US20070161064A1 - Antibodies as a cancer diagnostic - Google Patents

Antibodies as a cancer diagnostic Download PDF

Info

Publication number
US20070161064A1
US20070161064A1 US11/716,792 US71679207A US2007161064A1 US 20070161064 A1 US20070161064 A1 US 20070161064A1 US 71679207 A US71679207 A US 71679207A US 2007161064 A1 US2007161064 A1 US 2007161064A1
Authority
US
United States
Prior art keywords
epha2
cells
antibody
cell population
detecting
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US11/716,792
Inventor
Michael Kinch
Nicole Zantek
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Purdue Research Foundation
Original Assignee
Purdue Research Foundation
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 Purdue Research Foundation filed Critical Purdue Research Foundation
Priority to US11/716,792 priority Critical patent/US20070161064A1/en
Publication of US20070161064A1 publication Critical patent/US20070161064A1/en
Priority to US13/210,672 priority patent/US20120122112A1/en
Abandoned legal-status Critical Current

Links

Images

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
    • G01N33/57484Immunoassay; Biospecific binding assay; Materials therefor for cancer involving compounds serving as markers for tumor, cancer, neoplasia, e.g. cellular determinants, receptors, heat shock/stress proteins, A-protein, oligosaccharides, metabolites
    • G01N33/57496Immunoassay; Biospecific binding assay; Materials therefor for cancer involving compounds serving as markers for tumor, cancer, neoplasia, e.g. cellular determinants, receptors, heat shock/stress proteins, A-protein, oligosaccharides, metabolites involving intracellular compounds
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • C12Q1/6886Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material for cancer
    • 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
    • G01N33/57407Specifically defined cancers
    • 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
    • G01N33/57407Specifically defined cancers
    • G01N33/57415Specifically defined cancers of breast
    • 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
    • G01N33/57407Specifically defined cancers
    • G01N33/57419Specifically defined cancers of colon
    • 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
    • G01N33/57407Specifically defined cancers
    • G01N33/57423Specifically defined cancers of lung
    • 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
    • G01N33/57407Specifically defined cancers
    • G01N33/57434Specifically defined cancers of prostate
    • 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
    • G01N33/57407Specifically defined cancers
    • G01N33/57438Specifically defined cancers of liver, pancreas or kidney
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/112Disease subtyping, staging or classification
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/158Expression markers
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S436/00Chemistry: analytical and immunological testing
    • Y10S436/811Test for named disease, body condition or organ function
    • Y10S436/813Cancer

Definitions

  • the present invention relates to diagnosis of metastatic disease. More particularly, this invention relates to reagents that can detect a specific epithelial cell tyrosine kinase that is overexpressed in metastatic tumor cells. Most particularly, this invention relates to reagents which bond to an intracellular epitope of the epithelial cell tyrosine kinase, and the use of these reagents for cancer diagnosis.
  • Cancer cell metastasis requires cellular capacity to 1) detach from a primary tumor, 2) migrate and invade through local tissues, 3) translocate to distant sites in the body (via lymph or blood), 4) colonize a foreign site, and 5) grow and survive in this foreign environment. All of these behaviors are linked to cell adhesions.
  • Cell adhesions control the physical interactions of cells with their microenvironment. Cell adhesions also initiate signals that dictate tumor cell growth, death, and differentiation.
  • Various cancer cells including breast cancer cells, are known to exhibit altered cell adhesion. As compared to normal breast epithelia, transformed human breast epithelial cells have decreased cell-cell contacts and increased interactions with the surrounding extracellular matrix. These changes facilitate increased detachment and migration of cancer cells away from cell colonies and are directly linked with alteration in tyrosine phosphorylation of cell membrane proteins. Tyrosine phosphorylation is a potent form of cell signal transduction, and alteration in levels of tyrosine phosphorylation is believed to be important for tumor cell invasiveness. Thus, regulation of tyrosine phosphorylation represents a promising target for therapeutic intervention against metastatic cancer. Tyrosine phosphorylation is controlled by cell membrane tyrosine kinases, and increased expression of tyrosine kinases is known to occur in metastatic cancer cells.
  • EphA2 A member of the Eph family of tyrosine kinases known as Eplirins, EphA2 is a transmembrane receptor tyrosinie kinase with a cell-bound ligand.
  • EphA2 an improved method for generating a panel of monoclonal antibodies specific for tyrosine phosphorylated proteins has been developed. Using this method, a multiplicity of EphA2 recognizing monoclonal antibodies has been generated. These antibodies have been used to show that EphA2 is overexpressed in metastatic breast, lung, colon, and prostate cells. Because EphA2 is expressed differently in normal and metastatic cells, EphA2-specific antibodies are useful in the diagnosis of metastatic disease. Antibodies produced by one particular hybridoma recognize an intracellular epitope of EphA2 and have been shown to be highly specific in binding to EphA2.
  • one aspect of this invention is a compound which specifically binds to an intracellular epitope of EphA2.
  • the compound is an antibody specific for a domain of the EphA2 protein.
  • natural or artificial ligands, peptides, anti-sense, ATP analogies, or other small molecules capable of specifically targeting EphA2 may be employed.
  • a second aspect of this invention is a method for generating antibodies which recognize EphA2 intracellular epitopes.
  • Another aspect of this invention is the use of EphA2-specific antibodies in the diagnosis of metastatic disease.
  • An additional aspect of this invention is a diagnostic reagent specific for detecting EphA2, any fragment thereof, or DNA or RNA coding for the EphA2 protein.
  • FIG. 1A -C showN a series of western blots showing EphA2 expression in cell lines derived from human prostate cells;
  • FIG. 1A is a western blot showing EphA2 expression in various human prostate cancer cell lines
  • FIG. 1B is a western blot showing EphA2 expression in human prostatic epithelial cell line MLC and expression in that cell line after transformation by oncogenic K-Ras or X-irradiation;
  • FIG. 1C is similar to FIG. 1B , except showing expression in human prostatic epithelial cell line 267B1 and expression in that cell line after transformation by oncogenic K-Ras or X-irradiation;
  • FIG. 2 is a western blot showing EphA2 expression in various human mammary epithelial cell lines
  • FIG. 3A -B shows EphA2 localization in the cell membranes of various mammary epithelial cell lines, as seen by immunofluorescence microscopy;
  • FIG. 3A shows EphA2 localization in sites of cell adhesion in normal MCF-10A cells.
  • FIG. 3B shows EphA2 redistribution in malignant cells.
  • Antibodies specific for EphA2 have been isolated through an improved method.
  • the method employed is designed for increased sensitivity and diversity of responding hybridomas.
  • tyrosine phosphorylated proteins from Ras-transformed human epithelial cells are isolated by affinity chromatography using existing phosphotyrosine-specific antibodies.
  • the tyrosine phosphorylated proteins are then used as an immunogen for producing monoclonal antibodies.
  • Low-dose amounts of tyrosine phosphorylated proteins are injected proximal to lymph nodes, every other day, over a ten day period (the RIMMS strategy).
  • B cells from engorged lymph nodes are then isolated and fused with a Bcl-2-overexpressing myeloma, to minimize apoptosis after fusion.
  • This method results in increased diversity, specificity, and cost-effectiveness of hybridoma production.
  • the hybridomas are first screened to identify those hybridomas producing antibodies capable of distinguishing malignant from normal cancer cells. To date, at least 450 such hybridomas have been identified.
  • Hybridomas which are specific to EphA2 have been selected. Use of the RIMMS strategy has resulted in the production of various monoclonal antibodies that specifically bind EphA2. Of the first four hybridomas characterized, two recognize independent epitopes on EphA2. The first, D7, recognizes an intracellular epitope. The second, B2D6, binds to an extracellular epitope. D7 has proven to be highly specific for an intracellular epitope of EphA2 and this specificity provides much of the current basis for diagnosis of metastatic tumors.
  • EphA2-specific antibodies of this invention may be used to detect this overexpression and, thus, to detect metastatic disease.
  • Such techiques include but are not limited to western blotting, dot blotting, precipitation, agglutination, ELISA assays, immunohistochemistry, in situ hybridization, flow cytometry on a variety of tissues or bodily fluids, and a variety of sandwich assays.
  • EphA2 antibodies specific for an intracellular epitope of EphA2
  • the cells must be lysed and incubated with the antibody.
  • the above techniques may be performed on whole-cell lysates, or EphA2 may be separated out for testing, such as by immunoprecipitation.
  • the D7 antibodies of this invention are highly specific for an intracellular epitope of EphA2 and have proven to be sensitive to differential expression of EphA2 in metastatic cells.
  • Other techniques, such as immunohistological staining require whole cells, and may further require cell layers of a particular cell density. Such tests require an antibody specific for an extracellular epitope of EphA2.
  • the antibodies of this invention may be used to detect metastatic disease in a wide variety of tissue samples. For instance, research using EphA2-specific antibodies has revealed that altered EphA2 expression occurs in breast, kidney, prostate, lung, and colon cells, and it is believed that altered EphA2 expression occurs in other types of cell metastasis, particularly epithelial malignancies. EphA2-specific antibodies may be used to detect metastasis in biopsied tumor tissue. Also, samples of a variety of body fluid samples, such as blood, plasma, spinal fluid, saliva, and urine, can be tested with the antibodies of the present invention. Altered EphA2 expression in these samples indicates the presence of metastatic disease.
  • the EphA2 of metastatic cells exhibits altered tyrosine phosphorylation.
  • EphA2 is expressed and is tyrosine phosphorylated.
  • EphA2 is overexpressed, and the EphA2 is not tyrosine phosphorylated.
  • a test quantizing EphA2 expression sometimes may lead to an ambiguous result, it may be desirable to determine tyrosine phosphorylation, as well as the magnitude of EphA2 expression.
  • a method of diagnosis using the antibodies of this invention in combination with phosphotyrosine-specific antibodies provides data for determining the state of metastatic disease.
  • EphA2-specific antibodies of this invention can be exploited to detect changes in EphA2 localization which are associated with metastasis.
  • EphA2 is enriched in within cites of cell adhesion.
  • EphA2 is diffusely distributed, and in metastatic breast cancer cells EphA2 is redistributed into the membrane ruffles.
  • Techniques such as immunohistological staining or immunofluorescent microscopy are well known in the art and may be used to visualize EphA2 distribution. See, for example, U.S. Pat. No. 5,514,554, hereby incorporated by reference.
  • EphA2 expression can be detected by using antibodies capable of detecting whole EphA2 or fragments of the EphA2 protein.
  • Other methods of detecting altered EphA2 expression include detecting DNA or RNA sequences coding for the EphA2 protein.
  • the EphA2-specific antibodies may be labeled covalently or non-covalently with any of a number of knowfn detectable labels, such fluorescent, radioactive, or enzymatic substances, as is known in the art.
  • a secondary antibody specific for the antibodies of this invention is labeled with a known detectable label and used to detect the EphA2-specific antibodies in the above techniques.
  • Preferred labels include chromogens dyes. Among the most commonly used are 3-amino-9-ethylcarbazole (AEC) and 3,3′-diaminobenzidine tetrahydroclholoride (DAB). These can be detected using light microscopy. Also preferred are fluorescent labels. Among the most commonly used fluorescent labeling compounds are fluorescein isothiocyanate, rhodamine, phycoerythrin, phycocyanin, allophycocyanin, o-phtlhaldelhyde and fluorescamine.
  • Chemiluminescent and bioluminescent compounds such as luminol, isoluminol, theromatic acridinium ester, imidazole, acridinium salt, oxalate ester, luciferin, luciferase, and aequorin also may be used.
  • the fluorescent-labeled antibody is exposed to light of the proper wavelenigtlh, its presence can be detected due to its fluorescence.
  • Radioactive isotopes which are particularly useful for labeling the antibodies of the present invention include 3 H, 125 I, 131 I, 35 S, 32 P, and 14 C.
  • the radioactive isotope can be detected by such means as the use of a gamma counter, a scintillation counter, or by autoradiography.
  • Another method in which the antibodies can be detectably labeled is by linking the antibodies to an enzyme and subsequently using the antibodies in an enzyme immunoassay (EIA) or enzyme-linked immunosorbent assay (ELISA).
  • EIA enzyme immunoassay
  • ELISA enzyme-linked immunosorbent assay
  • the enzyme when subsequently exposed to its substrate, reacts with the substrate and generates a chemical moiety which can be detected, for example, by spectrophotometric, fluorometric, or visual means.
  • Enzymes which can be used to detectably label antibodies include, but are not limited to malate dehydrogenase, staphylococcal nuclease, delta-5-steroid isomerase, yeast alcohol dehydrogenase, alpha-glycerophosphate dehydrogenase, triose phosphate isomerase, horseradish peroxidase, alkaline phosphatase, asparaginase, glucose oxidase, beta-galactosidase, ribonuclease, urease, catalase, glucose-6-phosphate dehydrogenase, glucoamylase, and acetylcholinesterase.
  • Other methods of labeling and detecting antibodies are known in the art and are within the scope of this invention.
  • FIG. 1 shows EphA2 expression in various human prostate cell lines.
  • LNCAP three metastatic cell lines, LNCAP, DU145, and PC3, are tested for levels of EphA2 expression. It is known that, of these three cell lines, LNCAP is the least invasive, DU145 is somewhat more invasive, and PC3 is the most invasive.
  • EphA2 expression is determined by western blotting with D7 antibodies. As can be seen in FIG. 1A , EphA2 expression positively correlates with invasiveness.
  • FIG. 1B D7 antibodies are used to test EphA2 expression in normal MLC cells as compared to expression in transformed cells.
  • Normal MLC cells, MLC cells which have been transformed by K-Ras, and MLC cells with have been transformed by X-irradiation are studied.
  • EphA2 is overexpressed in both of the transformed cell lines.
  • FIG. 1C shows results similar to FIG. 1B , except the normal cell line is 267B1.
  • FIG. 1C shows that EphA2 is overexpressed in the transformed cells.
  • FIG. 1 demonstrates that EphA2-specific antibodies detect changes in metastatic cells, and that tests using these antibodies indicate the level of metastatic invasiveness.
  • EphA2 antibodies are used to detect altered EphA2 expression in metastatic mammary cells. EphA2 is expressed in normal mammary epithelial cells. FIG. 2 illustrates altered EphA2 expression in mammary tumor cell lines. As can be seen in FIG. 2 , western blots from whole cell lysates using D7 antibodies reveal that EphA2 expression is completely absent in cells derived from non-metastatic breast tumors (ZR75-1, BT474, SKBR3, MDA-MB-435). By contrast, EphA2 is overexpressed in metastatic breast cancer cell lines (MDA-MB-435, MDA-MB-231). Thus, EphA2 antibodies detect altered EphA2 expression in breast cancer cells, which can be used to diagnose metastasis.
  • EphA2 occurs early in the disease, and testing with EphA2-specific antibodies provide information relevant to invasiveness even when other known markers remain normal.
  • D7 antibodies are useful as a diagnostic, even in early stages of disease.
  • EphA2 antibodies in combination wvitlh other antibodies are used to detect further alterations in EphA2 expression.
  • wvesterni blots using D7 can distinguish between non-metastatic and metastatic tumors, with non-metastatic tumors failing to express EphA2, and metastatic cells overexpressing EphA2.
  • tyrosine phosphorylation is studied.
  • Using a phosphotyrosine-specific antibody it has been found that EphA2 is phosphorylated in normal cells, but it is not phosphorylated in metastatic cells.
  • EphA2 specific antibodies can qualitatively detect a difference between metastatic and non-metastatic mammary tumor cells
  • diagnostics incorporating both an EphA2-specific antibody and a phosphotyrosine-specific antibody provides a sensitive test for distinguishing between normal, non-metastatic, and metastatic mammary cells.
  • EphA2-specific antibodies detect redistribution of EphA2 expression in transformed cells.
  • the EphA2-specific antibodies used in this example are produced by a cell line known as B2D6, and these antibodies are specific for an extracellular epitope of EphA2.
  • B2D6 a cell line known as B2D6
  • FIG. 3A immunofluorescence with B2D6 demonstrates that EphA2 is found within sites of cell-cell contact in normal cells.
  • FIG. 3B EphA2 is redistributed.
  • EphA2 is found in the membrane ruffles.
  • EphA2 is found within sites of cell-cell adhesion, but in metastatic prostate epithelial cells, EphA2 is overexpressed and the expression is diffusely distributed. Therefore, immunofluorescence using EphA2-specific antibodies provides an additional means for diagnosing the transformation and metastatic state of tumor cells.
  • overexpression, redistribution, and phosplhornlation of EphA2 in metastatic cells provide various bases for diagnosis of metastatic tumors using EphA2-specific antibodies.
  • Immuniohistochemistry or Western blotting may be used to monitor the change of EphA2 expression in biopsied samples of patient breast tissue, prostate tissue, or tissue from other tumors.
  • D7 and other EphA2-specific antibodies can be used to monitor plasma, urine, and other body fluids to detect altered expression of EphA2, which would signal metastasis. Detection of altered tyrosine phosphorylation of EphA2 in combination with information concerning an alteration of EphA2 expression further aids in diagnosis of metastatic disease.

Abstract

Method and kits are provided for the detection and diagnosis of metastatic disease. More particularly, the methods and kits employ compounds that can detect EphA2, a specific epithelial cell tyrosine kinase that is overexpressed in metastatic tumor cells. In one embodiment the compound is an antibody capable of binding to an epitope of EphA2.

Description

    RELATED APPLICATIONS
  • This application claims priority under 35 U.S.C. § 119(e) to U.S. Provisional Application No. 60/149,259, filed Aug. 17, 1999, which is expressly incorporated by reference herein.
  • FIELD OF THE INVENTION
  • The present invention relates to diagnosis of metastatic disease. More particularly, this invention relates to reagents that can detect a specific epithelial cell tyrosine kinase that is overexpressed in metastatic tumor cells. Most particularly, this invention relates to reagents which bond to an intracellular epitope of the epithelial cell tyrosine kinase, and the use of these reagents for cancer diagnosis.
  • BACKGROUND AND SUMMARY OF THE INVENTION
  • Cancer cell metastasis requires cellular capacity to 1) detach from a primary tumor, 2) migrate and invade through local tissues, 3) translocate to distant sites in the body (via lymph or blood), 4) colonize a foreign site, and 5) grow and survive in this foreign environment. All of these behaviors are linked to cell adhesions. Cell adhesions control the physical interactions of cells with their microenvironment. Cell adhesions also initiate signals that dictate tumor cell growth, death, and differentiation.
  • Various cancer cells, including breast cancer cells, are known to exhibit altered cell adhesion. As compared to normal breast epithelia, transformed human breast epithelial cells have decreased cell-cell contacts and increased interactions with the surrounding extracellular matrix. These changes facilitate increased detachment and migration of cancer cells away from cell colonies and are directly linked with alteration in tyrosine phosphorylation of cell membrane proteins. Tyrosine phosphorylation is a potent form of cell signal transduction, and alteration in levels of tyrosine phosphorylation is believed to be important for tumor cell invasiveness. Thus, regulation of tyrosine phosphorylation represents a promising target for therapeutic intervention against metastatic cancer. Tyrosine phosphorylation is controlled by cell membrane tyrosine kinases, and increased expression of tyrosine kinases is known to occur in metastatic cancer cells.
  • Identification of increased expression of cell membrane tyrosine kinases would aid in the diagnosis and treatment of metastatic diseases. One such tyrosine kinase is EphA2. A member of the Eph family of tyrosine kinases known as Eplirins, EphA2 is a transmembrane receptor tyrosinie kinase with a cell-bound ligand.
  • Although cloned a decade ago, see Lindberg, R. A. and Hunter, T., “cDNA Cloning and Characterization of Eck, an Epithelial Cell Receptor Protein-tyrosine Kinase in the Eph/elk Family of Protein Kinases,” Mo. Cell. Biol. 10 (12), 6316-6324 (1990), rather little is known about EphA2 function, largely because EphA2-specific antibodies previously have been difficult to generate.
  • To facilitate research on EphA2,an improved method for generating a panel of monoclonal antibodies specific for tyrosine phosphorylated proteins has been developed. Using this method, a multiplicity of EphA2 recognizing monoclonal antibodies has been generated. These antibodies have been used to show that EphA2 is overexpressed in metastatic breast, lung, colon, and prostate cells. Because EphA2 is expressed differently in normal and metastatic cells, EphA2-specific antibodies are useful in the diagnosis of metastatic disease. Antibodies produced by one particular hybridoma recognize an intracellular epitope of EphA2 and have been shown to be highly specific in binding to EphA2.
  • Thus, one aspect of this invention is a compound which specifically binds to an intracellular epitope of EphA2. In a preferred embodiment, the compound is an antibody specific for a domain of the EphA2 protein. However, natural or artificial ligands, peptides, anti-sense, ATP analogies, or other small molecules capable of specifically targeting EphA2 may be employed. A second aspect of this invention is a method for generating antibodies which recognize EphA2 intracellular epitopes. Another aspect of this invention is the use of EphA2-specific antibodies in the diagnosis of metastatic disease. An additional aspect of this invention is a diagnostic reagent specific for detecting EphA2, any fragment thereof, or DNA or RNA coding for the EphA2 protein.
  • Additional features of the present invention will become apparent to those skilled in the art upon consideration of the following detailed description of preferred embodiments exemplifying the best mode of carrying out the invention as presently perceived.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1A-C showN a series of western blots showing EphA2 expression in cell lines derived from human prostate cells;
  • FIG. 1A is a western blot showing EphA2 expression in various human prostate cancer cell lines;
  • FIG. 1B is a western blot showing EphA2 expression in human prostatic epithelial cell line MLC and expression in that cell line after transformation by oncogenic K-Ras or X-irradiation;
  • FIG. 1C is similar to FIG. 1B, except showing expression in human prostatic epithelial cell line 267B1 and expression in that cell line after transformation by oncogenic K-Ras or X-irradiation;
  • FIG. 2 is a western blot showing EphA2 expression in various human mammary epithelial cell lines;
  • FIG. 3A-B shows EphA2 localization in the cell membranes of various mammary epithelial cell lines, as seen by immunofluorescence microscopy;
  • FIG. 3A shows EphA2 localization in sites of cell adhesion in normal MCF-10A cells; and
  • FIG. 3B shows EphA2 redistribution in malignant cells.
  • DETAILED DESCRIPTION OF THE INVENTION
  • Antibodies specific for EphA2 have been isolated through an improved method. The method employed is designed for increased sensitivity and diversity of responding hybridomas. According to this method, tyrosine phosphorylated proteins from Ras-transformed human epithelial cells are isolated by affinity chromatography using existing phosphotyrosine-specific antibodies. The tyrosine phosphorylated proteins are then used as an immunogen for producing monoclonal antibodies. Low-dose amounts of tyrosine phosphorylated proteins are injected proximal to lymph nodes, every other day, over a ten day period (the RIMMS strategy). B cells from engorged lymph nodes are then isolated and fused with a Bcl-2-overexpressing myeloma, to minimize apoptosis after fusion. This method results in increased diversity, specificity, and cost-effectiveness of hybridoma production. The hybridomas are first screened to identify those hybridomas producing antibodies capable of distinguishing malignant from normal cancer cells. To date, at least 450 such hybridomas have been identified.
  • Hybridomas which are specific to EphA2 have been selected. Use of the RIMMS strategy has resulted in the production of various monoclonal antibodies that specifically bind EphA2. Of the first four hybridomas characterized, two recognize independent epitopes on EphA2. The first, D7, recognizes an intracellular epitope. The second, B2D6, binds to an extracellular epitope. D7 has proven to be highly specific for an intracellular epitope of EphA2 and this specificity provides much of the current basis for diagnosis of metastatic tumors.
  • It is known in the art to use antibodies to detect the presence or overexpression of a specific protein. Because EphA2 is overexpressed in metastatic cells, EphA2-specific antibodies of this invention may be used to detect this overexpression and, thus, to detect metastatic disease. Such techiques include but are not limited to western blotting, dot blotting, precipitation, agglutination, ELISA assays, immunohistochemistry, in situ hybridization, flow cytometry on a variety of tissues or bodily fluids, and a variety of sandwich assays. These techniques are well known in the art. See, for example, U.S. Pat. No. 5,876,949, hereby incorporated by reference. When antibodies specific for an intracellular epitope of EphA2 are used, the cells must be lysed and incubated with the antibody. The above techniques may be performed on whole-cell lysates, or EphA2 may be separated out for testing, such as by immunoprecipitation. The D7 antibodies of this invention are highly specific for an intracellular epitope of EphA2 and have proven to be sensitive to differential expression of EphA2 in metastatic cells. Other techniques, such as immunohistological staining, require whole cells, and may further require cell layers of a particular cell density. Such tests require an antibody specific for an extracellular epitope of EphA2.
  • The antibodies of this invention may be used to detect metastatic disease in a wide variety of tissue samples. For instance, research using EphA2-specific antibodies has revealed that altered EphA2 expression occurs in breast, kidney, prostate, lung, and colon cells, and it is believed that altered EphA2 expression occurs in other types of cell metastasis, particularly epithelial malignancies. EphA2-specific antibodies may be used to detect metastasis in biopsied tumor tissue. Also, samples of a variety of body fluid samples, such as blood, plasma, spinal fluid, saliva, and urine, can be tested with the antibodies of the present invention. Altered EphA2 expression in these samples indicates the presence of metastatic disease.
  • Additionally, other antibodies may be used in combination with the antibodies of the present invention to provide further information concerning metastatic disease state. For example, the EphA2 of metastatic cells exhibits altered tyrosine phosphorylation. In normal breast epithelial cells, EphA2 is expressed and is tyrosine phosphorylated. However, in metastatic breast epithelial cells, EphA2 is overexpressed, and the EphA2 is not tyrosine phosphorylated. Because a test quantizing EphA2 expression sometimes may lead to an ambiguous result, it may be desirable to determine tyrosine phosphorylation, as well as the magnitude of EphA2 expression. Thus, a method of diagnosis using the antibodies of this invention in combination with phosphotyrosine-specific antibodies provides data for determining the state of metastatic disease.
  • Moreover, the EphA2-specific antibodies of this invention can be exploited to detect changes in EphA2 localization which are associated with metastasis. In normal breast and prostate epithelial cells, EphA2 is enriched in within cites of cell adhesion. Conversely, in metastatic prostate cells EphA2 is diffusely distributed, and in metastatic breast cancer cells EphA2 is redistributed into the membrane ruffles. Techniques such as immunohistological staining or immunofluorescent microscopy are well known in the art and may be used to visualize EphA2 distribution. See, for example, U.S. Pat. No. 5,514,554, hereby incorporated by reference. EphA2 expression can be detected by using antibodies capable of detecting whole EphA2 or fragments of the EphA2 protein. Other methods of detecting altered EphA2 expression include detecting DNA or RNA sequences coding for the EphA2 protein.
  • In order to detect overexpression or altered distribution of EphA2, the EphA2-specific antibodies may be labeled covalently or non-covalently with any of a number of knowfn detectable labels, such fluorescent, radioactive, or enzymatic substances, as is known in the art. Alternatively, a secondary antibody specific for the antibodies of this invention is labeled with a known detectable label and used to detect the EphA2-specific antibodies in the above techniques.
  • Preferred labels include chromogens dyes. Among the most commonly used are 3-amino-9-ethylcarbazole (AEC) and 3,3′-diaminobenzidine tetrahydroclholoride (DAB). These can be detected using light microscopy. Also preferred are fluorescent labels. Among the most commonly used fluorescent labeling compounds are fluorescein isothiocyanate, rhodamine, phycoerythrin, phycocyanin, allophycocyanin, o-phtlhaldelhyde and fluorescamine. Chemiluminescent and bioluminescent compounds such as luminol, isoluminol, theromatic acridinium ester, imidazole, acridinium salt, oxalate ester, luciferin, luciferase, and aequorin also may be used. When the fluorescent-labeled antibody is exposed to light of the proper wavelenigtlh, its presence can be detected due to its fluorescence.
  • Also preferred are radioactive labels. Radioactive isotopes which are particularly useful for labeling the antibodies of the present invention include 3H, 125I, 131I, 35S, 32P, and 14C. The radioactive isotope can be detected by such means as the use of a gamma counter, a scintillation counter, or by autoradiography.
  • Another method in which the antibodies can be detectably labeled is by linking the antibodies to an enzyme and subsequently using the antibodies in an enzyme immunoassay (EIA) or enzyme-linked immunosorbent assay (ELISA). The enzyme, when subsequently exposed to its substrate, reacts with the substrate and generates a chemical moiety which can be detected, for example, by spectrophotometric, fluorometric, or visual means. Enzymes which can be used to detectably label antibodies include, but are not limited to malate dehydrogenase, staphylococcal nuclease, delta-5-steroid isomerase, yeast alcohol dehydrogenase, alpha-glycerophosphate dehydrogenase, triose phosphate isomerase, horseradish peroxidase, alkaline phosphatase, asparaginase, glucose oxidase, beta-galactosidase, ribonuclease, urease, catalase, glucose-6-phosphate dehydrogenase, glucoamylase, and acetylcholinesterase. Other methods of labeling and detecting antibodies are known in the art and are within the scope of this invention.
  • EXAMPLE 1
  • The antibodies produced by the D7 hybridoma are used to detect differential expression of EphA2 between normal prostate epithelial cells and metastatic cells. FIG. 1 shows EphA2 expression in various human prostate cell lines. Referring first to FIG. 1A, three metastatic cell lines, LNCAP, DU145, and PC3, are tested for levels of EphA2 expression. It is known that, of these three cell lines, LNCAP is the least invasive, DU145 is somewhat more invasive, and PC3 is the most invasive. EphA2 expression is determined by western blotting with D7 antibodies. As can be seen in FIG. 1A, EphA2 expression positively correlates with invasiveness.
  • In FIG. 1B, D7 antibodies are used to test EphA2 expression in normal MLC cells as compared to expression in transformed cells. Normal MLC cells, MLC cells which have been transformed by K-Ras, and MLC cells with have been transformed by X-irradiation are studied. As can be seen in FIG. 1B, EphA2 is overexpressed in both of the transformed cell lines. FIG. 1C shows results similar to FIG. 1B, except the normal cell line is 267B1. As with FIG. 1B, FIG. 1C shows that EphA2 is overexpressed in the transformed cells. In sum, FIG. 1 demonstrates that EphA2-specific antibodies detect changes in metastatic cells, and that tests using these antibodies indicate the level of metastatic invasiveness.
  • EXAMPLE 2
  • EphA2 antibodies are used to detect altered EphA2 expression in metastatic mammary cells. EphA2 is expressed in normal mammary epithelial cells. FIG. 2 illustrates altered EphA2 expression in mammary tumor cell lines. As can be seen in FIG. 2, western blots from whole cell lysates using D7 antibodies reveal that EphA2 expression is completely absent in cells derived from non-metastatic breast tumors (ZR75-1, BT474, SKBR3, MDA-MB-435). By contrast, EphA2 is overexpressed in metastatic breast cancer cell lines (MDA-MB-435, MDA-MB-231). Thus, EphA2 antibodies detect altered EphA2 expression in breast cancer cells, which can be used to diagnose metastasis. Moreover, in non-metastatic breast epithelial cells, loss of EphA2 occurs early in the disease, and testing with EphA2-specific antibodies provide information relevant to invasiveness even when other known markers remain normal. Thus, D7 antibodies are useful as a diagnostic, even in early stages of disease.
  • EXAMPLE 3
  • EphA2 antibodies in combination wvitlh other antibodies are used to detect further alterations in EphA2 expression. As discussed above in Example 2, wvesterni blots using D7 can distinguish between non-metastatic and metastatic tumors, with non-metastatic tumors failing to express EphA2, and metastatic cells overexpressing EphA2. However, different results are found when tyrosine phosphorylation is studied. Using a phosphotyrosine-specific antibody, it has been found that EphA2 is phosphorylated in normal cells, but it is not phosphorylated in metastatic cells. Thus, while EphA2 specific antibodies can qualitatively detect a difference between metastatic and non-metastatic mammary tumor cells, diagnostics incorporating both an EphA2-specific antibody and a phosphotyrosine-specific antibody provides a sensitive test for distinguishing between normal, non-metastatic, and metastatic mammary cells.
  • EXAMPLE 4
  • Immunofluorescently labeled EphA2-specific antibodies detect redistribution of EphA2 expression in transformed cells. The EphA2-specific antibodies used in this example are produced by a cell line known as B2D6, and these antibodies are specific for an extracellular epitope of EphA2. As seen in FIG. 3A, immunofluorescence with B2D6 demonstrates that EphA2 is found within sites of cell-cell contact in normal cells. However, in transformed cells, shown in FIG. 3B, EphA2 is redistributed. Furthermore, in metastatic cells EphA2 is found in the membrane ruffles. Similarly, in normal prostate epithelial cells, EphA2 is found within sites of cell-cell adhesion, but in metastatic prostate epithelial cells, EphA2 is overexpressed and the expression is diffusely distributed. Therefore, immunofluorescence using EphA2-specific antibodies provides an additional means for diagnosing the transformation and metastatic state of tumor cells.
  • As shown in Examples 1-4, overexpression, redistribution, and phosplhornlation of EphA2 in metastatic cells provide various bases for diagnosis of metastatic tumors using EphA2-specific antibodies. Immuniohistochemistry or Western blotting may be used to monitor the change of EphA2 expression in biopsied samples of patient breast tissue, prostate tissue, or tissue from other tumors. Additionally, D7 and other EphA2-specific antibodies can be used to monitor plasma, urine, and other body fluids to detect altered expression of EphA2, which would signal metastasis. Detection of altered tyrosine phosphorylation of EphA2 in combination with information concerning an alteration of EphA2 expression further aids in diagnosis of metastatic disease.
  • Although the invention has been described in detail with reference to preferred embodiments, variations and modifications exist within the scope and spirit of the invention as described and defined in the following claims.

Claims (79)

1. A method for detecting the presence of metastatic cells in a cell population comprising the steps of
lysing at least a portion of the cell population,
incubating the lysed cells , with a reagent capable of specific binding to an epitope of EphA2 to allow, antibody binding to said epitope, and
detecting compound-epitope binding.
2. The method of claim 1 wherein the reagent is an antibody.
3. The method of claim 2 wherein the epitope of EphA2 is and intracellular epitope of EphA2.
4. The method of claim 3 wherein the antibody is produced by hybridoma cell line D7.
5. The method of claim 2 wherein the antibody is labeled with a detectable label, and the detecting step includes detecting the label.
6. The method of claim 5 wherein the antibody is labeled wvitlh a fluorescent label and the detecting step comprises detecting the fluorescent label.
7. The method of claim 5 wherein the antibody is labeled with a radioactive label and the detecting step comprises detecting the radioactive label.
8. The method of claim 1 wherein the cell population comprises cells from a breast or prostate tissue biopsy.
9. The method of claim 1 wherein the cell population is harvested from a body fluid selected from the group consisting of blood, plasma, spinal fluid, saliva, and urine.
10. The method of claim 9 wherein the detecting step includes a diagnostic method selected from the group consisting of ELISA assays and flow cytometry.
11. The method of claim 1 wherein the incubating and detecting steps comprise western blotting methodology.
12. The method of claim 11 further comprising the steps of providing a second antibody having phosphotyrosine specificity, and western blotting with the second antibody.
13. The method of claim 1 wherein the metastatic cells are selected from the group consisting of breast, prostate, lung, and colon cancers.
14. An antibody which specifically binds to an intracellular epitope of EphA2.
15. The antibody of claim 14 bound to a detectable label.
16. The antibody of claim 14 which is a monoclonal antibody.
17. A method for detecting the presence of metastatic cells in a cell population comprising the steps of:
incubating the cells with a reagent capable of specific binding to a compound associated with EphA2 expression, and
detecting reagent-compound binding.
18. The method of claim 17 wherein the reagent is an antibody.
19. The method of claim 17 wherein the compound is selected from the group consisting of EphA2, a fragment of EphA2, DNA coding for the EphA2 protein, and RNA coding for the EphA2 protein.
20. The method of claim 17 further comprising the step of fixing the cells on a slide, and the detecting step comprises immunofluorescence staining.
21. A kit for detecting the presence of metastatic cells in a cell population comprising
an antibody capable of specific binding to an epitope of EphA2, and
means for detecting antibody-epitope binding.
22. The kit of claim 21 wherein the means for detecting antibody-epitope binding is a label bound to the antibody.
23. The kit of claim 21 further comprising an antibody having phosphotyrosine specificity.
24. A method for detecting the presence of metastatic or potentially metastatic cells in a cell population comprising:
lysing at least a portion of the cell population;
incubating the lysed cells with a reagent capable of specific binding to EphA2 to allow binding of the reagent to EphA2; and
detecting reagent-EphA2 binding, wherein reagent-EphA2 binding is indicative of the presence of metastatic or potentially metastatic cells in the cell population.
25. The method of claim 24 wherein the reagent comprises an antibody, and wherein detecting reagent-EphA2 binding comprises detecting antibody-EphA2 binding.
26. The method of claim 25 wherein the antibody binds to an intracellular epitope of EphA2.
27. The method of claim 25 wherein the antibody is produced by hybridoma cell line D7.
28. The method of claim 25 wherein the antibody is labeled with a detectable label, and wherein detecting reagent-EphA2 binding comprises detecting the label.
29. The method of claim 28 wherein the antibody comprises at least one of a fluorescent label, a chemiluminescent label, a bioluminescent label, an enzymatic label, a chromogenic label and a radiolabel, wherein detecting reagent-EphA2 binding comprises detecting at least one detectable label.
30. The method of claim 24 wherein the cell population comprises cells selected from the group consisting of breast cells, kidney cells, prostate cells, lung cells and colon cells.
31. The method of claim 24 wherein the cell population comprises epithelial cells.
32. The method of claim 24 wherein the cell population comprises cells selected from the group consisting of breast cancer cells, kidney cancer cells, prostate cancer cells, lung cancer cells and colon cancer cells.
33. The method of claim 24 wherein the cell population comprises epithelial cancer cells.
34. The method of claim 24 wherein the cell population comprises metastatic or potentially metastatic cancer cells.
35. The method of claim 34 wherein the metastatic or potentially metastatic cancer cells comprise cells selected from the group consisting of breast cancer cells, kidney cancer cells, prostate cancer cells, lung cancer cells, and colon cancer cells.
36. The method of claim 34 wherein the metastatic or potentially metastatic cancer cells comprise epithelial cancer cells.
37. The method of claim 24 wherein the cell population comprises cells from a tissue biopsy.
38. The method of claim 37 wherein the tissue comprises breast tissue or prostate tissue.
39. The method of claim 24 wherein the cell population comprises cells from a body fluid.
40. The method of claim 39 wherein the body fluid is selected from the group consisting of blood, plasma, spinal fluid, saliva, and urine.
41. The method of claim 24 wherein detecting reagent-EphA2 binding comprises utilizing a diagnostic method selected from the group consisting of an ELISA assay, a Western blot, and flow cytometry.
42. The method of claim 24 wherein detecting reagent-EphA2 binding comprises utilizing a Western blot; the method further comprising Western blotting with a second antibody having phosphotyrosine specificity.
43. A method for detecting the presence of metastatic or potentially metastatic cells in a cell population comprising:
incubating at least a portion of the cell population with a reagent capable of binding to EphA2 to allow binding of the reagent to EphA2; and
detecting reagent-EphA2 binding, wherein reagent-EphA2 binding is indicative of the presence of metastatic or potentially metastatic cells in the cell population.
44. The method of claim 43 wherein the reagent comprises an antibody and wherein detecting reagent-EphA2 binding comprises detecting antibody-EphA2 binding.
45. The method of claim 44 wherein the antibody binds to an intracellular epitope of EphA2.
46. The method of claim 44 wherein the antibody is produced by hybridoma, cell line D7.
47. The method of claim 44 wherein the antibody binds to an extracellular epitope of EphA2.
48. The method of claim of claim 44 wherein antibody-EphA2 binding yields a bound complex comprising a whole cell.
49. The method of claim 48 wherein detecting antibody-EphA2 binding comprises subjecting the bound complex to immunohistochemical staining.
50. The method of claim 44 wherein the antibody is produced by hybridoma cell line B2D6.
51. The method of claim 44 wherein the bound antibody comprises a detectable label; and wherein detecting antibody-EphA2 binding comprises detecting the label.
52. The method of claim 44 wherein the bound antibody comprises at least one of a fluorescent label, a chemiluminescent label, a bioluminescent label, an enzymatic label, a chromogenic label and a radiolabel; and wherein detecting antibody-Eph2 binding comprises detecting, at least one detectable label.
53. The method of claim 43 wherein the cell population comprises cells selected from the group consisting of breast cells, kidney cells, prostate cells, lung cells and colon cells.
54. The method of claim 43 wherein the cell population comprises epithelial cells.
55. The method of claim 43 wherein the cell population comprises cells selected from the group consisting of breast cancer cells, kidney cancer cells, prostate cancer cells, lung cancer cells and colon cancer cells.
56. The method of claim 43 wherein the cell population comprises epithelial cancer cells.
57. The method of claim 43 wherein the cell population comprises metastatic or potentially metastatic cancer cells.
58. The method of claim 57 wherein the metastatic or potentially metastatic cells comprise cells selected from the group consisting of breast cancer cells, kidney cancer cells, prostate cancer cells, lung cancer cells, and colon cancer cells.
59. The method of claim 43 wherein the metastatic or potentially metastatic cells comprise epithelial cancer cells.
60. The method of claim 43 wherein the cell population comprises cells from a tissue biopsy.
61. The method of claim 60 wherein the tissue comprises breast tissue or prostate tissue.
62. The method of claim 43 wherein the cell population comprises cells from a body fluid.
63. The method of claim 62 wherein the body fluid is selected from the group consisting of blood, plasma, spinal fluid, saliva, and urine.
64. The method of claim 43 wherein detecting reagent-EphA2 binding comprises utilizing a diagnostic method selected from the group consisting of and ELISA assay, a Western blot, anal flow cytometry.
65. The method of claim 43 wherein detecting reagent-EphA2 binding comprises utilizing a Western blot; the method further comprising Western blotting with a second antibody having phosphotyrosine specificity.
66. A method for detecting the presence of metastatic or potentially metastatic cells in a cell population comprising:
lysing at least a portion of the cell population;
incubating the lysed cells with a reagent capable of specific binding to nucleic acid comprising RNA or DNA encoding at least a portion of an EphA2 protein to allow binding of the reagent to the nucleic acid; and
detecting reagent-nucleic acid binding, wherein reagent-nucleic acid binding is indicative of the presence of metastatic or potentially metastatic cells in the cell population.
67. The method of claim 66 wherein the nucleic acid comprises RNA.
68. A method for detecting the presence of cancer cells in a selected cell population comprising:
assaying at least a portion of the selected cell population for at least one of
a change in EphA2 expression level;
a change in EphA2 localization pattern; and
a change in EphA2 phosphorylation content
as compared to the EphA2 expression level, localization pattern and phosphorylation content in an analogous normal cell population;
wherein the change is indicative of the presence of a cancer cell in the selected cell population.
69. The method of claim 68 wherein a change in EphA2 expression level, localization pattern or phosphorylation content is indicative of the presence of metastatic cancer cells in the cell population.
70. The method of claim 68 wherein a change in EphA2 expression level is indicative of the presence of nonmetastatic cancer cells in the cell population.
71. The method of claim 68 wherein assaying the cell population comprises incubating at least a portion of the selected cell population with a reagent capable of binding to EphA2 to allow binding of the reagent to EphA2; and detecting reagent-EphA2 binding.
72. The method of claim 71 wherein the reagent is an antibody.
73. The method of claim 72 wherein the antibody is produced by hybridoma D7 or B2D6.
74. A method for determining the disease stage in a cell population comprising cancer cells, the method comprising:
assaying at least a portion of the cell population for at least one of
EphA2 expression level;
EphA2 localization; and
EphA2 phosphorylation content; and
determining the disease stage of the cancer cells.
75. The method of claim 74 wherein assaying the cell population comprises incubating at least a portion of the cancer cell population with a reagent capable of binding to EphA2 to allow binding of the reagent to EphA2; and detecting reagent-EphA2 binding.
76. The method of claim 75 wherein the reagent is an antibody.
77. The method of claim 76 wherein the antibody is produced by hybridoma D7 or B2D6.
78. An isolated antibody which specifically binds to an intracellular epitope of EphA2.
79. The isolated antibody of claim 78 which is a monoclonal antibody.
US11/716,792 1999-08-17 2007-03-12 Antibodies as a cancer diagnostic Abandoned US20070161064A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US11/716,792 US20070161064A1 (en) 1999-08-17 2007-03-12 Antibodies as a cancer diagnostic
US13/210,672 US20120122112A1 (en) 1999-08-17 2011-08-16 Antibodies as a cancer diagnostic

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US14925999P 1999-08-17 1999-08-17
US09/640,952 US7192698B1 (en) 1999-08-17 2000-08-17 EphA2 as a diagnostic target for metastatic cancer
US11/716,792 US20070161064A1 (en) 1999-08-17 2007-03-12 Antibodies as a cancer diagnostic

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US09/640,952 Continuation US7192698B1 (en) 1999-08-17 2000-08-17 EphA2 as a diagnostic target for metastatic cancer

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US13/210,672 Continuation US20120122112A1 (en) 1999-08-17 2011-08-16 Antibodies as a cancer diagnostic

Publications (1)

Publication Number Publication Date
US20070161064A1 true US20070161064A1 (en) 2007-07-12

Family

ID=37863795

Family Applications (3)

Application Number Title Priority Date Filing Date
US09/640,952 Expired - Fee Related US7192698B1 (en) 1999-08-17 2000-08-17 EphA2 as a diagnostic target for metastatic cancer
US11/716,792 Abandoned US20070161064A1 (en) 1999-08-17 2007-03-12 Antibodies as a cancer diagnostic
US13/210,672 Abandoned US20120122112A1 (en) 1999-08-17 2011-08-16 Antibodies as a cancer diagnostic

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US09/640,952 Expired - Fee Related US7192698B1 (en) 1999-08-17 2000-08-17 EphA2 as a diagnostic target for metastatic cancer

Family Applications After (1)

Application Number Title Priority Date Filing Date
US13/210,672 Abandoned US20120122112A1 (en) 1999-08-17 2011-08-16 Antibodies as a cancer diagnostic

Country Status (1)

Country Link
US (3) US7192698B1 (en)

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050169931A1 (en) * 1999-08-17 2005-08-04 Purdue Research Foundation EphA2 as a therapeutic target for cancer
US7402298B1 (en) 2000-09-12 2008-07-22 Purdue Research Foundation EphA2 monoclonal antibodies and methods of making and using same
CN102169121A (en) * 2010-02-25 2011-08-31 北京诺赛基因组研究中心有限公司 New application of human kinase SBK1 (SH3-binding domain kinase 1)
US8137912B2 (en) 2006-06-14 2012-03-20 The General Hospital Corporation Methods for the diagnosis of fetal abnormalities
US8168389B2 (en) 2006-06-14 2012-05-01 The General Hospital Corporation Fetal cell analysis using sample splitting
US8195415B2 (en) 2008-09-20 2012-06-05 The Board Of Trustees Of The Leland Stanford Junior University Noninvasive diagnosis of fetal aneuploidy by sequencing
US8921102B2 (en) 2005-07-29 2014-12-30 Gpb Scientific, Llc Devices and methods for enrichment and alteration of circulating tumor cells and other particles
CN104749174A (en) * 2014-12-18 2015-07-01 东莞星显家具有限公司 Urine tyrosine detecting method
US10591391B2 (en) 2006-06-14 2020-03-17 Verinata Health, Inc. Diagnosis of fetal abnormalities using polymorphisms including short tandem repeats
US10704090B2 (en) 2006-06-14 2020-07-07 Verinata Health, Inc. Fetal aneuploidy detection by sequencing
CN111579788A (en) * 2020-05-29 2020-08-25 郑州大学 Simoa kit of high-sensitivity tumor marker EPHA2 and application thereof

Families Citing this family (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7192698B1 (en) * 1999-08-17 2007-03-20 Purdue Research Foundation EphA2 as a diagnostic target for metastatic cancer
JP2006501153A (en) * 2002-05-23 2006-01-12 パーデュー・リサーチ・ファウンデーション Low molecular weight protein tyrosine phosphatase (LMW-PTP) as a diagnostic and therapeutic target
CA2525929A1 (en) * 2003-05-13 2004-11-25 Chiron Corporation Methods of modulating metastasis and skeletal related events resulting from metastases
WO2006023420A2 (en) * 2004-08-16 2006-03-02 Medimmune, Inc. Integrin antagonists with enhanced antibody dependent cell-mediated cytotoxicity activity
JP5875054B2 (en) * 2013-02-13 2016-03-02 国立大学法人 東京大学 Cancer testing method and testing kit
US10443054B2 (en) 2016-03-06 2019-10-15 Massachusetts Institute Of Technology Methods for identifying and treating invasive/metastatic breast cancers
JP6729917B2 (en) * 2016-08-19 2020-07-29 国立大学法人 東京大学 EphA2 N-terminal fragment antibody
MX2019009967A (en) 2017-02-24 2019-12-02 Macrogenics Inc Bispecific binding molecules that are capable of binding cd137 and tumor antigens, and uses thereof.

Citations (31)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4472371A (en) * 1979-10-29 1984-09-18 Summa Medical Corporation Radiolabeled antibody to anti-tumor associated antigen and process
US4704692A (en) * 1986-09-02 1987-11-03 Ladner Robert C Computer based system and method for determining and displaying possible chemical structures for converting double- or multiple-chain polypeptides to single-chain polypeptides
US4816567A (en) * 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US5001225A (en) * 1986-12-08 1991-03-19 Georgetown University Monoclonal antibodies to a pan-malarial antigen
US5447936A (en) * 1993-12-22 1995-09-05 Bionumerik Pharmaceuticals, Inc. Lactone stable formulation of 10-hydroxy 7-ethyl camptothecin and methods for uses thereof
US5457048A (en) * 1993-12-03 1995-10-10 La Jolla Cancer Research Foundation Eph-related tyrosine kinases, nucleotide sequences and methods of use
US5514554A (en) * 1991-08-22 1996-05-07 Becton Dickinson And Company Methods and compositions for cancer therapy and for prognosticating responses to cancer therapy
US5585089A (en) * 1988-12-28 1996-12-17 Protein Design Labs, Inc. Humanized immunoglobulins
US5635177A (en) * 1992-01-22 1997-06-03 Genentech, Inc. Protein tyrosine kinase agonist antibodies
US5770195A (en) * 1988-01-12 1998-06-23 Genentech, Inc. Monoclonal antibodies directed to the her2 receptor
US5795734A (en) * 1994-09-19 1998-08-18 President And Fellows Of Harvard College EPH receptor ligands, and uses related thereto
US5811098A (en) * 1992-11-24 1998-09-22 Bristol-Myers Squibb Company Antibodies to HER4, human receptor tyrosine kinase
US5824303A (en) * 1992-11-13 1998-10-20 Amgen Inc. Eck receptor ligands
US5872223A (en) * 1994-08-19 1999-02-16 Regents Of The University Of Minnesota Immunoconjugates comprising tyrosine kinase inhibitors
US5876949A (en) * 1995-05-31 1999-03-02 The Trustees Of The University Of Pennsylvania Antibodies specific for fragile X related proteins and method of using the same
US5876950A (en) * 1995-01-26 1999-03-02 Bristol-Myers Squibb Company Monoclonal antibodies specific for different epitopes of human GP39 and methods for their use in diagnosis and therapy
US5981245A (en) * 1994-04-15 1999-11-09 Amgen Inc. EPH-like receptor protein tyrosine kinases
US6057105A (en) * 1995-03-17 2000-05-02 Ngi/Cancer Tech Company, Llc Detection of melanoma or breast metastasis with a multiple marker assay
US6174670B1 (en) * 1996-06-04 2001-01-16 University Of Utah Research Foundation Monitoring amplification of DNA during PCR
US20010031262A1 (en) * 1999-12-06 2001-10-18 Michael Caplan Controlled delivery of antigens
US20010031252A1 (en) * 2000-03-31 2001-10-18 Low Philip Stewart Method of treatment using ligand-immunogen conjugates
US20040028685A1 (en) * 2002-05-10 2004-02-12 Kinch Michael S. EphA2 monoclonal antibodies and methods of use thereof
US20040091486A1 (en) * 2002-05-10 2004-05-13 Kinch Michael S. EphA2 agonistic monoclonal antibodies and methods of use thereof
US20050152899A1 (en) * 2002-05-10 2005-07-14 Kinch Michael S. EphA2 agonistic monoclonal antibodies and methods of use thereof
US20050169931A1 (en) * 1999-08-17 2005-08-04 Purdue Research Foundation EphA2 as a therapeutic target for cancer
US7033574B1 (en) * 1990-04-02 2006-04-25 Bracco International B.V. Stable microbubbles suspensions injectable into living organisms
US7101976B1 (en) * 2000-09-12 2006-09-05 Purdue Research Foundation EphA2 monoclonal antibodies and methods of making and using same
US7192698B1 (en) * 1999-08-17 2007-03-20 Purdue Research Foundation EphA2 as a diagnostic target for metastatic cancer
US20090220527A1 (en) * 2005-12-21 2009-09-03 Medlmmune, Llc Affinity optimized epha2 agonistic antibodies and methods of use thereof
US7662770B2 (en) * 2002-05-23 2010-02-16 Purdue Research Foundation Low molecular weight protein tyrosine phosphatase (LMW-PTP) as a diagnostic and therapeutic target
US7776328B2 (en) * 2005-02-04 2010-08-17 Macrogenics, Inc. Antibodies that bind to EphA2 and methods of use thereof

Family Cites Families (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1994004679A1 (en) 1991-06-14 1994-03-03 Genentech, Inc. Method for making humanized antibodies
EP0590030B1 (en) 1991-06-21 2004-09-29 The Walter and Eliza Hall Institute of Medical Research A receptor-type tyrosine kinase [human eph/elk-like kinase - hek] and use thereof
JPH08505765A (en) 1992-11-13 1996-06-25 アムジエン・インコーポレーテツド ECK receptor ligand
AU678483B2 (en) 1993-06-03 1997-05-29 Protherics Inc. Antibody fragments in therapy
US5969101A (en) * 1995-10-27 1999-10-19 Duke University ABL-interactor protein
UA73073C2 (en) 1997-04-03 2005-06-15 Уайт Холдінгз Корпорейшн Substituted 3-cyan chinolines
AU766077C (en) 1998-11-20 2004-10-07 Genentech Inc. Uses for Eph receptor antagonists and agonists to treat vascular disorders
WO2000037500A1 (en) 1998-12-18 2000-06-29 Mount Sinai Hospital Three dimensional structure of a sterile alpha motif domain
DE60027768T2 (en) 1999-08-17 2007-04-05 Purdue Research Foundation, West Lafayette TREATMENT OF METASTATIC DISEASE
ATE483978T1 (en) 1999-08-17 2010-10-15 Purdue Research Foundation ANTI-EPHA2 ANTIBODIES AS A CANCER DIAGNOSTIC
DE60033316T2 (en) 1999-12-29 2007-11-22 Wyeth TRICYCLIC PROTEIN KINASE INHIBITORS
EP1689346A4 (en) 2003-11-20 2007-11-21 Medimmune Inc Epha2 agonistic monoclonal antibodies and methods of use thereof

Patent Citations (39)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4472371A (en) * 1979-10-29 1984-09-18 Summa Medical Corporation Radiolabeled antibody to anti-tumor associated antigen and process
US4816567A (en) * 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US4704692A (en) * 1986-09-02 1987-11-03 Ladner Robert C Computer based system and method for determining and displaying possible chemical structures for converting double- or multiple-chain polypeptides to single-chain polypeptides
US5001225A (en) * 1986-12-08 1991-03-19 Georgetown University Monoclonal antibodies to a pan-malarial antigen
US5770195A (en) * 1988-01-12 1998-06-23 Genentech, Inc. Monoclonal antibodies directed to the her2 receptor
US5585089A (en) * 1988-12-28 1996-12-17 Protein Design Labs, Inc. Humanized immunoglobulins
US7033574B1 (en) * 1990-04-02 2006-04-25 Bracco International B.V. Stable microbubbles suspensions injectable into living organisms
US5514554A (en) * 1991-08-22 1996-05-07 Becton Dickinson And Company Methods and compositions for cancer therapy and for prognosticating responses to cancer therapy
US5635177A (en) * 1992-01-22 1997-06-03 Genentech, Inc. Protein tyrosine kinase agonist antibodies
US5824303A (en) * 1992-11-13 1998-10-20 Amgen Inc. Eck receptor ligands
US5811098A (en) * 1992-11-24 1998-09-22 Bristol-Myers Squibb Company Antibodies to HER4, human receptor tyrosine kinase
US5457048A (en) * 1993-12-03 1995-10-10 La Jolla Cancer Research Foundation Eph-related tyrosine kinases, nucleotide sequences and methods of use
US5447936A (en) * 1993-12-22 1995-09-05 Bionumerik Pharmaceuticals, Inc. Lactone stable formulation of 10-hydroxy 7-ethyl camptothecin and methods for uses thereof
US5981245A (en) * 1994-04-15 1999-11-09 Amgen Inc. EPH-like receptor protein tyrosine kinases
US5872223A (en) * 1994-08-19 1999-02-16 Regents Of The University Of Minnesota Immunoconjugates comprising tyrosine kinase inhibitors
US5795734A (en) * 1994-09-19 1998-08-18 President And Fellows Of Harvard College EPH receptor ligands, and uses related thereto
US5876950A (en) * 1995-01-26 1999-03-02 Bristol-Myers Squibb Company Monoclonal antibodies specific for different epitopes of human GP39 and methods for their use in diagnosis and therapy
US6057105A (en) * 1995-03-17 2000-05-02 Ngi/Cancer Tech Company, Llc Detection of melanoma or breast metastasis with a multiple marker assay
US5876949A (en) * 1995-05-31 1999-03-02 The Trustees Of The University Of Pennsylvania Antibodies specific for fragile X related proteins and method of using the same
US6174670B1 (en) * 1996-06-04 2001-01-16 University Of Utah Research Foundation Monitoring amplification of DNA during PCR
US20060088541A1 (en) * 1999-08-17 2006-04-27 Purdue Research Foundation EphA2 as a therapeutic target for cancer
US7192698B1 (en) * 1999-08-17 2007-03-20 Purdue Research Foundation EphA2 as a diagnostic target for metastatic cancer
US20050169931A1 (en) * 1999-08-17 2005-08-04 Purdue Research Foundation EphA2 as a therapeutic target for cancer
US6927203B1 (en) * 1999-08-17 2005-08-09 Purdue Research Foundation Treatment of metastatic disease
US20010031262A1 (en) * 1999-12-06 2001-10-18 Michael Caplan Controlled delivery of antigens
US20010031252A1 (en) * 2000-03-31 2001-10-18 Low Philip Stewart Method of treatment using ligand-immunogen conjugates
US7033594B2 (en) * 2000-03-31 2006-04-25 Purdue Research Foundation Method of treatment using ligand-immunogen conjugates
US7101976B1 (en) * 2000-09-12 2006-09-05 Purdue Research Foundation EphA2 monoclonal antibodies and methods of making and using same
US20100278838A1 (en) * 2002-05-10 2010-11-04 Medimmune, Llc Epha2 monoclonal antibodies and methods of use thereof
US20040028685A1 (en) * 2002-05-10 2004-02-12 Kinch Michael S. EphA2 monoclonal antibodies and methods of use thereof
US20070086943A1 (en) * 2002-05-10 2007-04-19 Purdue Research Foundation And Medimmune, Inc. EphA2 agonistic monoclonal antibodies and methods of use thereof
US20040091486A1 (en) * 2002-05-10 2004-05-13 Kinch Michael S. EphA2 agonistic monoclonal antibodies and methods of use thereof
US20070134254A1 (en) * 2002-05-10 2007-06-14 Medimmune, Inc. EphA2 agonistic monoclonal antibodies and methods of use thereof
US20070166314A1 (en) * 2002-05-10 2007-07-19 Medimmune, Inc. EpA2 monoclonal antibodies and methods of use thereof
US20100143345A1 (en) * 2002-05-10 2010-06-10 Medimmune, Llc Epha2 agonistic monoclonal antibodies and methods of use thereof
US20050152899A1 (en) * 2002-05-10 2005-07-14 Kinch Michael S. EphA2 agonistic monoclonal antibodies and methods of use thereof
US7662770B2 (en) * 2002-05-23 2010-02-16 Purdue Research Foundation Low molecular weight protein tyrosine phosphatase (LMW-PTP) as a diagnostic and therapeutic target
US7776328B2 (en) * 2005-02-04 2010-08-17 Macrogenics, Inc. Antibodies that bind to EphA2 and methods of use thereof
US20090220527A1 (en) * 2005-12-21 2009-09-03 Medlmmune, Llc Affinity optimized epha2 agonistic antibodies and methods of use thereof

Cited By (27)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050169931A1 (en) * 1999-08-17 2005-08-04 Purdue Research Foundation EphA2 as a therapeutic target for cancer
US20060088541A1 (en) * 1999-08-17 2006-04-27 Purdue Research Foundation EphA2 as a therapeutic target for cancer
US8591887B2 (en) 1999-08-17 2013-11-26 Purdue Research Foundation EPHA2 as a therapeutic target for cancer
US20110150898A1 (en) * 1999-08-17 2011-06-23 Purdue Research Foundation EPHA2 as a therapeutic target for cancer
US7776327B2 (en) 1999-08-17 2010-08-17 Purdue Research Foundation EphA2 as a therapeutic target for cancer
US7402298B1 (en) 2000-09-12 2008-07-22 Purdue Research Foundation EphA2 monoclonal antibodies and methods of making and using same
US8921102B2 (en) 2005-07-29 2014-12-30 Gpb Scientific, Llc Devices and methods for enrichment and alteration of circulating tumor cells and other particles
US8168389B2 (en) 2006-06-14 2012-05-01 The General Hospital Corporation Fetal cell analysis using sample splitting
US9347100B2 (en) 2006-06-14 2016-05-24 Gpb Scientific, Llc Rare cell analysis using sample splitting and DNA tags
US8137912B2 (en) 2006-06-14 2012-03-20 The General Hospital Corporation Methods for the diagnosis of fetal abnormalities
US11781187B2 (en) 2006-06-14 2023-10-10 The General Hospital Corporation Rare cell analysis using sample splitting and DNA tags
US8372584B2 (en) 2006-06-14 2013-02-12 The General Hospital Corporation Rare cell analysis using sample splitting and DNA tags
US10155984B2 (en) 2006-06-14 2018-12-18 The General Hospital Corporation Rare cell analysis using sample splitting and DNA tags
US10704090B2 (en) 2006-06-14 2020-07-07 Verinata Health, Inc. Fetal aneuploidy detection by sequencing
US9017942B2 (en) 2006-06-14 2015-04-28 The General Hospital Corporation Rare cell analysis using sample splitting and DNA tags
US10591391B2 (en) 2006-06-14 2020-03-17 Verinata Health, Inc. Diagnosis of fetal abnormalities using polymorphisms including short tandem repeats
US9273355B2 (en) 2006-06-14 2016-03-01 The General Hospital Corporation Rare cell analysis using sample splitting and DNA tags
US11674176B2 (en) 2006-06-14 2023-06-13 Verinata Health, Inc Fetal aneuploidy detection by sequencing
US8682594B2 (en) 2008-09-20 2014-03-25 The Board Of Trustees Of The Leland Stanford Junior University Noninvasive diagnosis of fetal aneuploidy by sequencing
US9404157B2 (en) 2008-09-20 2016-08-02 The Board Of Trustees Of The Leland Stanford Junior University Noninvasive diagnosis of fetal aneuploidy by sequencing
US9353414B2 (en) 2008-09-20 2016-05-31 The Board Of Trustees Of The Leland Stanford Junior University Noninvasive diagnosis of fetal aneuploidy by sequencing
US10669585B2 (en) 2008-09-20 2020-06-02 The Board Of Trustees Of The Leland Stanford Junior University Noninvasive diagnosis of fetal aneuploidy by sequencing
US8296076B2 (en) 2008-09-20 2012-10-23 The Board Of Trustees Of The Leland Stanford Junior University Noninvasive diagnosis of fetal aneuoploidy by sequencing
US8195415B2 (en) 2008-09-20 2012-06-05 The Board Of Trustees Of The Leland Stanford Junior University Noninvasive diagnosis of fetal aneuploidy by sequencing
CN102169121A (en) * 2010-02-25 2011-08-31 北京诺赛基因组研究中心有限公司 New application of human kinase SBK1 (SH3-binding domain kinase 1)
CN104749174A (en) * 2014-12-18 2015-07-01 东莞星显家具有限公司 Urine tyrosine detecting method
CN111579788A (en) * 2020-05-29 2020-08-25 郑州大学 Simoa kit of high-sensitivity tumor marker EPHA2 and application thereof

Also Published As

Publication number Publication date
US7192698B1 (en) 2007-03-20
US20120122112A1 (en) 2012-05-17

Similar Documents

Publication Publication Date Title
US20070161064A1 (en) Antibodies as a cancer diagnostic
EP1210603B1 (en) Anti-epha2 antibodies as a cancer diagnostic
US7090983B1 (en) Methods for detecting early cancer
EP1930445B1 (en) Kit and method for detection of urothelial cancer
KR20050114604A (en) Use of nicotinamide n-methyltransferase as a marker for colorectal cancer
US20060188949A1 (en) Use of protein PLST as a marker for colorectal cancer
EP2449133B1 (en) Method for testing a subject thought to be predisposed to having metastatic cancer using delta133p53 isoforms
WO2005102938A2 (en) Antibodies specific for bcr-abl fusion protein and uses thereof
WO2008028066A2 (en) Boris isoforms and methods of detecting and treating disease
AU2011253563A1 (en) Anti-epha2 antibodies as a cancer diagnostic
AU2007202091A1 (en) Anti-epha2 antibodies as a cancer diagnostic
US20130203059A1 (en) Method for Diagnosis of Bladder Cancer and Related Kits
US20130095483A1 (en) Predictive biomarkers for breast cancer
WO2009053354A1 (en) Use of tenascin-w as a biomarker for colon cancer
US20100284923A1 (en) Methods of diagnosing latent and active malignancies
US20070218510A1 (en) Use of protein PSA3 as a marker for colorectal cancer
WO2005015234A1 (en) Use of protein sahh as a marker for colorectal cancer

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION