US20090117038A1 - Breast Endothelial Cell Expression Patterns - Google Patents

Breast Endothelial Cell Expression Patterns Download PDF

Info

Publication number
US20090117038A1
US20090117038A1 US12/269,418 US26941808A US2009117038A1 US 20090117038 A1 US20090117038 A1 US 20090117038A1 US 26941808 A US26941808 A US 26941808A US 2009117038 A1 US2009117038 A1 US 2009117038A1
Authority
US
United States
Prior art keywords
protein
alpha
type
collagen
homolog
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
US12/269,418
Inventor
Saraswati Sukumar
Stephen L. Madden
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.)
Genzyme Corp
Johns Hopkins University
Original Assignee
Genzyme Corp
Johns Hopkins University
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 Genzyme Corp, Johns Hopkins University filed Critical Genzyme Corp
Priority to US12/269,418 priority Critical patent/US20090117038A1/en
Publication of US20090117038A1 publication Critical patent/US20090117038A1/en
Priority to US13/176,222 priority patent/US8568985B2/en
Priority to US14/065,587 priority patent/US20140056911A1/en
Priority to US14/800,457 priority patent/US20150315269A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/39558Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against tumor tissues, cells, antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K51/00Preparations containing radioactive substances for use in therapy or testing in vivo
    • A61K51/02Preparations containing radioactive substances for use in therapy or testing in vivo characterised by the carrier, i.e. characterised by the agent or material covalently linked or complexing the radioactive nucleus
    • A61K51/04Organic compounds
    • A61K51/08Peptides, e.g. proteins, carriers being peptides, polyamino acids, proteins
    • A61K51/10Antibodies or immunoglobulins; Fragments thereof, the carrier being an antibody, an immunoglobulin or a fragment thereof, e.g. a camelised human single domain antibody or the Fc fragment of an antibody
    • A61K51/1018Antibodies or immunoglobulins; Fragments thereof, the carrier being an antibody, an immunoglobulin or a fragment thereof, e.g. a camelised human single domain antibody or the Fc fragment of an antibody against material from animals or humans
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • 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
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5011Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics for testing antineoplastic activity
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/34Identification of a linear epitope shorter than 20 amino acid residues or of a conformational epitope defined by amino acid residues
    • CCHEMISTRY; METALLURGY
    • 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
    • C12Q2600/136Screening for pharmacological compounds

Definitions

  • This invention is related to the area of angiogenesis and anti-angiogenesis. In particular, it relates to genes which are characteristically expressed in breast tumor endothelial cells.
  • a method is provided to aid in diagnosing breast tumors.
  • An expression product (protein or RNA) of at least one gene in a first breast tissue sample suspected of being neoplastic is detected.
  • the at least one gene is selected from the group consisting of hypothetical protein DKFZp434G171; heat shock 70 kDa protein 1A; jagged 1 (Alagille syndrome); cyclin-dependent kinase 3; 6-phosphogluconolactonase; likely homolog of rat and mouse retinoid-inducible serine carboxypeptidase; plasmalemma vesicle associated protein; NADH:ubiquinone oxidoreductase MLRQ subunit homolog; HIF-1 responsive RTP801; ribosomal protein L27; secreted protein, acidic, cysteine-rich (osteonectin); hexokinase 1; ribosomal protein L13a; collagen, type IV, alpha 1; insulin-like growth
  • elegans major histocompatibility complex, class I, C; hypoxia up-regulated 1; complement component 4B; prefoldin 2; cytoskeleton-associated protein 1; Rho GTPase activating protein 4; Homo sapiens clone FLC1492 PRO3121 mRNA, complete cds; transducin-like enhancer of split 2 (E(spl) homolog, Drosophila); ribosomal protein L37; hypothetical protein MGC4677; ESTs, Highly similar to MT1A_HUMAN METALLOTHIONEIN-IA (MT-1A) [ H.
  • TEM11 nidogen (enactin); guanine nucleotide binding protein (G protein), gamma 5; matrix Gla protein; heat shock 105 kD; GNAS complex locus; Homo sapiens cDNA FLJ11658 fis, clone HEMBA1004577; H19, imprinted maternally expressed untranslated mRNA; protein tyrosine phosphatase type IVA, member 3; snail homolog 1 (Drosophila); integrin-binding sialoprotein (bone sialoprotein, bone sialoprotein II); tissue inhibitor of metalloproteinase 1 (erythroid potentiating activity, collagenase inhibitor); peptidylprolyl isomerase B (cyclophilin B); MARCKS-like protein; FAST kinase; protease, serine, 11 (IGF binding); beta-2-microglobulin; delta sleep inducing peptide, immunoreactor; collagen, type IV, alpha 2; immediate
  • Expression of the at least one gene in the first breast tissue sample is compared to expression of the at least one gene in a second breast tissue sample which is normal. Increased expression of the at least one gene in the first breast endothelial tissue sample relative to the second tissue sample identifies the first breast tissue sample as likely to be neoplastic.
  • a method is provided of treating a breast tumor.
  • Cells of the breast tumor are contacted with an antibody.
  • the antibody specifically binds to an extracellular epitope of a protein selected from the group consisting of benzodiazapine receptor (peripheral); cadherin 5, type 2, VE-cadherin (vascular epithelium); calcium channel, voltage-dependent, alpha 1H subunit; CD74 antigen (invariant polypeptide of major histocompatibility complex, class II antigen-associated); CD9 antigen (p24); dysferlin, limb girdle muscular dystrophy 2B (autosomal recessive); ectonucleoside triphosphate diphosphohydrolase 1; G protein-coupled receptor 4; hypothetical protein FLJ20898; hypoxia up-regulated 1; immediate early response 3; interferon, alpha-inducible protein (clone IFI-6-16); jagged 1 (Alagille syndrome); KIAA0152 gene product; Lysosomal-
  • a method for identifying a test compound as a potential anti-cancer or anti-breast tumor drug is contacted with a cell which expresses at least one gene selected from the group consisting of: hypothetical protein DKFZp434G171; heat shock 70 kDa protein 1A; jagged 1 (Alagille syndrome); cyclin-dependent kinase 3; 6-phosphogluconolactonase; likely homolog of rat and mouse retinoid-inducible serine carboxypeptidase; plasmalemma vesicle associated protein; NADH:ubiquinone oxidoreductase MLRQ subunit homolog; HIF-1 responsive RTP801; ribosomal protein L27; secreted protein, acidic, cysteine-rich (osteonectin); hexokinase 1; ribosomal protein L13a; collagen, type IV, alpha 1; insulin-like growth factor binding
  • elegans major histocompatibility complex, class I, C; hypoxia up-regulated 1; complement component 4B; prefoldin 2; cytoskeleton-associated protein 1; Rho GTPase activating protein 4; Homo sapiens clone FLC1492 PRO3121 mRNA, complete cds; transducin-like enhancer of split 2 (E(spl) homolog, Drosophila); ribosomal protein L37; hypothetical protein MGC4677; ESTs, Highly similar to MT1A_HUMAN METALLOTHIONEIN-IA (MT-1A) [ H.
  • Still another embodiment of the invention is a method to induce an immune response to a breast tumor.
  • a protein or nucleic acid encoding a protein is administered to a mammal, preferably a human.
  • the protein is selected from the group consisting of: hypothetical protein DKFZp434G171; heat shock 70 kDa protein 1A; jagged 1 (Alagille syndrome); cyclin-dependent kinase 3; 6-phosphogluconolactonase; likely homolog of rat and mouse retinoid-inducible serine carboxypeptidase; plasmalemma vesicle associated protein; NADH:ubiquinone oxidoreductase MLRQ subunit homolog; HIF-1 responsive RTP801; ribosomal protein L27; secreted protein, acidic, cysteine-rich (osteonectin); hexokinase 1; ribosomal protein L13a; collagen, type IV, alpha 1; insulin-like growth factor
  • elegans major histocompatibility complex, class I, C; hypoxia up-regulated 1; complement component 4B; prefoldin 2; cytoskeleton-associated protein 1; Rho GTPase activating protein 4 ; Homo sapiens clone FLC1492 PRO3121 mRNA, complete cds; transducin-like enhancer of split 2 (E(spl) homolog, Drosophila); ribosomal protein L37; hypothetical protein MGC4677; ESTs, Highly similar to MT1A_HUMAN METALLOTHIONEIN-IA (MT-1A) [ H.
  • TEM11 nidogen (enactin); guanine nucleotide binding protein (G protein), gamma 5; matrix Gla protein; heat shock 105 kD; GNAS complex locus; Homo sapiens cDNA FLJ11658 fis, clone HEMBA1004577; H19, imprinted maternally expressed untranslated mRNA; protein tyrosine phosphatase type IVA, member 3; snail homolog 1 (Drosophila); integrin-binding sialoprotein (bone sialoprotein, bone sialoprotein II); tissue inhibitor of metalloproteinase 1 (erythroid potentiating activity, collagenase inhibitor); peptidylprolyl isomerase B (cyclophilin B); MARCKS-like protein; FAST kinase; protease, serine, 11 (IGF binding); beta-2-microglobulin; delta sleep inducing peptide, immunoreactor; collagen, type IV, alpha 2; immediate
  • the present invention thus provides the art with methods of diagnosing and treating breast tumors.
  • angiogenesis-specific markers that discriminate between non-proliferative and pathologic endothelial cells.
  • angiogenesis-specific markers from other tumor types (colon and/or brain) were found to be expressed in breast tumor endothelium as well.
  • BEMs breast tumor endothelial Markers
  • BEMs that are expressed in both colon and breast tumor epithelium are identified in Table 3.
  • BEMs that are expressed in both brain and breast tumor epithelium are identified in Table 4.
  • BEMs that are expressed in each of brain, colon, and breast tumor epithelium are identified in Table 5.
  • Hs.277477 major histocompatibility complex, class I C 142840 NP_002108 Hs.277704 hypoxia up-regulated 1 601746 NP_006380 Hs.278625 complement component 4B 120820 NP_000583 Hs.298229 prefoldin 2 NP_036526 Hs.31053 cytoskeleton-associated protein 1 601303 NP_001272 Hs.3109 Rho GTPase activating protein 4 300023 NP_001657 Hs.327412 Homo sapiens clone FLC1492 PRO3121 mRNA, complete cds Hs.332173 transducin-like enhancer of split 2 (E(sp1) homolog, 601041 NP_003251 Drosophila ) Hs.337445 ribosomal protein L37 604181 NP_000988 Hs.337986 hypothetical protein MGC4677 NP_443103 Hs.353882
  • Endothelial cells represent only a minor fraction of the total cells within normal or tumor tissues, and only those EC transcripts expressed at the highest levels would be expected to be represented in libraries constructed from unfractionated tissues.
  • the genes described in the current study should therefore provide a valuable resource for basic and clinical studies of human breast angiogenesis in the future.
  • Isolated and purified nucleic acids are those which are not linked to those genes to which they are linked in the human genome. Moreover, they are not present in a mixture such as a library containing a multitude of distinct sequences from distinct genes. They may be, however, linked to other genes such as vector sequences or sequences of other genes to which they are not naturally adjacent.
  • the nucleic acids may represent either the sense or the anti-sense strand.
  • Nucleic acids and proteins although disclosed herein with sequence particularity, may be derived from a single individual. Allelic variants which occur in the population of humans are included within the scope of such nucleic acids and proteins. Those of skill in the art are well able to identify allelic variants as being the same gene or protein. Given a nucleic acid, one of ordinary skill in the art can readily determine an open reading frame present, and consequently the sequence of a polypeptide encoded by the open reading frame and, using techniques well known in the art, express such protein in a suitable host.
  • Proteins comprising such polypeptides can be the naturally occurring proteins, fusion proteins comprising exogenous sequences from other genes from humans or other species, epitope tagged polypeptides, etc. Isolated and purified proteins are not in a cell, and are separated from the normal cellular constituents, such as nucleic acids, lipids, etc. Typically the protein is purified to such an extent that it comprises the predominant species of protein in the composition, such as greater than 50, 60 70, 80, 90, or even 95% of the proteins present.
  • antibodies which specifically bind to the proteins.
  • Such antibodies can be monoclonal or polyclonal. They can be chimeric, humanized, or totally human. Any functional fragment or derivative of an antibody can be used including Fab, Fab′, Fab2, Fab′2, and single chain variable regions. So long as the fragment or derivative retains specificity of binding for the endothelial marker protein it can be used.
  • Antibodies can be tested for specificity of binding by comparing binding to appropriate antigen to binding to irrelevant antigen or antigen mixture under a given set of conditions. If the antibody binds to the appropriate antigen at least 2, 5, 7, and preferably 10 times more than to irrelevant antigen or antigen mixture then it is considered to be specific.
  • fully human antibody sequences are made in a transgenic mouse which has been engineered to express human heavy and light chain antibody genes. Multiple strains of such transgenic mice have been made which can produce different classes of antibodies. B cells from transgenic mice which are producing a desirable antibody can be fused to make hybridoma cell lines for continuous production of the desired antibody. See for example, Nina D. Russel, Jose R. F. Corvalan, Michael L. Gallo, C. Geigery Davis, Liise-Anne Pirofski.
  • Antibody engineering via genetic engineering of the mouse XenoMouse strains are a vehicle for the facile generation of therapeutic human monoclonal antibodies Journal of Immunological Methods 231 11-23, 1999; Yang X-D, Corvalan J R F, Wang P, Roy CM-N and Davis CG. Fully Human Anti-interleukin-8 Monoclonal Antibodies: Potential Therapeutics for the Treatment of Inflammatory Disease States. Journal of Leukocyte Biology Vol. 66, pp 401-410 (1999); Yang X-D, Jia X-C, Corvalan J R F, Wang P, C G Davis and Jakobovits A.
  • Monoclonal Antibodies The Evolution from '80s Magic Bullets To Mature, Mainstream Applications as Clinical Therapeutics. Genetic Engineering News Vol. 17, Number 14 (March 1997); Mendez M, Green L, Corvalan J, Jia X-C, Maynard-Currie C, Yang X-d, Gallo M, Louie D, Lee D, Erickson K, Luna J, Roy C, Abderrahim H, Kirschenbaum F, Noguchi M, Smith D, Fukushima A, Hales J, Finer M, Davis C, Zsebo K, Jakobovits A. Functional transplant of megabase human immunoglobulin loci recapitulates human antibody response in mice. Nature Genetics Vol.
  • Antibodies can also be made using phage display techniques. Such techniques can be used to isolate an initial antibody or to generate variants with altered specificity or avidity characteristics. Single chain Fv can also be used as is convenient. They can be made from vaccinated transgenic mice, if desired. Antibodies can be produced in cell culture, in phage, or in various animals, including but not limited to cows, rabbits, goats, mice, rats, hamsters, guinea pigs, sheep, dogs, cats, monkeys, chimpanzees, apes.
  • Antibodies can be labeled with a detectable moiety such as a radioactive atom, a chromophore, a fluorophore, or the like. Such labeled antibodies can be used for diagnostic techniques, either in vivo, or in an isolated test sample. Antibodies can also be conjugated, for example, to a pharmaceutical agent, such as chemotherapeutic drug or a toxin. They can be linked to a cytokine, to a ligand, to another antibody.
  • a detectable moiety such as a radioactive atom, a chromophore, a fluorophore, or the like.
  • Such labeled antibodies can be used for diagnostic techniques, either in vivo, or in an isolated test sample.
  • Antibodies can also be conjugated, for example, to a pharmaceutical agent, such as chemotherapeutic drug or a toxin. They can be linked to a cytokine, to a ligand, to another antibody.
  • Suitable agents for coupling to antibodies to achieve an anti-tumor effect include cytokines, such as interleukin 2 (IL-2) and Tumor Necrosis Factor (TNF); photosensitizers, for use in photodynamic therapy, including aluminum (III) phthalocyanine tetrasulfonate, hematoporphyrin, and phthalocyanine; radionuclides, such as iodine-131 ( 131 I), yttrium-90 ( 90 Y), bismuth-212 ( 212 Bi), bismuth-213 ( 213 Bi), technetium-99m ( 99m Tc), rhenium-186 ( 186 Re), and rhenium-188 ( 188 Re); antibiotics, such as doxorubicin, adriamycin, daunorubicin, methotrexate, daunomycin, neocarzinostatin, and carboplatin; bacterial, plant, and other toxins, such as diphtheria
  • the antibodies may be cytotoxic on their own, or they may be used to deliver cytotoxic agents to particular locations in the body.
  • the antibodies can be administered to individuals in need thereof as a form of passive immunization.
  • Characterization of extracellular regions for the cell surface and secreted proteins from the protein sequence is based on the prediction of signal sequence, transmembrane domains and functional domains.
  • Antibodies are preferably specifically immunoreactive with membrane associated proteins, particularly to extracellular domains of such proteins or to secreted proteins. Such targets are readily accessible to antibodies, which typically do not have access to the interior of cells or nuclei. However, in some applications, antibodies directed to intracellular proteins may be useful as well. Moreover, for diagnostic purposes, an intracellular protein may be an equally good target since cell lysates may be used rather than a whole cell assay.
  • Computer programs can be used to identify extracellular domains of proteins whose sequences are known. Such programs include SMART software (Schultz et al., Proc. Natl. Acad. Sci. USA 95: 5857-5864, 1998) and Pfam software (BaBEMan et al., Nucleic acids Res. 28: 263-266, 2000) as well as PSORTII. Typically such programs identify transmembrane domains; the extracellular domains are identified as immediately adjacent to the transmembrane domains. Prediction of extracellular regions and the signal cleavage sites are only approximate. It may have a margin of error + or ⁇ 5 residues.
  • Putative functions or functional domains of novel proteins can be inferred from homologous regions in the database identified by BLAST searches (Altschul et. al. Nucleic Acid Res. 25: 3389-3402, 1997) and/or from a conserved domain database such as Pfam (BaBEMan et. al, Nucleic Acids Res. 27:260-262 1999) BLOCKS (Henikoff, et. al, Nucl. Acids Res. 28:228-230, 2000) and SMART (Ponting, et. al, Nucleic Acid Res. 27,229-232, 1999).
  • Extracellular domains include regions adjacent to a transmembrane domain in a single transmembrane domain protein (out-in or type I class).
  • the extracellular domain also includes those regions between two adjacent transmembrane domains (in-out and out-in).
  • regions following the transmembrane domain is generally extracellular.
  • Secreted proteins on the other hand do not have a transmembrane domain and hence the whole protein is considered as extracellular.
  • Membrane associated proteins can be engineered to delete the transmembrane domains, thus leaving the extracellular portions which can bind to ligands.
  • Such soluble forms of transmembrane receptor proteins can be used to compete with natural forms for binding to ligand.
  • Such soluble forms act as inhibitors and can be used therapeutically as anti-angiogenic agents, as diagnostic tools for the quantification of natural ligands, and in assays for the identification of small molecules which modulate or mimic the activity of a BEM:ligand complex.
  • the endothelial markers themselves can be used as vaccines to raise an immune response in the vaccinated animal or human.
  • a protein, or immunogenic fragment of such protein corresponding to the intracellular, extracellular or secreted BEM of interest is administered to a subject.
  • the immogenic agent may be provided as a purified preparation or in an appropriately expressing cell.
  • the administration may be direct, by the delivery of the immunogenic agent to the subject, or indirect, through the delivery of a nucleic acid encoding the immunogenic agent under conditions resulting in the expression of the immunogenic agent of interest in the subject.
  • the BEM of interest may be delivered in an expressing cell, such as a purified population of breast tumor endothelial cells or a population of fused breast tumor endothelial and dendritic cells.
  • Nucleic acids encoding the BEM of interest may be delivered in a viral or non-viral delivery vector or vehicle.
  • Non-human sequences encoding the human BEM of interest or other mammalian homolog can be used to induce the desired immunologic response in a human subject.
  • mouse, rat or other ortholog sequences can be obtained from the literature or using techniques well within the skill of the art.
  • Endothelial cells can be identified using the markers which are disclosed herein as being endothelial cell specific. Antibodies specific for such markers can be used to identify such cells, by contacting the antibodies with a population of cells containing some endothelial cells. The presence of cross-reactive material with the antibodies identifies particular cells as endothelial. Similarly, lysates of cells can be tested for the presence of cross-reactive material. Any known format or technique for detecting cross-reactive material can be used including, immunoblots, radioimmunoassay, ELISA, immunoprecipitation, and immunohistochemistry. In addition, nucleic acid probes for these markers can also be used to identify endothelial cells. Any hybridization technique known in the art including Northern blotting, RT-PCR, microarray hybridization, and in situ hybridization can be used.
  • Endothelial cells can also be made using the antibodies to endothelial markers of the invention.
  • the antibodies can be used to purify cell populations according to any technique known in the art, including but not limited to fluorescence activated cell sorting. Such techniques permit the isolation of populations which are at least 50, 60, 70, 80, 90, 92, 94, 95, 96, 97, 98, and even 99% the type of endothelial cell desired, whether normal, tumor, or pan-endothelial.
  • Antibodies can be used to both positively select and negatively select such populations. Preferably at least 1, 5, 10, 15, 20, or 25 of the appropriate markers are expressed by the endothelial cell population.
  • Populations of endothelial cells made as described herein, can be used for screening drugs to identify those suitable for inhibiting the growth of tumors by virtue of inhibiting the growth of the tumor vasculature.
  • Endothelial cells made as described herein can be used for screening candidate drugs to identify those suitable for modulating angiogenesis, such as for inhibiting the growth of tumors by virtue of inhibiting the growth of endothelial cells, such as inhibiting the growth of the tumor or other undesired vasculature, or alternatively, to promote the growth of endothelial cells and thus stimulate the growth of new or additional large vessel or microvasculature.
  • Inhibiting the growth of endothelial cells means either regression of vasculature which is already present, or the slowing or the absence of the development of new vascularization in a treated system as compared with a control system.
  • By stimulating the growth of endothelial cells one can influence development of new (neovascularization) or additional vasculature development (revascularization).
  • a variety of model screening systems are available in which to test the angiogenic and/or anti-angiogenic properties of a given candidate drug. Typical tests involve assays measuring the endothelial cell response, such as proliferation, migration, differentiation and/or intracellular interaction with a given candidate drug. By such tests, one can study the signals and effects of the test stimuli.
  • Some common screens involve measurement of the inhibition of heparanase, endothelial tube formation on Matrigel, scratch induced motility of endothelial cells, platelet-derived growth factor driven proliferation of vascular smooth muscle cells, and the rat aortic ring assay (which provides an advantage of capillary formation rather than just one cell type).
  • Drugs can be screened for the ability to mimic or modulate, inhibit or stimulate, growth of tumor endothelium cells and/or normal endothelial cells. Drugs can be screened for the ability to inhibit tumor endothelium growth but not normal endothelium growth or survival.
  • human cell populations such as normal endothelium populations or breast tumor endothelial cell populations, can be contacted with test substances and the expression of breast tumor endothelial markers and/or normal endothelial markers determined.
  • Test substances that decrease the expression of breast tumor endothelial markers are candidates for inhibiting angiogenesis and the growth of tumors. In cases where the activity of a BEM is known, agents can be screened for their ability to decrease or increase the activity.
  • the identification of drug candidates capable of binding to the BEM receptors found at the cell surface For those breast tumor endothelial markers identified as containing transmembrane regions, it is desirable to identify drug candidates capable of binding to the BEM receptors found at the cell surface. For some applications, the identification of drug candidates capable of blocking the BEM receptor from its native ligand will be desired. For some applications, the identification of a drug candidate capable of binding to the BEM receptor may be used as a means to deliver a therapeutic or diagnostic agent. For other applications, the identification of drug candidates capable of mimicking the activity of the native ligand will be desired. Thus, by manipulating the binding of a transmembrane BEM receptor:ligand complex, one may be able to promote or inhibit further development of endothelial cells and hence, vascularization.
  • Expression can be monitored according to any convenient method. Protein or mRNA can be monitored. Any technique known in the art for monitoring specific genes' expression can be used, including but not limited to ELISAs, SAGE, microarray hybridization, Western blots. Changes in expression of a single marker may be used as a criterion for significant effect as a potential pro-angiogenic, anti-angiogenic or anti-tumor agent. However, it also may be desirable to screen for test substances that are able to modulate the expression of at least 5, 10, 15, or 20 of the relevant markers, such as the tumor or normal endothelial markers. Inhibition of BEM protein activity can also be used as a drug screen.
  • Test substances for screening can come from any source. They can be libraries of natural products, combinatorial chemical libraries, biological products made by recombinant libraries, etc.
  • the source of the test substances is not critical to the invention.
  • the present invention provides means for screening compounds and compositions that may previously have been overlooked in other screening schemes.
  • Nucleic acids and the corresponding encoded proteins of the markers of the present invention can be used therapeutically in a variety of modes. BEMs can be used to stimulate the growth of vasculature, such as for wound healing or to circumvent a blocked vessel.
  • the nucleic acids and encoded proteins can be administered by any means known in the art. Such methods include, using liposomes, nanospheres, viral vectors, non-viral vectors comprising polycations, etc.
  • Suitable viral vectors include adenovirus, retroviruses, and Sindbis virus.
  • Administration modes can be any known in the art, including parenteral, intravenous, intramuscular, intraperitoneal, topical, intranasal, intrarectal, intrabronchial, etc.
  • Specific biological antagonists of BEMs can also be used to therapeutic benefit.
  • antibodies, T cells specific for a BEM, antisense to a BEM, interference RNA to a BEM, and ribozymes specific for a BEM can be used to restrict, inhibit, reduce, and/or diminish tumor or other abnormal or undesirable vasculature growth.
  • Such antagonists can be administered as is known in the art for these classes of antagonists generally.
  • Anti-angiogenic drugs and agents can be used to inhibit tumor growth, as well as to treat diabetic retinopathy, rheumatoid arthritis, psoriasis, polycystic kidney disease (PKD), and other diseases requiring angiogenesis for their pathologies.
  • Mouse counterparts to human BEMs can be used in mouse cancer models or in cell lines or in vitro to evaluate potential anti-angiogenic or anti-tumor compounds or therapies. Their expression can be monitored as an indication of effect.
  • Mouse BEMs can be used as antigens for raising antibodies which can be tested in mouse tumor models.
  • Mouse BEMs with transmembrane domains are particularly preferred for this purpose.
  • Mouse BEMs can also be used as vaccines to raise an immunological response in a human to the human ortholog.
  • BEM proteins Function of BEM proteins was determined using bioinformatics tools. BEMs that are putative functional receptors with short cytoplasmic tails make particularly interesting targets.
  • Protein kinases were identified among the BEMs. These are particularly good druggable targets, especially for small molecules.
  • OMIMID Protein Hs.91143 jagged 1 (Alagille syndrome) 601920 NP_000205 Hs.119206 insulin-like growth factor 602867 NP_001544 binding protein 7 Hs.1516 insulin-like growth factor 146733 NP_001543 binding protein 4 Hs.211573 heparan sulfate proteoglycan 2 142461 NP_005520 (perlecan) Hs.75111 protease, serine, 11 (IGF 602194 NP_002766 binding) Hs.8546 Notch homolog 3 ( Drosophila ) 600276 NP_000426
  • Phosphatases like kinases, are readily amenable to screening for inhibitors, especially small molecule inhibitors:
  • the cellular location of the BEMs was determined to be either cytoplasmic, extracellular, membrane, or nuclear, as shown below.
  • NP_000705 3 107-129, 78-100, OUT 133-155 Hs.76206 cadherin 5, type 2, VE-cadherin (vascular epithelium)
  • NP_001786 1 598-620 Unsure Hs.122359 calcium channel, voltage-dependent, alpha 1H subunit NP_066921 19 1370-1392, IN 1614-1636, 1533-1555, 141-163, 915-937, 396-418, 1651-1673, 1745-1767, 990-1012, 234-256, 1430-1452, 1333-1355, 1680-1702, 855-877, 1295-1316, 826-848, 100-122, 1840-1862, 364-386 Hs.84298 CD74 antigen (invariant polypeptide of major NP_004346 1 49-71 IN histocomp
  • Cytoplasmic proteins Unigene ID Function OMIMID Protein Hs.184367 Ca2+-promoted Ras inactivator BAA25464 Hs.2575 calpain 1, (mu/l) large subunit 114220 NP_005177 Hs.100009 cyclin-dependent kinase 3 123828 Hs.31053 cytoskeleton-associated protein 1 601303 NP_001272 Hs.82646 DnaJ (Hsp40) homolog, subfamily B, member 1 604572 NP_006136 Hs.169476 glyceraldehyde-3-phosphate dehydrogenase 138400 NP_002037 Hs.77890 guanylate cyclase 1, soluble, beta 3 139397 NP_000848 Hs.36927 heat shock 105 Kd NP_006635 Hs.1197 heat shock 10 kDa protein 1 (chaperonin 10) 600141 NP_002148 Hs.

Abstract

To gain a better understanding of breast tumor angiogenesis, breast endothelial cells (ECs) were isolated and evaluated for gene expression patterns. When transcripts from breast ECs derived from normal and malignant breast tissues were compared, genes that were specifically elevated in tumor-associated breast endothelium were revealed. These results confirm that neoplastic and normal endothelium in human breast are distinct at the molecular level, and have significant implications for the development of anti-angiogenic therapies in the future.

Description

  • This application is continuation application of Ser. No. 10/551,217 filed Dec. 12, 2006, which is a National Stage application of co-pending PCT application PCT/US2004/009704 filed 31 Mar. 2004, which was published in the English language under PCT Article 21(2) on Oct. 28, 2004, and claims the benefit of the U.S. Provisional Application No. 60/458,960 filed 1 Apr. 2003.
  • TECHNICAL FIELD OF THE INVENTION
  • This invention is related to the area of angiogenesis and anti-angiogenesis. In particular, it relates to genes which are characteristically expressed in breast tumor endothelial cells.
  • BACKGROUND OF THE INVENTION
  • To date, global gene expression profiles from endothelial cell-specific populations is limited to normal and tumorigenic colon tissue [St Croix, 2000]. There is a need in the art for analysis of endothelial cells from other tissue, so that diagnostic and therapeutic agents for non-colonic tumors can be developed.
  • SUMMARY OF THE INVENTION
  • According to one embodiment of the invention a method is provided to aid in diagnosing breast tumors. An expression product (protein or RNA) of at least one gene in a first breast tissue sample suspected of being neoplastic is detected. The at least one gene is selected from the group consisting of hypothetical protein DKFZp434G171; heat shock 70 kDa protein 1A; jagged 1 (Alagille syndrome); cyclin-dependent kinase 3; 6-phosphogluconolactonase; likely homolog of rat and mouse retinoid-inducible serine carboxypeptidase; plasmalemma vesicle associated protein; NADH:ubiquinone oxidoreductase MLRQ subunit homolog; HIF-1 responsive RTP801; ribosomal protein L27; secreted protein, acidic, cysteine-rich (osteonectin); hexokinase 1; ribosomal protein L13a; collagen, type IV, alpha 1; insulin-like growth factor binding protein 7; collagen, type III, alpha 1 (Ehlers-Danlos syndrome type IV, autosomal dominant); heat shock 10 kDa protein 1 (chaperonin 10); calcium channel, voltage-dependent, alpha 1H subunit; CD9 antigen (p24); TEM17; TEM13, Thy-1 cell surface antigen; Tax interaction protein 1; dysferlin, limb girdle muscular dystrophy 2B (autosomal recessive); hypothetical protein MGC34648; putative translation initiation factor; insulin-like growth factor binding protein 4; matrix metalloproteinase 9 (gelatinase B, 92 kDa gelatinase, 92 kDa type IV collagenase); heterogeneous nuclear ribonucleoprotein R; bHLH factor Hes4; collagen, type VI, alpha 2; T-box 2; glyceraldehyde-3-phosphate dehydrogenase; G protein-coupled receptor 4; collagen, type I, alpha 1; ras-related C3 botulinum toxin substrate 1 (rho family, small GTP binding protein Rac1); ribosomal protein, large, P1; TEM10, COL1A2 involved in tissue remodeling; heat shock 70 kDa protein 8; KIAA0152 gene product; Ca2+-promoted Ras inactivator; serine/arginine repetitive matrix 2; hypoxia-inducible factor 1, alpha subunit (basic helix-loop-helix transcription factor); benzodiazapine receptor (peripheral); ectonucleoside triphosphate diphosphohydrolase 1; heparan sulfate proteoglycan 2 (perlecan); fibromodulin; hairy/enhancer-of-split related with YRPW motif 1; collagen, type V, alpha 3; hairy/enhancer-of-split related with YRPW motif-like; hypothetical protein MGC2731; amino-terminal enhancer of split; mitogen-activated protein kinase 9; regulator of G-protein signalling 5; prothymosin, alpha (gene sequence 28); tubulin, beta, 2; protease, serine, 23; hypothetical protein FLJ20898; calpain 1, (mu/I) large subunit; interferon, alpha-inducible protein (clone IFI-6-16); ESTs, Weakly similar to T25031 hypothetical protein T20D3.3—Caenorhabditis elegans [C. elegans]; major histocompatibility complex, class I, C; hypoxia up-regulated 1; complement component 4B; prefoldin 2; cytoskeleton-associated protein 1; Rho GTPase activating protein 4; Homo sapiens clone FLC1492 PRO3121 mRNA, complete cds; transducin-like enhancer of split 2 (E(spl) homolog, Drosophila); ribosomal protein L37; hypothetical protein MGC4677; ESTs, Highly similar to MT1A_HUMAN METALLOTHIONEIN-IA (MT-1A) [H. sapiens]; TEM11, nidogen (enactin); guanine nucleotide binding protein (G protein), gamma 5; matrix Gla protein; heat shock 105 kD; GNAS complex locus; Homo sapiens cDNA FLJ11658 fis, clone HEMBA1004577; H19, imprinted maternally expressed untranslated mRNA; protein tyrosine phosphatase type IVA, member 3; snail homolog 1 (Drosophila); integrin-binding sialoprotein (bone sialoprotein, bone sialoprotein II); tissue inhibitor of metalloproteinase 1 (erythroid potentiating activity, collagenase inhibitor); peptidylprolyl isomerase B (cyclophilin B); MARCKS-like protein; FAST kinase; protease, serine, 11 (IGF binding); beta-2-microglobulin; delta sleep inducing peptide, immunoreactor; collagen, type IV, alpha 2; immediate early response 3; cadherin 5, type 2, VE-cadherin (vascular epithelium); RGC32 protein; guanylate cyclase 1, soluble, beta 3; major histocompatibility complex, class I, B; ribonuclease, RNase A family, 1 (pancreatic); collagen, type XVIII, alpha 1; v-jun sarcoma virus 17 oncogene homolog (avian); Homo sapiens mRNA; cDNA DKFZp686G1610 (from clone DKFZp686G1610); nucleolin; lectin, galactoside-binding, soluble, 3 binding protein; Lysosomal-associated multispanning membrane protein-5; ribosomal protein S16; guanine nucleotide binding protein (G protein), gamma 12; serine (or cysteine) proteinase inhibitor, clade E (nexin, plasminogen activator inhibitor type 1), member 1; biglycan; DnaJ (Hsp40) homolog, subfamily B, member 1; tumor rejection antigen (gp96) 1; interferon, alpha-inducible protein (clone IFI-15K); solute carrier family 21 (prostaglandin transporter), member 2; CD74 antigen (invariant polypeptide of major histocompatibility complex, class II antigen-associated); serum/glucocorticoid regulated kinase; mitogen-activated protein kinase; receptor (calcitonin) activity modifying protein 3; sema domain, immunoglobulin domain (Ig); benzodiazapine receptor (peripheral)—mitochondrial; C1 domain-containing phosphatase & tensin-like; and Notch homolog 3 (Drosophila). Expression of the at least one gene in the first breast tissue sample is compared to expression of the at least one gene in a second breast tissue sample which is normal. Increased expression of the at least one gene in the first breast endothelial tissue sample relative to the second tissue sample identifies the first breast tissue sample as likely to be neoplastic.
  • According to another embodiment of the invention a method is provided of treating a breast tumor. Cells of the breast tumor are contacted with an antibody. The antibody specifically binds to an extracellular epitope of a protein selected from the group consisting of benzodiazapine receptor (peripheral); cadherin 5, type 2, VE-cadherin (vascular epithelium); calcium channel, voltage-dependent, alpha 1H subunit; CD74 antigen (invariant polypeptide of major histocompatibility complex, class II antigen-associated); CD9 antigen (p24); dysferlin, limb girdle muscular dystrophy 2B (autosomal recessive); ectonucleoside triphosphate diphosphohydrolase 1; G protein-coupled receptor 4; hypothetical protein FLJ20898; hypoxia up-regulated 1; immediate early response 3; interferon, alpha-inducible protein (clone IFI-6-16); jagged 1 (Alagille syndrome); KIAA0152 gene product; Lysosomal-associated multispanning membrane protein-5; major histocompatibility complex, class I, B; major histocompatibility complex, class I, C; NADH:ubiquinone oxidoreductase MLRQ subunit homolog; Notch homolog 3 (Drosophila); plasmalemma vesicle associated protein; solute carrier family 21 (prostaglandin transporter), member 2; TEM13, Thy-1 cell surface antigen; receptor (calcitonin) activity modifying protein 3; sema domain, immunoglobulin domain (Ig); benzodiazapine receptor (peripheral)-mitochondrial; and TEM17. Immune destruction of cells of the breast tumor is thereby triggered.
  • According to still another embodiment of the invention a method is provided for identifying a test compound as a potential anti-cancer or anti-breast tumor drug. A test compound is contacted with a cell which expresses at least one gene selected from the group consisting of: hypothetical protein DKFZp434G171; heat shock 70 kDa protein 1A; jagged 1 (Alagille syndrome); cyclin-dependent kinase 3; 6-phosphogluconolactonase; likely homolog of rat and mouse retinoid-inducible serine carboxypeptidase; plasmalemma vesicle associated protein; NADH:ubiquinone oxidoreductase MLRQ subunit homolog; HIF-1 responsive RTP801; ribosomal protein L27; secreted protein, acidic, cysteine-rich (osteonectin); hexokinase 1; ribosomal protein L13a; collagen, type IV, alpha 1; insulin-like growth factor binding protein 7; collagen, type III, alpha 1 (Ehlers-Danlos syndrome type IV, autosomal dominant); heat shock 10 kDa protein 1 (chaperonin 10); calcium channel, voltage-dependent, alpha 1H subunit; CD9 antigen (p24); TEM17; TEM13, Thy-1 cell surface antigen; Tax interaction protein 1; dysferlin, limb girdle muscular dystrophy 2B (autosomal recessive); hypothetical protein MGC34648; putative translation initiation factor; insulin-like growth factor binding protein 4; matrix metalloproteinase 9 (gelatinase B, 92 kDa gelatinase, 92 kDa type IV collagenase); heterogeneous nuclear ribonucleoprotein R; bHLH factor Hes4; collagen, type VI, alpha 2; T-box 2; glyceraldehyde-3-phosphate dehydrogenase; G protein-coupled receptor 4; collagen, type I, alpha 1; ras-related C3 botulinum toxin substrate 1 (rho family, small GTP binding protein Rac1); ribosomal protein, large, P1; TEM10, COL1A2 involved in tissue remodeling; heat shock 70 kDa protein 8; KIAA0152 gene product; Ca2+-promoted Ras inactivator; serine/arginine repetitive matrix 2; hypoxia-inducible factor 1, alpha subunit (basic helix-loop-helix transcription factor); benzodiazapine receptor (peripheral); ectonucleoside triphosphate diphosphohydrolase 1; heparan sulfate proteoglycan 2 (perlecan); fibromodulin; hairy/enhancer-of-split related with YRPW motif 1; collagen, type V, alpha 3; hairy/enhancer-of-split related with YRPW motif-like; hypothetical protein MGC2731; amino-terminal enhancer of split; mitogen-activated protein kinase 9; regulator of G-protein signalling 5; prothymosin, alpha (gene sequence 28); tubulin, beta, 2; protease, serine, 23; hypothetical protein FLJ20898; calpain 1, (mu/I) large subunit; interferon, alpha-inducible protein (clone IFI-6-16); ESTs, Weakly similar to T25031 hypothetical protein T20D3.3—Caenorhabditis elegans [C. elegans]; major histocompatibility complex, class I, C; hypoxia up-regulated 1; complement component 4B; prefoldin 2; cytoskeleton-associated protein 1; Rho GTPase activating protein 4; Homo sapiens clone FLC1492 PRO3121 mRNA, complete cds; transducin-like enhancer of split 2 (E(spl) homolog, Drosophila); ribosomal protein L37; hypothetical protein MGC4677; ESTs, Highly similar to MT1A_HUMAN METALLOTHIONEIN-IA (MT-1A) [H. sapiens]; TEM11, nidogen (enactin); guanine nucleotide binding protein (G protein), gamma 5; matrix Gla protein; heat shock 105 kD; GNAS complex locus; Homo sapiens cDNA FLJ11658 fis, clone HEMBA1004577; H19, imprinted maternally expressed untranslated mR NA; protein tyrosine phosphatase type IVA, member 3; snail homolog 1 (Drosophila); integrin-binding sialoprotein (bone sialoprotein, bone sialoprotein II); tissue inhibitor of metalloproteinase 1 (erythroid potentiating activity, collagenase inhibitor); peptidylprolyl isomerase B (cyclophilin B); MARCKS-like protein; FAST kinase; protease, serine, 11 (IGF binding); beta-2-microglobulin; delta sleep inducing peptide, immunoreactor; collagen, type IV, alpha 2; immediate early response 3; cadherin 5, type 2, VE-cadherin (vascular epithelium); RGC32 protein; guanylate cyclase 1, soluble, beta 3; major histocompatibility complex, class I, B; ribonuclease, RNase A family, 1 (pancreatic); collagen, type XVIII, alpha 1; v-jun sarcoma virus 17 oncogene homolog (avian); Homo sapiens mRNA; cDNA DKFZp686G 1610 (from clone DKFZp686G 1610); nucleolin; lectin, galactoside-binding, soluble, 3 binding protein; Lysosomal-associated multispanning membrane protein-5; ribosomal protein S16; guanine nucleotide binding protein (G protein), gamma 12; serine (or cysteine) proteinase inhibitor, clade E (nexin, plasminogen activator inhibitor type 1), member 1; biglycan; DnaJ (Hsp40) homolog, subfamily B, member 1; tumor rejection antigen (gp96) 1; interferon, alpha-inducible protein (clone IFI-15K); solute carrier family 21 (prostaglandin transporter), member 2; CD74 antigen (invariant polypeptide of major histocompatibility complex, class II antigen-associated); serum/glucocorticoid regulated kinase; mitogen-activated protein kinase; receptor (calcitonin) activity modifying protein 3; sema domain, immunoglobulin domain (Ig); benzodiazapine receptor (peripheral)—mitochondrial; C1 domain-containing phosphatase & tensin-like; and Notch homolog 3 (Drosophila). An expression product of the at least one gene is monitored. The test compound is identified as a potential anti-cancer drug if it decreases the expression of the at least one gene.
  • Still another embodiment of the invention is a method to induce an immune response to a breast tumor. A protein or nucleic acid encoding a protein is administered to a mammal, preferably a human. The protein is selected from the group consisting of: hypothetical protein DKFZp434G171; heat shock 70 kDa protein 1A; jagged 1 (Alagille syndrome); cyclin-dependent kinase 3; 6-phosphogluconolactonase; likely homolog of rat and mouse retinoid-inducible serine carboxypeptidase; plasmalemma vesicle associated protein; NADH:ubiquinone oxidoreductase MLRQ subunit homolog; HIF-1 responsive RTP801; ribosomal protein L27; secreted protein, acidic, cysteine-rich (osteonectin); hexokinase 1; ribosomal protein L13a; collagen, type IV, alpha 1; insulin-like growth factor binding protein 7; collagen, type III, alpha 1 (Ehlers-Danlos syndrome type IV, autosomal dominant); heat shock 10 kDa protein 1 (chaperonin 10); calcium channel, voltage-dependent, alpha 1H subunit; CD9 antigen (p24); TEM17; TEM13, Thy-1 cell surface antigen; Tax interaction protein 1; dysferlin, limb girdle muscular dystrophy 2B (autosomal recessive); hypothetical protein MGC34648; putative translation initiation factor; insulin-like growth factor binding protein 4; matrix metalloproteinase 9 (gelatinase B, 92 kDa gelatinase, 92 kDa type IV collagenase); heterogeneous nuclear ribonucleoprotein R; bHLH factor Hes4; collagen, type VI, alpha 2; T-box 2; glyceraldehyde-3-phosphate dehydrogenase; G protein-coupled receptor 4; collagen, type I, alpha 1; ras-related C3 botulinum toxin substrate 1 (rho family, small GTP binding protein Rac1); ribosomal protein, large, P1; TEM10, COL1A2 involved in tissue remodeling; heat shock 70 kDa protein 8; KIAA0152 gene product; Ca2+-promoted Ras inactivator; serine/arginine repetitive matrix 2; hypoxia-inducible factor 1, alpha subunit (basic helix-loop-helix transcription factor); benzodiazapine receptor (peripheral); ectonucleoside triphosphate diphosphohydrolase 1; heparan sulfate proteoglycan 2 (perlecan); fibromodulin; hairy/enhancer-of-split related with YRPW motif 1; collagen, type V, alpha 3; hairy/enhancer-of-split related with YRPW motif-like; hypothetical protein MGC2731; amino-terminal enhancer of split; mitogen-activated protein kinase 9; regulator of G-protein signalling 5; prothymosin, alpha (gene sequence 28); tubulin, beta, 2; protease, serine, 23; hypothetical protein FLJ20898; calpain 1, (mu/I) large subunit; interferon, alpha-inducible protein (clone IFI-6-16); ESTs, Weakly similar to T25031 hypothetical protein T20D3.3—Caenorhabditis elegans [C. elegans]; major histocompatibility complex, class I, C; hypoxia up-regulated 1; complement component 4B; prefoldin 2; cytoskeleton-associated protein 1; Rho GTPase activating protein 4; Homo sapiens clone FLC1492 PRO3121 mRNA, complete cds; transducin-like enhancer of split 2 (E(spl) homolog, Drosophila); ribosomal protein L37; hypothetical protein MGC4677; ESTs, Highly similar to MT1A_HUMAN METALLOTHIONEIN-IA (MT-1A) [H. sapiens]; TEM11, nidogen (enactin); guanine nucleotide binding protein (G protein), gamma 5; matrix Gla protein; heat shock 105 kD; GNAS complex locus; Homo sapiens cDNA FLJ11658 fis, clone HEMBA1004577; H19, imprinted maternally expressed untranslated mRNA; protein tyrosine phosphatase type IVA, member 3; snail homolog 1 (Drosophila); integrin-binding sialoprotein (bone sialoprotein, bone sialoprotein II); tissue inhibitor of metalloproteinase 1 (erythroid potentiating activity, collagenase inhibitor); peptidylprolyl isomerase B (cyclophilin B); MARCKS-like protein; FAST kinase; protease, serine, 11 (IGF binding); beta-2-microglobulin; delta sleep inducing peptide, immunoreactor; collagen, type IV, alpha 2; immediate early response 3; cadherin 5, type 2, VE-cadherin (vascular epithelium); RGC32 protein; guanylate cyclase 1, soluble, beta 3; major histocompatibility complex, class I, B; ribonuclease, RNase A family, 1 (pancreatic); collagen, type XVIII, alpha 1; v-jun sarcoma virus 17 oncogene homolog (avian); Homo sapiens mRNA; cDNA DKFZp686G1610 (from clone DKFZp686G1610); nucleolin; lectin, galactoside-binding, soluble, 3 binding protein; Lysosomal-associated multispanning membrane protein-5; ribosomal protein S16; guanine nucleotide binding protein (G protein), gamma 12; serine (or cysteine) proteinase inhibitor, clade E (nexin, plasminogen activator inhibitor type 1), member 1; biglycan; DnaJ (Hsp40) homolog, subfamily B, member 1; tumor rejection antigen (gp96) 1; interferon, alpha-inducible protein (clone IFI-15K); solute carrier family 21 (prostaglandin transporter), member 2; CD74 antigen (invariant polypeptide of major histocompatibility complex, class II antigen-associated); serum/glucocorticoid regulated kinase; mitogen-activated protein kinase; receptor (calcitonin) activity modifying protein 3; sema domain, immunoglobulin domain (Ig); benzodiazapine receptor (peripheral)—mitochondrial; C1 domain-containing phosphatase & tensin-like; and Notch homolog 3 (Drosophila). An immune response to the protein is thereby induced.
  • The present invention thus provides the art with methods of diagnosing and treating breast tumors.
  • DETAILED DESCRIPTION OF THE INVENTION
  • Using SAGE (Serial Analysis of Gene Expression) profiling, the present inventors were able to identify previously unrecognized, angiogenesis-specific markers that discriminate between non-proliferative and pathologic endothelial cells. In addition, a set of previously identified angiogenesis-specific markers from other tumor types (colon and/or brain) were found to be expressed in breast tumor endothelium as well. We identified 111 human genes that were expressed at significantly higher levels in breast tumor endothelium than in normal breast endothelium. See Table 1. Additional such genes which can be used similarly to the 11 human genes are shown in Table 2. We have named these markers BEMs (breast tumor endothelial Markers). BEMs that are expressed in both colon and breast tumor epithelium are identified in Table 3. BEMs that are expressed in both brain and breast tumor epithelium are identified in Table 4. BEMs that are expressed in each of brain, colon, and breast tumor epithelium are identified in Table 5.
  • TABLE 1
    111 Breast Markers
    Unigene ID Function OMIMID Protein
    Hs.8728 hypothetical protein DKFZp434G171 CAB61365
    Hs.8997 heat shock 70 kDa protein 1A 140550 NP_005336
    Hs.91143 jagged 1 (Alagille syndrome) 601920 NP_000205
    Hs.100009 cyclin-dependent kinase 3 123828
    Hs.100071 6-phosphogluconolactonase 604951 NP_036220
    Hs.106747 likely homolog of rat and mouse retinoid-inducible NP_067639
    serine carboxypeptidase
    Hs.107125 plasmalemma vesicle associated protein NP_112600
    Hs.110024 NADH: ubiquinone oxidoreductase MLRQ subunit NP_064527
    homolog
    Hs.111244 HIF-1 responsive RTP801 NP_061931
    Hs.111611 ribosomal protein L27 607526 NP_000979
    Hs.111779 secreted protein, acidic, cysteine-rich (osteonectin) 182120 NP_003109
    Hs.118625 hexokinase 1 142600 NP_277035
    Hs.119122 ribosomal protein L13a
    Hs.119129 collagen, type IV, alpha 1 120130 NP_001836
    Hs.119206 insulin-like growth factor binding protein 7 602867 NP_001544
    Hs.119571 collagen, type III, alpha 1 (Ehlers-Danlos syndrome 120180 NP_000081
    type IV, autosomal dominant)
    Hs.1197 heat shock 10 kDa protein 1 (chaperonin 10) 600141 NP_002148
    Hs.122359 calcium channel, voltage-dependent, alpha 1H NP_066921
    subunit
    Hs.1244 CD9 antigen (p24) 143030 NP_001760
    Hs.125036 TEM17 606826 NP_065138
    Hs.125359 TEM13, Thy-1 cell surface antigen 188230 NP_006279
    Hs.12956 Tax interaction protein 1 NP_055419
    Hs.143897 dysferlin, limb girdle muscular dystrophy 2B 603009 NP_003485
    (autosomal recessive)
    Hs.146360 hypothetical protein MGC34648 NP_689873
    Hs.150580 putative translation initiation factor NP_005792
    Hs.1516 insulin-like growth factor binding protein 4 146733 NP_001543
    Hs.151738 matrix metalloproteinase 9 (gelatinase B, 92 kDa 120361 NP_004985
    gelatinase, 92 kDa type IV collagenase)
    Hs.15265 heterogeneous nuclear ribonucleoprotein R 607201 NP_005817
    Hs.154029 bHLH factor Hes4 NP_066993
    Hs.159263 collagen, type VI, alpha 2 120240 NP_001840
    Hs.168357 T-box 2 600747 NP_005985
    Hs.169476 glyceraldehyde-3-phosphate dehydrogenase 138400 NP_002037
    Hs.17170 G protein-coupled receptor 4 600551 NP_005273
    Hs.172928 collagen, type I, alpha 1 120150 NP_000079
    Hs.173737 ras-related C3 botulinum toxin substrate 1 (rho family, 602048
    small GTP binding protein Rac1)
    Hs.177592 ribosomal protein, large, P1 180520
    Hs.179573 TEM10, COL1A2 involved in tissue remodeling 120160 NP_000080
    Hs.180414 heat shock 70 kDa protein 8 600816 NP_006588
    Hs.181418 KIAA0152 gene product NP_055545
    Hs.184367 Ca2+-promoted Ras inactivator BAA25464
    Hs.197114 serine/arginine repetitive matrix 2 606032 NP_057417
    Hs.197540 hypoxia-inducible factor 1, alpha subunit (basic helix- 603348 NP_001521
    loop-helix transcription factor)
    Hs.202 benzodiazapine receptor (peripheral) 109610 NP_000705
    Hs.205353 ectonucleoside triphosphate diphosphohydrolase 1 601752 NP_001767
    Hs.211573 heparan sulfate proteoglycan 2 (perlecan) 142461 NP_005520
    Hs.230 Fibromodulin 600245 NP_002014
    Hs.234434 hairy/enhancer-of-split related with YRPW motif 1 602953 NP_036390
    Hs.235368 collagen, type V, alpha 3 120216 NP_056534
    Hs.23823 hairy/enhancer-of-split related with YRPW motif-like NP_055386
    Hs.240170 hypothetical protein MGC2731 NP_076973
    Hs.244 amino-terminal enhancer of split 600188
    Hs.246857 mitogen-activated protein kinase 9 602896 NP_620708
    Hs.24950 regulator of G-protein signalling 5 603276 NP_003608
    Hs.250655 prothymosin, alpha (gene sequence 28) 188390 NP_002814
    Hs.251653 tubulin, beta, 2 602660 NP_006079
    Hs.25338 protease, serine, 23
    Hs.25549 hypothetical protein FLJ20898 NP_078876
    Hs.2575 calpain 1, (mu/l) large subunit 114220 NP_005177
    Hs.265827 interferon, alpha-inducible protein (clone IFI-6-16) 147572 NP_075011
    Hs.267200 ESTs, Weakly similar to T25031 hypothetical protein
    T20D3.3 - Caenorhabditis elegans [C. elegans]
    Hs.277477 major histocompatibility complex, class I, C 142840 NP_002108
    Hs.277704 hypoxia up-regulated 1 601746 NP_006380
    Hs.278625 complement component 4B 120820 NP_000583
    Hs.298229 prefoldin 2 NP_036526
    Hs.31053 cytoskeleton-associated protein 1 601303 NP_001272
    Hs.3109 Rho GTPase activating protein 4 300023 NP_001657
    Hs.327412 Homo sapiens clone FLC1492 PRO3121 mRNA,
    complete cds
    Hs.332173 transducin-like enhancer of split 2 (E(sp1) homolog, 601041 NP_003251
    Drosophila)
    Hs.337445 ribosomal protein L37 604181 NP_000988
    Hs.337986 hypothetical protein MGC4677 NP_443103
    Hs.353882 ESTs, Highly similar to MT1A_HUMAN
    METALLOTHIONEIN-IA (MT-1A) [H. sapiens]
    Hs.356624 TEM11, nidogen (enactin) 131390 NP_002499
    Hs.356668 guanine nucleotide binding protein (G protein), 600874 NP_005265
    gamma 5
    Hs.365706 matrix Gla protein 154870 NP_000891
    Hs.36927 heat shock 105 Kd NP_006635
    Hs.374523 GNAS complex locus 139320 NP_536350
    Hs.380824 Homo sapiens cDNA FLJ11658 fis, clone
    HEMBA1004577
    Hs.406410 H19, imprinted maternally expressed untranslated 103280 BAB71280
    mRNA
    Hs.43666 protein tyrosine phosphatase type IVA, member 3 606449 NP_116000
    Hs.48029 snail homolog 1 (Drosophila) 604238 NP_005976
    Hs.49215 integrin-binding sialoprotein (bone sialoprotein, bone 147563 NP_004958
    sialoprotein II)
    Hs.5831 tissue inhibitor of metalloproteinase 1 (erythroid 305370 NP_003245
    potentiating activity, collagenase inhibitor)
    Hs.699 peptidylprolyl isomerase B (cyclophilin B) 123841 NP_000933
    Hs.75061 MARCKS-like protein 602940 NP_075385
    Hs.75087 FAST kinase 606965 NP_079372
    Hs.75111 protease, serine, 11 (IGF binding) 602194 NP_002766
    Hs.75415 beta-2-microglobulin 109700 NP_004039
    Hs.75450 delta sleep inducing peptide, immunoreactor 602960
    Hs.75617 collagen, type IV, alpha 2 120090 NP_001837
    Hs.76095 immediate early response 3 602996 NP_434702
    Hs.76206 cadherin 5, type 2, VE-cadherin (vascular epithelium) 601120 NP_001786
    Hs.76640 RGC32 protein
    Hs.77890 guanylate cyclase 1, soluble, beta 3 139397 NP_000848
    Hs.77961 major histocompatibility complex, class I, B 142830 NP_005505
    Hs.78224 ribonuclease, RNase A family, 1 (pancreatic) 180440 AAH05324
    Hs.78409 collagen, type XVIII, alpha 1 120328 NP_085059
    Hs.78465 v-jun sarcoma virus 17 oncogene homolog (avian) 165160 NP_002219
    Hs.7869 Homo sapiens mRNA; cDNA DKFZp686G1610 (from
    clone DKFZp686G1610)
    Hs.79110 Nucleolin 164035 NP_005372
    Hs.79339 lectin, galactoside-binding, soluble, 3 binding protein 600626 NP_005558
    Hs.79356 Lysosomal-associated multispanning membrane 601476 NP_006753
    protein-5
    Hs.80617 ribosomal protein S16 603675
    Hs.8107 guanine nucleotide binding protein (G protein),
    gamma 12
    Hs.82085 serine (or cysteine) proteinase inhibitor, clade E 173360 NP_000593
    (nexin, plasminogen activator inhibitor type 1),
    member 1
    Hs.821 Biglycan 301870 NP_001702
    Hs.82646 DnaJ (Hsp40) homolog, subfamily B, member 1 604572 NP_006136
    Hs.82689 tumor rejection antigen (gp96) 1 191175 NP_003290
    Hs.833 interferon, alpha-inducible protein (clone IFI-15K) 147571 NP_005092
    Hs.83974 solute carrier family 21 (prostaglandin transporter), 601460 NP_005621
    member 2
    Hs.84298 CD74 antigen (invariant polypeptide of major 142790 NP_004346
    histocompatibility complex, class II antigen-
    associated)
    Hs.8546 Notch homolog 3 (Drosophila) 600276 NP_000426
  • TABLE 2
    Additional Tumor Endothelial Markers in Breast
    Unigene ID Function OMIMID Protein
    Hs.296323 serum/glucocorticoid regulated 602958 NP_005618
    kinase
    Hs.246857 mitogen-activated protein kinase 602896 NP_620708
    Hs.25691 receptor (calcitonin) activity 605155 NP_005847
    modifying protein 3
    Hs.9598 sema domain, immunoglobulin BAB21836
    domain (Ig)
    Hs.202 benzodiazapine receptor 109610 NP_000715
    (peripheral) - mitochondrial
    Hs.6147 C1 domain-containing NP_056134
    phosphatase &
    tensin-like
  • TABLE 3
    Markers in Colon and Breast Tumor Epithelium
    Unigene ID Function OMIMID Protein
    Hs.8997 heat shock 70 kDa protein 1A 140550 NP_005336
    Hs.110024 NADH: ubiquinone oxidoreductase MLRQ subunit homolog NP_064527
    Hs.111779 secreted protein, acidic, cysteine-rich (osteonectin) 182120 NP_003109
    Hs.119129 collagen, type IV, alpha 1 120130 NP_001836
    Hs.119206 insulin-like growth factor binding protein 7 602867 NP_001544
    Hs.119571 collagen, type III, alpha 1 (Ehlers-Danlos syndrome type 120180 NP_000081
    IV, autosomal dominant)
    Hs.1197 heat shock 10 kDa protein 1 (chaperonin 10) 600141 NP_002148
    Hs.125036 TEM17 606826 NP_065138
    Hs.125359 TEM13, Thy-1 cell surface antigen 188230 NP_006279
    Hs.151738 matrix metalloproteinase 9 (gelatinase B, 92 kDa 120361 NP_004985
    gelatinase, 92 kDa type IV collagenase)
    Hs.159263 collagen, type VI, alpha 2 120240 NP_001840
    Hs.168357 T-box 2 600747 NP_005985
    Hs.172928 collagen, type I, alpha 1 120150 NP_000079
    Hs.179573 TEM10, COL1A2 involved in tissue remodeling 120160 NP_000080
    Hs.230 Fibromodulin 600245 NP_002014
    Hs.23823 hairy/enhancer-of-split related with YRPW motif-like NP_055386
    Hs.24950 regulator of G-protein signalling 5 603276 NP_003608
    Hs.265827 interferon, alpha-inducible protein (clone IFI-6-16) 147572 NP_075011
    Hs.327412 Homo sapiens clone FLC1492 PRO3121 mRNA,
    complete cds
    Hs.337986 hypothetical protein MGC4677 NP_443103
    Hs.356624 TEM11, nidogen (enactin) 131390 NP_002499
    Hs.36927 heat shock 105 kD NP_006635
    Hs.43666 protein tyrosine phosphatase type IVA, member 3 606449 NP_116000
    Hs.5831 tissue inhibitor of metalloproteinase 1 (erythroid 305370 NP_003245
    potentiating activity, collagenase inhibitor)
    Hs.699 peptidylprolyl isomerase B (cyclophilin B) 123841 NP_000933
    Hs.75617 collagen, type IV, alpha 2 120090 NP_001837
    Hs.77890 guanylate cyclase 1, soluble, beta 3 139397 NP_000848
    Hs.78409 collagen, type XVIII, alpha 1 120328 NP_085059
    Hs.78465 v-jun sarcoma virus 17 oncogene homolog (avian) 165160 NP_002219
    Hs.821 Biglycan 301870 NP_001702
    Hs.82646 DnaJ (Hsp40) homolog, subfamily B, member 1 604572 NP_006136
    Hs.8546 Notch homolog 3 (Drosophila) 600276 NP_000426
  • TABLE 4
    Markers in Brain and Breast Tumor Epithelium
    Unigene ID Function OMIMID Protein
    Hs.107125 plasmalemma vesicle associated protein NP_112600
    Hs.111611 ribosomal protein L27 607526 NP_000979
    Hs.111779 Secreted protein, acidic, cysteine-rich 182120 NP_003109
    (osteonectin)
    Hs.119129 Collagen, type IV, alpha 1 120130 NP_001836
    Hs.119571 Collagen, type III, alpha 1 (Ehlers- 120180 NP_000081
    Danlos syndrome type IV, autosomal
    dominant)
    Hs.125359 TEM13, Thy-1 cell surface antigen 188230 NP_006279
    Hs.143897 Dysferlin, limb girdle muscular dystrophy 603009 NP_003485
    2B (autosomal recessive)
    Hs.151738 matrix metalloproteinase 9 (gelatinase 120361 NP_004985
    B, 92 kDa gelatinase, 92 kDa type IV
    collagenase)
    Hs.159263 Collagen, type VI, alpha 2 120240 NP_001840
    Hs.172928 Collagen, type I, alpha 1 120150 NP_000079
    Hs.179573 TEM10, COL1A2 involved in tissue 120160 NP_000080
    remodeling
    Hs.211573 Heparan sulfate proteoglycan 2 142461 NP_005520
    (perlecan)
    Hs.277477 major histocompatibility complex, class 142840 NP_002108
    I, C
    Hs.327412 Homo sapiens clone FLC1492
    PRO3121 mRNA, complete cds
    Hs.332173 transducin-like enhancer of split 2 601041 NP_003251
    (E(sp1) homolog, Drosophila)
    Hs.337986 hypothetical protein MGC4677 NP_443103
    Hs.365706 matrix Gla protein 154870 NP_000891
    Hs.75061 MARCKS-like protein 602940 NP_075385
    Hs.75111 Protease, serine, 11 (IGF binding) 602194 NP_002766
    Hs.75617 collagen, type IV, alpha 2 120090 NP_001837
    Hs.77961 major histocompatibility complex, class 142830 NP_005505
    I, B
    Hs.79356 Lysosomal-associated multispanning 601476 NP_006753
    membrane protein-5
    Hs.82085 serine (or cysteine) proteinase inhibitor, 173360 NP_000593
    clade E (nexin, plasminogen activator inhibitor
    type 1), member 1
    Hs.821 Biglycan 301870 NP_001702
  • TABLE 5
    Breast, Brain, and Colon Tumor Endothelial Markers
    Unigene ID Function OMIMID Protein
    Hs.111779 secreted protein, acidic, 182120 NP_003109
    cysteine-rich (osteonectin)
    Hs.119129 collagen, type IV, alpha 1 120130 NP_001836
    Hs.119571 collagen, type III, alpha 1 120180 NP_000081
    (Ehlers-Danlos syndrome
    type IV, autosomal
    dominant)
    Hs.125359 TEM13, Thy-1 cell surface 188230 NP_006279
    antigen
    Hs.151738 matrix metalloproteinase 9 120361 NP_004985
    (gelatinase B, 92 kDa
    gelatinase, 92 kDa type IV
    collagenase)
    Hs.159263 collagen, type VI, alpha 2 120240 NP_001840
    Hs.172928 collagen, type I, alpha 1 120150 NP_000079
    Hs.179573 TEM10, COL1A2 involved 120160 NP_000080
    in tissue remodeling
    Hs.327412 Homo sapiens clone
    FLC1492 PRO3121 mRNA,
    complete cds
    Hs.337986 hypothetical protein NP_443103
    MGC4677
    Hs.75617 collagen, type IV, alpha 2 120090 NP_001837
    Hs.821 biglycan 301870 NP_001702
  • Endothelial cells (ECs) represent only a minor fraction of the total cells within normal or tumor tissues, and only those EC transcripts expressed at the highest levels would be expected to be represented in libraries constructed from unfractionated tissues. The genes described in the current study should therefore provide a valuable resource for basic and clinical studies of human breast angiogenesis in the future.
  • Isolated and purified nucleic acids, according to the present invention are those which are not linked to those genes to which they are linked in the human genome. Moreover, they are not present in a mixture such as a library containing a multitude of distinct sequences from distinct genes. They may be, however, linked to other genes such as vector sequences or sequences of other genes to which they are not naturally adjacent.
  • The nucleic acids may represent either the sense or the anti-sense strand. Nucleic acids and proteins although disclosed herein with sequence particularity, may be derived from a single individual. Allelic variants which occur in the population of humans are included within the scope of such nucleic acids and proteins. Those of skill in the art are well able to identify allelic variants as being the same gene or protein. Given a nucleic acid, one of ordinary skill in the art can readily determine an open reading frame present, and consequently the sequence of a polypeptide encoded by the open reading frame and, using techniques well known in the art, express such protein in a suitable host. Proteins comprising such polypeptides can be the naturally occurring proteins, fusion proteins comprising exogenous sequences from other genes from humans or other species, epitope tagged polypeptides, etc. Isolated and purified proteins are not in a cell, and are separated from the normal cellular constituents, such as nucleic acids, lipids, etc. Typically the protein is purified to such an extent that it comprises the predominant species of protein in the composition, such as greater than 50, 60 70, 80, 90, or even 95% of the proteins present.
  • Using the proteins according to the invention, one of ordinary skill in the art can readily generate antibodies which specifically bind to the proteins. Such antibodies can be monoclonal or polyclonal. They can be chimeric, humanized, or totally human. Any functional fragment or derivative of an antibody can be used including Fab, Fab′, Fab2, Fab′2, and single chain variable regions. So long as the fragment or derivative retains specificity of binding for the endothelial marker protein it can be used. Antibodies can be tested for specificity of binding by comparing binding to appropriate antigen to binding to irrelevant antigen or antigen mixture under a given set of conditions. If the antibody binds to the appropriate antigen at least 2, 5, 7, and preferably 10 times more than to irrelevant antigen or antigen mixture then it is considered to be specific.
  • Techniques for making such partially to fully human antibodies are known in the art and any such techniques can be used. According to one particularly preferred embodiment, fully human antibody sequences are made in a transgenic mouse which has been engineered to express human heavy and light chain antibody genes. Multiple strains of such transgenic mice have been made which can produce different classes of antibodies. B cells from transgenic mice which are producing a desirable antibody can be fused to make hybridoma cell lines for continuous production of the desired antibody. See for example, Nina D. Russel, Jose R. F. Corvalan, Michael L. Gallo, C. Geoffrey Davis, Liise-Anne Pirofski. Production of Protective Human Antipneumococcal Antibodies by Transgenic Mice with Human Immunoglobulin Loci Infection and Immunity April 2000, p. 1820-1826; Michael L. Gallo, Vladimir E. Ivanov, Aya Jakobovits, and C. Geoffrey Davis. The human immunoglobulin loci introduced into mice: V (D) and J gene segment usage similar to that of adult humans European Journal of Immunology 30: 534-540, 2000; Larry L. Green. Antibody engineering via genetic engineering of the mouse: XenoMouse strains are a vehicle for the facile generation of therapeutic human monoclonal antibodies Journal of Immunological Methods 231 11-23, 1999; Yang X-D, Corvalan J R F, Wang P, Roy CM-N and Davis CG. Fully Human Anti-interleukin-8 Monoclonal Antibodies: Potential Therapeutics for the Treatment of Inflammatory Disease States. Journal of Leukocyte Biology Vol. 66, pp 401-410 (1999); Yang X-D, Jia X-C, Corvalan J R F, Wang P, C G Davis and Jakobovits A. Eradication of Established Tumors by a Fully Human Monoclonal Antibody to the Epidermal Growth Factor Receptor without Concomitant Chemotherapy. Cancer Research Vol. 59, Number 6, pp 1236-1243 (1999); Jakobovits A. Production and selection of antigen-specific fully human monoclonal antibodies from mice engineered with human Ig loci. Advanced Drug Delivery Reviews Vol. 31, pp: 33-42 (1998); Green L and Jakobovits A. Regulation of B cell development by variable gene complexity in mice reconstituted with human immunoglobulin yeast artificial chromosomes. J. Exp. Med. Vol. 188, Number 3, pp: 483-495 (1998); Jakobovits A. The long-awaited magic bullets: therapeutic human monoclonal antibodies from transgenic mice. Exp. Opin. Invest. Drugs Vol. 7(4), pp: 607-614 (1998); Tsuda H, Maynard-Currie K, Reid L, Yoshida T, Edamura K, Maeda N, Smithies O, Jakobovits A. Inactivation of Mouse HPRT locus by a 203-bp retrotransposon insertion and a 55-kb gene-targeted deletion: establishment of new HPRT-Deficient mouse embryonic sBEM cell lines. Genomics Vol. 42, pp: 413-421 (1997); Sherman-Gold, R. Monoclonal Antibodies: The Evolution from '80s Magic Bullets To Mature, Mainstream Applications as Clinical Therapeutics. Genetic Engineering News Vol. 17, Number 14 (August 1997); Mendez M, Green L, Corvalan J, Jia X-C, Maynard-Currie C, Yang X-d, Gallo M, Louie D, Lee D, Erickson K, Luna J, Roy C, Abderrahim H, Kirschenbaum F, Noguchi M, Smith D, Fukushima A, Hales J, Finer M, Davis C, Zsebo K, Jakobovits A. Functional transplant of megabase human immunoglobulin loci recapitulates human antibody response in mice. Nature Genetics Vol. 15, pp: 146-156 (1997); Jakobovits A. Mice engineered with human immunoglobulin YACs: A new technology for production of fully human antibodies for autoimmunity therapy. Weir's Handbook of Experimental Immunology, The Integrated Immune System Vol. IV, pp: 194.1-194.7 (1996); Jakobovits A. Production of fully human antibodies by transgenic mice. Current Opinion in Biotechnology Vol. 6, No. 5, pp: 561-566 (1995); Mendez M, Abderrahim H, Noguchi M, David N, Hardy M, Green L, Tsuda H, Yoast S, Maynard-Currie C, Garza D, BEMmill R, Jakobovits A, Klapholz S. Analysis of the structural integrity of YACs comprising human immunoglobulin genes in yeast and in embryonic sBEM cells. Genomics Vol. 26, pp: 294-307 (1995); Jakobovits A. YAC Vectors: Humanizing the mouse genome. Current Biology Vol. 4, No. 8, pp: 761-763 (1994); Arbones M, Ord D, Ley K, Ratech H, Maynard-Curry K, Otten G, Capon D, Tedder T. Lymphocyte homing and leukocyte rolling and migration are impaired in L-selectin-deficient mice. Immunity Vol. 1, No. 4, pp: 247-260 (1994); Green L, Hardy M, Maynard-Curry K, Tsuda H, Louie D, Mendez M, Abderrahim H, Noguchi M, Smith D, Zeng Y, et. al. Antigen-specific human monoclonal antibodies from mice engineered with human Ig heavy and light chain YACs. Nature Genetics Vol. 7, No. 1, pp: 13-21 (1994); Jakobovits A, Moore A, Green L, Vergara G, Maynard-Curry K, Austin H, Klapholz S. Germ-line transmission and expression of a human-derived yeast artificial chromosome. Nature Vol. 362, No. 6417, pp: 255-258 (1993); Jakobovits A, Vergara G, Kennedy J, Hales J, McGuinness R, Casentini-Borocz D, Brenner D, Otten G. Analysis of homozygous mutant chimeric mice: deletion of the immunoglobulin heavy-chain joining region blocks B-cell development and antibody production. Proceedings of the National Academy of Sciences USA Vol. 90, No. 6, pp: 2551-2555 (1993); Kucherlapati et al., U.S. Pat. No. 6,1075,181.
  • Antibodies can also be made using phage display techniques. Such techniques can be used to isolate an initial antibody or to generate variants with altered specificity or avidity characteristics. Single chain Fv can also be used as is convenient. They can be made from vaccinated transgenic mice, if desired. Antibodies can be produced in cell culture, in phage, or in various animals, including but not limited to cows, rabbits, goats, mice, rats, hamsters, guinea pigs, sheep, dogs, cats, monkeys, chimpanzees, apes.
  • Antibodies can be labeled with a detectable moiety such as a radioactive atom, a chromophore, a fluorophore, or the like. Such labeled antibodies can be used for diagnostic techniques, either in vivo, or in an isolated test sample. Antibodies can also be conjugated, for example, to a pharmaceutical agent, such as chemotherapeutic drug or a toxin. They can be linked to a cytokine, to a ligand, to another antibody. Suitable agents for coupling to antibodies to achieve an anti-tumor effect include cytokines, such as interleukin 2 (IL-2) and Tumor Necrosis Factor (TNF); photosensitizers, for use in photodynamic therapy, including aluminum (III) phthalocyanine tetrasulfonate, hematoporphyrin, and phthalocyanine; radionuclides, such as iodine-131 (131I), yttrium-90 (90Y), bismuth-212 (212Bi), bismuth-213 (213Bi), technetium-99m (99mTc), rhenium-186 (186Re), and rhenium-188 (188Re); antibiotics, such as doxorubicin, adriamycin, daunorubicin, methotrexate, daunomycin, neocarzinostatin, and carboplatin; bacterial, plant, and other toxins, such as diphtheria toxin, pseudomonas exotoxin A, staphylococcal enterotoxin A, abrin-A toxin, ricin A (deglycosylated ricin A and native ricin A), TGF-alpha toxin, cytotoxin from chinese cobra (naja naja atra), and gelonin (a plant toxin); ribosome inactivating proteins from plants, bacteria and fungi, such as restrictocin (a ribosome inactivating protein produced by Aspergillus restrictus), saporin (a ribosome inactivating protein from Saponaria officinalis), and RNase; tyrosine kinase inhibitors; ly207702 (a difluorinated purine nucleoside); liposomes containing antitumor agents (e.g., antisense oligonucleotides, plasmids which encode for toxins, methotrexate, etc.); and other antibodies or antibody fragments, such as F(ab).
  • Those of skill in the art will readily understand and be able to make such antibody derivatives, as they are well known in the art. The antibodies may be cytotoxic on their own, or they may be used to deliver cytotoxic agents to particular locations in the body. The antibodies can be administered to individuals in need thereof as a form of passive immunization.
  • Characterization of extracellular regions for the cell surface and secreted proteins from the protein sequence is based on the prediction of signal sequence, transmembrane domains and functional domains. Antibodies are preferably specifically immunoreactive with membrane associated proteins, particularly to extracellular domains of such proteins or to secreted proteins. Such targets are readily accessible to antibodies, which typically do not have access to the interior of cells or nuclei. However, in some applications, antibodies directed to intracellular proteins may be useful as well. Moreover, for diagnostic purposes, an intracellular protein may be an equally good target since cell lysates may be used rather than a whole cell assay.
  • Computer programs can be used to identify extracellular domains of proteins whose sequences are known. Such programs include SMART software (Schultz et al., Proc. Natl. Acad. Sci. USA 95: 5857-5864, 1998) and Pfam software (BaBEMan et al., Nucleic acids Res. 28: 263-266, 2000) as well as PSORTII. Typically such programs identify transmembrane domains; the extracellular domains are identified as immediately adjacent to the transmembrane domains. Prediction of extracellular regions and the signal cleavage sites are only approximate. It may have a margin of error + or −5 residues. Signal sequence can be predicted using three different methods (Nielsen et al, Protein Engineering 10: 1-6, 1997, Jagla et. al, Bioinformatics 16: 245-250, 2000, Nakai, K and Horton, P. Trends in Biochem. Sci. 24:34-35, 1999) for greater accuracy. Similarly transmembrane (TM) domains can be identified by multiple prediction methods. (Pasquier, et. al, Protein Eng. 12:381-385, 1999, Sonnhammer et al., In Proc. of Sixth Int. Conf. on Intelligent Systems for Molecular Biology, p. 175-182, Ed J. Glasgow, T. Littlejohn, F. Major, R. Lathrop, D. Sankoff, and C. Sensen Menlo Park, Calif.: AAAI Press, 1998, Klein, et. al, Biochim. Biophys. Acta, 815:468, 1985, Nakai and Kanehisa Genomics, 14: 897-911, 1992). In ambiguous cases, locations of functional domains in well characterized proteins are used as a guide to assign a cellular localization.
  • Putative functions or functional domains of novel proteins can be inferred from homologous regions in the database identified by BLAST searches (Altschul et. al. Nucleic Acid Res. 25: 3389-3402, 1997) and/or from a conserved domain database such as Pfam (BaBEMan et. al, Nucleic Acids Res. 27:260-262 1999) BLOCKS (Henikoff, et. al, Nucl. Acids Res. 28:228-230, 2000) and SMART (Ponting, et. al, Nucleic Acid Res. 27,229-232, 1999). Extracellular domains include regions adjacent to a transmembrane domain in a single transmembrane domain protein (out-in or type I class). For multiple transmembrane domains proteins, the extracellular domain also includes those regions between two adjacent transmembrane domains (in-out and out-in). For type II transmembrane domain proteins, for which the N-terminal region is cytoplasmic, regions following the transmembrane domain is generally extracellular. Secreted proteins on the other hand do not have a transmembrane domain and hence the whole protein is considered as extracellular.
  • Membrane associated proteins can be engineered to delete the transmembrane domains, thus leaving the extracellular portions which can bind to ligands. Such soluble forms of transmembrane receptor proteins can be used to compete with natural forms for binding to ligand. Thus such soluble forms act as inhibitors and can be used therapeutically as anti-angiogenic agents, as diagnostic tools for the quantification of natural ligands, and in assays for the identification of small molecules which modulate or mimic the activity of a BEM:ligand complex.
  • Alternatively, the endothelial markers themselves can be used as vaccines to raise an immune response in the vaccinated animal or human. For such uses, a protein, or immunogenic fragment of such protein, corresponding to the intracellular, extracellular or secreted BEM of interest is administered to a subject. The immogenic agent may be provided as a purified preparation or in an appropriately expressing cell. The administration may be direct, by the delivery of the immunogenic agent to the subject, or indirect, through the delivery of a nucleic acid encoding the immunogenic agent under conditions resulting in the expression of the immunogenic agent of interest in the subject. The BEM of interest may be delivered in an expressing cell, such as a purified population of breast tumor endothelial cells or a population of fused breast tumor endothelial and dendritic cells. Nucleic acids encoding the BEM of interest may be delivered in a viral or non-viral delivery vector or vehicle. Non-human sequences encoding the human BEM of interest or other mammalian homolog can be used to induce the desired immunologic response in a human subject. For several of the BEMs of the present invention, mouse, rat or other ortholog sequences can be obtained from the literature or using techniques well within the skill of the art.
  • Endothelial cells can be identified using the markers which are disclosed herein as being endothelial cell specific. Antibodies specific for such markers can be used to identify such cells, by contacting the antibodies with a population of cells containing some endothelial cells. The presence of cross-reactive material with the antibodies identifies particular cells as endothelial. Similarly, lysates of cells can be tested for the presence of cross-reactive material. Any known format or technique for detecting cross-reactive material can be used including, immunoblots, radioimmunoassay, ELISA, immunoprecipitation, and immunohistochemistry. In addition, nucleic acid probes for these markers can also be used to identify endothelial cells. Any hybridization technique known in the art including Northern blotting, RT-PCR, microarray hybridization, and in situ hybridization can be used.
  • One can identify breast tumor endothelial cells for diagnostic purposes, testing cells suspected of containing one or more BEMs. One can test both tissues and bodily fluids of a subject. For example, one can test a patient's blood for evidence of intracellular and membrane associated BEMs, as well as for secreted BEMs. Of particular interest in this context is the testing of breast duct fluid. Intracellular and/or membrane associated BEMs may be present in bodily fluids as the result of high levels of expression of these factors and/or through lysis of cells expressing the BEMs.
  • Populations of various types of endothelial cells can also be made using the antibodies to endothelial markers of the invention. The antibodies can be used to purify cell populations according to any technique known in the art, including but not limited to fluorescence activated cell sorting. Such techniques permit the isolation of populations which are at least 50, 60, 70, 80, 90, 92, 94, 95, 96, 97, 98, and even 99% the type of endothelial cell desired, whether normal, tumor, or pan-endothelial. Antibodies can be used to both positively select and negatively select such populations. Preferably at least 1, 5, 10, 15, 20, or 25 of the appropriate markers are expressed by the endothelial cell population.
  • Populations of endothelial cells made as described herein, can be used for screening drugs to identify those suitable for inhibiting the growth of tumors by virtue of inhibiting the growth of the tumor vasculature.
  • Populations of endothelial cells made as described herein, can be used for screening candidate drugs to identify those suitable for modulating angiogenesis, such as for inhibiting the growth of tumors by virtue of inhibiting the growth of endothelial cells, such as inhibiting the growth of the tumor or other undesired vasculature, or alternatively, to promote the growth of endothelial cells and thus stimulate the growth of new or additional large vessel or microvasculature.
  • Inhibiting the growth of endothelial cells means either regression of vasculature which is already present, or the slowing or the absence of the development of new vascularization in a treated system as compared with a control system. By stimulating the growth of endothelial cells, one can influence development of new (neovascularization) or additional vasculature development (revascularization). A variety of model screening systems are available in which to test the angiogenic and/or anti-angiogenic properties of a given candidate drug. Typical tests involve assays measuring the endothelial cell response, such as proliferation, migration, differentiation and/or intracellular interaction with a given candidate drug. By such tests, one can study the signals and effects of the test stimuli. Some common screens involve measurement of the inhibition of heparanase, endothelial tube formation on Matrigel, scratch induced motility of endothelial cells, platelet-derived growth factor driven proliferation of vascular smooth muscle cells, and the rat aortic ring assay (which provides an advantage of capillary formation rather than just one cell type).
  • Drugs can be screened for the ability to mimic or modulate, inhibit or stimulate, growth of tumor endothelium cells and/or normal endothelial cells. Drugs can be screened for the ability to inhibit tumor endothelium growth but not normal endothelium growth or survival. Similarly, human cell populations, such as normal endothelium populations or breast tumor endothelial cell populations, can be contacted with test substances and the expression of breast tumor endothelial markers and/or normal endothelial markers determined. Test substances that decrease the expression of breast tumor endothelial markers (BEMs) are candidates for inhibiting angiogenesis and the growth of tumors. In cases where the activity of a BEM is known, agents can be screened for their ability to decrease or increase the activity.
  • For those breast tumor endothelial markers identified as containing transmembrane regions, it is desirable to identify drug candidates capable of binding to the BEM receptors found at the cell surface. For some applications, the identification of drug candidates capable of blocking the BEM receptor from its native ligand will be desired. For some applications, the identification of a drug candidate capable of binding to the BEM receptor may be used as a means to deliver a therapeutic or diagnostic agent. For other applications, the identification of drug candidates capable of mimicking the activity of the native ligand will be desired. Thus, by manipulating the binding of a transmembrane BEM receptor:ligand complex, one may be able to promote or inhibit further development of endothelial cells and hence, vascularization.
  • For those breast tumor endothelial markers identified as being secreted proteins, i.e., extracellular, it is desirable to identify drug candidates capable of binding to the secreted BEM protein. For some applications, the identification of drug candidates capable of interfering with the binding of the secreted BEM it is native receptor. For other applications, the identification of drug candidates capable of mimicking the activity of the native receptor will be desired. Thus, by manipulating the binding of the secreted BEM:receptor complex, one may be able to promote or inhibit further development of endothelial cells, and hence, vascularization.
  • Expression can be monitored according to any convenient method. Protein or mRNA can be monitored. Any technique known in the art for monitoring specific genes' expression can be used, including but not limited to ELISAs, SAGE, microarray hybridization, Western blots. Changes in expression of a single marker may be used as a criterion for significant effect as a potential pro-angiogenic, anti-angiogenic or anti-tumor agent. However, it also may be desirable to screen for test substances that are able to modulate the expression of at least 5, 10, 15, or 20 of the relevant markers, such as the tumor or normal endothelial markers. Inhibition of BEM protein activity can also be used as a drug screen.
  • Test substances for screening can come from any source. They can be libraries of natural products, combinatorial chemical libraries, biological products made by recombinant libraries, etc. The source of the test substances is not critical to the invention. The present invention provides means for screening compounds and compositions that may previously have been overlooked in other screening schemes. Nucleic acids and the corresponding encoded proteins of the markers of the present invention can be used therapeutically in a variety of modes. BEMs can be used to stimulate the growth of vasculature, such as for wound healing or to circumvent a blocked vessel. The nucleic acids and encoded proteins can be administered by any means known in the art. Such methods include, using liposomes, nanospheres, viral vectors, non-viral vectors comprising polycations, etc. Suitable viral vectors include adenovirus, retroviruses, and sindbis virus. Administration modes can be any known in the art, including parenteral, intravenous, intramuscular, intraperitoneal, topical, intranasal, intrarectal, intrabronchial, etc.
  • Specific biological antagonists of BEMs can also be used to therapeutic benefit. For example, antibodies, T cells specific for a BEM, antisense to a BEM, interference RNA to a BEM, and ribozymes specific for a BEM can be used to restrict, inhibit, reduce, and/or diminish tumor or other abnormal or undesirable vasculature growth. Such antagonists can be administered as is known in the art for these classes of antagonists generally. Anti-angiogenic drugs and agents can be used to inhibit tumor growth, as well as to treat diabetic retinopathy, rheumatoid arthritis, psoriasis, polycystic kidney disease (PKD), and other diseases requiring angiogenesis for their pathologies.
  • Mouse counterparts to human BEMs can be used in mouse cancer models or in cell lines or in vitro to evaluate potential anti-angiogenic or anti-tumor compounds or therapies. Their expression can be monitored as an indication of effect. Mouse BEMs can be used as antigens for raising antibodies which can be tested in mouse tumor models. Mouse BEMs with transmembrane domains are particularly preferred for this purpose. Mouse BEMs can also be used as vaccines to raise an immunological response in a human to the human ortholog.
  • The above disclosure generally describes the present invention. All references disclosed herein are expressly incorporated by reference in their entireties. A more complete understanding can be obtained by reference to the following specific examples which are provided herein for purposes of illustration only, and are not intended to limit the scope of the invention.
  • EXAMPLE 1
  • Function of BEM proteins was determined using bioinformatics tools. BEMs that are putative functional receptors with short cytoplasmic tails make particularly interesting targets.
  • Breast Tumor Endothelial Putative Functional Receptors with
    Short Cytoplasmic Tails
    Unigene ID Function OMIMID Protein
    Hs.181418 KIAA0152 gene product 055545
    Hs.25691 receptor (calcitonin) activity 605155 005847
    modifying protein 3
    Hs.9598 sema domain BAB212835
  • EXAMPLE 2
  • Protein kinases were identified among the BEMs. These are particularly good druggable targets, especially for small molecules.
  • Protein Kinases
    Unigene ID Function OMIMID Protein
    Hs.100009 cyclin-dependent 123828
    kinase 3
    Hs.143897 dysferlin, limb girdle 603009 NP_003485
    muscular dystrophy 2B
    (autosomal recessive)
    Hs.184367 Ca2+-promoted Ras BAA25464
    inactivator
    Hs.246857 mitogen-activated 602896 NP_620708
    protein kinase 9
    Hs.75087 FAST kinase 606965 NP-079372
    Hs.296323 serum/glucocorticoid 602958 NP_005618
    regulated kinase
    Hs.246857 mitogen-activated 602986 NP_620708
    protein kinase
  • EXAMPLE 3
  • Kinases with non-protein substrates were also identified. These similarly are believed to be exceedingly good druggable targets.
  • Kinases with non-protein substrates
    Unigene ID Function OMIMID Protein
    Hs.118625 hexokinase 1 142600 NP_277035
    Hs.82689 tumor rejection antigen (gp96) 1 191175 NP_003290
  • EXAMPLE 4
  • Growth factors were identified among the BEMs:
  • Growth factors
    Unigene ID Function OMIMID Protein
    Hs.91143 jagged 1 (Alagille syndrome) 601920 NP_000205
    Hs.119206 insulin-like growth factor 602867 NP_001544
    binding protein 7
    Hs.1516 insulin-like growth factor 146733 NP_001543
    binding protein 4
    Hs.211573 heparan sulfate proteoglycan 2 142461 NP_005520
    (perlecan)
    Hs.75111 protease, serine, 11 (IGF 602194 NP_002766
    binding)
    Hs.8546 Notch homolog 3 (Drosophila) 600276 NP_000426
  • EXAMPLE 5
  • Phosphatases, like kinases, are readily amenable to screening for inhibitors, especially small molecule inhibitors:
  • Phosphatases
    Unigene ID Function OMIMID Protein
    Hs.8997
    Figure US20090117038A1-20090507-P00899
     a protein 1A
    140550 NP_005336
    Hs.205353
    Figure US20090117038A1-20090507-P00899
    riphosphate
    601752 NP_001767
    diphosphohydrolase
    Hs.43666 phosphatase type IVA, member 606449 NP_116000
    Hs.6147
    Figure US20090117038A1-20090507-P00899
    e C1 domain-containing
    NP_056134
    Figure US20090117038A1-20090507-P00899
    ophosphatase &tensin-like
    Figure US20090117038A1-20090507-P00899
    indicates data missing or illegible when filed
  • EXAMPLE 6
  • GPCRs were identified among the BEMs:
  • GPCRs
    Unigene
    ID Function OMIMID Protein
    Hs.17170 G protein-coupled receptor 4 600551 NP_005273
  • EXAMPLE 7
  • The cellular location of the BEMs was determined to be either cytoplasmic, extracellular, membrane, or nuclear, as shown below.
  • Extracellular Proteins
    Unigene ID Function OMIMID Protein
    Hs.75415 Beta-2-microglobulin 109700 NP_004039
    Hs.821 Biglycan 301870 NP_001702
    Hs.172928 collagen, type I, alpha 1 120150 NP_000079
    Hs.119571 collagen, type III, alpha 1 (Ehlers-Danlos syndrome 120180 NP_000081
    type IV, autosomal dominant)
    Hs.119129 collagen, type IV, alpha 1 120130 NP_001836
    Hs.75617 collagen, type IV, alpha 2 120090 NP_001837
    Hs.235368 collagen, type V, alpha 3 120216 NP_056534
    Hs.159263 collagen, type VI, alpha 2 120240 NP_001840
    Hs.78409 collagen, type XVIII, alpha 1 120328 NP_085059
    Hs.278625 complement component 4B 120820 NP_000583
    Hs.230 Fibromodulin 600245 NP_002014
    Hs.211573 heparan sulfate proteoglycan 2 (perlecan) 142461 NP_005520
    Hs.1516 insulin-like growth factor binding protein 4 146733 NP_001543
    Hs.119206 insulin-like growth factor binding protein 7 602867 NP_001544
    Hs.49215 integrin-binding sialoprotein (bone sialoprotein, bone 147563 NP_004958
    sialoprotein II)
    Hs.79339 lectin, galactoside-binding, soluble, 3 binding protein 600626 NP_005558
    Hs.106747 likely homolog of rat and mouse retinoid-inducible NP_067639
    serine carboxypeptidase
    Hs.365706 matrix Gla protein 154870 NP_000891
    Hs.151738 matrix metalloproteinase 9 (gelatinase B, 92 kDa 120361 NP_004985
    gelatinase, 92 kDa type IV collagenase)
    Hs.699 peptidylprolyl isomerase B (cyclophilin B) 123841 NP_000933
    Hs.75111 protease, serine, 11 (IGF binding) 602194 NP_002766
    Hs.25338 protease, serine, 23
    Hs.78224 ribonuclease, RNase A family, 1 (pancreatic) 180440 AAH05324
    Hs.111779 secreted protein, acidic, cysteine-rich (osteonectin) 182120 NP_003109
    Hs.82085 serine (or cysteine) proteinase inhibitor, clade E 173360 NP_000593
    (nexin, plasminogen activator inhibitor type 1),
    member 1
    Hs.179573 TEM10, COL1A2 involved in tissue remodeling 120160 NP_000080
    Hs.356624 TEM11, nidogen (enactin) 131390 NP_002499
    Hs.5831 tissue inhibitor of metalloproteinase 1 (erythroid 305370 NP_003245
    potentiating activity, collagenase inhibitor)
    Hs.82689 tumor rejection antigen (gp96) 1 191175 NP_003290
  • Membrane Proteins
    Orientation
    TM of N-
    Unigene ID Function Protein Domains TM Location terminus
    Hs.202 benzodiazapine receptor (peripheral) NP_000705 3 107-129, 78-100, OUT
    133-155
    Hs.76206 cadherin 5, type 2, VE-cadherin (vascular epithelium) NP_001786 1 598-620 Unsure
    Hs.122359 calcium channel, voltage-dependent, alpha 1H subunit NP_066921 19 1370-1392, IN
    1614-1636, 1533-1555,
    141-163, 915-937,
    396-418, 1651-1673,
    1745-1767, 990-1012,
    234-256, 1430-1452,
    1333-1355, 1680-1702,
    855-877, 1295-1316,
    826-848, 100-122,
    1840-1862, 364-386
    Hs.84298 CD74 antigen (invariant polypeptide of major NP_004346 1 49-71 IN
    histocompatibility complex, class II antigen-
    associated)
    Hs.1244 CD9 antigen (p24) NP_001760 4 59-81, 88-110, IN
    12-34, 194-216
    Hs.143897 dysferlin, limb girdle muscular dystrophy 2B NP_003485 1 2045-2067 Unsure
    (autosomal recessive)
    Hs.205353 ectonucleoside triphosphate diphosphohydrolase 1 NP_001767 1 477-499 IN
    Hs.17170 G protein-coupled receptor 4 NP_005273 5 55-77, 92-113, OUT
    20-42, 225-244,
    183-205
    Hs.25549 hypothetical protein FLJ20898 NP_078876 3 102-124, 139-161, Unsure
    168-190
    Hs.277704 hypoxia up-regulated 1 NP_006380 1 13-35 IN
    Hs.76095 Immediate early response 3 NP_434702 1 123-145 Unsure
    Hs.265827 interferon, alpha-inducible protein (clone IFI-6-16) NP_075011 2 5-24, 44-66 IN
    Hs.91143 jagged 1 (Alagille syndrome) NP_000205 1 1069-1091 Unsure
    Hs.181418 KIAA0152 gene product NP_055545 1 271-290 OUT
    Hs.79356 Lysosomal-associated multispanning membrane NP_006753 5 63-85, 100-121, Unsure
    protein-5 142-164, 15-37,
    184-206
    Hs.77961 major histocompatibility complex, class I, B NP_005505 1 308-330 OUT
    Hs.277477 major histocompatibility complex, class I, C NP_002108 1 308-330 OUT
    Hs.110024 NADH: ubiquinone oxidoreductase MLRQ subunit NP_064527 1 20-42 Unsure
    homolog
    Hs.8546 Notch homolog 3 (Drosophila) NP_000426 3 1641-1663, 1496-1518, Unsure
    20-42
    Hs.107125 plasmalemma vesicle associated protein NP_112600 1 42-64 IN
    Hs.83974 solute carrier family 21 (prostaglandin transporter), NP_005621 12 256-278, 363-385, Unsure
    member 2 397-419, 100-122,
    208-230, 326-348,
    173-195, 514-536,
    71-93, 557-576,
    606-628, 25-47
    Hs.125359 TEM13, Thy-1 cell surface antigen NP_006279 1 140-161 Unsure
    Hs.125036 TEM17 NP_065138 1 425-447 OUT
    Hs.9598 sema domain, immunoglobulin domain (Ig) BAB21836 1 727-794 OUT
    Hs.202 Benzodiazapine receptor (peripheral)-mitochondrial NP_00715 3 107-129, 78-100, OUT
    133-155
  • Nuclear Proteins
    Unigene ID Function OMIMID Protein
    Hs.244 amino-terminal enhancer of split 600188
    Hs.154029 bHLH factor Hes4 NP_066993
    Hs.75450 delta sleep inducing peptide, immunoreactor 602960
    Hs.75087 FAST kinase 606965 NP_079372
    Hs.356668 guanine nucleotide binding protein (G protein), gamma 5 600874 NP_005265
    Hs.406410 H19, imprinted maternally expressed untranslated mRNA 103280 BAB71280
    Hs.234434 hairy/enhancer-of-split related with YRPW motif 1 602953 NP_036390
    Hs.23823 hairy/enhancer-of-split related with YRPW motif-like NP_055386
    Hs.15265 heterogeneous nuclear ribonucleoprotein R 607201 NP_005817
    Hs.8728 hypothetical protein DKFZp434G171 CAB61365
    Hs.240170 hypothetical protein MGC2731 NP_076973
    Hs.146360 hypothetical protein MGC34648 NP_689873
    Hs.337986 hypothetical protein MGC4677 NP_443103
    Hs.197540 hypoxia-inducible factor 1, alpha subunit (basic helix- 603348 NP_001521
    loop-helix transcription factor)
    Hs.75061 MARCKS-like protein 602940 NP_075385
    Hs.246857 mitogen-activated protein kinase 9 602896 NP_620708
    Hs.79110 Nucleolin 164035 NP_005372
    Hs.298229 prefoldin 2 NP_036526
    Hs.250655 prothymosin, alpha (gene sequence 28) 188390 NP_002814
    Hs.24950 regulator of G-protein signalling 5 603276 NP_003608
    Hs.76640 RGC32 protein
    Hs.3109 Rho GTPase activating protein 4 300023 NP_001657
    Hs.337445 ribosomal protein L37 604181 NP_000988
    Hs.197114 serine/arginine repetitive matrix 2 606032 NP_057417
    Hs.48029 snail homolog 1 (Drosophila) 604238 NP_005976
    Hs.168357 T-box 2 600747 NP_005985
    Hs.332173 transducin-like enhancer of split 2 (E(sp1) homolog, 601041 NP_003251
    Drosophila)
    Hs.78465 v-jun sarcoma virus 17 oncogene homolog (avian) 165160 NP_002219
  • Cytoplasmic proteins
    Unigene ID Function OMIMID Protein
    Hs.184367 Ca2+-promoted Ras inactivator BAA25464
    Hs.2575 calpain 1, (mu/l) large subunit 114220 NP_005177
    Hs.100009 cyclin-dependent kinase 3 123828
    Hs.31053 cytoskeleton-associated protein 1 601303 NP_001272
    Hs.82646 DnaJ (Hsp40) homolog, subfamily B, member 1 604572 NP_006136
    Hs.169476 glyceraldehyde-3-phosphate dehydrogenase 138400 NP_002037
    Hs.77890 guanylate cyclase 1, soluble, beta 3 139397 NP_000848
    Hs.36927 heat shock 105 Kd NP_006635
    Hs.1197 heat shock 10 kDa protein 1 (chaperonin 10) 600141 NP_002148
    Hs.8997 heat shock 70 kDa protein 1A 140550 NP_005336
    Hs.180414 heat shock 70 kDa protein 8 600816 NP_006588
    Hs.118625 hexokinase 1 142600 NP_277035
    Hs.327412 Homo sapiens clone FLC1492 PRO3121 mRNA,
    complete cds
    Hs.833 interferon, alpha-inducible protein (clone IFI-15K) 147571 NP_005092
    Hs.150580 putative translation initiation factor NP_005792
    Hs.173737 ras-related C3 botulinum toxin substrate 1 (rho 602048
    family, small GTP binding protein Rac1)
    Hs.119122 ribosomal protein L13a
    Hs.111611 ribosomal protein L27 607526 NP_000979
    Hs.177592 ribosomal protein, large, P1 180520
    Hs.12956 Tax interaction protein 1 NP_055419
    Hs.251653 tubulin, beta, 2 602660 NP_006079
  • REFERENCES
    • Abounader, R., Lal, B., Luddy, C., Koe, G., Davidson, B., Rosen, E. M., and Laterra, J. (2002). In vivo targeting of SF/HGF and c-met expression via U1snRNA/ribozymes inhibits breast tumor growth and angiogenesis and promotes apoptosis. Faseb J 16, 108-10.
    • Bart, J., Groen, H. J., Hendrikse, N. H., van der Graaf, W. T., Vaalburg, W., and de Vries, E. G. (2000). The blood-brain barrier and oncology: new insights into function and modulation. Cancer Treat Rev 26, 449-62.
    • Bernsen, H. J., Rijken, P. F., Oostendorp, T., and van der Kogel, A. J. (1995). Vascularity and perfusion of human breast tumors xenografted in the athymic nude mouse. Br J Cancer 71, 721-6.
    • Bowers, D. C., Fan, S., Walter, K. A., Abounader, R., Williams, J. A., Rosen, E. M., and Laterra, J. (2000). Scatter factor/hepatocyte growth factor protects against cytotoxic death in human glioblastoma via phosphatidylinositol 3-kinase- and AKT-dependent pathways. Cancer Res 60, 4277-83.
    • Chen, H., Centola, M., Altschul, S. F., and Metzger, H. (1998). Characterization of gene expression in resting and activated mast cells. J Exp Med 188, 1657-68.
    • Guerin, C., Wolff, J. E., Laterra, J., Drewes, L. R., Brem, H., and Goldstein, G. W. (1992). Vascular differentiation and glucose transporter expression in rat breast tumors: effects of steroids. Ann Neurol 31, 481-7.
    • Hobbs, S. K., Monsky, W. L., Yuan, F., Roberts, W. G., Griffith, L., Torchilin, V. P., and Jain, R. K. (1998). Regulation of transport pathways in tumor vessels: role of tumor type and microenvironment. Proc Natl Acad Sci USA 95, 4607-12.
    • Holash, J., Maisonpierre, P. C., Compton, D., Boland, P., Alexander, C. R., Zagzag, D., Yancopoulos, G. D., and Wiegand, S. J. (1999). Vessel cooption, regression, and growth in tumors mediated by angiopoietins and VEGF. Science 284, 1994-8.
    • Huminiecki, L., and Bicknell, R. (2000). In silico cloning of novel endothelial-specific genes. Genome Res 10, 1796-806.
    • Lamszus, K., Laterra, J., Westphal, M., and Rosen, E. M. (1999). Scatter factor/hepatocyte growth factor (SF/HGF) content and function in human breast tumors. Int J Dev Neurosci 17, 517-30.
    • Marx, J. (2001). Caveolae: a once-elusive structure gets some respect. Science 294, 1862-5.
    • Roberts, W. G., and Palade, G. E. (1997). Neovasculature induced by vascular endothelial growth factor is fenestrated. Cancer Res 57, 765-72.
    • Shinoura, N., Shamraj, O. I., Hugenholz, H., Zhu, J. G., McBlack, P., Warnick, R., Tew, J. J., Wani, M. A., and Menon, A. G. (1995). Identification and partial sequence of a cDNA that is differentially expressed in human brain tumors. Cancer Lett 89, 215-21.
    • Smith, R. M., Jarret, L. (1988). Lab. Invest. 58, 613-629.
    • St Croix, B., Rago, C., Velculescu, V., Traverso, G., Romans, K. E., Montgomery, E., Lal, A., Riggins, G. J., Lengauer, C., Vogelstein, B., and Kinzler, K. W. (2000). Genes expressed in human tumor endothelium. Science 289, 1197-202.
    • Stan, R. V., Arden, K. C., and Palade, G. E. (2001). cDNA and protein sequence, genomic organization, and analysis of cis regulatory elements of mouse and human PLVAP genes. Genomics 72, 304-13.
    • Tamagnone, L., Artigiani, S., Chen, H., He, Z., Ming, G. I., Song, H., Chedotal, A., Winberg, M. L., Goodman, C. S., Poo, M., Tessier-Lavigne, M., and Comoglio, P. M. (1999). Plexins are a large family of receptors for transmembrane, secreted, and GPI-anchored semaphorins in vertebrates. Cell 99, 71-80.
    • Vajkoczy, P., and Menger, M. D. (2000). Vascular microenvironment in breast tumors. J Neurooncol 50, 99-108.
    • Vajkoczy, P., Schilling, L., Ullrich, A., Schmiedek, P., and Menger, M. D. (1998). Characterization of angiogenesis and microcirculation of high-grade breast tumor: an intravital multifluorescence microscopic approach in the athymic nude mouse. J Cereb Blood Flow Metab 18, 510-20.
    • Vick, N. A., and Bigner, D. D. (1972). Microvascular abnormalities in virally-induced canine BREAST tumors. Structural bases for altered blood-brain barrier function. J Neurol Sci 17, 29-39.

Claims (42)

1. A method to aid in diagnosing breast tumor, comprising the steps of:
detecting an expression product of at least one gene in a first brain tissue sample suspected of being neoplastic wherein said at least one gene is selected from the group consisting of hypothetical protein DKFZp434G171; heat shock 70 kDa protein 1A; jagged 1 (Alagille syndrome); cyclin-dependent kinase 3; 6-phosphogluconolactonase; likely homolog of rat and mouse retinoid-inducible serine carboxypeptidase; plasmalemma vesicle associated protein; NADH:ubiquinone oxidoreductase MLRQ subunit homolog; HIF-1 responsive RTP801; ribosomal protein L27; secreted protein, acidic, cysteine-rich (osteonectin); hexokinase 1; ribosomal protein L13a; collagen, type IV, alpha 1; insulin-like growth factor binding protein 7; collagen, type III, alpha 1 (Ehlers-Danlos syndrome type IV, autosomal dominant); heat shock 10 kDa protein 1 (chaperonin 10); calcium channel, voltage-dependent, alpha 1H subunit; CD9 antigen (p24); TEM17; TEM13, Thy-1 cell surface antigen; Tax interaction protein 1; dysferlin, limb girdle muscular dystrophy 2B (autosomal recessive); hypothetical protein MGC34648; putative translation initiation factor; insulin-like growth factor binding protein 4; matrix metalloproteinase 9 (gelatinase B, 92 kDa gelatinase, 92 kDa type IV collagenase); heterogeneous nuclear ribonucleoprotein R; bHLH factor Hes4; collagen, type VI, alpha 2; T-box 2; glyceraldehyde-3-phosphate dehydrogenase; G protein-coupled receptor 4; collagen, type I, alpha 1; ras-related C3 botulinum toxin substrate 1 (rho family, small GTP binding protein Rac1); ribosomal protein, large, P1; TEM10, COL1A2 involved in tissue remodeling; heat shock 70 kDa protein 8; KIAA0152 gene product; Ca2+-promoted Ras inactivator; serine/arginine repetitive matrix 2; hypoxia-inducible factor 1, alpha subunit (basic helix-loop-helix transcription factor); benzodiazapine receptor (peripheral); ectonucleoside triphosphate diphosphohydrolase 1; heparan sulfate proteoglycan 2 (perlecan); fibromodulin; hairy/enhancer-of-split related with YRPW motif 1; collagen, type V, alpha 3; hairy/enhancer-of-split related with YRPW motif-like; hypothetical protein MGC2731; amino-terminal enhancer of split; mitogen-activated protein kinase 9; regulator of G-protein signalling 5; prothymosin, alpha (gene sequence 28); tubulin, beta, 2; protease, serine, 23; hypothetical protein FLJ20898; calpain 1, (mu/I) large subunit; interferon, alpha-inducible protein (clone IFI-6-16); ESTs, Weakly similar to T25031 hypothetical protein T20D3.3—Caenorhabditis elegans [C. elegans]; major histocompatibility complex, class I, C; hypoxia up-regulated 1; complement component 4B; prefoldin 2; cytoskeleton-associated protein 1; Rho GTPase activating protein 4; Homo sapiens clone FLC1492 PRO3121 mRNA, complete cds; transducin-like enhancer of split 2 (E(spl) homolog, Drosophila); ribosomal protein L37; hypothetical protein MGC4677; ESTs, Highly similar to MT1A_HUMAN METALLOTHIONEIN-IA (MT-1A) [H. sapiens]; TEM11, nidogen (enactin); guanine nucleotide binding protein (G protein), gamma 5; matrix Gla protein; heat shock 105 kD; GNAS complex locus; Homo sapiens cDNA FLJ11658 fis, clone HEMBA1004577; H19, imprinted maternally expressed untranslated mRNA; protein tyrosine phosphatase type IVA, member 3; snail homolog 1 (Drosophila); integrin-binding sialoprotein (bone sialoprotein, bone sialoprotein II); tissue inhibitor of metalloproteinase 1 (erythroid potentiating activity, collagenase inhibitor); peptidylprolyl isomerase B (cyclophilin B); MARCKS-like protein; FAST kinase; protease, serine, 11 (IGF binding); beta-2-microglobulin; delta sleep inducing peptide, immunoreactor; collagen, type IV, alpha 2; immediate early response 3; cadherin 5, type 2, VE-cadherin (vascular epithelium); RGC32 protein; guanylate cyclase 1, soluble, beta 3; major histocompatibility complex, class I, B; ribonuclease, RNase A family, 1 (pancreatic); collagen, type XVIII, alpha 1; v-jun sarcoma virus 17 oncogene homolog (avian); Homo sapiens mRNA; cDNA DKFZp686G1610 (from clone DKFZp686G1610); nucleolin; lectin, galactoside-binding, soluble, 3 binding protein; Lysosomal-associated multispanning membrane protein-5; ribosomal protein S16; guanine nucleotide binding protein (G protein), gamma 12; serine (or cysteine) proteinase inhibitor, clade E (nexin, plasminogen activator inhibitor type 1), member 1; biglycan; DnaJ (Hsp40) homolog, subfamily B, member 1; tumor rejection antigen (gp96) 1; interferon, alpha-inducible protein (clone IFI-15K); solute carrier family 21 (prostaglandin transporter), member 2; CD74 antigen (invariant polypeptide of major histocompatibility complex, class II antigen-associated); serum/glucocorticoid regulated kinase; mitogen-activated protein kinase; receptor (calcitonin) activity modifying protein 3; sema domain, immunoglobulin domain (Ig); benzodiazapine receptor (peripheral)—mitochondrial; C1 domain-containing phosphatase & tensin-like; and Notch homolog 3 (Drosophila); and
comparing expression of the at least one gene in the first breast tissue sample with expression of the at least one gene in a second breast tissue sample which is normal, wherein increased expression of the at least one gene in the first breast tissue sample relative to the second tissue sample identifies the first breast tissue sample as likely to be neoplastic.
2. The method of claim 1 wherein the increased expression of the at least one gene in the first breast tissue sample relative to the second tissue sample is at least two-fold higher.
3. The method of claim 1 wherein the increased expression of the at least one gene in the first breast tissue sample relative to the second tissue sample is at least five-fold higher.
4. The method of claim 1 wherein the increased expression of the at least one gene in the first breast tissue sample relative to the second tissue sample is at least ten-fold higher.
5. The method of claim 1 wherein the expression product is RNA.
6. The method of claim 1 wherein the expression product is protein.
7. The method of claim 1 wherein the first and second tissue samples are from a human.
8. The method of claim 1 wherein the first and second tissue samples are from the same human.
9. The method of claim 1 wherein the step of detecting is performed using a Western blot.
10. The method of claim 1 wherein the step of detecting is performed using an immunoassay.
11. The method of claim 1 wherein the step of detecting is performed using an immunohistochemical assay.
12. The method of claim 1 wherein the step of detecting is performed using SAGE.
13. The method of claim 1 wherein the step of detecting is performed using hybridization to a microarray.
14. A method of treating a breast tumor, comprising the step of:
contacting cells of the breast tumor with an antibody, wherein the antibody specifically binds to an extracellular epitope of a protein selected from the group consisting of benzodiazapine receptor (peripheral); cadherin 5, type 2, VE-cadherin (vascular epithelium); calcium channel, voltage-dependent, alpha 1H subunit; CD74 antigen (invariant polypeptide of major histocompatibility complex, class II antigen-associated); CD9 antigen (p24); dysferlin, limb girdle muscular dystrophy 2B (autosomal recessive); ectonucleoside triphosphate diphosphohydrolase 1; G protein-coupled receptor 4; hypothetical protein FLJ20898; hypoxia up-regulated 1; immediate early response 3; interferon, alpha-inducible protein (clone IFI-6-16); jagged 1 (Alagille syndrome); KIAA0152 gene product; Lysosomal-associated multispanning membrane protein-5; major histocompatibility complex, class I, B; major histocompatibility complex, class I, C; NADH:ubiquinone oxidoreductase MLRQ subunit homolog; Notch homolog 3 (Drosophila); plasmalemma vesicle associated protein; solute carrier family 21 (prostaglandin transporter), member 2; TEM13, Thy-1 cell surface antigen; receptor (calcitonin) activity modifying protein 3; sema domain, immunoglobulin domain (Ig); benzodiazapine receptor (peripheral)—mitochondrial; and TEM17; whereby immune destruction of cells of the breast tumor is triggered.
15. The method of claim 14 wherein the antibody is conjugated to a diagnostic or therapeutic reagent.
16. The method of claim 14 wherein the breast tumor is multidrug-sensitive.
17. The method of claim 14 wherein the reagent is a chemotherapeutic agent.
18. The method of claim 14 wherein the reagent is a cytotoxin.
19. The method of claim 14 wherein the reagent is a non-radioactive label.
20. The method of claim 14 wherein the reagent is a radioactive compound.
21. The method of claim 14 wherein the breast tumor is in a human.
22. A method of identifying a test compound as a potential anti-cancer or anti-breast tumor drug, comprising the step of:
contacting a test compound with a cell which expresses at least one gene selected from the group consisting of hypothetical protein DKFZp434G171; heat shock 70 kDa protein 1A; jagged 1 (Alagille syndrome); cyclin-dependent kinase 3; 6-phosphogluconolactonase; likely homolog of rat and mouse retinoid-inducible serine carboxypeptidase; plasmalemma vesicle associated protein; NADH:ubiquinone oxidoreductase MLRQ subunit homolog; HIF-1 responsive RTP801; ribosomal protein L27; secreted protein, acidic, cysteine-rich (osteonectin); hexokinase 1; ribosomal protein L13a; collagen, type IV, alpha 1; insulin-like growth factor binding protein 7; collagen, type III, alpha 1 (Ehlers-Danlos syndrome type IV, autosomal dominant); heat shock 10 kDa protein 1 (chaperonin 10); calcium channel, voltage-dependent, alpha 1H subunit; CD9 antigen (p24); TEM17; TEM13, Thy-1 cell surface antigen; Tax interaction protein 1; dysferlin, limb girdle muscular dystrophy 2B (autosomal recessive); hypothetical protein MGC34648; putative translation initiation factor; insulin-like growth factor binding protein 4; matrix metalloproteinase 9 (gelatinase B, 92 kDa gelatinase, 92 kDa type IV collagenase); heterogeneous nuclear ribonucleoprotein R; bHLH factor Hes4; collagen, type VI, alpha 2; T-box 2; glyceraldehyde-3-phosphate dehydrogenase; G protein-coupled receptor 4; collagen, type I, alpha 1; ras-related C3 botulinum toxin substrate 1 (rho family, small GTP binding protein Rac1); ribosomal protein, large, P1; TEM10, COL1A2 involved in tissue remodeling; heat shock 70 kDa protein 8; KIAA0152 gene product; Ca2+-promoted Ras inactivator; serine/arginine repetitive matrix 2; hypoxia-inducible factor 1, alpha subunit (basic helix-loop-helix transcription factor); benzodiazapine receptor (peripheral); ectonucleoside triphosphate diphosphohydrolase 1; heparan sulfate proteoglycan 2 (perlecan); fibromodulin; hairy/enhancer-of-split related with YRPW motif 1; collagen, type V, alpha 3; hairy/enhancer-of-split related with YRPW motif-like; hypothetical protein MGC2731; amino-terminal enhancer of split; mitogen-activated protein kinase 9; regulator of G-protein signalling 5; prothymosin, alpha (gene sequence 28); tubulin, beta, 2; protease, serine, 23; hypothetical protein FLJ20898; calpain 1, (mu/I) large subunit; interferon, alpha-inducible protein (clone IFI-6-16); ESTs, Weakly similar to T25031 hypothetical protein T20D3.3—Caenorhabditis elegans [C. elegans]; major histocompatibility complex, class I, C; hypoxia up-regulated 1; complement component 4B; prefoldin 2; cytoskeleton-associated protein 1; Rho GTPase activating protein 4; Homo sapiens clone FLC1492 PRO3121 mRNA, complete cds; transducin-like enhancer of split 2 (E(spl) homolog, Drosophila); ribosomal protein L37; hypothetical protein MGC4677; ESTs, Highly similar to MT1A_HUMAN METALLOTHIONEIN-IA (MT-1A) [H. sapiens]; TEM11, nidogen (enactin); guanine nucleotide binding protein (G protein), gamma 5; matrix Gla protein; heat shock 105 kD; GNAS complex locus; Homo sapiens cDNA FLJ11658 fis, clone HEMBA1004577; H19, imprinted maternally expressed untranslated mRNA; protein tyrosine phosphatase type IVA, member 3; snail homolog 1 (Drosophila); integrin-binding sialoprotein (bone sialoprotein, bone sialoprotein II); tissue inhibitor of metalloproteinase 1 (erythroid potentiating activity, collagenase inhibitor); peptidylprolyl isomerase B (cyclophilin B); MARCKS-like protein; FAST kinase; protease, serine, 11 (IGF binding); beta-2-microglobulin; delta sleep inducing peptide, immunoreactor; collagen, type IV, alpha 2; immediate early response 3; cadherin 5, type 2, VE-cadherin (vascular epithelium); RGC32 protein; guanylate cyclase 1, soluble, beta 3; major histocompatibility complex, class I, B; ribonuclease, RNase A family, 1 (pancreatic); collagen, type XVIII, alpha 1; v-jun sarcoma virus 17 oncogene homolog (avian); Homo sapiens mRNA; cDNA DKFZp686G1610 (from clone DKFZp686G1610); nucleolin; lectin, galactoside-binding, soluble, 3 binding protein; Lysosomal-associated multispanning membrane protein-5; ribosomal protein S16; guanine nucleotide binding protein (G protein), gamma 12; serine (or cysteine) proteinase inhibitor, clade E (nexin, plasminogen activator inhibitor type 1), member 1; biglycan; DnaJ (Hsp40) homolog, subfamily B, member 1; tumor rejection antigen (gp96) 1; interferon, alpha-inducible protein (clone IFI-15K); solute carrier family 21 (prostaglandin transporter), member 2; CD74 antigen (invariant polypeptide of major histocompatibility complex, class II antigen-associated); serum/glucocorticoid regulated kinase; mitogen-activated protein kinase; receptor (calcitonin) activity modifying protein 3; sema domain, immunoglobulin domain (Ig); benzodiazapine receptor (peripheral)—mitochondrial; C1 domain-containing phosphatase & tensin-like; and Notch homolog 3 (Drosophila);
monitoring an expression product of the at least one gene; and
identifying the test compound as a potential anti-cancer drug if it decreases the expression of the at least one gene.
23. The method of claim 22 wherein the cell is a human cell.
24. The method of claim 22 wherein the cell is a breast tumor cell.
25. The method of claim 22 wherein the cell is a human breast tumor cell.
26. The method of claim 22 wherein the expression product is RNA.
27. The method of claim 22 wherein the expression product is protein.
28. The method of claim 22 wherein the cell overexpresses the at least one gene relative to a normal cell of the same tissue.
29. The method of claim 22 wherein expression of at least two of said genes is monitored.
30. The method of claim 22 wherein expression of at least three of said genes is monitored.
31. The method of claim 22 wherein expression of at least four of said genes is monitored.
32. The method of claim 22 wherein the test compound is identified if the decrease in expression is at least two-fold.
33. The method of claim 22 wherein the test compound is identified if the decrease in expression is at least five-fold.
34. The method of claim 22 wherein the decrease in expression is at least ten-fold.
35. The method of claim 22 wherein the test compound is identified as an anti-breast tumor drug.
36. A method to induce an immune response to a breast tumor, comprising:
administering to a mammal a protein or nucleic acid encoding a protein selected from the group consisting of: hypothetical protein DKFZp434G171; heat shock 70 kDa protein 1A; jagged 1 (Alagille syndrome); cyclin-dependent kinase 3; 6-phosphogluconolactonase; likely homolog of rat and mouse retinoid-inducible serine carboxypeptidase; plasmalemma vesicle associated protein; NADH:ubiquinone oxidoreductase MLRQ subunit homolog; HIF-1 responsive RTP801; ribosomal protein L27; secreted protein, acidic, cysteine-rich (osteonectin); hexokinase 1; ribosomal protein L13a; collagen, type IV, alpha 1; insulin-like growth factor binding protein 7; collagen, type III, alpha 1 (Ehlers-Danlos syndrome type IV, autosomal dominant); heat shock 10 kDa protein 1 (chaperonin 10); calcium channel, voltage-dependent, alpha 1H subunit; CD9 antigen (p24); TEM17; TEM13, Thy-1 cell surface antigen; Tax interaction protein 1; dysferlin, limb girdle muscular dystrophy 2B (autosomal recessive); hypothetical protein MGC34648; putative translation initiation factor; insulin-like growth factor binding protein 4; matrix metalloproteinase 9 (gelatinase B, 92 kDa gelatinase, 92 kDa type IV collagenase); heterogeneous nuclear ribonucleoprotein R; bHLH factor Hes4; collagen, type VI, alpha 2; T-box 2; glyceraldehyde-3-phosphate dehydrogenase; G protein-coupled receptor 4; collagen, type I, alpha 1; ras-related C3 botulinum toxin substrate 1 (rho family, small GTP binding protein Rac1); ribosomal protein, large, P1; TEM10, COL1A2 involved in tissue remodeling; heat shock 70 kDa protein 8; KIAA0152 gene product; Ca2+-promoted Ras inactivator; serine/arginine repetitive matrix 2; hypoxia-inducible factor 1, alpha subunit (basic helix-loop-helix transcription factor); benzodiazapine receptor (peripheral); ectonucleoside triphosphate diphosphohydrolase 1; heparan sulfate proteoglycan 2 (perlecan); fibromodulin; hairy/enhancer-of-split related with YRPW motif 1; collagen, type V, alpha 3; hairy/enhancer-of-split related with YRPW motif-like; hypothetical protein MGC2731; amino-terminal enhancer of split; mitogen-activated protein kinase 9; regulator of G-protein signalling 5; prothymosin, alpha (gene sequence 28); tubulin, beta, 2; protease, serine, 23; hypothetical protein FLJ20898; calpain 1, (mu/I) large subunit; interferon, alpha-inducible protein (clone IFI-6-16); ESTs, Weakly similar to T25031 hypothetical protein T20D3.3—Caenorhabditis elegans [C. elegans]; major histocompatibility complex, class I, C; hypoxia up-regulated 1; complement component 4B; prefoldin 2; cytoskeleton-associated protein 1; Rho GTPase activating protein 4; Homo sapiens clone FLC1492 PRO3121 mRNA, complete cds; transducin-like enhancer of split 2 (E(spl) homolog, Drosophila); ribosomal protein L37; hypothetical protein MGC4677; ESTs, Highly similar to MT1A HUMAN METALLOTHIONEIN-IA (MT-1A) [H. sapiens]; TEM11, nidogen (enactin); guanine nucleotide binding protein (G protein), gamma 5; matrix Gla protein; heat shock 105 kD; GNAS complex locus; Homo sapiens cDNA FLJ11658 fis, clone HEMBA1004577; H19, imprinted maternally expressed untranslated mRNA; protein tyrosine phosphatase type IVA, member 3; snail homolog 1 (Drosophila); integrin-binding sialoprotein (bone sialoprotein, bone sialoprotein II); tissue inhibitor of metalloproteinase 1 (erythroid potentiating activity, collagenase inhibitor); peptidylprolyl isomerase B (cyclophilin B); MARCKS-like protein; FAST kinase; protease, serine, 11 (IGF binding); beta-2-microglobulin; delta sleep inducing peptide, immunoreactor; collagen, type IV, alpha 2; immediate early response 3; cadherin 5, type 2, VE-cadherin (vascular epithelium); RGC32 protein; guanylate cyclase 1, soluble, beta 3; major histocompatibility complex, class I, B; ribonuclease, RNase A family, 1 (pancreatic); collagen, type XVIII, alpha 1; v-jun sarcoma virus 17 oncogene homolog (avian); Homo sapiens mRNA; cDNA DKFZp686G1610 (from clone DKFZp686G1610); nucleolin; lectin, galactoside-binding, soluble, 3 binding protein; Lysosomal-associated multispanning membrane protein-5; ribosomal protein S16; guanine nucleotide binding protein (G protein), gamma 12; serine (or cysteine) proteinase inhibitor, clade E (nexin, plasminogen activator inhibitor type 1), member 1; biglycan; DnaJ (Hsp40) homolog, subfamily B, member 1; tumor rejection antigen (gp96) 1; interferon, alpha-inducible protein (clone IFI-15K); solute carrier family 21 (prostaglandin transporter), member 2; CD74 antigen (invariant polypeptide of major histocompatibility complex, class II antigen-associated); serum/glucocorticoid regulated kinase; mitogen-activated protein kinase; receptor (calcitonin) activity modifying protein 3; sema domain, immunoglobulin domain (Ig); benzodiazapine receptor (peripheral)—mitochondrial; C1 domain-containing phosphatase & tensin-like; and Notch homolog 3 (Drosophila), whereby an immune response to the protein is induced.
37. The method of claim 36 wherein a protein is administered.
38. The method of claim 36 wherein a nucleic acid is administered.
39. The method of claim 38 wherein the nucleic acid is administered intramuscularly.
40. The method of claim 36 further comprising administering an immune adjuvant to the mammal.
41. The method of claim 36 wherein the mammal has a breast tumor.
42. The method of claim 36 wherein the mammal has had a breast tumor surgically removed.
US12/269,418 2003-04-01 2008-11-12 Breast Endothelial Cell Expression Patterns Abandoned US20090117038A1 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
US12/269,418 US20090117038A1 (en) 2003-04-01 2008-11-12 Breast Endothelial Cell Expression Patterns
US13/176,222 US8568985B2 (en) 2003-04-01 2011-07-05 Breast endothelial cell expression patterns
US14/065,587 US20140056911A1 (en) 2003-04-01 2013-10-29 Breast Endothelial Cell Expression Patterns
US14/800,457 US20150315269A1 (en) 2003-04-01 2015-07-15 Breast Endothelial Cell Expression Patterns

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US45896003P 2003-04-01 2003-04-01
PCT/US2004/009704 WO2004091383A2 (en) 2003-04-01 2004-03-31 Breast endothelial cell expression patterns
US10/551,217 US20070178090A1 (en) 2003-04-01 2004-03-31 Breast endothelial cell expression patterns
US12/269,418 US20090117038A1 (en) 2003-04-01 2008-11-12 Breast Endothelial Cell Expression Patterns

Related Parent Applications (3)

Application Number Title Priority Date Filing Date
US10/551,217 Continuation US20070178090A1 (en) 2003-04-01 2004-03-31 Breast endothelial cell expression patterns
PCT/US2004/009704 Continuation WO2004091383A2 (en) 2003-04-01 2004-03-31 Breast endothelial cell expression patterns
US55121706A Continuation 2003-04-01 2006-12-12

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US13/176,222 Continuation US8568985B2 (en) 2003-04-01 2011-07-05 Breast endothelial cell expression patterns

Publications (1)

Publication Number Publication Date
US20090117038A1 true US20090117038A1 (en) 2009-05-07

Family

ID=33299657

Family Applications (5)

Application Number Title Priority Date Filing Date
US10/551,217 Abandoned US20070178090A1 (en) 2003-04-01 2004-03-31 Breast endothelial cell expression patterns
US12/269,418 Abandoned US20090117038A1 (en) 2003-04-01 2008-11-12 Breast Endothelial Cell Expression Patterns
US13/176,222 Expired - Lifetime US8568985B2 (en) 2003-04-01 2011-07-05 Breast endothelial cell expression patterns
US14/065,587 Abandoned US20140056911A1 (en) 2003-04-01 2013-10-29 Breast Endothelial Cell Expression Patterns
US14/800,457 Abandoned US20150315269A1 (en) 2003-04-01 2015-07-15 Breast Endothelial Cell Expression Patterns

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US10/551,217 Abandoned US20070178090A1 (en) 2003-04-01 2004-03-31 Breast endothelial cell expression patterns

Family Applications After (3)

Application Number Title Priority Date Filing Date
US13/176,222 Expired - Lifetime US8568985B2 (en) 2003-04-01 2011-07-05 Breast endothelial cell expression patterns
US14/065,587 Abandoned US20140056911A1 (en) 2003-04-01 2013-10-29 Breast Endothelial Cell Expression Patterns
US14/800,457 Abandoned US20150315269A1 (en) 2003-04-01 2015-07-15 Breast Endothelial Cell Expression Patterns

Country Status (4)

Country Link
US (5) US20070178090A1 (en)
EP (3) EP2233926A3 (en)
JP (1) JP2007525167A (en)
WO (1) WO2004091383A2 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9127053B2 (en) 2012-06-22 2015-09-08 Cytomx Therapeutics, Inc. Anti-jagged 1/jagged 2 cross-reactive antibodies, activatable anti-jagged antibodies and methods of use thereof
US9260517B2 (en) 2009-11-17 2016-02-16 Musc Foundation For Research Development Human monoclonal antibodies to human nucleolin

Families Citing this family (28)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6984522B2 (en) 2000-08-03 2006-01-10 Regents Of The University Of Michigan Isolation and use of solid tumor stem cells
AU2006249235B2 (en) * 2004-05-14 2010-11-11 Abraxis Bioscience, Llc Sparc and methods of use thereof
US8420603B2 (en) 2004-05-14 2013-04-16 Abraxis Bioscience, Llc SPARC and methods of use thereof
CA2566571A1 (en) * 2004-05-14 2005-12-15 Abraxis Bioscience, Inc. Treatment methods utilizing albumin-binding proteins as targets
UA92465C2 (en) * 2004-08-16 2010-11-10 Кварк Фармасьютикалз Инк. Therapeutic uses of inhibitors of rtp801
CN101123994B (en) 2004-08-16 2012-11-14 夸克医药公司 Therapeutic uses of inhibitors of rtp801
WO2006081473A2 (en) * 2005-01-28 2006-08-03 Children's Medical Center Corporation Methods for diagnosis and prognosis of epithelial cancers
CA2598510C (en) * 2005-02-18 2011-12-20 Abraxis Bioscience, Inc. Q3 sparc deletion mutant and uses thereof
CN1862258B (en) 2005-05-12 2012-05-30 清华大学 Nucleolin assisted method for diagnosing and curing cancer
NL2000439C2 (en) 2006-01-20 2009-03-16 Quark Biotech Therapeutic applications of inhibitors of RTP801.
WO2008054534A2 (en) 2006-05-11 2008-05-08 Quark Pharmaceuticals, Inc. Screening systems utilizing rtp801
WO2007141796A2 (en) 2006-06-09 2007-12-13 Quark Pharmaceuticals, Inc. Therapeutic uses of inhibitors of rtp801l
JP2009539403A (en) 2006-06-13 2009-11-19 オンコメッド ファーマシューティカルズ インコーポレイテッド Compositions and methods for diagnosing and treating cancer
WO2008091781A1 (en) * 2007-01-22 2008-07-31 Mayo Foundation For Medical Education And Research Reducing tumor growth
US8088617B2 (en) 2007-01-24 2012-01-03 Oncomed Pharmaceuticals, Inc. Antibodies that bind the glutamate ligand binding region of Notch1
US7872119B2 (en) 2007-02-26 2011-01-18 Quark Pharmaceuticals, Inc. Inhibitors of RTP801 and their use in disease treatment
EA200700940A1 (en) 2007-04-27 2007-12-28 Петр Генриевич ЛОХОВ METHOD FOR OBTAINING ANTITUMENT VACCINE ON THE BASIS OF SURFACE ANTIGENES OF ENDOTHELIAL CELLS
EP2155249A4 (en) 2007-05-15 2011-11-16 Oncomed Pharm Inc Compositions and methods for diagnosing and treating cancer
EP2227693B1 (en) * 2007-11-30 2015-08-19 Clarient Diagnostic Services, Inc. Tle3 as a marker for chemotherapy
US8614311B2 (en) 2007-12-12 2013-12-24 Quark Pharmaceuticals, Inc. RTP801L siRNA compounds and methods of use thereof
US8435513B2 (en) 2008-07-08 2013-05-07 Oncomed Pharmaceuticals, Inc. NOTCH1 receptor antibodies and methods of treatment
US9132189B2 (en) 2008-07-08 2015-09-15 Oncomed Pharmaceuticals, Inc. Notch1 binding agents and methods of use thereof
ES2363669B1 (en) * 2009-06-10 2012-08-09 Fundacio Privada Institut D'investigacio Biomedica De Bellvitge (Idibell) METHOD FOR DETERMINING THE RISK OF DEVELOPING BRAIN METASTASIS AND A KIT TO CARRY OUT THIS PROCEDURE
WO2011088215A2 (en) 2010-01-13 2011-07-21 Oncomed Pharmaceuticals, Inc. Notch1 binding agents and methods of use thereof
EP2742358A4 (en) * 2011-09-08 2015-08-12 Univ Ben Gurion Methods for diagnosing cancer
WO2016022914A1 (en) * 2014-08-08 2016-02-11 Moderna Therapeutics, Inc. Compositions and methods for the treatment of ophthalmic diseases and conditions
CN114606309A (en) 2014-11-05 2022-06-10 威拉赛特公司 Diagnostic system and method using machine learning and high-dimensional transcription data
US11217329B1 (en) 2017-06-23 2022-01-04 Veracyte, Inc. Methods and systems for determining biological sample integrity

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6811776B2 (en) * 2000-12-27 2004-11-02 The Regents Of The University Of Michigan Process for ex vivo formation of mammalian bone and uses thereof
US6855554B2 (en) * 2001-09-21 2005-02-15 Board Of Regents, The University Of Texas Systems Methods and compositions for detection of breast cancer
US20050249666A1 (en) * 2003-07-10 2005-11-10 Central Institute For Experimental Animals Establishment of human cancer cell lines with metastatic potential using NOD/SCID

Family Cites Families (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6075181A (en) 1990-01-12 2000-06-13 Abgenix, Inc. Human antibodies derived from immunized xenomice
EP0950097A2 (en) * 1996-12-27 1999-10-20 Instituto de Investigaciones Bioquimicas Fundacion Campomar Compositions and methods for tumour therapy
AU2001271365A1 (en) * 2000-06-21 2002-01-02 Wyeth Cyr61 as a target for treatment and diagnosis of breast cancer
JP2005503760A (en) * 2001-01-24 2005-02-10 プロテイン デザイン ラブス, インコーポレイテッド Breast cancer diagnosis method, composition and breast cancer modulator screening method
WO2002079492A2 (en) * 2001-02-14 2002-10-10 Protein Design Labs, Inc. Methods of diagnosis of angiogenesis, compositions and methods of screening for angiogenesis modulators
JP2003043046A (en) * 2001-07-31 2003-02-13 Chemo Sero Therapeut Res Inst Marker for judgment of mammary cancer and judgment method for mammary cancer by using the same
WO2003042246A2 (en) * 2001-11-14 2003-05-22 Lorantis Limited Inhibitors of the notch signalling pathway for use in the treatment of cancer
AU2003205913A1 (en) * 2002-02-20 2003-09-09 Ncc Technology Ventures Pte Limited Materials and methods relating to cancer diagnosis
US20070054271A1 (en) * 2003-03-20 2007-03-08 Dana-Farber Cancer Institute, Inc. Gene expression in breast cancer
JP4286085B2 (en) * 2003-07-28 2009-06-24 Okiセミコンダクタ株式会社 Amplifier and semiconductor memory device using the same

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6811776B2 (en) * 2000-12-27 2004-11-02 The Regents Of The University Of Michigan Process for ex vivo formation of mammalian bone and uses thereof
US6855554B2 (en) * 2001-09-21 2005-02-15 Board Of Regents, The University Of Texas Systems Methods and compositions for detection of breast cancer
US20050249666A1 (en) * 2003-07-10 2005-11-10 Central Institute For Experimental Animals Establishment of human cancer cell lines with metastatic potential using NOD/SCID

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9260517B2 (en) 2009-11-17 2016-02-16 Musc Foundation For Research Development Human monoclonal antibodies to human nucleolin
US10385128B2 (en) 2009-11-17 2019-08-20 Musc Foundation For Research Development Nucleolin antibodies
US9127053B2 (en) 2012-06-22 2015-09-08 Cytomx Therapeutics, Inc. Anti-jagged 1/jagged 2 cross-reactive antibodies, activatable anti-jagged antibodies and methods of use thereof
US9688748B2 (en) 2012-06-22 2017-06-27 Cytomx Therapeutics, Inc. Anti-jagged 1/jagged 2 cross-reactive antibodies, activatable anti-jagged antibodies and methods of use thereof
US10301380B2 (en) 2012-06-22 2019-05-28 Cytomx Therapeutics, Inc. Anti-jagged 1/jagged 2 cross-reactive antibodies, activatable anti-jagged antibodies and methods of use thereof

Also Published As

Publication number Publication date
US8568985B2 (en) 2013-10-29
EP2233926A3 (en) 2011-01-12
EP1608255A2 (en) 2005-12-28
EP2060918A2 (en) 2009-05-20
WO2004091383A2 (en) 2004-10-28
WO2004091383A3 (en) 2007-01-18
US20140056911A1 (en) 2014-02-27
EP2060918A3 (en) 2009-08-26
EP1608255A4 (en) 2008-06-25
JP2007525167A (en) 2007-09-06
US20070178090A1 (en) 2007-08-02
EP2233926A2 (en) 2010-09-29
US20150315269A1 (en) 2015-11-05
US20110262350A1 (en) 2011-10-27

Similar Documents

Publication Publication Date Title
US8568985B2 (en) Breast endothelial cell expression patterns
EP2365004B1 (en) Membrane associated tumor endothelium markers
ES2869167T3 (en) PIK3CA gene mutations in human cancers
US20120308479A1 (en) Brain Endothelial Cell Expression Patterns
JP2007518399A (en) Compositions and methods for diagnosing and treating lung cancer
WO2013033629A2 (en) Methods and compositions for the treatment and diagnosis of colorectal cancer
EP2780472A1 (en) Methods and compositions for the treatment and diagnosis of bladder cancer
CN101133167A (en) GITR antibodies for the diagnosis of NSCLC
WO2013106844A2 (en) Methods and compositions for the treatment and diaginosis of pancreatic cancer
US20180074062A1 (en) Compositions and methods for the identification, assessment, prevention and therapy of cancer
RU2144675C1 (en) Method for diagnosing, treating and predicting solid nonlymphoid tumors and their metastases
WO2004005883A2 (en) Secreted and cytoplasmic tumor endothelial markers
US20070065889A1 (en) Compositions and methods to diagnose and treat lung cancer
US6858386B1 (en) Method of diagnosing, monitoring, staging, imaging and treating colon cancer
JP2004514909A (en) Diagnosis method for cancer patients by measuring PIBF concentration
EP1705487A2 (en) Method of diagnosing, monitoring, staging, imaging and treating colon cancer
Suckow et al. Tissue vaccines for cancer
JP2004161776A (en) New method for diagnosing, monitoring, staging, imaging and treating cancer

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

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