and is preferably, part of a series of calibrators in which each of the calibrators differs from the other calibrators in the series by the concentration of scMet.
  • the method can further comprise diagnosing, prognosticating, or assessing the efficacy of a therapeutic/prophylactic treatment of a patient from whom the test sample was obtained. If the method further comprises assessing the efficacy of a therapeutic/prophylactic treatment of the patient from whom the test sample was obtained, the method optionally further comprises modifying the therapeutic/prophylactic treatment of the patient as needed to improve efficacy.
  • the method allows for more diseases to be more accurately diagnosed, and subsequently treated more successfully, compared to scMet immunoassays that measure scMet levels using a different combination of antibodies.
  • the method can be adapted for use in an automated system or a semi-automated system.
  • a predetermined level can be employed as a benchmark against which to assess results obtained upon assaying a test sample for scMet or scMet fragment.
  • the predetermined level is obtained by running a particular assay a sufficient number of times and under appropriate conditions such that a linkage or association of analyte presence, amount or concentration with a particular stage or endpoint of a disease, disorder or condition (e.g., liver disease or cancer) or with particular indicia can be made.
  • the predetermined level is obtained with assays of reference subjects (or populations of subjects).
  • the scMet measured can include scMet fragments thereof, degradation products thereof, and/or enzymatic cleavage products thereof.
  • the amount or concentration of scMet or scMet fragment may be "unchanged,” “favorable” (or “favorably altered”), or “unfavorable” (or “unfavorably altered”).
  • “Elevated” or “increased” refers to an amount or a concentration in a test sample that is higher or greater than a typical or normal level or range (e.g., predetermined level), or is higher or greater than another reference level or range (e.g., earlier or baseline sample).
  • lowered or reduced refers to an amount or a concentration in a test sample that is lower or less than a typical or normal level or range (e.g., predetermined level), or is lower or less than another reference level or range (e.g., earlier or baseline sample).
  • altered refers to an amount or a concentration in a sample that is altered (increased or decreased) over a typical or normal level or range (e.g., predetermined level), or over another reference level or range (e.g., earlier or baseline sample).
  • the typical or normal level or range for scMet is defined in accordance with standard practice.
  • a so-called altered level or alteration can be considered to have occurred when there is any net change as compared to the typical or normal level or range, or reference level or range that cannot be explained by experimental error or sample variation.
  • the level measured in a particular sample will be compared with the level or range of levels determined in similar samples from a so-called normal subject.
  • a "normal subject” is an individual with no detectable disease or disorder
  • a "normal” (sometimes termed “control") patient or population is/are one(s) that exhibit(s) no detectable disease or disorder, respectively, for example.
  • An "apparently normal subject” is one in which scMet has not been or is being assessed.
  • the level of an analyte is said to be "elevated" when the analyte is normally undetectable (e.g., the normal level is zero, or within a range of from about 25 to about 75 percentiles of normal populations), but is detected in a test sample, as well as when the analyte is present in the test sample at a higher than normal level.
  • the disclosure provides a method of screening for a subject having, or at risk of having, liver disease or cancer.
  • the method described herein can be used to provide a diagnosis of a subject having disease by determining the levels of scMet in a subject.
  • the method showed that scMet concentrations were higher in patients with liver disease or cancer.
  • the method may be used to detect disease in a subject using an anti-cMet antibody, such as the 11E1 antibody and 224D10 antibody, or antibody fragments thereof.
  • the method includes the steps of (a) obtaining a biological sample from a subject, (b) determining the level of scMet in the biological sample using the 11E1 antibody and 224D10 antibody, or antibody fragments thereof, as described above, (c) comparing the level of scMet in the biological sample to a reference level of scMet, (d) identifying the subject as having disease if the level of scMet in the biological sample is greater than the reference level of scMet, and (e) administering a treatment regimen to the subject identified as having disease.
  • the method described herein can be used to provide a diagnosis of a subject having liver disease by determining the levels of scMet in a subject.
  • the method showed that scMet concentrations were higher in patients with liver disease compared to healthy patients.
  • the method may be used to detect liver disease in a subject using an anti-cMet antibody, such as the 11E1 antibody and 224D10 antibody, or antibody fragments thereof.
  • the method includes the steps of (a) obtaining a biological sample from a subject, (b) determining the level of scMet in the biological sample using the 11E1 antibody and 224D10 antibody, or antibody fragments thereof, as described above, (c) comparing the level of scMet in the biological sample to a reference level of scMet, (d) identifying the subject as having liver disease if the level of scMet in the biological sample is greater than the reference level of scMet, and (e) administering a treatment regimen to the subject identified as having liver disease.
  • the reference level in this method can be the level of scMet in a patient having liver disease. Levels higher than or equal to 110 ng/mL, 111 ng/mL, 112 ng/mL, 113 ng/mL, 114 ng/mL, 115 ng/mL, 116 ng/mL, 117 ng/mL, 118 ng/mL, 119 ng/mL, 120 ng/mL, 121 ng/mL, 122 ng/mL, 123 ng/mL, 124 ng/mL, 125 ng/mL, 126 ng/mL, 127 ng/mL, 128 ng/mL, 129 ng/mL, 130 ng/mL, 131 ng/mL, 132 ng/mL, 133 ng/mL, 134 ng/mL, 135 ng/mL, 140 ng/mL, 145 ng/mL, 150
  • the method described herein can be used to provide a diagnosis of a subject having liver fibrosis by determining the levels of scMet in a subject.
  • the method showed that scMet concentrations were higher in patients with liver fibrosis compared to healthy patients.
  • the method may be used to detect liver fibrosis in a subject using an anti-cMet antibody, such as the 11E1 antibody and 224D10 antibody, or antibody fragments thereof.
  • the method includes the steps of (a) obtaining a biological sample from a subject, (b) determining the level of scMet in the biological sample using the 11E1 antibody and 224D10 antibody, or antibody fragments thereof, as described above, (c) comparing the level of scMet in the biological sample to a reference level of scMet, (d) identifying the subject as having liver fibrosis if the level of scMet in the biological sample is greater than the reference level of scMet, and (e) administering a treatment regimen to the subject identified as having liver fibrosis.
  • the reference level in this method can be the level of scMet in a patient having liver fibrosis. Levels higher than or equal to 120 ng/mL, 121 ng/mL, 122 ng/mL, 123 ng/mL, 124 ng/mL, 125 ng/mL, 126 ng/mL, 127 ng/mL, 128 ng/mL, 129 ng/mL, 130 ng/mL, 135 ng/mL, 140 ng/mL, 145 ng/mL, 150 ng/mL, 155 ng/mL, 160 ng/mL, 165 ng/mL, 170 ng/mL, 175 ng/mL, 180 ng/mL, 181 ng/mL, 182 ng/mL, 183 ng/mL, 184 ng/mL, 185 ng/mL, 186 ng/mL, 187 ng/mL,
  • the method described herein can be used to provide a diagnosis of a subject having liver cancer by determining the levels of scMet in a subject.
  • the method showed that scMet concentrations were higher in patients with liver cancer compared to healthy normal patients.
  • the method may be used to detect liver cancer in a subject using an anti-cMet antibody, such as the 11E1 antibody and 224D10 antibody, or antibody fragments thereof.
  • the method includes the steps of (a) obtaining a biological sample from a subject, (b) determining the level of scMet in the biological sample using the 11E1 antibody and 224D10 antibody, or antibody fragments thereof, as described above, (c) comparing the level of scMet in the biological sample to a reference level of scMet, (d) identifying the subject as having liver cancer if the level of scMet in the biological sample is greater than the reference level of scMet, and (e) administering a treatment regimen to the subject identified as having liver cancer.
  • the reference level in this method can be the level of scMet in a patient having liver cancer.
  • the method described herein can be used to provide a diagnosis of a subject having cancer by determining the levels of scMet in a subject.
  • the method showed that scMet concentrations were higher in patients with cancer.
  • the method may be used to detect cancer in a subject using an anti-cMet antibody, such as the 11E1 antibody and 224D10 antibody, or antibody fragments thereof.
  • the method includes the steps of (a) obtaining a biological sample from a subject, (b) determining the level of scMet in the biological sample using the 11E1 antibody and 224D10 antibody, or antibody fragments thereof, as described above, (c) comparing the level of scMet in the biological sample to a reference level of scMet, (d) identifying the subject as having cancer if the level of scMet in the biological sample is greater than the reference level of scMet, and (e) administering a treatment regimen to the subject identified as having cancer.
  • the reference level in this method can be the level of scMet in a patient having cancer. Levels higher than or equal to 105 ng/mL, 106 ng/mL, 107 ng/mL, 108 ng/mL, 109 ng/mL, 110 ng/mL, 111 ng/mL, 112 ng/mL, 113 ng/mL, 114 ng/mL, 115 ng/mL, 116 ng/mL, 117 ng/mL, 118 ng/mL, 119 ng/mL, 120 ng/mL, 121 ng/mL, 122 ng/mL, 123 ng/mL, 124 ng/mL, 125 ng/mL, 126 ng/mL, 127 ng/mL, 128 ng/mL, 129 ng/mL, 130 ng/mL, 131 ng/mL, 132 ng/mL,
  • the method described herein can be used to provide a diagnosis of a subject having gastric cancer by determining the levels of scMet in a subject.
  • scMet was measured using the 11E1 antibody as the capture antibody and the 224D10 F(ab') 2 fragment antibody as the detection antibody, each of which binds to cMet.
  • the decreased amounts of scMet detected by the combination of 224G11 and 11E1 antibodies may be due to the different epitope recognized by the 224G11 antibody, which may indicate that more scMet is being bound to its ligand HGF, even though there are high levels of scMet in patients with gastric cancer.
  • the method may be used to detect gastric cancer in a subject using an anti-cMet antibody, such as the 11E1 antibody and 224D10 antibody, or antibody fragments thereof.
  • the method includes the steps of (a) obtaining a biological sample from a subject, (b) determining the level of scMet in the biological sample using the 11E1 antibody and 224D10 antibody, or antibody fragments thereof, as described above, (c) comparing the level of scMet in the biological sample to a reference level of scMet, (d) identifying the subject as having gastric cancer if the level of scMet in the biological sample is greater than the reference level of scMet, and (e) administering a treatment regimen to the subject identified as having gastric cancer.
  • the reference level in this method can be the level of scMet in a patient having gastric cancer. Levels higher than or equal to 135 ng/niL, 136 ng/niL, 137 ng/niL, 138 ng/niL, 139 ng/mL, 140 ng/mL, 141 ng/niL, 142 ng/mL, 143 ng/mL, 144 ng/mL, 145 ng/niL, 150 ng/niL, 155 ng/mL, 160 ng/mL, 165 ng/mL, 170 ng/mL, 175 ng/mL, 180 ng/mL, 185 ng/mL, 190 ng/mL, 195 ng/mL, 196 ng/mL, 197 ng/mL, 198 ng/mL, 199 ng/mL, 200 ng/mL, 201 ng/mL, 202
  • the method described herein can be used to provide a diagnosis of a subject having liver cancer by determining the levels of scMet in a subject.
  • the method showed that scMet concentrations were higher in patients with liver cancer compared to healthy normal patients.
  • the method may be used to detect liver cancer in a subject using an anti-cMet antibody, such as the 11E1 antibody and 224D10 antibody, or antibody fragments thereof.
  • the method includes the steps of (a) obtaining a biological sample from a subject, (b) determining the level of scMet in the biological sample using the 11E1 antibody and 224D10 antibody, or antibody fragments thereof, as described above, (c) comparing the level of scMet in the biological sample to a reference level of scMet, (d) identifying the subject as having liver cancer if the level of scMet in the biological sample is greater than the reference level of scMet, and (e) administering a treatment regimen to the subject identified as having liver cancer.
  • the reference level in this method can be the level of scMet in a patient having liver cancer.
  • the method described herein can be used to provide a diagnosis of a subject having prostate cancer by determining the levels of scMet in a subject.
  • the method showed that scMet concentrations were higher in patients with prostate cancer compared to healthy normal patients.
  • the method may be used to detect prostate cancer in a subject using an anti-cMet antibody, such as the 11E1 antibody and 224D10 antibody, or antibody fragments thereof.
  • the method includes the steps of (a) obtaining a biological sample from a subject, (b) determining the level of scMet in the biological sample using the 11E1 antibody and 224D10 antibody, or antibody fragments thereof, as described above, (c) comparing the level of scMet in the biological sample to a reference level of scMet, (d) identifying the subject as having prostate cancer if the level of scMet in the biological sample is greater than the reference level of scMet, and (e) administering a treatment regimen to the subject identified as having prostate cancer.
  • the reference level in this method can be the level of scMet in a patient having prostate cancer. Levels higher than or equal to 107 ng/mL, 108 ng/mL, 109 ng/mL, 110 ng/mL, 111 ng/mL, 112 ng/mL, 113 ng/mL, 114 ng/mL, 115 ng/mL, 116 ng/mL, 117 ng/mL, 120 ng/mL, 125 ng/mL, 130 ng/mL, 135 ng/mL, 140 ng/mL, 145 ng/mL, 150 ng/mL, 155 ng/mL, 160 ng/mL, 161 ng/mL, 162 ng/mL, 163 ng/mL, 164 ng/mL, 165 ng/mL, 166 ng/mL, 167 ng/mL, 168 ng/mL, 169
  • the method described herein can be used to provide a diagnosis of a subject having breast cancer by determining the levels of scMet in a subject using.
  • the method showed that scMet concentrations were higher in patients with breast cancer compared to healthy normal patients.
  • the method may be used to detect breast cancer in a subject using an anti-cMet antibody, such as the 11E1 antibody and 224D10 antibody, or antibody fragments thereof.
  • the method includes the steps of (a) obtaining a biological sample from a subject, (b) determining the level of scMet in the biological sample using the 11E1 antibody and 224D10 antibody, or antibody fragments thereof, as described above, (c) comparing the level of scMet in the biological sample to a reference level of scMet, (d) identifying the subject as having breast cancer if the level of scMet in the biological sample is greater than the reference level of scMet, and (e) administering a treatment regimen to the subject identified as having breast cancer.
  • the reference level in this method can be the level of scMet in a patient having breast cancer. Levels higher than or equal to 110 ng/mL, 111 ng/mL, 112 ng/mL, 113 ng/mL, 114 ng/mL, 115 ng/mL, 116 ng/mL, 117 ng/mL, 118 ng/mL, 119 ng/mL, 120 ng/mL, 121 ng/mL, 122 ng/mL, 123 ng/mL, 124 ng/mL, 125 ng/mL, 130 ng/mL, 135 ng/mL, 140 ng/mL, 145 ng/mL, 150 ng/mL, 155 ng/mL, 160 ng/mL, 161 ng/mL, 162 ng/mL, 163 ng/mL, 164 ng/mL, 165 ng/mL, 166
  • the method described herein can be used to provide a diagnosis of a subject having colon cancer by determining the levels of scMet in a subject.
  • the method showed that scMet concentrations were higher in patients with colon cancer compared to healthy normal patients.
  • the method may be used to detect colon cancer in a subject using an anti-cMet antibody, such as the 11E1 antibody and 224D10 antibody, or antibody fragments thereof.
  • the method includes the steps of (a) obtaining a biological sample from a subject, (b) determining the level of scMet in the biological sample using the 11E1 antibody and 224D10 antibody, or antibody fragments thereof, as described above, (c) comparing the level of scMet in the biological sample to a reference level of scMet, (d) identifying the subject as having colon cancer if the level of scMet in the biological sample is greater than the reference level of scMet, and (e) administering a treatment regimen to the subject identified as having colon cancer.
  • the reference level in this method can be the level of scMet in a patient having colon cancer.
  • the methods described herein also can be used to determine whether or not a subject has or is at risk of developing liver disease by determining the levels of scMet in a subject using an anti-cMet antibody, such as the 11E1 antibody and 224D10 antibody, or antibody fragments thereof.
  • an anti-cMet antibody such as the 11E1 antibody and 224D10 antibody, or antibody fragments thereof.
  • the disclosure also provides a method for determining whether a subject having, or at risk for, liver disease, as discussed herein and known in the art, is a candidate for therapy or treatment.
  • the subject is one who has experienced some symptom of the disease or who has actually been diagnosed as having, or being at risk for, such a disease, and/or who demonstrates an unfavorable concentration or amount of scMet or scMet fragment, as described herein.
  • such a method can comprise the steps of: (a) determining the
  • the liver disease may be liver fibrosis or liver cancer.
  • the methods described herein also can be used to determine whether or not a subject has or is at risk of developing cancer by determining the levels of scMet in a subject using an anti-cMet antibody, such as the 11E1 antibody and 224D10 antibody, or antibody fragments thereof.
  • an anti-cMet antibody such as the 11E1 antibody and 224D10 antibody, or antibody fragments thereof.
  • the disclosure also provides a method for determining whether a subject having, or at risk for, cancer, as discussed herein and known in the art, is a candidate for therapy or treatment.
  • the subject is one who has experienced some symptom of the disease or who has actually been diagnosed as having, or being at risk for, such a disease, and/or who demonstrates an unfavorable concentration or amount of scMet or scMet fragment, as described herein.
  • such a method can comprise the steps of: (a) determining the concentration or amount in a test sample from a subject of scMet or scMet fragment using the methods described herein, or methods known in the art), and (b) comparing the concentration or amount of scMet or scMet fragment determined in step (a) with a predetermined level, wherein, if the concentration or amount of scMet determined in step (a) is favorable with respect to a predetermined level, then the subject is determined not to have or be at risk for cancer as discussed herein and known in the art.
  • the subject is determined to have or be at risk for cancer as discussed herein and known in the art.
  • the cancer may be gastric cancer, liver cancer, prostate cancer, breast cancer, or colon cancer.
  • the present invention is also directed to a method of monitoring the progression of disease, such as liver disease or cancer, in a subject by determining the levels of scMet in a subject using an anti-cMet antibody, such as the 11E1 antibody and 224D10 antibody, or antibody fragments thereof.
  • an anti-cMet antibody such as the 11E1 antibody and 224D10 antibody, or antibody fragments thereof.
  • the method includes the steps of (a) determining the concentration or amount of scMet or scMet fragment in a test sample from a subject using the 11E1 antibody and 224D10 antibody, or antibody fragments thereof, as described above, (b) determining the concentration or amount of scMet or scMet fragment in a later test sample from a subject using the 11E1 antibody and 224D10 antibody, or antibody fragments thereof, antibodies as described above, and (c) comparing the concentration or amount of scMet as determined in step (b) with the concentration or amount of scMet determined in step (a), wherein if the concentration or amount determined in step (b) is unchanged or is unfavorable when compared to the concentration or amount of scMet determined in step (a), then the disease in the subject is determined to have continued, progressed or worsened.
  • the concentration or amount of scMet as determined in step (b) is favorable when compared to the concentration or amount of scMet as
  • the method further comprises comparing the concentration or amount of scMet as determined in step (b), for example, with a predetermined level. Further, optionally the method comprises treating the subject with one or more pharmaceutical compositions for a period of time if the comparison shows that the concentration or amount of scMet as determined in step (b), for example, is unfavorably altered with respect to the predetermined level.
  • the methods can be used to monitor treatment in a subject receiving treatment with one or more pharmaceutical compositions.
  • such methods involve providing a first test sample from a subject before the subject has been administered one or more pharmaceutical compositions.
  • concentration or amount in a first test sample from a subject of scMet is determined (e.g., using the methods described herein or as known in the art).
  • concentration or amount of scMet is then compared with a predetermined level. If the concentration or amount of scMet as determined in the first test sample is lower than the predetermined level, then the subject is not treated with one or more pharmaceutical compositions or alternatively, the subject may be treated with one or more pharmaceutical compositions.
  • the subject is treated with one or more pharmaceutical compositions for a period of time or alternatively, the subject is not treated with one or more pharmaceutical compositions.
  • the period of time that the subject is treated with the one or more pharmaceutical compositions can be determined by one skilled in the art (for example, the period of time can be from about seven (7) days to about two years, preferably from about fourteen (14) days to about one (1) year).
  • second and subsequent test samples are then obtained from the subject.
  • the number of test samples and the time in which said test samples are obtained from the subject are not critical. For example, a second test sample could be obtained seven (7) days after the subject is first administered the one or more pharmaceutical compositions, a third test sample could be obtained two (2) weeks after the subject is first administered the one or more pharmaceutical
  • a fourth test sample could be obtained three (3) weeks after the subject is first administered the one or more pharmaceutical compositions, a fifth test sample could be obtained four (4) weeks after the subject is first administered the one or more pharmaceutical
  • the concentration or amount of scMet or scMet fragment is determined in the second or subsequent test sample is determined (e.g., using the methods described herein or as known in the art).
  • the concentration or amount of scMet as determined in each of the second and subsequent test samples is then compared with the concentration or amount of scMet as determined in the first test sample (e.g., the test sample that was originally optionally compared to the predetermined level).
  • step (c) If the concentration or amount of scMet as determined in step (c) is favorable when compared to the concentration or amount of scMet as determined in step (a), then the disease in the subject is determined to have discontinued, regressed or improved, and the subject should continue to be administered the one or pharmaceutical compositions of step (b).
  • the concentration or amount determined in step (c) is unchanged or is unfavorable when compared to the concentration or amount of scMet as determined in step (a)
  • the disease in the subject is determined to have continued, progressed or worsened, and the subject should be treated with a higher concentration of the one or more pharmaceutical compositions administered to the subject in step (b) or the subject should be treated with one or more pharmaceutical compositions that are different from the one or more pharmaceutical compositions administered to the subject in step (b).
  • the subject can be treated with one or more pharmaceutical compositions that are different from the one or more pharmaceutical compositions that the subject had previously received to decrease or lower said subject's scMet level.
  • a second or subsequent test sample is obtained at a period in time after the first test sample has been obtained from the subject.
  • a second test sample from the subject can be obtained minutes, hours, days, weeks or years after the first test sample has been obtained from the subject.
  • the second test sample can be obtained from the subject at a time period of about 1 minute, about 5 minutes, about 10 minutes, about 15 minutes, about 30 minutes, about 45 minutes, about 60 minutes, about 2 hours, about 3 hours, about 4 hours, about 5 hours, about 6 hours, about 7 hours, about 8 hours, about 9 hours, about 10 hours, about 11 hours, about 12 hours, about 13 hours, about 14 hours, about 15 hours, about 16 hours, about 17 hours, about 18 hours, about 19 hours, about 20 hours, about 21 hours, about 22 hours, about 23 hours, about 24 hours, about 2 days, about 3 days, about 4 days, about 5 days, about 6 days, about 7 days, about 2 weeks, about 3 weeks, about 4 weeks, about 5 weeks, about 6 weeks, about 7 weeks, about 8 weeks, about 9 weeks, about 10 weeks, about 11 weeks, about 12 weeks, about 13 weeks, about 14 weeks, about 15 weeks, about 16 weeks, about 17 weeks, about 18 weeks, about 19 weeks, about 20 weeks, about 21 weeks, about 22 weeks, about 23 weeks, about 24 weeks, about 2
  • Acute conditions also known as critical care conditions, refer to acute, life-threatening diseases or other critical medical conditions involving, for example, the cardiovascular system or excretory system.
  • critical care conditions refer to those conditions requiring acute medical intervention in a hospital-based setting (including, but not limited to, the emergency room, intensive care unit, trauma center, or other emergent care setting) or administration by a paramedic or other field- based medical personnel.
  • repeat monitoring is generally done within a shorter time frame, namely, minutes, hours or days (e.g., about 1 minute, about 5 minutes, about 10 minutes, about 15 minutes, about 30 minutes, about 45 minutes, about 60 minutes, about 2 hours, about 3 hours, about 4 hours, about 5 hours, about 6 hours, about 7 hours, about 8 hours, about 9 hours, about 10 hours, about 11 hours, about 12 hours, about 13 hours, about 14 hours, about 15 hours, about 16 hours, about 17 hours, about 18 hours, about 19 hours, about 20 hours, about 21 hours, about 22 hours, about 23 hours, about 24 hours, about 2 days, about 3 days, about 4 days, about 5 days, about 6 days or about 7 days), and the initial assay likewise is generally done within a shorter timeframe, e.g., about minutes, hours or days of the onset of the disease or condition.
  • minutes, hours or days e.g., about 1 minute, about 5 minutes, about 10 minutes, about 15 minutes, about 30 minutes, about 45 minutes, about 60 minutes, about 2 hours,
  • the assays also can be used to monitor the progression of disease in subjects suffering from chronic or non-acute conditions.
  • Non-critical care conditions or non-acute conditions refers to conditions other than acute, life-threatening disease or other critical medical conditions involving, for example, the cardiovascular system and/or excretory system.
  • non-acute conditions include those of longer-term or chronic duration.
  • repeat monitoring generally is done with a longer timeframe, e.g., hours, days, weeks, months or years (e.g., about 1 hour, about 2 hours, about 3 hours, about 4 hours, about 5 hours, about 6 hours, about 7 hours, about 8 hours, about 9 hours, about 10 hours, about 11 hours, about 12 hours, about 13 hours, about 14 hours, about 15 hours, about 16 hours, about 17 hours, about 18 hours, about 19 hours, about 20 hours, about 21 hours, about 22 hours, about 23 hours, about 24 hours, about 2 days, about 3 days, about 4 days, about 5 days, about 6 days, about 7 days, about 2 weeks, about 3 weeks, about 4 weeks, about 5 weeks, about 6 weeks, about 7 weeks, about 8 weeks, about 9 weeks, about 10 weeks, about 11 weeks, about 12 weeks, about 13 weeks, about 14 weeks, about 15 weeks, about 16 weeks, about 17 weeks, about 18 weeks, about 19 weeks, about 20 weeks, about 21 weeks, about 22 weeks, about 23 weeks, about 24 weeks, about 2 days, about 3 days, about
  • the initial assay likewise generally is done within a longer time frame, e.g., about hours, days, months or years of the onset of the disease or condition.
  • the above assays can be performed using a first test sample obtained from a subject where the first test sample is obtained from one source, such as urine, serum, or plasma.
  • the above assays can then be repeated using a second test sample obtained from the subject where the second test sample is obtained from another source.
  • the first test sample was obtained from urine
  • the second test sample can be obtained from serum or plasma.
  • the results obtained from the assays using the first test sample and the second test sample can be compared. The comparison can be used to assess the status of a disease or condition in the subject.
  • the present disclosure also relates to methods for determining whether a subject predisposed to or suffering from a disease (e.g., liver disease or cancer, as discussed herein and known in the art) will benefit from treatment.
  • a disease e.g., liver disease or cancer, as discussed herein and known in the art
  • the disclosure relates to scMet companion diagnostic methods and products.
  • the method of "monitoring the treatment of disease in a subject" as described herein further optimally also can encompass selecting or identifying candidates for liver disease treatments, such as liver resection and liver transplant, or for cancer treatments, such as surgery, radiation therapy, targeted therapy, and chemotherapy.
  • the present invention is also directed to a method for determining if a subject is responding to the administration of one or more pharmaceutical compositions by determining the levels of scMet in the subject using an anti-cMet antibody, such as the 11E1 antibody and
  • the method optionally comprises an assay as described herein, where the level of scMet is assessed before and following treatment of the subject with one or more pharmaceutical compositions (e.g., particularly with a pharmaceutical related to a mechanism of action involving scMet), or where the level of scMet is assessed following such treatment and the concentration or the level of scMet is compared against a predetermined level.
  • An unfavorable concentration of amount of scMet observed following treatment confirms that the subject will not benefit from receiving further or continued treatment, whereas a favorable concentration or amount of scMet observed following treatment confirms that the subject will benefit from receiving further or continued treatment. This confirmation assists with management of clinical studies, and provision of improved patient care.
  • the method includes the steps of (a) obtaining a biological sample from a subject, (b) determining the level of scMet in the biological sample using the 11E1 antibody and 224D10 antibody, or antibody fragments thereof, described above, (c) comparing the level of scMet in the biological sample to a reference level of scMet, wherein an altered concentration of scMet indicates that the subject is not responding to the administration of one or more pharmaceutical compositions, and (d) adjusting the treatment of the subject if the subject is not responding to the administration of one or more pharmaceutical compositions.
  • the antibodies and methods described above may be used to detect and measure levels and concentrations of scMet in combination with one or more biomarkers or immunoassays specific for disease.
  • the combination of scMet with one or more biomarkers or immunoassays specific for disease may provide a greater discrimination between healthy controls and individuals with disease compared to measuring scMet alone. For example, measure a panel of scMet and HGF biomarkers provide a greater discrimination between healthy controls and individuals with disease compared to a panel of scMet alone.
  • the combination of scMet with at least one or more biomarkers may provide greater discrimination between healthy controls and individuals who have liver disease and/or individuals who have cancer.
  • Examples of the one or more biomarker and/or immunoassay include cMet, including scMet, and/or immunoassays that measure cMet and/or scMet, such as the ARCHITECT® scMet A Assay.
  • the ARCHITECT® scMet A Assay measures scMet concentration using the 224G11 antibody as the capture antibody and an 11E1 antibody as the detection antibody, each of which binds to cMet.
  • the 224G11 antibody is an antibody produced by hybridoma cell line deposited at the CNCM, Institut Pasteur, Paris, on March 14, 2007 under the number 1-3731.
  • the 11E1 antibody is an antibody produced by hybridoma cell line deposited at the CNCM, Institut Pasteur, Paris, on March 14, 2007 under the number 1-3724.
  • the 224G11 antibody recognizes and binds to an epitope that is different from the epitopes recognized by the 11E1 and 224D10 antibodies.
  • Another example includes the biomarker HGF and/or immunoassays that measure HGF, such as the ARCHITECT® HGF Assay.
  • the ARCHITECT® HGF Assay measures HGF concentration using a 64.4.1 antibody as the capture antibody and an H98C4 antibody as the detection antibody, each of which binds to HGF.
  • the 64.4.1 antibody is an antibody produced by hybridoma cell line that will be deposited with the American Type Culture Collection
  • the H98C4 antibody is an antibody produced by hybridoma cell line that will be deposited with the American Type Culture Collection (A.T.C.C.), 10801 University Boulevard., Manassas, VA 20110-2209 under the terms of the Budapest Treaty. 5. Treatment of Subjects Suffering from Liver Disease
  • liver disease The subject identified in the methods described above having levels of scMet greater than or equal to the values discussed above is identified as a patient suffering from liver disease. The subject is then treated for the liver disease. Treatment of liver disease may include small molecule cMet inhibitors, such as K252a, SU11274, PHA-665752, SGX523, MP470,
  • INCB28060 EMD 1214063, EMD 1214831, AMG-458, PF-04217903, crizotinib (PF- 02341066), E7050, MK-2461, BMS-777607, JNJ-38877605, tivantinib (ARQ197), foretinib (GSK/1363089 XL880), and cabozantinib (XL 184), anti-cMet neutralizing antibodies, such as onartuzumab, HGF inhibitors, such as NK4, and anti-HGF neutralizing antibodies, such TAK- 701, rilotumumab (AMG102) and ficlatuzumab (AV-299).
  • anti-cMet neutralizing antibodies such as onartuzumab
  • HGF inhibitors such as NK4
  • anti-HGF neutralizing antibodies such TAK- 701, rilotumumab (AMG102) and ficlatuzumab (AV-299).
  • the subject identified in the methods described above having levels of scMet greater than or equal to the values discussed above is identified as a patient suffering from liver fibrosis.
  • the subject is then treated for liver fibrosis.
  • Treatment may include medications. These medications may include cMet inhibitors, HGF inhibitors, angiotensin inhibitors, corticosteroids, endothelin inhibitors, interferon-alpha, interleukin 10, pentoxifylline, peroxisome proliferator- activated receptor (PPAR) antagonists, TGF-betal inhibitors, antioxidants, such as S-adenosyl- methionine, phosphatidylcholine, silymarin, and tocopherol, lamivudine, or herbal compounds, such as Sho-saiko-to, glycyrrhizin, colchicine, and salvia miltiorrhiza.
  • cMet inhibitors such as HGF inhibitors, angiotensin inhibitors, corticosteroids
  • the subject identified in the methods described above having levels of scMet greater than or equal to the values discussed above is identified as a patient suffering from liver cancer.
  • the subject is then treated for liver cancer.
  • Treatment may include surgical removal of the cancer (liver resection) with or without liver transplant, chemotherapy, such as doxorubicin (Adriamycin), 5-fluorouracil (5 FU), tamoxifen (Nolvadex), Octreotide (Sandostatin), gemcitabine, cisplatin, and oxaliplatin, biotherapy, such as bevacizumab, chemoembolization (trans-arterial chemoembolization or TACE), radioembolization (selective internal radiotherapy; "SIPvT”), ablation, such as radiofrequency ablation (RFA) therapy, percutaneous ethanol (alcohol) injection, and cryoablation, stereotactic radiosurgery, and proton beam therapy.
  • RFA radiofrequency ablation
  • Treatment may also include drugs that block components of the angiogenesis pathway, such as sorafenib (Nexavar). Treatment may also include cMet inhibitors and HGF inhibitors. The drugs may be delivered via the hepatic artery or portal vein.
  • the subject identified in the methods described above having levels of scMet greater than or equal to the values discussed above is identified as a patient suffering from cancer.
  • the subject is then treated for the cancer.
  • Treatment of cancer may include small molecule cMet inhibitors, such as K252a, SU11274, PHA-665752, SGX523, MP470, INCB28060, EMD 1214063, EMD 1214831, AMG-458, PF-04217903, crizotinib (PF-02341066), E7050, MK- 2461, BMS-777607, JNJ-38877605, tivantinib (ARQ197), foretinib (GSK/1363089/XL880), and cabozantinib (XL 184), anti-cMet neutralizing antibodies, such as onartuzumab, HGF inhibitors, such as NK4, and anti-HGF neutralizing antibodies, such TAK-701, rilotumumab (AMG
  • the subject identified in the methods described above having levels of scMet greater than or equal to the values discussed above is identified as a patient suffering from gastric cancer.
  • the subject is then treated for gastric cancer.
  • Treatment may include surgery, including endoscopic mucosal resection and endoscopic submucosal dissection, radiation (X-rays), and chemotherapy, including 5-FU (fluorouracil) or its analog capecitabine, BCNU (carmustine), methyl-CCNU (Semustine), doxorubicin (Adriamycin), Mitomycin C, cisplatin and taxotere.
  • Treatment may also include imatinib (Gleevec) and sunitinib (Sutent).
  • the subject identified in the methods described above having levels of scMet greater than or equal to the values discussed above is identified as a patient suffering from liver cancer. The subject is then treated for the liver cancer, as described above.
  • the subject identified in the methods described above having levels of scMet greater than or equal to the values discussed above is identified as a patient suffering from prostate cancer.
  • the subject is then treated for the prostate cancer.
  • Treatments may include watchful waiting or active surveillance, surgery, cryosurgery, high-intensity focused ultrasound, radiation, hormone therapy, chemotherapy, and targeted therapy.
  • surgery include pelvic lymphadenectomy, radical prostatectomy (retropubic prostatectomy and perineal prostatectomy), transurethral resection of the prostate, and laparoscopic prostatectomy.
  • radiation therapy include high-energy x-rays (external and internal radiation therapy), proton beam radiation, and intensity-modulated radiation therapy.
  • hormone therapy examples include luteinizing hormone-releasing hormone agonists, such as leuprolide (Lupron, Eligard), goserelin, triptorelin (Trelstar), histrelin (Vantas), and buserelin, antiandrogens, such as flutamide, bicalutamide (Casodex), and nilutamide (Nilandron), ketoconazole, orchiectomy, estrogen, and aminoglutethimide.
  • leuprolide Liupron, Eligard
  • goserelin triptorelin (Trelstar)
  • histrelin Vantas
  • buserelin antiandrogens
  • antiandrogens such as flutamide, bicalutamide (Casodex), and nilutamide (Nilandron)
  • ketoconazole orchiectomy
  • estrogen aminoglutethimide
  • chemotherapeutic drugs include abiraterone acetate (Zytiga), cabazitaxel, degarelix, docetaxel, enzalutamide (Xtandi), cabazitaxel (Jevtana), leuprolide acetate (Lupron, Lupron Depot, Lupron Depot-3 Month, Lupron Depot-4 Month, Lupron Depot- Ped, Viadur), prednisone, sipuleucel-T (Provenge), estramustine (Emcyt), mitoxantrone
  • the subject identified in the methods described above having levels of scMet greater than or equal to the values discussed above is identified as a patient suffering from breast cancer.
  • the subject is then treated for the breast cancer.
  • Treatment may include surgery, radiation therapy, bone-directed therapy, chemotherapy, hormone therapy, and targeted therapy.
  • Examples of surgery include lumpectomy, quadrantectomy, mastectomy, such as simple mastectomy, skin-sparing mastectomy, modified radical mastectomy, prophylactic mastectomy, and radical mastectomy, prophylactic ovary removal, cryotherapy, and lymph node surgery, such as axillary lymph node dissection and sentinel lymph node biopsy.
  • Examples of radiation therapy include external beam radiation, such as accelerated breast irradiation and 3D-conformal radiotherapy, and brachytherapy (internal radiation), such as interstitial brachytherapy, intracavitary brachytherapy, and intraoperative radiation.
  • Examples of bone-directed therapy include bisphosphonates and denosumab.
  • chemotherapy examples include anthracyclines, such as doxorubicin (Adriamycin, Doxil), epirubicin (Ellence), and daunorubicin (Cerubidine, DaunoXome), capecitabine (Xeloda), carboplatin (Paraplatin), cisplatin, cyclophosphamide (Cytoxan), eribulin (Halaven), fluorouracil (also called 5-fluorouracil or 5-FU; Adrucil), gemcitabine (Gemzar), ixabepilone (Ixempra), methotrexate (Amethopterin, Mexate, Folex), mitoxantrone (Novantrone), mutamycin (Mitomycin), taxanes, such as paclitaxel (Taxol, Abraxane), and docetaxel (Taxotere), thiotepa (Thioplex), vincristine (Oncovin
  • hormone therapy examples include aromatase inhibitors, such as anastrozole (Arimidex), exemestane (Aromasin), and letrozole (Femara), selective estrogen receptor modulators (SERMs), such as tamoxifen (Nolvadex), raloxifene (Evista), and toremifene (Fareston), and estrogen-receptor downregulators, such as fulvestrant (Faslodex).
  • SERMs selective estrogen receptor modulators
  • targeted therapy include trastuzumab (Herceptin), lapatinib (Tykerb), bevacizumab (Avastin), pertuzumab (Perjeta), and everolimus (Afinitor).
  • the subject identified in the methods described above having levels of scMet greater than or equal to the values discussed above is identified as a patient suffering from colon cancer.
  • the subject is then treated for the colon cancer.
  • Treatment may include surgery, including local excision, resection with or without colostomy, radiofrequency ablation, and cryosurgery, radiation, such as high-energy X-rays, chemotherapy, including fluorouracil (5-FU), capecitabine (Xeloda), irinotecan hydrochloride (Camptosar), oxaliplatin (Eloxatin), leucovorin calcium (Wellcovorin), and ziv-aflibercept (Zaltrap), and targeted therapy, such as bevacizumab
  • kits which may be used for assaying a test sample for scMet or scMet fragment.
  • the kit comprises at least one component for assaying the test sample for scMet or scMet fragment and instructions for assaying the test sample for scMet or scMet fragment.
  • the kit can comprise instructions for assaying the test sample for scMet or scMet fragment by immunoassay, e.g., chemiluminescent microparticle immunoassay.
  • kits can be affixed to packaging material or can be included as a package insert. While the instructions are typically written or printed materials they are not limited to such. Any medium capable of storing such instructions and communicating them to an end user is contemplated by this disclosure. Such media include, but are not limited to, electronic storage media (e.g., magnetic discs, tapes, cartridges, chips), optical media (e.g., CD ROM), and the like. As used herein, the term "instructions" can include the address of an internet site that provides the instructions.
  • the at least one component may include at least one composition comprising one or more isolated antibodies or antibody fragments thereof that specifically bind to scMet or scMet fragment.
  • the antibody may be a scMet capture antibody and/or a scMet detection antibody.
  • the antibody may include the 11E1 antibody, 224D10 antibody, or antibody fragments thereof. The antibody is optionally detectably labeled.
  • the kit can comprise a calibrator or control, e.g., purified, and optionally lyophilized, scMet or scMet fragment, and/or at least one container (e.g., tube, microtiter plates or strips, which can be already coated with an anti-cMet monoclonal antibody) for conducting the assay, and/or a buffer, such as an assay buffer or a wash buffer, either one of which can be provided as a concentrated solution, a substrate solution for the detectable label (e.g., an enzymatic label), or a stop solution.
  • the kit comprises all components, i.e., reagents, standards, buffers, diluents, etc., which are necessary to perform the assay.
  • the instructions also can include instructions for generating a standard curve.
  • the kit may further comprise reference standards for quantifying scMet.
  • the reference standards may be employed to establish standard curves for interpolation and/or extrapolation of scMet concentrations.
  • the reference standards may include a high scMet concentration level, for example, about 100 ng/mL, about 125 ng/mL, about 150 ng/mL, about 175 ng/mL, about 200 ng/mL, about 225 ng/mL, about 250 ng/mL, about 275 ng/mL, or about 300 ng/mL; a medium scMet concentration level, for example, about 25 ng/mL, about 40 ng/mL, about 45 ng/mL, about 50 ng/mL, about 55 ng/mL, about 60 ng/mL, about 75 ng/mL or about 100 ng/mL; and/or a low scMet concentration level, for example, about 1 ng/mL, about 5 ng/mL, about 10
  • any antibodies, which are provided in the kit can incorporate a detectable label, such as a fluorophore, radioactive moiety, enzyme, biotin/avidin label, chromophore, chemiluminescent label, or the like, or the kit can include reagents for labeling the antibodies or reagents for detecting the antibodies (e.g., detection antibodies) and/or for labeling the analytes or reagents for detecting the analyte.
  • the antibodies, calibrators, and/or controls can be provided in separate containers or pre-dispensed into an appropriate assay format, for example, into microtiter plates.
  • the kit includes quality control components (for example, sensitivity panels, calibrators, and positive controls).
  • quality control components for example, sensitivity panels, calibrators, and positive controls.
  • Preparation of quality control reagents is well-known in the art and is described on insert sheets for a variety of immunodiagnostic products.
  • Sensitivity panel members optionally are used to establish assay performance characteristics, and further optionally are useful indicators of the integrity of the immunoassay kit reagents, and the standardization of assays.
  • the kit can also optionally include other reagents required to conduct a diagnostic assay or facilitate quality control evaluations, such as buffers, salts, enzymes, enzyme co-factors, substrates, detection reagents, and the like.
  • Other components such as buffers and solutions for the isolation and/or treatment of a test sample (e.g., pretreatment reagents), also can be included in the kit.
  • the kit can additionally include one or more other controls.
  • One or more of the components of the kit can be lyophilized, in which case the kit can further comprise reagents suitable for the reconstitution of the lyophilized components.
  • kits for holding or storing a sample (e.g., a container or cartridge for a urine, plasma, or serum sample).
  • a sample e.g., a container or cartridge for a urine, plasma, or serum sample.
  • the kit optionally also can contain reaction vessels, mixing vessels, and other components that facilitate the preparation of reagents or the test sample.
  • the kit can also include one or more instrument for assisting with obtaining a test sample, such as a syringe, pipette, forceps, measured spoon, or the like.
  • the kit can comprise at least one acridinium-9-carboxamide, at least one acridinium-9-carboxylate aryl ester, or any combination thereof. If the detectable label is at least one acridinium compound, the kit also can comprise a source of hydrogen peroxide, such as a buffer, solution, and/or at least one basic solution. If desired, the kit can contain a solid phase, such as a magnetic particle, bead, test tube, microtiter plate, cuvette, membrane, scaffolding molecule, film, filter paper, disc, or chip.
  • the kit can further comprise one or more components, alone or in further combination with instructions, for assaying the test sample for another analyte, which can be a biomarker, such as a biomarker of liver disease or disorder.
  • another analyte which can be a biomarker, such as a biomarker of liver disease or disorder.
  • concentration of scMet in a test sample by an immunoassay as described herein can be adapted for use in a variety of automated and semi-automated systems (including those wherein the solid phase comprises a microparticle), as described, e.g., in U.S. Pat. Nos. 5,089,424 and 5,006,309, and as commercially marketed, e.g., by Abbott Laboratories (Abbott Park, IL) as
  • Some of the differences between an automated or semi-automated system as compared to a non-automated system include the substrate to which the first specific binding partner (e.g., analyte antibody or capture antibody) is attached (which can affect sandwich formation and analyte reactivity), and the length and timing of the capture, detection, and/or any optional wash steps.
  • the first specific binding partner e.g., analyte antibody or capture antibody
  • an automated or semi-automated format e.g., ARCHITECT® and any successor platform, Abbott Laboratories
  • an automated or semi-automated format may have a relatively shorter incubation time (e.g., approximately 18 minutes for ARCHITECT®).
  • an automated or semi-automated format e.g., ARCHITECT® and any successor platform
  • a relatively shorter incubation time e.g., approximately 4 minutes for the ARCHITECT® and any successor platform.
  • the assays, kits, and kit components can be employed in other formats, for example, on electrochemical or other hand-held or point-of-care assay systems.
  • the present disclosure is, for example, applicable to the commercial Abbott Point of Care (i-STAT®, Abbott Laboratories) electrochemical immunoassay system that performs sandwich immunoassays.
  • Immunosensors and their methods of manufacture and operation in single-use test devices are described, for example in, U.S. Pat. No. 5,063,081, U.S. Pat. App. Pub. No. 2003/0170881, U.S. Pat. App. Pub. No. 2004/0018577, U.S. Pat. App. Pub. No. 2005/0054078, and U.S. Pat. App. Pub. No. 2006/0160164, which are incorporated in their entireties by reference for their teachings regarding same.
  • a microfabricated silicon chip is manufactured with a pair of gold amperometric working electrodes and a silver-silver chloride reference electrode. On one of the working electrodes, polystyrene beads (0.2 mm diameter) with immobilized capture antibody are adhered to a polymer coating of patterned polyvinyl alcohol over the electrode. This chip is assembled into an I-STAT® cartridge with a fluidics format suitable for
  • an aqueous reagent that includes p-aminophenol phosphate.
  • a sample suspected of containing scMet is added to the holding chamber of the test cartridge and the cartridge is inserted into the I-STAT® reader.
  • a pump element within the cartridge forces the sample into a conduit containing the chip.
  • fluid is forced out of the pouch and into the conduit to wash the sample off the chip and into a waste chamber.
  • the alkaline phosphatase label reacts with p-aminophenol phosphate to cleave the phosphate group and permit the liberated p-aminophenol to be electrochemically oxidized at the working electrode.
  • the reader is able to calculate the amount of analyte scMet in the sample by means of an embedded algorithm and factory-determined calibration curve.
  • kits as described herein necessarily encompass other reagents and methods for carrying out the immunoassay.
  • various buffers such as are known in the art and/or which can be readily prepared or optimized to be employed, e.g., for washing, as a conjugate diluent, and/or as a calibrator diluent.
  • An exemplary conjugate diluent is ARCHITECT® conjugate diluent employed in certain kits (Abbott Laboratories, Abbott Park, IL) and containing 2-(N-morpholino)ethanesulfonic acid (MES), a salt, a protein blocker, an antimicrobial agent, and a detergent.
  • MES 2-(N-morpholino)ethanesulfonic acid
  • An exemplary calibrator diluent is
  • ARCHITECT® human calibrator diluent employed in certain kits (Abbott Laboratories, Abbott Park, IL), which comprises a buffer containing MES, other salt, a protein blocker, and an antimicrobial agent. Additionally, as described in U.S. Pat. Application No. 61/142,048 filed December 31, 2008, improved signal generation may be obtained, e.g., in an I-STAT® cartridge format, using a nucleic acid sequence linked to the signal antibody as a signal amplifier. [00161] While certain embodiments herein are advantageous when employed to assess disease, such as liver disease or cancer, the assays and kits also optionally can be employed to assess scMet in other diseases, disorders, and conditions as appropriate.
  • the method of assay also can be used to identify a compound that ameliorates diseases, such as liver disease or cancer.
  • a cell that expresses scMet can be contacted with a candidate compound.
  • the level of expression of scMet in the cell contacted with the compound can be compared to that in a control cell using the method of assay described herein.
  • the ARCHITECT® sandwich immunoassay was used to determine all possible combination of antibody pairs of scMet antibodies (see Table 2). Briefly, as shown in FIG. 1, an antibody coated on a microparticle captures the analyte of interest, then a second antibody conjugated to acridinium binds to a second epitope on the analyte, then a separation of the particles from the label and subsequent read is performed. The antibody pairs of scMet antibodies were tested against soluble cMet (scMet) and HGF antigens using antibody arrays (see e.g. FIG. 2) to determine which antibodies could pair together as potential microparticles and/or conjugates.
  • the binding affinity (Kd) was determined for each of the individual antibodies against the cMet antigen using fluorescent resonance energy transfer (FRET) according to the method outlined in: Q. Ruan, J.P. Skinner, and S.Y. Tetin, Using nonfluorescent Forster resonance energy transfer acceptors in protein binding studies.
  • FRET fluorescent resonance energy transfer
  • the antibody array work and the FRET analyses were utilized to choose the best antibodies pairs for initial assay prototyping on the ARCHITECT®, where the candidate antibodies were coated on magnetic microparticles or labeled with acridinium as conjugates and tested in a sandwich against the cMet antigen.
  • the highest affinity antibodies have the lowest Kd versus the cMet antigen.
  • anti-scMet monoclonal antibody candidates were coated on microparticles or conjugated to acridinium for further evaluation on the ARCHITECT® instrument.
  • Combinations of antibodies were assessed for their ability to bind and produce a signal on the ARCHITECT instrument using recombinant human scMet antigen ("cMet (25-932)-HIEGRMD-6His"; SEQ ID NO:33), which includes the soluble part of cMet, i.e., amino acids 25-932 of cMet, an HIEGRMD linker (SEQ ID NO:36) and a 6-histidine tag (SEQ ID NO: 37) as the antigen.
  • cMet 25-932
  • HIEGRMD linker SEQ ID NO:36
  • 6-histidine tag SEQ ID NO: 37
  • Table 3 shows the antibody pairs (see bolded) that were tested on the ARCHITECT® as either microparticles (uP) or conjugates (e.g., uP coated with mAb 11E1 was tested separately with mAb 5D5, mAb 223C4 and mAb 224D10, each conjugated to acridinium, in a sandwich fashion against the scMet antigen). There were a total of 12 combinations for the scMet assay on the ARCHITECT®. mAbs were coated onto two types of magnetic microparticles (uP) (Dynal and Polymer Labs).
  • scMet (SEQ ID NO:34) was cloned into a pHybE vector generating an expression vector (SEQ ID NO:35) that was expressed in HEK293 6E cell lines.
  • SEQ ID NO:35 an expression vector that was expressed in HEK293 6E cell lines.
  • the best uP was selected based on cMet Calibrator A calibration uses ⁇ see e.g., Table 4) and accelerated stability (37°C x 7 days).
  • Table 4 shows the background of the microparticle/conjugate pair of 11E1/223C4 with Calibrator A (scMet concentration of 0 ng/mL) using Dynal and Polymer Lab microparticles.
  • the Dynal microparticles were better than the Polymer Labs because of the lower initial background when testing potential formats. All of the formats had lower background with the Dynal microparticles.
  • the robust pairs of microparticle/conjugate were uP/conjugate: 11E1/223C4 and 223C4/11E1.
  • the final microparticle/conjugate combination of anti-cMet 11E1 antibody/anti- cMet 224D10 F(ab') 2 fragment antibody was used and examined for the ARCHITECT® scMet Assay ("ARCHITECT® scMet B Assay").
  • the anti-scMet 224D10 antibody was used as an F(ab') 2 fragment to avoid aggregation and to avoid interference from rheumatoid factor (RF), HAMA, and heterophilic interactions.
  • the ARCHITECT® scMet B Assay demonstrated equimolarity (the ability to measure the same amount of scMet) in the presence of Hepatocyte growth factor (HGF) and mouse anticreatine kinase (MAK33), an irrelevant antibody (see FIG. 5).
  • HGF Hepatocyte growth factor
  • MAK33 mouse anticreatine kinase
  • Table 5 shows the results of the ARCHITECT® scMet B Assay ("ARCHITECT® soluble cMet B") compared to R&D SYSTEMS® cMet ELISA (Human HGF R/c-MET, Catalog Number DY358).
  • the time to the first test result using the ARCHITECT® scMet B Assay was 20 min and the throughput was 200 tests per hour.
  • scMet B Sample The stability of scMet B Sample was determined using recombinant human scMet antigen (amino acids 25-932) ("scMet B Sample”; SEQ ID NO:33) for calibrators and controls. Six matched serum and EDTA plasma samples were drawn and processed. Samples were tested in triplicate, per the storage conditions below, with endogenous and spiked levels at
  • FIG.6 shows a standard curve of the ARCHITECT® scMet B Assay where the circle on the graph represents a range of current samples tested. 6 matched serum and EDTA plasma samples were drawn and processed. Samples were tested per the storage conditions in Table 6 with endogenous and spiked levels.
  • the scMet B Samples were stable at room temperature for 24 hrs, 2-8°C for 72 hrs, and -80°C for 12 days.
  • FIGS. 7A-7B show the stability of scMet B Samples at room temperature at 4 and 24 hrs.
  • FIG. 7A shows the endogenous sample stability at room temperature at 4 and 24 hrs.
  • FIG. 7B shows the spiked sample (100 ng) stability at room temperature at 4 and 24 hrs. The average difference between base line and room temperature at 24 hr was 4.4%.
  • FIGS. 8A-8B show the stability of scMet B Samples at 2-8°C at 24 and 72 hrs.
  • FIG. 8A shows the endogenous sample stability at 2-8°C at 24 and 72 hrs.
  • FIG. 8B shows the spiked sample (100 ng) stability at 2-8°C at 24 and 72 hrs. The average difference between base line and 2-8°C at 72 hr was -1.2%.
  • FIGS. 9A-9B show the stability of scMet B Samples at -80°C at 24 and 72 hrs and day 12 and day 12 with 3 freeze/thaw cycles.
  • FIG. 8A shows the stability of scMet B Samples at -80°C at 24 and 72 hrs and day 12 and day 12 with 3 freeze/thaw cycles.
  • FIG. 9A shows the endogenous sample stability at - 80°C at 24 and 72 hrs and day 12 and day 12 with 3 freeze/thaw cycles.
  • FIG. 9B shows the spiked sample (100 ng) stability at -80°C at 24 and 72 hrs and day 12 and day 12 with 3 freeze/thaw cycles.
  • the average difference between base line and -80°C at 12 days was -0.6%.
  • LOD testing was performed on three days with two runs per day on two instruments each day. Six runs were performed on each of two ARCHITECT® instruments for a total of twelve runs. The LOD was determined using twelve reps of Calibrator A (0 ng/mL) and four reps of Calibrator B (6.25 ng/mL) per run. The calculation for LOD was as follows:
  • LOQ testing was performed on three days with two runs per day on two instruments each day. Eight reps per run of each sample were used to calculate the LOQ.
  • the Cal B (6.25 ng/mL) was diluted 1 :2 in serum to 3.13 ng/mL, 1.56 ng/mL, 0.78 ng/mL and 0.39 ng/mL for the LOQ samples.
  • Table 7 shows the limit of detection and limit of quantification of the ARCHITECT® scMet B Assay.
  • the average LOD was 0.191 ng/mL and the average LOQ was 3.13 ng/mL at 30% total error.
  • the precision study duration was five days with two runs per day, with five reps per run each on two instruments of the low (12.5 ng/mL), medium (50 ng/mL) and high (200 ng/mL) scMet concentration controls.
  • the overall precision (%CV) was calculated based on the mean and standard deviation of each sample tested in each run based on Clinical Laboratory Standards Institute (CLSI) guidelines found in EP5-A2 at the URL of www.CLSI.org. Table 8 shows the 5 day precision using the ARCHITECT® scMet B Assay.
  • the total percent coefficient of variation (CV) for the Low to High controls was less than or equal to 10% except for the high control on one instrument which had about 12%.
  • soluble cMet Known amounts of soluble cMet were spiked into five serum specimens at 50 and 400 ng/mL. The soluble cMet was then measured in the serum control and the two spike samples.
  • the ARCHITECT® scMet B Assay used an initial 18 minute incubation time with 16 ⁇ ⁇ sample (SMP), 50 of microparticles (MP), 32 of the analyte specific diluent (ASD). The automated dilution of 1 : 1.5 was used for the specimens. After the first wash, 50 of conjugate was added and washed. The RLUs were then read. The percent recovery was calculated for each spiked serum sample based on the control serum without a spike. The range and the mean percent recovery for the five specimens are reported (Table 9).
  • Table 9 shows the spike recovery using the ARCHITECT® scMet B Assay.
  • Proteins that may interact with cMet such as RON, HGF-A, HAI-1 , FKBP12, angiostatin, and plasminogen, were tested at twice the levels typically found in human serum to test for cross-reactivity. Table 12 shows that there was less than 0.05% cross-reactivity for all of the tested antigens using the ARCHITECT® scMet B Assay.
  • HAMA human anti-mouse antibodies
  • RF rheumatoid factor
  • Table 13 shows the results of the ARCHITECT® scMet B Assay for ten HAMA specimens, ten RF specimens, and ten heterophilic specimens. The percentage difference was calculated without and with Mouse IgG at 0.15 mg/mL. Table 13 shows the results of the HAMA, RF, and Heterophilic Specimens. The assay goal of attaining a mean difference of less than or equal to about 10% was accomplished by the ARCHITECT® scMet B Assay.
  • Sample carry over is performed to determine if any cMet is carried over on the sample pipettor from a high sample of 1600 ng/mL to the next sample of Calibrator A ("Cal A"; 0 ng/mL) by comparing the detection of Cal A before and after the high sample.
  • Table 14 shows that sample carryover of the ARCHITECT® scMet B Assay is not detectable at 1600 ng/mL.
  • FIG. 10 shows the levels of scMet in human patients with various diseases as measured by the ARCHITECT® scMet B Assay.
  • Table 16 shows the number of normal, breast cancer, colon cancer, prostate cancer, gastric cancer, esophageal cancer, liver fibrosis, liver cirrhosis, liver non-viral disease, and liver cancer specimens used in FIG. 10.
  • Table 17 shows the AUROC univariate summary where the results in bold have AUROC greater than 0.80.
  • the ARCHITECT® scMet B Assay differentiated between normal samples and prostate cancer (see FIG. 11 and Table 18); normal samples and breast cancer (see FIG. 12 and Table 19); normal samples and gastric cancer (see FIG. 13 and Table 20), normal samples and liver fibrosis (Table 21), normal samples and colon cancer (Table 22), and normal samples and liver cancer (Table 23).
  • Table 18 Prostate Cancer vs Normal AUROC
  • the ARCHITECT® scMet A Assay measures scMet concentration using the 224G11 antibody as the capture antibody and an 11E1 antibody as the detection antibody, each of which binds to cMet.
  • the 224G11 antibody is an antibody produced by hybridoma cell line deposited at the CNCM, Institut Pasteur, Paris, on March 14, 2007 under the number 1-3731.
  • the 11E1 antibody is an antibody produced by hybridoma cell line deposited at the CNCM, Institut Pasteur, Paris, on March 14, 2007 under the number 1-3724.
  • the 224G11 antibody binds an epitope different from the 11E1 and 224D10 antibodies, hence different amounts of soluble cMet may be detected using the ARCHITECT® scMet A Assay compared to the ARCHITECT® scMet B Assay.
  • the ARCHITECT® HGF Assay measures HGF concentration using a 64.4.1 antibody as the capture antibody and an H98C4 antibody as the detection antibody, each of which binds to HGF.
  • the 64.4.1 antibody is an antibody produced by hybridoma cell line that will be deposited with the American Type Culture Collection (A.T.C.C.), 10801 University Boulevard. , Manassas, VA 20110-2209 under the terms of the Budapest Treaty.
  • the H98C4 antibody is an antibody produced by hybridoma cell line that will be deposited with the A.T.C.C., 10801 University Boulevard., Manassas, VA 20110-2209 under the terms of the Budapest Treaty.
  • Table 21 Data from the Effects Likelihood Ratio test within Fit Model in JMP Version 9.0.0 are shown in Table 21 for the ARCHITECT® scMet A Assay, ARCHITECT® scMet B Assay and ARCHITECT® HGF Assay.
  • Table 21 shows the multivariate summary where the p-values shown in bold represent the biomarkers in the panel that contribute significantly (i.e., less than 0.05) to the discrimination between normal healthy individuals and individuals with cancer.
  • the multivariate analysis showed multiple markers were significant in the model for breast cancer, colon cancer, prostate cancer, gastric cancer, esophageal cancer, liver fibrosis, and liver cancer.
  • the panel of these three biomarkers had greater discrimination than one biomarker by itself.
  • Table 24 AUROC Multivariate Summary

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Immunology (AREA)
  • Engineering & Computer Science (AREA)
  • Oncology (AREA)
  • Chemical & Material Sciences (AREA)
  • Biomedical Technology (AREA)
  • Urology & Nephrology (AREA)
  • Molecular Biology (AREA)
  • Hematology (AREA)
  • Microbiology (AREA)
  • Cell Biology (AREA)
  • Biotechnology (AREA)
  • Hospice & Palliative Care (AREA)
  • Food Science & Technology (AREA)
  • Medicinal Chemistry (AREA)
  • Physics & Mathematics (AREA)
  • Analytical Chemistry (AREA)
  • Biochemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • General Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Peptides Or Proteins (AREA)

Abstract

La présente invention concerne des procédés d'utilisation d'anticorps pour détecter cMet soluble (scMet) dans un échantillon. Les procédés peuvent être utilisés pour diagnostiquer une maladie ou un cancer hépatiques chez un patient par détection de la présence et/ou de la quantité de scMet dans un échantillon du patient en utilisant des anticorps anti-cMet. Les procédés peuvent être utilisés pour mettre au point un pronostic précis pour un patient atteint d'une maladie ou d'un cancer hépatiques. Les procédés peuvent être utilisés pour identifier et/ou classifier un patient en tant que candidat pour une thérapie contre une maladie hépatique et/ou pour une thérapie contre un cancer du foie.
PCT/US2014/053181 2013-08-28 2014-08-28 Dosage de cmet soluble WO2015031626A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201361871184P 2013-08-28 2013-08-28
US61/871,184 2013-08-28

Publications (1)

Publication Number Publication Date
WO2015031626A1 true WO2015031626A1 (fr) 2015-03-05

Family

ID=51542462

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2014/053181 WO2015031626A1 (fr) 2013-08-28 2014-08-28 Dosage de cmet soluble

Country Status (1)

Country Link
WO (1) WO2015031626A1 (fr)

Citations (29)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5006309A (en) 1988-04-22 1991-04-09 Abbott Laboratories Immunoassay device with liquid transfer between wells by washing
US5063081A (en) 1988-11-14 1991-11-05 I-Stat Corporation Method of manufacturing a plurality of uniform microfabricated sensing devices having an immobilized ligand receptor
US5089424A (en) 1988-06-14 1992-02-18 Abbott Laboratories Method and apparatus for heterogeneous chemiluminescence assay
US5241070A (en) 1988-09-26 1993-08-31 Ciba Corning Diagnostics Corp. Nucleophilic polysubstituted aryl acridinium esters and uses thereof
US5294404A (en) 1991-06-03 1994-03-15 Abbott Laboratories Reagent pack for immunoassays
US5352803A (en) 1992-03-30 1994-10-04 Abbott Laboratories 5(6)-methyl substituted fluorescein derivatives
US5468646A (en) 1986-10-22 1995-11-21 Abbott Laboratories Chemiluminescent acridinium salts
US5480792A (en) 1990-09-14 1996-01-02 Biosite Diagnostics, Inc. Antibodies to complexes of ligand receptors and ligands and their utility in ligand-receptor assays
US5525524A (en) 1991-04-10 1996-06-11 Biosite Diagnostics, Inc. Crosstalk inhibitors and their uses
US5593896A (en) 1992-03-30 1997-01-14 Abbott Laboratories Reagents and methods for the detection and quantification of thyroxine in fluid samples
US5679526A (en) 1989-01-10 1997-10-21 Biosite Diagnostics Incorporated Threshold ligand-receptor assay
US5824799A (en) 1993-09-24 1998-10-20 Biosite Diagnostics Incorporated Hybrid phthalocyanine derivatives and their uses
US5851776A (en) 1991-04-12 1998-12-22 Biosite Diagnostics, Inc. Conjugates and assays for simultaneous detection of multiple ligands
US5885527A (en) 1992-05-21 1999-03-23 Biosite Diagnostics, Inc. Diagnostic devices and apparatus for the controlled movement of reagents without membrances
US5922615A (en) 1990-03-12 1999-07-13 Biosite Diagnostics Incorporated Assay devices comprising a porous capture membrane in fluid-withdrawing contact with a nonabsorbent capillary network
US5939272A (en) 1989-01-10 1999-08-17 Biosite Diagnostics Incorporated Non-competitive threshold ligand-receptor assays
US5947124A (en) 1997-03-11 1999-09-07 Biosite Diagnostics Incorporated Diagnostic for determining the time of a heart attack
US6113855A (en) 1996-11-15 2000-09-05 Biosite Diagnostics, Inc. Devices comprising multiple capillarity inducing surfaces
US6143576A (en) 1992-05-21 2000-11-07 Biosite Diagnostics, Inc. Non-porous diagnostic devices for the controlled movement of reagents
WO2001058956A2 (fr) 2000-02-10 2001-08-16 Abbott Laboratories Anticorps de liaison de l'interleukine 18 humaine et procedes de preparation et d'utilisation
US20030170881A1 (en) 2002-03-05 2003-09-11 I-Stat Corporation Apparatus and methods for analyte measurement and immuno assay
US20040018577A1 (en) 2002-07-29 2004-01-29 Emerson Campbell John Lewis Multiple hybrid immunoassay
US20050054078A1 (en) 2003-09-10 2005-03-10 Miller Cary James Immunoassay device with improved sample closure
US20060160164A1 (en) 2003-09-10 2006-07-20 Miller Cary J Immunoassay device with immuno-reference electrode
WO2009007427A2 (fr) 2007-07-12 2009-01-15 Pierre Fabre Medicament Nouveaux anticorps inhibant la dimérisation de c-met et utilisations de ceux-ci
EP2143441A1 (fr) * 2008-07-08 2010-01-13 Pierre Fabre Medicament Combinaison d'antagoniste c-MET et composé d'aminihétéroaryl pour le traitement du cancer
EP2287197A1 (fr) * 2009-08-21 2011-02-23 Pierre Fabre Medicament Anticorps anti-cMET et son utilisation pour la détection et le diagnostic du cancer
US20110097262A1 (en) * 2008-12-02 2011-04-28 Liliane Goetsch NOVEL ANTI-cMET ANTIBODY
WO2012031027A1 (fr) * 2010-08-31 2012-03-08 Genentech, Inc. Biomarqueurs et procédés de traitement

Patent Citations (37)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5468646A (en) 1986-10-22 1995-11-21 Abbott Laboratories Chemiluminescent acridinium salts
US5783699A (en) 1986-10-22 1998-07-21 Abbott Laboratories Chemiluminescent acridinium salts
US5543524A (en) 1986-10-22 1996-08-06 Abbott Laboratories Chemiluminescent acridinium salts
US5006309A (en) 1988-04-22 1991-04-09 Abbott Laboratories Immunoassay device with liquid transfer between wells by washing
US5089424A (en) 1988-06-14 1992-02-18 Abbott Laboratories Method and apparatus for heterogeneous chemiluminescence assay
US5241070A (en) 1988-09-26 1993-08-31 Ciba Corning Diagnostics Corp. Nucleophilic polysubstituted aryl acridinium esters and uses thereof
US5063081A (en) 1988-11-14 1991-11-05 I-Stat Corporation Method of manufacturing a plurality of uniform microfabricated sensing devices having an immobilized ligand receptor
US5679526A (en) 1989-01-10 1997-10-21 Biosite Diagnostics Incorporated Threshold ligand-receptor assay
US5939272A (en) 1989-01-10 1999-08-17 Biosite Diagnostics Incorporated Non-competitive threshold ligand-receptor assays
US5922615A (en) 1990-03-12 1999-07-13 Biosite Diagnostics Incorporated Assay devices comprising a porous capture membrane in fluid-withdrawing contact with a nonabsorbent capillary network
US5985579A (en) 1990-09-14 1999-11-16 Biosite Diagnostics, Inc. Antibodies to complexes of ligand receptors and ligands and their utility in ligand-receptor assays
US5480792A (en) 1990-09-14 1996-01-02 Biosite Diagnostics, Inc. Antibodies to complexes of ligand receptors and ligands and their utility in ligand-receptor assays
US5525524A (en) 1991-04-10 1996-06-11 Biosite Diagnostics, Inc. Crosstalk inhibitors and their uses
US5851776A (en) 1991-04-12 1998-12-22 Biosite Diagnostics, Inc. Conjugates and assays for simultaneous detection of multiple ligands
US5294404A (en) 1991-06-03 1994-03-15 Abbott Laboratories Reagent pack for immunoassays
US5593896A (en) 1992-03-30 1997-01-14 Abbott Laboratories Reagents and methods for the detection and quantification of thyroxine in fluid samples
US5573904A (en) 1992-03-30 1996-11-12 Abbott Laboratories 5(6)-Methyl substituted fluorescein derivatives
US5496925A (en) 1992-03-30 1996-03-05 Abbott Laboratories 5(6)-methyl substituted fluorescein derivatives
US5359093A (en) 1992-03-30 1994-10-25 Abbott Laboratories Reagents and methods for the detection and quantification of thyroxine in fluid samples
US5352803A (en) 1992-03-30 1994-10-04 Abbott Laboratories 5(6)-methyl substituted fluorescein derivatives
US5885527A (en) 1992-05-21 1999-03-23 Biosite Diagnostics, Inc. Diagnostic devices and apparatus for the controlled movement of reagents without membrances
US6019944A (en) 1992-05-21 2000-02-01 Biosite Diagnostics, Inc. Diagnostic devices and apparatus for the controlled movement of reagents without membranes
US6143576A (en) 1992-05-21 2000-11-07 Biosite Diagnostics, Inc. Non-porous diagnostic devices for the controlled movement of reagents
US5824799A (en) 1993-09-24 1998-10-20 Biosite Diagnostics Incorporated Hybrid phthalocyanine derivatives and their uses
US6113855A (en) 1996-11-15 2000-09-05 Biosite Diagnostics, Inc. Devices comprising multiple capillarity inducing surfaces
US5947124A (en) 1997-03-11 1999-09-07 Biosite Diagnostics Incorporated Diagnostic for determining the time of a heart attack
WO2001058956A2 (fr) 2000-02-10 2001-08-16 Abbott Laboratories Anticorps de liaison de l'interleukine 18 humaine et procedes de preparation et d'utilisation
US20030170881A1 (en) 2002-03-05 2003-09-11 I-Stat Corporation Apparatus and methods for analyte measurement and immuno assay
US20040018577A1 (en) 2002-07-29 2004-01-29 Emerson Campbell John Lewis Multiple hybrid immunoassay
US20050054078A1 (en) 2003-09-10 2005-03-10 Miller Cary James Immunoassay device with improved sample closure
US20060160164A1 (en) 2003-09-10 2006-07-20 Miller Cary J Immunoassay device with immuno-reference electrode
WO2009007427A2 (fr) 2007-07-12 2009-01-15 Pierre Fabre Medicament Nouveaux anticorps inhibant la dimérisation de c-met et utilisations de ceux-ci
EP2143441A1 (fr) * 2008-07-08 2010-01-13 Pierre Fabre Medicament Combinaison d'antagoniste c-MET et composé d'aminihétéroaryl pour le traitement du cancer
US20110097262A1 (en) * 2008-12-02 2011-04-28 Liliane Goetsch NOVEL ANTI-cMET ANTIBODY
EP2287197A1 (fr) * 2009-08-21 2011-02-23 Pierre Fabre Medicament Anticorps anti-cMET et son utilisation pour la détection et le diagnostic du cancer
US20120149031A1 (en) 2009-08-21 2012-06-14 Liliane Goetsch Anti-cmet antibody and its use for the detection and the diagnosis of cancer
WO2012031027A1 (fr) * 2010-08-31 2012-03-08 Genentech, Inc. Biomarqueurs et procédés de traitement

Non-Patent Citations (21)

* Cited by examiner, † Cited by third party
Title
ABBOTT DIAGNOSTICS: "Architect i100SR", XP002734296, Retrieved from the Internet <URL:https://www.abbottdiagnostics.com/en-us/products/ARCHITECT-i1000SR.html#test-menu> [retrieved on 20150112] *
ADAMCZYK ET AL., ANAL. CHIM. ACTA, vol. 579, no. 1, 2006, pages 61 - 67
ADAMCZYK ET AL., BIOCONJUGATE CHEM., vol. 11, 2000, pages 714 - 724
ADAMCZYK ET AL., BIOORG. MED. CHEM. LETT., vol. 16, 2006, pages 1324 - 1328
ADAMCZYK ET AL., BIOORG. MED. CHEM. LETT., vol. 4, 2004, pages 2313 - 2317
ADAMCZYK ET AL., BIORG. MED. CHEM. LETT., vol. 14, 2004, pages 3917 - 3921
ADAMCZYK ET AL., J. ORG. CHEM., vol. 63, 1998, pages 5636 - 5639
ADAMCZYK ET AL., ORG. LETT., vol. 1, 1999, pages 779 - 781
ADAMCZYK ET AL., ORG. LETT., vol. 5, 2003, pages 3779 - 3782
ADAMCZYK ET AL., TETRAHEDRON, vol. 55, 1999, pages 10899 - 10914
HAUGLAND: "Handbook offluorescent Probes and Research Chemicals", 1996, MOLECULAR PROBES, INC.
KAMIYA: "Human c-Met ELISA For the quantitative determination of c-Met in human serum, EDTA-plasma or cell culture media", 20080201, no. KT-444, 1 February 2008 (2008-02-01), pages 1 - 6, XP007919726 *
MATTINGLY ET AL.: "Luminescence Biotechnology: Instruments and Applications", 2002, CRC PRESS, pages: 77 - 105
MATTINGLY, J., BIOLUMIN. CHEMILUMIN., vol. 6, 1991, pages 107 - 114
MCCAPRA ET AL., PHOTOCHEM. PHOTOBIOL., vol. 4, 1965, pages 1111 - 21
POLAK; VAN NOORDEN: "Introduction to Immunocytochemistry", 1997, SPRINGER VERLAG
Q. RUAN; J.P. SKINNER; S.Y. TETIN: "Using nonfluorescent Forster resonance energy transfer acceptors in protein binding studies", ANAL. BIOCHEM., vol. 393, 2009, pages 196 - 204, XP026468113, DOI: doi:10.1016/j.ab.2009.06.033
RAZAVI ET AL., LUMINESCENCE, vol. 15, 2000, pages 239 - 244
RAZAVI ET AL., LUMINESCENCE, vol. 15, 2000, pages 245 - 249
WELLS: "Methods and Automation Strategies", 2003, ELSEVIER, article "High Throughput Bioanalytical Sample Preparation"
WU, C. ET AL., NATURE BIOTECHNOLOGY, vol. 25, no. 11, 2007, pages 1290 - 1297

Similar Documents

Publication Publication Date Title
US11421023B2 (en) Anti-GP73 monoclonal antibodies and methods of obtaining the same
JP7269183B2 (ja) 心臓トロポニンiを使用する、ヒト対象における軽度外傷性脳損傷を診断及び査定する一助となるための方法
EP3721234A1 (fr) Procédés d&#39;aide au diagnostic et à l&#39;évaluation d&#39;un lésion cérébrale traumatique chez un sujet humain au moyen d&#39;une combinaison de gfap et d&#39;uch-l1
EP3721233A2 (fr) Procédés d&#39;aide au diagnostic et à l&#39;évaluation d&#39;un sujet qui a subi une lésion orthopédique et qui a subi ou peut avoir subi une lésion à la tête, telle qu&#39;une lésion cérébrale traumatique légère (tbi), à l&#39;aide d&#39;une protéine acide fibrillaire gliale (gfap) et/ou d&#39;hydrolase carboxy-terminale d&#39;ubiquitine l1 (uch-l1)
JP7174385B2 (ja) 膵臓がんを診断するためのラミニン2の使用
JP6328103B2 (ja) がんの予後判定および診断方法
JP6940505B2 (ja) 組成物および癌の発生リスクの評価方法
EP3183578A1 (fr) Méthodes de détection précoce du cancer colorectal
WO2015031626A1 (fr) Dosage de cmet soluble
WO2015031614A1 (fr) Dosage de la cmet soluble
US11079395B2 (en) Methods for predicting major adverse cardiovascular events in subjects with coronary artery disease
WO2015031578A1 (fr) Dosage de hgf
AU2022279156A9 (en) Methods of evaluating brain injury in a pediatric subject
JP2024045038A (ja) 胃癌の高度リンパ節転移を検出するためのバイオマーカー、それを用いた検出方法及び検出試薬
AU2022423989A1 (en) Use of biomarkers to determine sub-acute traumatic brain injury (tbi) in a subject having received a head computerized tomography (ct) scan that is negative for a tbi or no head ct scan
WO2023129942A1 (fr) Utilisation de biomarqueurs pour déterminer une lésion cérébrale traumatique (tbi) subaiguë chez un sujet ayant subi un tomodensitogramme (ct) de la tête négatif pour une tbi ou n&#39;ayant pas subi de tomodensitogramme

Legal Events

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

Ref document number: 14766593

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 14766593

Country of ref document: EP

Kind code of ref document: A1