US20090017030A1 - Endothelial Cell Expression Patterns - Google Patents

Endothelial Cell Expression Patterns Download PDF

Info

Publication number
US20090017030A1
US20090017030A1 US11/930,528 US93052807A US2009017030A1 US 20090017030 A1 US20090017030 A1 US 20090017030A1 US 93052807 A US93052807 A US 93052807A US 2009017030 A1 US2009017030 A1 US 2009017030A1
Authority
US
United States
Prior art keywords
tem
cells
seq
protein
patient
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US11/930,528
Inventor
Brad St.Croix
Bert Vogelstein
Kenneth W. Kinzler
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.)
Johns Hopkins University
Original Assignee
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 Johns Hopkins University filed Critical Johns Hopkins University
Priority to US11/930,528 priority Critical patent/US20090017030A1/en
Publication of US20090017030A1 publication Critical patent/US20090017030A1/en
Assigned to NATIONAL INSTITUTES OF HEALTH - DIRECTOR DEITR reassignment NATIONAL INSTITUTES OF HEALTH - DIRECTOR DEITR CONFIRMATORY LICENSE (SEE DOCUMENT FOR DETAILS). Assignors: THE JOHNS HOPKINS UNIVERSITY
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • 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
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies

Definitions

  • This invention is related to the area of angiogenesis and anti-angiogenesis. In particular, it relates to genes which are characteristically expressed in tumor endothelial and normal endothelial cells.
  • tumors require a blood supply for expansive growth. This recognition has stimulated a profusion of research on tumor angiogenesis, based on the idea that the vasculature in tumors represents a potential therapeutic target.
  • tumor endothelium remains unanswered. For example, are vessels of tumors qualitatively different from normal vessels of the same tissue? What is the relationship of tumor endothelium to endothelium of healing wounds or other physiological or pathological forms of angiogenesis? The answers to these questions critically impact on the potential for new therapeutic approaches to inhibit angiogenesis in a specific manner.
  • serial analysis of gene expression is termed serial analysis of gene expression (SAGE).
  • SAGE serial analysis of gene expression
  • SAGE Tags short defined sequence tags
  • Each Tag is a short nucleotide sequences (9-17 base pairs in length) from a defined position in the transcript.
  • the Tags are dimerized to reduce bias inherent in cloning or amplification reactions.
  • SAGE is particularly suited to the characterization of genes associated with vasculature stimulation or inhibition because it is capable of detecting rare sequences, evaluating large numbers of sequences at one time, and to provide a basis for the identification of previously unknown genes.
  • One embodiment of the invention provides an isolated molecule comprising an antibody variable region which specifically binds to an extracellular domain of a TEM protein selected from the group consisting of: 1, 3, 9, 17, 19, and 44, as shown in SEQ ID NO: 196, 200, 212, 230, 232, and 271, respectively.
  • the molecule can be, for example, an in tact antibody molecule, a single chain variable region (ScFv), a monoclonal antibody, a humanized antibody, or a human antibody.
  • the molecule can optionally be bound to a cytotoxic moiety, bound to a therapeutic moiety, bound to a detectable moiety, or bound to an anti-tumor agent.
  • a method of inhibiting neoangiogenesis is provided.
  • An effective amount of an isolated molecule comprising an antibody variable region which specifically binds to an extracellular domain of a TEM protein selected from the group consisting of: 1, 3, 9, 17, 19, 22, and 44, as shown in SEQ ID NO: 196, 200, 212, 230, 232, 238, and 271, respectively, is administered to a subject in need thereof.
  • Neoangiogenesis is consequently inhibited.
  • the subject may bear a vascularized tumor, may have polycystic kidney disease, may have diabetic retinopathy, may have rheumatoid arthritis, may have psoriasis, for example.
  • Another aspect of the invention is a method of inhibiting tumor growth.
  • An effective amount of an isolated molecule comprising an antibody variable region which specifically binds to an extracellular domain of a TEM protein selected from the group consisting of: 1, 3, 9, 17, 19, 22, and 44, as shown in SEQ ID NO: 196, 200, 212, 230, 232, 238, and 271, respectively, is administered to a human subject bearing a tumor. The growth of the tumor is consequently inhibited.
  • Still another aspect of the invention provides an isolated molecule comprising an antibody variable region which specifically binds to a TEM protein selected from the group consisting of: 3, 9, 17, 19, and 44, as shown in SEQ ID NO: 200, 212, 230, 232, and 271, respectively.
  • the molecule can be, for example, an in tact antibody molecule, a single chain variable region (ScFv), a monoclonal antibody, a humanized antibody, or a human antibody.
  • the molecule can optionally be bound to a cytotoxic moiety, bound to a therapeutic moiety, bound to a detectable moiety, or bound to an anti-tumor agent.
  • an isolated and purified human transmembrane protein is provided.
  • the protein is selected from the group consisting of: TEM 3, 9, 17, and 19 as shown in SEQ ID NO: 200, 212, 230, and 232, respectively.
  • Yet another aspect of the invention is an isolated and purified nucleic acid molecule comprising a coding sequence for a transmembrane TEM selected from the group consisting of:: TEM 3, 9, 17, and 19 as shown in SEQ ID NO: 200, 212, 230, and 232, respectively.
  • the isolated and purified nucleic acid molecule may optionally comprise a coding sequence selected from those shown in SEQ ID NO:: 199, 211, 229, and 231.
  • Still another aspect of the invention is a recombinant host cell which comprises a nucleic acid molecule.
  • the nucleic acid molecule comprises a coding sequence for a transmembrane TEM selected from the group consisting of:: TEM 3, 9, 17, and 19 as shown in SEQ ID NO: 200, 212, 230, and 232, respectively.
  • the recombinant host cell optionally comprises a coding sequence selected from those shown in SEQ ID NO: 199, 211, 229, and 231.
  • a method for inducing an immune response in a mammal.
  • a nucleic acid molecule comprising a coding sequence for a human transmembrane protein selected from the group consisting of: TEM 1, 3, 9, 13, 17, 19, 22, 30, and 44 as shown in SEQ ID NO: respectively, is administered to the mammal.
  • An immune response to the human transmembrane protein is thereby induced in the mammal.
  • the coding sequence is shown in SEQ ID NO: 196, 200, 212, 220, 230, 232, 238, 250 and 271.
  • a method of inducing an immune response in a mammal is provided.
  • a purified human transmembrane protein selected from the group consisting of: TEM 1, 3, 9, 13, 17, 19, 22, 30, and 44 as shown in SEQ ID NO: 196, 200, 212, 220, 230, 232, 238, 250 and 271, respectively, is administered to the mammal.
  • An immune response to the human transmembrane protein is thereby induced in the mammal.
  • Another aspect of the invention is a method for identification of a ligand involved in endothelial cell regulation.
  • a test compound is contacted with an isolated and purified human transmembrane protein selected from the group consisting of 1, 3, 9, 13, 17, 30, 19, and 44 as shown in SEQ ID NO: 196, 200, 212, 220, 230, 232, 250, and 271.
  • the isolated and purified human transmembrane protein is also contacted with a molecule comprising an antibody variable region which specifically binds to an extracellular domain of a TEM protein selected from the group consisting of: 1, 3, 9, 13, 17, 30, 19, and 44 as shown in SEQ ID NO: 196, 200, 212, 220, 230, 232, 250, and 271 respectively.
  • Binding of the molecule comprising an antibody variable region to the human transmembrane protein is determined.
  • a test compound which diminishes the binding of the molecule comprising an antibody variable region to the human transmembrane protein is identified as a ligand involved in endothelial cell regulation.
  • Yet another aspect of the invention is a method for identification of a ligand involved in endothelial cell regulation.
  • a test compound is contacted with a cell comprising a human transmembrane protein selected from the group consisting of 1, 3, 9, 17, and 19 as shown in SEQ ID NO: 196, 200, 212, 230, and 232.
  • the cell is also contacted with a molecule comprising an antibody variable region which specifically binds to an extracellular domain of a TEM protein selected from the group consisting of: 1, 3, 9, 17, and 19 as shown in SEQ ID NO: 196, 200, 212, 230, and 232, respectively. Binding of the molecule comprising an antibody variable region to the cell is determined.
  • a test compound which diminishes the binding of the molecule comprising an antibody variable region to the cell is identified as a ligand involved in endothelial cell regulation.
  • Yet another aspect of the invention is a method for identification of a ligand involved in endothelial cell regulation.
  • a test compound is contacted with a human transmembrane protein selected from the group consisting of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 14, 15, 16, 17, 19, 20, 21, 22, 24, 25, 27, 28, 29, 40, 31, 33, 35, 36, 37, 38, 39, 41, 42, 44, 45, and 46 as shown in SEQ ID NO: 196, 198, 200, 202, 204, 206, 208, 210, 212, 214, 216, 218, 223 & 224, 226, 228, 230, 232, 234, 236, 238, 240, 242, 244, 246, 248, 250, 252, 254, 358, 257, 259, 261, 263, 267, 269, 271, 273, and 275.
  • Binding of a test compound to the human transmembrane protein is determined.
  • a test compound which binds to the protein is identified as a ligand
  • Another embodiment of the present invention is a soluble form of a human transmembrane protein selected from the group consisting of: TEM 1, 3, 9, 17, 19, 22, 30, and 44 as shown in SEQ ID NO: 196, 200, 212, 230, 232, 238, 250, and 271 respectively.
  • the soluble forms lack transmembrane domains.
  • the soluble form may consist of an extracellular domain of the human transmembrane protein.
  • Also provided by the present invention is a method of inhibiting neoangiogenesis in a patient.
  • a soluble form of a human transmembrane protein is adminstered to the patient.
  • Neoangiogenesis in the patient is consequently inhibited.
  • the patient may bear a vascularized tumor, may have polycystic kidney disease, may have diabetic retinopathy, may have rheumatoid arthritis, or may have psoriasis, for example.
  • Another embodiment of the invention provides a method of inhibiting neoangiogenesis in a patient.
  • a soluble form of a human transmembrane protein is administered to the patient.
  • Neoangiogenesis in the patient is consequently inhibited.
  • the patient may bear a vascularized tumor, may have polycystic kidney disease, may have diabetic retinopathy, may have rheumatoid arthritis, or may have psoriasis, for example.
  • a method of identifying regions of neoangiogenesis in a patient is provided.
  • a molecule comprising an antibody variable region which specifically binds to an extracellular domain of a TEM protein selected from the group consisting of: 1, 3, 9, 13, 17, 19, 22, 30, and 44, as shown in SEQ ID NO: 196, 200, 212, 220, 230, 232, 238, 250, and 271, respectively, is administered to a patient.
  • the molecule is bound to a detectable moiety.
  • the detectable moiety is detected in the patient, thereby identifying neoangiogenesis.
  • a method for inducing an immune response to tumor endothelial cells in a patient.
  • a mouse TEM protein selected from the group consisting of: 1, 2, 3, 9, 13, 17, 19, 22, and 30 as shown in SEQ ID NO: 291, 293, 299, 295, 303, 297, 301, 305, and 307, is administered to a patient in need thereof.
  • An immune response to a human TEM protein is consequently induced.
  • Still another embodiment of the invention is a method of screening for neoangiogenesis in a patient.
  • a body fluid collected from the patient is contacted with a molecule comprising an antibody variable region which specifically binds to an extracellular domain of a TEM protein selected from the group consisting of: 1, 3, 9, 17, 19, and 44, as shown in SEQ ID NO: 196, 200, 212, 230, 232, and 271, respectively.
  • Detection of cross-reactive material in the body fluid with the molecule indicates neo-angiogenesis in the patient.
  • Still another embodiment of the invention provides a method of inhibiting neoangiogenesis in a patient.
  • a molecule comprising an antibody variable region which specifically binds to a TEM protein selected from the group consisting of: 4, 6, 7, 10, 12, 14, 20, 25, 27, 31, 36, 37, 38, 39, and 40 as shown in SEQ ID NO: 202, 206, 208, 214, 218, 223 and 224, 234, 242, 244, 252, 257, 259, 261. 263, and 265, is administered to the patient.
  • Neoangiogenesis in the patient consequently inhibited.
  • Yet another aspect of the invention is a method of screening for neoangiogenesis in a patient.
  • a body fluid collected from the patient is contacted with a molecule comprising an antibody variable region which specifically binds to a TEM protein selected from the group consisting of: 4, 6, 7, 10, 12, 14, 20, 25, 27, 31, 36, 37, 38, 39, and 40, as shown in SEQ ID NO: 202, 206, 208, 214, 218, 223 & 224, 234, 242, 244, 252, 257, 259, 261. 263, and 265, respectively.
  • Detection of cross-reactive material in the body fluid with the molecule indicates neoangiogenesis in the patient.
  • Also provided by the present invention is a method of promoting neoangiogenesis in a patient.
  • a TEM protein selected from the group consisting of: 4, 6, 7, 10, 12, 14, 20, 25, 27, 31, 36, 37, 38, 39, and 40, as shown in SEQ ID NO: 202, 206, 208, 214, 218, 223 & 224, 234, 242, 244, 252, 257, 259, 261. 263, and 265, is administered to a patient in need of neoangiogenesis. Neoangiogenesis in the patient is consequently stimulated.
  • One embodiment of the invention provides a method of promoting neoangiogenesis in a patient.
  • a nucleic acid molecule encoding a TEM protein selected from the group consisting of: 4, 6, 7, 10, 12, 14, 20, 25, 27, 31, 36, 37, 38, 39, and 40, as shown in SEQ ID NO: 201, 205, 207, 213, 217, 221 & 222, 233, 241, 243, 251, 256, 258, 260, 262, and 264, is administered to a patient in need of neoangiogenesis.
  • the TEM protein is consequently expressed and neoangiogenesis in the patient is stimulated.
  • Another embodiment of the invention provides a method of screening for neoangiogenesis in a patient.
  • a TEM protein selected from the group consisting of: 4, 6, 7, 10, 12, 14, 20, 25, 27, 31, 36, 37, 38, 39, and 40, as shown in SEQ ID NO:: 202, 206, 208, 214, 218, 223 & 224, 234, 242, 244, 252, 257, 259, 261. 263, and 265, respectively, is detected in a body fluid collected from the patient. Detection of the TEM protein indicates neoangiogenesis in the patient.
  • Another aspect of the invention is a method of screening for neoangiogenesis in a patient.
  • a nucleic acid encoding a TEM protein selected from the group consisting of: 4, 6, 7, 10, 12, 14, 20, 25, 27, 31, 36, 37, 38, 39, and 40 is detected in a body fluid collected from the patient.
  • the nucleic acid is selected from the group consisting of those shown in SEQ ID NO: 201, 205, 207, 213, 217, 221 & 222, 233, 241, 243, 251, 256, 258, 260, 262, and 264. Detection of the TEM protein indicates neoangiogenesis in the patient.
  • Yet another embodiment of the invention is an isolated and purified nucleic acid molecule which encodes a NEM protein selected from the group consisting of: 14, 22, 23, and 33 as shown in SEQ ID NO: 279, 283, 285, 286, 287, and 289.
  • the nucleic acid molecule optionally comprises a coding sequence as shown in SEQ ID NO: 278, 282, 284, and 288.
  • the nucleic acid may be maintained in a recombinant host cell.
  • the present invention also provides an isolated and purified NEM protein selected from the group consisting of: 14, 22, 23, and 33 as shown in SEQ ID NO: 279, 283, 285, 286, 287, and 289.
  • the present invention further provides an isolated molecule comprising an antibody variable region which specifically binds to a NEM protein selected from the group consisting of: 14, 22, 23, and 33, as shown in SEQ ID NO: 279, 283, 285, 286, 287, and 289.
  • An additional embodiment of the present invention is a method of inhibiting neoangiogenesis.
  • An effective amount of a NEM protein selected from the group consisting of: 14, 22, 23, and 33 as shown in SEQ ID NO: 279, 283, 285, 286, 287, and 289 is administered to a subject in need thereof. Neoangiogenesis is thereby inhibited.
  • a still further embodiment of the invention is a method to identify candidate drugs for treating tumors.
  • Cells which express one or more TEM genes selected from the group consisting of: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 14, 15, 16, 17, 19, 20, 21, 22, 24, 25, 27, 28, 29, 40, 31, 33, 35, 36, 37, 38, 39, 41, 42, 44, 45, and 46 as shown in SEQ ID NO:: 195, 197, 199, 201, 203, 205, 207, 209, 211, 213, 215, 217, 221 & 222, 225, 227, 229, 231, 233, 235, 237, 239, 241, 243, 245, 247, 249, 251, 253, 255, 256, 258, 260, 262, 266, 268, 270, 272, and 274, respectively, are contacted with a test compound.
  • Expression of said one or more TEM genes is determined by hybridization of mRNA of said cells to a nucleic acid probe which is complementary to said mRNA.
  • a test compound is identified as a candidate drug for treating tumors if it decreases expression of said one or more TEM genes.
  • the cells are endothelial cells.
  • the cells are recombinant host cells which are transfected with an expression construct which encodes said one or more TEMs. Test compounds which increase expression can be identified as candidates for promoting wound healing.
  • Yet another embodiment of the invention is a method to identify candidate drugs for treating tumors.
  • Cells which express one or more TEM proteins selected from the group consisting of: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 14, 15, 16, 17, 19, 20, 21, 22, 24, 25, 27, 28, 29, 40, 31, 33, 35, 36, 37, 38, 39, 41, 42, 44, 45, and 46 as shown in SEQ ID NO: 196, 198, 200, 202, 204, 206, 208, 210, 212, 214, 216, 218, 223 & 224, 226, 228, 230, 232, 234, 236, 238, 240, 242, 244, 246, 248, 250, 252, 254, 358, 257, 259, 261, 263, 267, 269, 271, 273, and 275, respectively, are contacted with a test compound.
  • the amount of said one or more TEM proteins in said cells is determined.
  • a test compound is identified as a candidate drug for treating tumors if it decreases the amount of one or more TEM proteins in said cells.
  • the cells are endothelial cells.
  • the cells are recombinant host cells which are transfected with an expression construct which encodes said one or more TEMs.
  • a test compound which increases the amount of one or more TEM proteins in said cells is identified as a candidate drug for treating wound healing.
  • a method is provided to identify candidate drugs for treating tumors.
  • Cells which express one or more TEM proteins selected from the group consisting of: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 14, 15, 16, 17, 19, 20, 21, 22, 24, 25, 27, 28, 29, 40, 31, 33, 35, 36, 37, 38, 39, 41, 42, 44, 45, and 46 as shown in SEQ ID NO: 196, 198, 200, 202, 204, 206, 208, 210, 212, 214, 216, 218, 223 & 224, 226, 228, 230, 232, 234, 236, 238, 240, 242, 244, 246, 248, 250, 252, 254, 358, 257, 259, 261, 263, 267, 269, 271, 273, and 275, respectively, are contacted with a test compound.
  • Activity of said one or more TEM proteins in said cells is determined.
  • a test compound is identified as a candidate drug for treating tumors if it decreases the activity of one more TEM proteins in said cells.
  • the cells are endothelial cells.
  • the cells are recombinant host cells which are transfected with an expression construct which encodes said one or more TEMs.
  • the cells are endothelial cells. If a test compound increases the activity of one more TEM proteins in said cells it can be identified as a candidate drug for treating wound healing.
  • An additional aspect of the invention is a method to identify candidate drugs for treating patients bearing tumors.
  • a test compound is contacted with recombinant host cells which are transfected with an expression construct which encodes one or more TEM proteins selected from the group consisting of 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 14, 15, 16, 17, 19, 20, 21, 22, 24, 25, 27, 28, 29, 40, 31, 33, 35, 36, 37, 38, 39, 41, 42, 44, 45, and 46 as shown in SEQ ID NO: 198, 200, 202, 204, 206, 208, 210, 212, 214, 216, 218, 223 & 224, 226, 228, 230, 232, 234, 236, 238, 240, 242, 244, 246, 248, 250, 252, 254, 358, 257, 259, 261, 263, 267, 269, 271, 273, and 275, respectively.
  • a test compound which inhibits proliferation of said cells is identified as a candidate drug for treating patients bearing tumors.
  • a test compound which stimulates proliferation of said cells is identified as a candidate drug for promoting neoangiogenesis, such as for use in wound healing.
  • Another embodiment of the invention provides a method to identify candidate drugs for treating tumors.
  • Cells which express one or more NEM genes selected from the group consisting of: 14, 22, 23, and 33 as shown in SEQ ID NO: 278, 282, 284, and 288, respectively, are contacted with a test compound.
  • Expression of said one or more NEM genes is determined by hybridization of mRNA of said cells to a nucleic acid probe which is complementary to said mRNA.
  • a test compound is identified as a candidate drug for treating tumors if it increases expression of said one or more NEM genes.
  • the cells are endothelial cells.
  • the cells are recombinant host cells which are transfected with an expression construct which encodes said one or more NEMs.
  • a method is provided to identify candidate drugs for treating tumors.
  • Cells which express one or more NEM proteins selected from the group consisting of: 14, 22, 23, and 33 as shown in SEQ ID NO: 279, 283, 285, 286, 287, and 289, are contacted with a test compound.
  • the amount of said one or more NEM proteins in said cells is determined.
  • a test compound is identified as a candidate drug for treating tumors if it increases the amount of one more NEM proteins in said cells.
  • the cells are endothelial cells.
  • the cells are recombinant host cells which are transfected with an expression construct which encodes said one or more NEMs.
  • An additional aspect of the invention is a method to identify candidate drugs for treating tumors.
  • Cells which express one or more NEM proteins selected from the group consisting of: 14, 22, 23, and 33 as shown in SEQ ID NO: 279, 283, 285, 286, 287, and 289, are contacted with a test compound.
  • Activity of said one or more NEM proteins in said cells is determined.
  • a test compound is identified as a candidate drug for treating tumors if it increases the activity of said one or more NEM proteins in said cells.
  • the cells are endothelial cells.
  • the cells are recombinant host cells which are transfected with an expression construct which encodes said one or more NEMs.
  • Still another embodiment of the invention provides a method to identify candidate drugs for treating patients bearing tumors.
  • a test compound is contacted with recombinant host cells which are transfected with an expression construct which encodes one or more NEM proteins selected from the group consisting of 14, 22, 23, and 33 as shown in SEQ ID NO: 279, 283, 285, 286, 287, and 289. Proliferation of said cells is determined.
  • a test compound which stimulates proliferation of said cells is identified as a candidate drug for treating patients bearing tumors.
  • Another aspect of the invention is a method for identifying endothelial cells.
  • Still another aspect of the invention is a method for identifying endothelial cells.
  • One or more nucleic acid hybridization probes which are complementary to a TEM or NEM gene nucleic acid sequence selected from the group consisting of TEM: 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 14, 15, 16, 17, 19, 20, 21, 22, 24, 25, 27, 28, 29, 30, 31, 33, 35, 36, 37, 38, 39, 41, 42, 44, 45, and 46 as shown in SEQ ID NO: 198, 200, 202, 204, 206, 208, 210, 212, 214, 216, 218, 223 & 224, 226, 228, 230, 232, 234, 236, 238, 240, 242, 244, 246, 248, 250, 252, 254, 358, 257, 259, 261, 263, 267, 269, 271, 273, and 275 and NEM 14, 22, 23, and 33 as shown in SEQ ID NO: 279, 283, 285, 286, 287, and 289, is contacted with nucleic acids of
  • Yet another embodiment of the invention is a method of inhibiting neoangiogenesis.
  • An effective amount of an isolated molecule comprising an antibody variable region which specifically binds to an extracellular domain of a mouse TEM protein selected from the group consisting of: 1, 2, 3, 9, 17, and 19, as shown in SEQ ID NO: 291, 293, 299, 295, 297, and 301, respectively, is administered to a subject in need thereof.
  • Neoangiogenesis is thereby inhibited.
  • the subject may be a mouse, may bear a vascularized tumor, may have polycystic kidney disease, may have diabetic retinopathy may have rheumatoid arthritis, or may have psoriasis, for example.
  • FIG. 1A-1B vWF expression in colorectal cancers.
  • vWF red stain
  • FIG. 1.A vessels were often surrounded by a perivascular cuff of viable cells (red arrows), with a ring of necrotic cells evident at the periphery (black arrows).
  • FIG. 1.B the expression of vWF (red) was clearly localized to the vessels. Sections were counterstained with methyl green.
  • FIG. 2A-2D Purification of Endothelial Cells (ECs) from human normal and malignant tissue.
  • FIG. 2A Vessels (red) of frozen sections were stained by immunofluorescence with the P1H12 monoclonal antibody (Chemicon, Temecula, Calif.) and detected using a biotinylated goat anti-mouse IgG secondary antibody followed by rhodamine-linked strepavidin. The region stained is from within the lamina intestinal of normal colonic mucosa. Note that the larger vessels (arrowheads) and capillaries (arrows) are positive, and staining of hematopoietic cells was undetectable.
  • E-cadherin positive epithelial cells (green) at the edge of the crypt were simultaneously visualized using a rabbit polyclonal antibody (Santa Cruz, Santa Cruz, Calif.), followed by a goat anti-rabbit IgG secondary antibody labelled with alexa (Molecular Probes, Eugene, Oreg.). Sections were imaged at 60 ⁇ magnification using confocal microscopy.
  • FIG. 2.B To isolate pure populations from collagenase dispersed tissues, the epithelial and hematopoietic cell fractions were sequentially removed by negative selection with magnetic beads. The remaining cells were stained with P1H12 and ECs were isolated by positive selection with magnetic beads.
  • RT-PCR analysis used to assess the purity of the EC preparations.
  • Semiquantitative PCR analysis was performed on cDNA generated either directly from colorectal cancer tissue (unfractionated tumor) or from purified ECs isolated from normal colonic mucosa (normal EC fraction) or colorectal cancer (tumor EC fraction).
  • PCR amplification of the epithelial specific marker cytokeratin 20 (CK20) demonstrated its expression was limited to the unfractionated tumor.
  • Two endothelial specific markers, vWF and VE-cadherin (VE-Cad) showed robust amplification only in the endothelial fractions, validating the purity and enrichment protocol shown in ( FIG. 2.B ).
  • the ubiquitous housekeeping enzyme GAPDH was observed in all samples. No signal was detected in the no-template (NT) control. cDNA templates were diluted 1:10, 1:100, 1:1000, 1:4000, and 1:40,000 as indicated by the declining wedge.
  • FIG. 2.D The relative expression level of select genes was determined by measuring the tag abundance from several SAGE libraries combined into four groups. The first was composed of ⁇ 193,000 tags from the two in vivo-derived EC preparations (Endothelial Cell Fraction) while the second contained a single library of ⁇ 57,000 tags containing macrophages and other leukocytes derived from the negative selection (Hematopoietic Fraction).
  • the fourth library contained ⁇ 401,000 tags from cultured HUVEC and HMVEC (Endothelial Cells in Culture), and the fourth consisted of 748,000 tags from 6 colon cancer cell lines in culture (Epithelial Cells). After normalization, the library with the highest tag number for each marker was given a value of 100%, and the corresponding relative expression levels of the remaining 3 libraries was plotted on the ordinate. Note the high level of CD31 present on hematopoietic cells, the likely cause of the impurity of the initial endothelial selection, compared with the selectivity of P1H12.
  • FIG. 3A-3E Expression of Pan-Endothelial Markers (PEMs) is limited to ECs. The endothelial origin of PEMs identified by SAGE was confirmed using a highly sensitive in situ hybridization assay. Localization of novel PEMs to the ECs was demonstrated by examining two representative PEMs, PEM3 ( FIG. 3A ) and PEM6 ( FIG. 3B ) in lung cancer and colon cancer, respectively. Hevin expression was readily detected in the ECs of a colon tumor ( FIG. 3C ) despite its low level of expression in cultured ECs. Expression of VEGFR2 was readily detectable in the ECs of both normal ( FIG. 3D ) and malignant colon tissue ( FIG. 3E ).
  • PEMs Pan-Endothelial Markers
  • FIG. 4A-4J Expression of Tumor Endothelial Markers (TEMs).
  • FIG. 4A RT-PCR analysis confirmed the tumor specific expression of selected novel TEMs.
  • Semiquantitative PCR analysis was performed on cDNA generated either from purified epithelial cells as a negative control (Control) or from purified ECs isolated from normal colonic mucosa (Normal ECs) or colorectal cancer (Tumor ECs) from two different patients.
  • Two endothelial specific markers, vWF and PEM6 showed robust amplification only in the endothelial fractions whereas the ubiquitous housekeeping enzyme GAPDH was observed in all samples.
  • TEM1 BSC-TEM1
  • BSC-TEM7 TEM 17
  • BSC-TEM9 TEM22
  • FIG. 4 B- 4 J The endothelial origin of TEMs identified by SAGE was confirmed using in situ hybridization as in FIG. 3 .
  • TEM17 (BSC-TEM7) was demonstrated to be highly specific to the ECs in colorectal cancers; sections were imaged in the absence of a counterstain to show the complete lack of detectable expression in the non-endothelial cells of the tumor.
  • Expression of TEM17 (BSC-TEM7) in ECs was demonstrated in a metastatic liver lesion from a primary colorectal cancer ( FIG. 4 D), a lung ( FIG. 4 E), breast ( FIG. 4 F), pancreatic ( FIG. 4 G) and brain cancer ( FIG. 4 H), as well as in a sarcoma ( FIG. 4 I).
  • TEM 17 (BSC-TEM7) was also localized to vessels during normal physiological angiogenesis of the corpus luteum ( FIG. 4 J).
  • the SAGE Tag BSC-TEM3 has been found to hybridize to an alternatively spliced form of the transcript encoding BSC-TEM7.
  • the proteins encoded by the two transcripts are the same; therefore they are cumulatively called TEM7.
  • a highly related sequence was found via homology searches, BSC-TEM7R. This paralog sequence is now called TEM3. See Table 2, which follows, showing tumor endothelial markers by order of prevalence (except for TEM 3). Column 1 indicates the prevalence number. Column 2 indicates the original nomenclature. Column 3 indicates the short tags. Column 4 indicates the long tags. Column 5 indicates the accession number in GenBank. Column 6 indicates the sequence identifiers for the short tag, the long tag, the full nucleic acid, and the protein. Column 7 provides a functional description, which is expanded below in the text.
  • TEM1 BSC-TEM1 GGGGCTGCCC GGGGCTGCCCAGCT NM020404 SEQ ID NO: tumor endothelial marker 1 precursor A GA 94, 309, 195, 196 TEM 2 BSC-TEM2 GATCTCCGTG SEQ ID NO: sapiens tumor endothelial marker 2 T 95, 197, (BSC-TEM2) mRNA/mouse Ras, dexa- 198 methasone-induced 1 (RASD1), mRNA TEM 3 BSC-TEM7R SEQ ID NO: human ortholog of mouse paralog of 199, 200 mouse TEM-7 TEM 4 CTTTCTTTGA CTTTCTTTGAGTTT AB034203 SEQ ID NO: Homo sapiens dickkopf-3 (DKK-3) mRNA, G TAA 97, 311, 201, 202 TEM 5 BSC-TEM4 TATTAACTCT TATTAACTCTCTTT SEQ ID NO: Tumor endothelial marker 4 C GGA 98, 312, 203
  • the genes expressed differentially in tumor endothelium are also expressed during other angiogenic processes such as corpus luteum formation and wound healing. It is therefore more appropriate to regard the formation of new vessels in tumors as “neoangiogenesis” rather than “tumor angiogenesis” per se. This distinction is important from a variety of perspectives, and is consistent with the idea that tumors recruit vasculature using much of, or basically the same signals elaborated during other physiologic or pathological processes. That tumors represent “unhealed wounds” is one of the oldest ideas in cancer biology.
  • TEMs Tumor Endothelial Markers
  • this may be due to the fact that the EST databases are heavily biased toward certain tissues, but moreover, may be due to the fact that even in highly vascularized tissues endothelial cells are still a relatively small proportion of the population.
  • the sensitivity of the SAGE method is a particularly appropriate tool.
  • TEM proteins have been identified which contain transmembrane regions. These include TEM 1, TEM 3, TEM 9, TEM 13, TEM 17, TEM 19, TEM 22, TEM 30, and TEM 44. TEM proteins have been identified which are secreted proteins, including TEM 4, TEM 6, TEM 7, TEM 10, TEM 12, TEM 14, TEM 20, TEM 25, TEM 27, TEM 31, TEM 36, TEM 37, TEM 38, and TEM 39.
  • HeyL (TEM 8) is a transcription factor which may be involved in regulating TEMs as one or more groups. The protein corresponding to the tag for TEM44 was found in the public databases, but no biological function has yet been ascribed to it.
  • TEM 1 has been named endosialin in the literature. It has a signal sequence at amino acids 1-17 and a transmembrare domain at amino acids 686-708. Thus it is a cell surface protein. Its extracellular domain is at resiudes 1-685. Endosialin may be involved in endocytosis.
  • the mouse ortholog is predicted to have a signal peptide at residues 1-21.
  • TEM 2 is a dexamethasone induced, ras related protein homolog of 266 amino acids. It has neither a signal sequence nor a transmembrane domain. Thus it is neither a cell surface nor a secreted protein. TEM 2 plays a role in signal transduction. It regulates alterations in cell morphology, proliferation, and cell-extracellular matrix interactions.
  • TEM 3 (originally termed TEM 7R) has both a signal sequence (at residues 1-24 or 1-30) and a transmembrane domain (at residues 456-477). Thus it is a cell surface protein. The portion of the protein which is extracellular is at amino acids 1-455. TEM 3 has domains with homology to integrins, plexin, and adhesion molecules. TEM 3 may regulate GTPases that control signal transduction pathways linking plasma membrane receptors to the actin cytoskeleton. In the mouse ortholog, the signal peptide is predicted to be residues 1-30.
  • TEM 4 is also known as DKK-3. It has a signal sequence (residues1-16), suggesting that is a secreted protein. TEM 4 regulates wnt signaling, and it may be involved in vasculogenesis and wnt-dependent signaling for endothelial growth. TEM 4 is an inhibitor of Wnt oncogene and such inhibition can be determined by assay. Tsuji et al., Biochem. Biophys. Res. Comm. 268:20-4, 2000.
  • TEM 5 appears to be neither secreted nor a cell surface protein. TEM 5 appears to be a component of a G protein—GTPase signaling pathway.
  • TEM 6 is also known as stromelysin-3/Matrix metalloproteinase 11 (MMP-11). It has a signal sequence at residues 1-31, but no transmembrane domain. It has an alternative signal peptide splice site at residues 108-109. Thus it appears to be a secreted protein.
  • TEM 6 belongs to the zinc metalloprotease family, also known as the matrixin subfamily. TEM 6 is expressed in most invasive carcinomas.
  • Alpha 1-protease inhibitor is a natural substrate of MMP 11.
  • TEM 6 degrades extracellular matrix proteins such as collagen and is involved in extracellular matrix remodeling and cell migration. Stromelysin can be assayed using a casein-resorufin substrate, for example. See Tortorella and Amer, Inflammation Research 46 Supp. 2:S 122-3, 1997.
  • TEM 7 is a protein of many names, also being known as matrix metalloproeinase 2, gelatinase A, and 72 KD type IV collagenase.
  • TEM 7 has a signal sequence at residues 1-26 and is a secreted protein.
  • TEM 7 belongs to the matrixin subfamily (zinc metalloproteinases).
  • TEM 7 cleaves gelatin type I, collagen type I, IV, V VII and X.
  • TEM 7 associates with integrin on the surface of endothelial cells and promotes vascular invasion.
  • TEM 7 is involved in tissue remodeling.
  • TEM 7 can be assayed using zymography or quenched fluorescent substrate hydrolysis, for example.
  • a fluorogenic matrix metalloproteinase substrate assay can also be used which employs methoxycoumarin containing septapeptide analog of the alpha2(I) collagen cleavage site. See Bhide et al., J. Periodontology 71:690-700, 2000.
  • TEM 8 is HEYL protein. It has neither a signal sequence nor a transmembrane domain. It is related to the hairy/Enhancer of split genes. TEM 8 is likely a nuclear protein, having a role as a transcription factor. TEM 8 belongs to a new class of Notch signal transducers and plays a key role in various developmental processes, such as vascular development, somatogenesis and neurogenesis. SNP's at residues 615 and 2201 have Cytosine bases. Notch 3 mutations underlie the CADASIL vascular disorder. See Mech Dev 2000 November; 98 (1-2):175
  • TEM 9 is a G-protein coupled receptor homolog, having both a signal sequence at residues 1-26 and 7 transmembrane domains. Thus it is a cell surface protein. Its extracellular region resides in amino acids 1-769. Its transmembrane domains are at residues 817-829 (TM2 and TM3), residues 899-929 (TM4 and TM5), and residues 1034-1040 (TM6 and TM7).
  • TEM 9 acts as a G-protein coupled receptor with extracellular domains characteristic of cell adhesion proteins. One of its splice variants may function as a soluble receptor. TEM 9 may regulate cell polarity and cell migration. It may be involved in exocytosis based on latrophilin function. The mouse ortholog has a predicted signal peptide at residues 1-29.
  • TEM 10 is collagen type I, alpha2 (COL1A2), which has a signal sequence at residues 1-22. It is an extracellular matrix (ECM) protein which is secreted subsequent to synthesis. TEM 10 interacts with a number of proteins including other ECM proteins, certain growth factors, and matrix metalloproteases. TEM 10 is required for the induction of endothelial tube formation and is involved in tissue remodeling. A variant at nucleotide 3233 which substitutes an A, is associated with osteogenesis imperfecta type IV. A variant at nucleotide 4321 substituting an A retains a wild type phenotype. Nucleotide 715 is a site of a polymorphism.
  • Nucleotides 695-748 are deleted in Ehlers-Danos syndrome. Other mutations are associated with idiopathic osteoporosis, and atypical Marfan syndrome. Variants are known at nucleotides 226(T,C), 314(A,C), 385(T,C), 868 (G,A), 907(C,T), 965(A,G), 970(T,A), 1784 (G,C), 2017(T,G), 2172(C,A), 2284(T,C), 2308(T,C), 2323(T,G), 2344(T,G), 2604(G,A), 2974(A,T), 2903(A,G), 2995(C,T), 3274(C,T), 3581(A,C), 3991(A,C), 4201(G,T), 4434(C,T), 4551(A,C), 4606(C,A), 4947(T,C), 4978(C,T), 4982(G,T), 5051
  • PolyA sites are located at nucleotides 4450, 4550, 4885, and 5082.
  • PolyA signals are located at 4420-4424, 4515-4520, 4529-4534, 4866-4871, 5032-5037, 5053-5058.
  • TEM 10, 20, and 40 derive from the same gene but are different isoforms having different lengths.
  • TEM 11 is Nidogen/Entactin. It is a secreted protein which has a signal sequence at residues 1-28.
  • TEM 11 is an extracellular matrix protein which is a component of a basement membrane. TEM 11 binds to laminin and collagen IV and other extracellular matrix proteins. TEM 11 regulates capillary formation and is involved in tissue remodelling. Variations have been observed at nucleotides 4265(T,C), 4267(G,C,T), and 4738(T,G). Nidogen can be assayed by its effect on the morphology of astrocytes. See Grimpe et al., GLIA 28:138-49, 1999.
  • TEM 12 is the alpha 3 chain of collagen type VI. It has a signal sequence at residues 1-25. A secreted protein, TEM 12 is an extracellular matrix protein. TEM 12 has a splice variant. TEM 12 is a major constituent of vascular subendothelium and is involved in tissue remodeling. It regulates platelet activation and aggregation. Alternatively spliced domains are located at nucleotides 347-964, 965-1567, 2153-3752, and 4541-5041.
  • TEM 13 is also known as Thy-1 glycoprotein. It has both a signal sequence (at residues 1-19) and a transmembrane domain (at residues 143-159). Residues 131-161 are removed in a matured form of the protein. The extracellular region of the protein is residues 1-142 or residues 1-130. TEM 13 has a glycosyl phosphatidylinositol (GPI) anchor at residue 130 anchoring it to the membrane. TEM 13 is detectable in its soluble form in human serum. TEM 13 is reported to be a marker for activated endothelial cells (a marker of adult but not embryonic angiogenesis).
  • GPI glycosyl phosphatidylinositol
  • TEM 13 on vascular endothelial cells may function as a possible vascular permeability modulator.
  • Antibody to Thy-1 is a mitogenic signal for the CD4+CD45+ and CD8+CD45+ cells, but fails to induce proliferation in the CD45 ⁇ T cells. Pingel et al., International Immunology 6:169-78, 1994. Thy-1 can be assayed as an inhibitor of such signal.
  • TEM 14 is also known as cystatin S. It is a secreted protein with a signal sequence at residues 1-20 and an extracellular region at residues 1-141. It is a cysteine protease inhibitor. TEM 14 may regulate cysteine protease function involved in angiogenesis and tissue remodeling. TEM14 is an inhibitor of the activity of papain and such inhibition can be assayed. Hiltke et al., J. Dental Research 78:1401-9, 1999.
  • TEM 15 is collagen type III, alpha 1 (COL3A1). It has a signal sequence (residues 1-23) and is secreted. Type III collagen binds to von Willebrand factor. It is involved in cell-cell adhesion, proliferation, and migration activities. Variants at nucloetides 2104(C,A), 2194(G,A), 2346(C,T), 2740(C,T), 3157(T), 3468(G), 3652(T), 3666(C), 3693(C), 3755(G), 3756(T), 3824(C), 4546(A, G), 4661(G), 4591(C,T), 4665(C), 5292(C), 5293(C), and 5451 (A) have been observed.
  • TEM 16 is a tensin homolog which is apparently an intracellular protein. It may have splice variants or isoforms. One form with 1704 amino acids has a region at the N-terminal domain which is similar to a tumor suppressor protein, phosphatase and tensin homolog (PTEN).
  • PTEN phosphatase and tensin homolog
  • Tensin is a focal adhesion molecule that binds to actins and phosphorylated proteins. It is involved in cell migration linking signal tranduction pathways to the cytoskeleton. PTEN regulates tumor induced angiogenesis.
  • TEM 17 (BSC-TEM 7) has a signal sequence which includes residues 1-18 and a transmembrane domain at residues 427-445. It is a cell surface marker with an extracellular region comprising residues 1-426. It has homologs in both mouse and C. elegans . Residues 137-244 share weak homology with nidogen; residues 280-344 share homology to PSI domains found in plexin, semaphorins and integrin beta subunits. Variants have been observed at nucleotides 1893(A,G), 1950(C,G), 2042(A,G), and 2220(G,A). In mouse TEM 17 the signal sequence includes residues 1-19.
  • TEM 19 was originally reported to be tumor endothelial marker 8, i.e., BSC-TEM 8. It has a signal sequence at residues 1-27 and a transmembrane domain at residues 322-343. It is a cell surface protein having an extracellular region at residues 1-321.
  • TEM 19 has a von Willebrand Factor (vWF) A domain at residues 44-216; a domain at residues 34-253 which is found in leukointegrin alpha D chain; and a domain at residues 408-560 found in PRAM-1 or adaptor molecule-1 of the vinculin family.
  • TEM 19's function is adhesion related. von Willebrand Factor domains are typically involved in a variety of functions including vascular processes.
  • TEM 19 may play a role in the migration of vascular endothelial cells.
  • the mouse ortholog has a predicted signal peptide at residues 1-27.
  • TEM 20 is collagen type I, alpha 2 (COL1A2). It has a signal sequence at residues 1-22 and is a secreted extracellular matrix protein. TEM 20 induces endothelial tube formation in vitro and is involved in tissue remodeling. Variants have been observed at nucleotides 226(T,C), 314(A,C), 385(T,C), 868 (G,A), 907(C,T), 965(A,G), 970(T,A), 1784(G,C), 2017(T,G), 2172(C,A), 2284(T,C), 2308(T,C), 2323(T,G), 2344(T,G), 2604(G,A), 2794(A,T), 2903(A,G), 2995(C,T), 3274(C,T), 3581(A,C), 3991(A,C), 4201(G,T), 4434(C,T), 4551(A,C), 4606(C,A), 4895-4901 (--
  • TEM 21 is a Formin-like protein homolog which is an intracellular protein.
  • Formin related proteins interact with Rho family small GTPases, profilin, and other actin associated proteins.
  • Formin-binding proteins bind to FH1 domains with their WW domains.
  • TEM 21 has a proline rich FH1 domain at residues 221-449.
  • Formin related proteins play crucial roles in morphogenesis, cell polarity, cytokinesis and reorganization of the actin cytoskeleton. They may also regulate apoptosis, cell adhesion and migration.
  • TEM 22 is an endocytic receptor in the macrophage mannose receptor family. It has both a signal sequence at residues 1-30 and a transmembrane domain at residues 1415-1435, and resides on the cell surface. Its extracellular domain is amino acids 1-1414.
  • TEM 22 may be present as a soluble (secreted) form and act as an inhibitor. It may bind secreted phopholipase A2 (sPLA2) and mediate biological responses elicited by sPLA2.
  • TEM 22 may have endocytic properties for sPLA2 and mediate endocytosis for endothelial related proteins. It may promote cell adhesion and be involved in cell-cell communication. Variations have been observed at nucleotide 5389 (A, G). TEM 22 mediates uptake of micro-organisms and host-derived glycoproteins. Groger et al., J. Immunology 165:5428-34, 2000.
  • TEM 24 is tensin, an intracellular protein. It is a focal adhesion molecule that binds to actin filaments and interacts with phosphotyrosine containing proteins. It may mediate kinase signaling activities and regulate cellular transformation. Variations have been observed at nucleotides 2502 (A, G), 2622(A, G), 6027(A, G).
  • TEM24 binds to actin filaments and interacts with phosphotyrosine-containing proteins. Chen et al., Biochem. J. 351 Pt 2:403-11, 2000. TEM24 also binds to phosphoinositide3-kinase. Auger et al., J. Bio. Chem. 271:23452-7, 1996 TEM 24 also binds to nuclear protein p130. Lo et al., Bioassays 16:817-23, 1994.
  • TEM 25 is Bone morphogenic protein 1 (BMP-1) which has a signal sequence at residues 1-22. It is a secreted protein. There are at least 6 isoforms of BMP-1 as well as splice variants which add carboxy terminal CUB domains and an additional EGF domain.
  • TEM 25 is a metalloprotease enzyme. It cleaves the C-terminal propeptide of collagen type I, II and III and laminin 5 gamma 2, proteins that are important for vascular processes. It is involved in cartilage formation. Variations have been observed at nucleotides 3106(C,T), 3248(G,A), 3369(G,A).
  • TEM 25 cleave probiglycan at a single site, removing the propeptide and producing a biglycan molecule with an NH(2) terminus identical to that of the mature form found in tissues.
  • Laminin alpha 3 and gamma2 short chains are substrates of TEM 25.
  • TEM 27 is known as Slit homolog 3, a secreted protein with a signal sequence at residues 1-27.
  • TEM 27 is a secreted guide protein involved in migration, repulsion and patterning. It interacts with “round about” receptors (Robo receptors).
  • TEM 27 may interact with extracellular matrix (ECM) proteins and is involved in cell adhesion. Variations have been observed at nucleotides 4772 (C,T)
  • TEM 28 is similar to mouse nadrin (neuron specific GTPase activating protein).
  • TEM 28 is an intracellular protein with a RhoGAP domain.
  • RhoGAP domain activates RhoA, Rac1, and Cdc42 GTPases. It is involved in the reorganization of actin filaments and enhancing exocytosis. It may also be involved in cell signalling. Variations have been observed at nucleotide 3969 (A,C),
  • TEM 29 is protein tyrosine phosphatase type IVA, member 3, isoform 1, an intracellular protein. It has alternate splice variants.
  • TEM 29 belongs to a small class of prenylated protein tyrosine phosphatases (PTPs). It may be membrane associated by prenylation. PTPs are cell signaling molecules and play regulatory roles in a variety of cellular processes and promote cell proliferation. PTP PRL-3 regulates angiotensin-II induced signaling events.
  • TEM 30 is integrin alpha 1, a cell surface protein having both a signal sequence (residues 1-28) and a transmembrane domain (residues 1142-1164). Its extracellular region includes amino acids 1-1141.
  • TEM 30 is a receptor for laminin and collagen. It mediates a variety of adhesive interactions. TEM 30 is abundantly expressed on microvascular endothelial cells. It stimulates endothelial cell proliferation and vascularization. TEM 30 may regulate angiostatin production. Variations have been observed at nucleotide 418 (C,T). TEM 30 activates the Ras/Shc/mitogen-activated protein kinase pathway promoting fibroblast cell proliferation. It also acts to inhibit collagen and metalloproteinase synthesis. Pozzi et al., Proc. Nat. Acad. Sci. USA 97:2202-7, 2000,
  • TEM 31 is Collagen IV alpha 1 (COL4A1) a secreted protein with a at residues 1-27.
  • TEM 31 is a component of the basement membrane. It binds to alpha3 beta lintegrin and promotes integrin mediated cell adhesion. Non-collagenous domains of type IV subunits are involved in tumoral angiogenesis.
  • TEM 31 is involved in tissue remodeling. Variations have been observed at nucleotide 4470 (C,T)
  • TEM 33 is methylmalonyl Co-A Mutase a protein which is localized in the mitochondrial matrix. It degrades several amino acids, odd-numbered-acid fatty acids, and cholesterol to the tricarboylic acid cycle. A defect in TEM 33 causes a fatal disorder in organic acid metabolism termed methylmalonic acidurea. Variations have been observed at nucleotides 1531(G,A), 1671(G,A), 2028(T,C), 2087(G,A), 2359(A,G), 2437(C,A), 2643(G,C), 2702(G,C). TEM 33 converts L-methylmalonyl CoA to succinyl CoA. This reaction can be assayed as is known in the art. See, e.g., Clin. Chem. 41(8 Pt I): 1164-70, 1995.
  • TEM 36 is collagen type XII, alpha1 (COL12A1), an extracellular matrix protein having a signal sequence at residues 1-23 or 24.
  • TEM 36 has von Willebrand Factor (vWF) type A domains, Fibronectin type III domains, and thrombospondin N-terminal like domain.
  • vWF von Willebrand Factor
  • TEM 36 is expressed in response to stress environment.
  • TEM 36 may organize extracellular matrix architecture and be involved in matrix remodeling. There are two isoforms of the protein, a long form and a short form. The short form is missing amino acids 25-1188, and therefore nucleotides 73 to 3564. Both forms share the signal sequence and are therefore both secreted.
  • TEM 37 is lumican, an extracellular matrix sulfated proteoglycan having a signal sequence at residues 1-18. Lumican interacts with proteins that are involved in matrix assembly such as collagen type I and type VI; it is involved in cell proliferation and tissue morphogenesis. Lumican plays an important role in the regulation of collagen fiber assembly. Variations have been observed at nucleotides 1021(G,T), 1035(A,G), 1209(A,G), 1259(A,C), 1418(C,A), 1519(T,A).
  • TEM 37 is a binding partner of TGF- ⁇ . See FASEB J. 15:559-61, 2000.
  • One assay that can be used to determine TEM 37 activity is a collagen fibril formation/sedimentation assay. Svensson et al., FEBS Letters 470:178-82, 2000.
  • TEM 38 is collagen type I, alpha 1 (COL1A1), an extracellular matrix protein having a signal sequence at residues 1-22.
  • Type I collagen promotes endothelial cell migration and vascularization and induces tube formation and is involved in tissue remodelling.
  • Telopeptide derivative is used as a marker for malignancy and invasion for certain cancer types.
  • TEM 39 is transforming growth factor ⁇ -3 (TGF-beta3). It has a signal sequence at residues 1-23. It is a secreted protein. TEM 39 regulates cell growth and differentiation. TGF-beta isoforms play a major role in vascular repair processes and remodeling. Variations have been observed at nucleotide 2020(G,T).
  • TEM 41 is similar to Olfactomedin like protein. It appears to be an intracellular protein, having no obvious predicted signal sequence. Olfactomedin is the major glycoprotein of the extracellular mucous matrix of olfactory neuroepithelium. TEM 41 shares homology with latrophilin (extracellular regions) which has cell-adhesive type domains. TEM 41 may be involved in adhesive function.
  • TEM 42 is MSTP032 protein, a cell surface protein having a transmembrane domain at residues 42-61. Its function is unknown and it shares little homology with other proteins. Variations have been observed at nucleotides 418(A,T), 724(C,A).
  • TEM 44 is a hypothetical protein FLJ1190 (NM — 018354) which has two predicted transmembrane domains at residues 121-143 and 176-197. Residues 144-175 may form an extracellular region. TEM 44's function is not known and shares no homology to other known proteins.
  • TEM 45 is tropomyosin 1 (alpha), a protein which is intracellular. It forms dimers with a beta subunit. It influences actin function. TEM 45 may be involved in endothelial cell cytoskeletal rearrangement. Variations have been observed at nucleotides 509(A,C), 621(A,C), 635(T,G), 642(C,G), 1059(G,T).
  • TEM 46 is peanut-like 1 protein/septin 5, which belongs to the septin family. Proteins in the septin family bind to GTP and phosphatidylinositol 4,5-bisphosphate. They are involved in the signal tranduction cascades controlling cytokinesis and cell division.
  • NEM 4 is a member of the small inducible cytokine subfamily A (cys-cys), member 14 (SCYA14). NEM4 is a secreted protein characterized by two adjacent cysteine residues. One isoform lacks internal 16 amino acids compared to isoform 2.
  • NEM 22 shares homology with guanylate kinase-interacting protein 1Maguin-1. It is a membrane associated protein.
  • NEM 23 is human signaling lymphocytic activation molecule (SLAM). It has a signal sequence at residues 1-20. The extracellular domain may reside at residues 21-237. There is a secreted isoform of the protein.
  • SLAM signaling lymphocytic activation molecule
  • NEM33 is netrin 4. It induces neurite outgrowth and promotes vascular development. At higher concentration, neurite outgrowth is inhibited.
  • 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 angiogenesis in the future.
  • Genes which have been identified as tumor endothelial markers (TEMs) correspond to tags shown in SEQ ID NOS: 94-139, 173-176, 180-186.
  • Genes which have been identified as normal endothelial markers (NEMs) correspond to tags shown in SEQ ID NOS: 140-172.
  • PEMs pan-endothelial markers
  • 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.
  • Tags disclosed herein because of the way that they were made, represent sequences which are 3′ of the 3′ most restriction enzyme recognition site for the tagging enzyme used to generate the SAGE tags. In this case, the tags are 3′ of the most 3′ most NlaIII site in the cDNA molecules corresponding to mRNA.
  • Nucleic acids corresponding to tags may be RNA, cDNA, or genomic DNA, for example. Such corresponding nucleic acids can be determined by comparison to sequence databases to determine sequence identities. Sequence comparisons can be done using any available technique, such as BLAST, available from the National Library of Medicine, National Center for Biotechnology Information. Tags can also be used as hybridization probes to libraries of genomic or cDNA to identify the genes from which they derive. Thus, using sequence comparisons or cloning, or combinations of these methods, one skilled in the art can obtain full-length nucleic acid sequences.
  • Genes corresponding to tags will contain the sequence of the tag at the 3′ end of the coding sequence or of the 3′ untranslated region (UTR), 3′ of the 3′ most recognition site in the cDNA for the restriction endonuclease which was used to make the tags.
  • 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 including within the scope of such nucleic acids and proteins.
  • allelic variants 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 JRF, 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 JRF, Wang P, CG 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 (Bateman 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 (Bateman 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. 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 TEM: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 TEM of interest is administered to a subject.
  • the immunogenic 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 TEM of interest may be delivered in an expressing cell, such as a purified population of tumor endothelial cells or a populations of fused tumor endothelial and dendritic cells.
  • Nucleic acids encoding the TEM of interest may be delivered in a viral or non-viral delivery vector or vehicle.
  • Non-human sequences encoding the human TEM 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 are described herein or 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. These include the human markers identified by SEQ ID NOS: 1-172, i.e., the normal, pan-endothelial, and the tumor endothelial markers. Homologous mouse markers include tumor endothelial markers of SEQ ID NO: 182-186 and 190-194. 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.
  • 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 screen 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 of 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 tumor endothelial cell populations, can be contacted with test substances and the expression of tumor endothelial markers and/or normal endothelial markers determined.
  • Test substances which decrease the expression of tumor endothelial markers are candidates for inhibiting angiogenesis and the growth of tumors.
  • markers which are only expressed in normal endothelium but not in tumor endothelium can be monitored.
  • Test substances which increase the expression of such NEMs in tumor endothelium and other human cells can be identified as candidate antitumor or anti-angiogenic drugs
  • agents can be screened for their ability to decrease or increase the activity.
  • the identification of drug candidates capable of binding to the TEM receptors found at the cell surface For those tumor endothelial markers identified as containing transmembrane regions, it is desirable to identify drug candidates capable of binding to the TEM receptors found at the cell surface. For some applications, the identification of drug candidates capable of blocking the TEM receptor from its native ligand will be desired. For some applications, the identification of a drug candidate capable of binding to the TEM 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 TEM receptor:ligand complex, one may be able to promote or inhibit further development of endothelial cells and hence, vascularization.
  • the identification of drug candidates capable of binding to the secreted TEM protein For those tumor endothelial markers identified as being secreted proteins, it is desirable to identify drug candidates capable of binding to the secreted TEM protein. For some applications, the identification of drug candidates capable of interfering with the binding of the secreted TEM 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 TEM: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 which 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 TEM protein activity can also be used as a drug screen. Human and mouse TEMS can be used for this purpose.
  • 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 which 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. NEMs, can be used to restrict, diminish, reduce, or inhibit proliferation of tumor or other abnormal or undesirable vasculature. TEMs 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 TEMs can also be used to therapeutic benefit.
  • antibodies, T cells specific for a TEM, antisense to a TEM, and ribozymes specific for a TEM 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.
  • PPD polycystic kidney disease
  • Mouse counterparts to human TEMS 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 TEMs are disclosed in SEQ ID NO: 182-186 and 190-194. Mouse TEMs can be used as antigens for raising antibodies which can be tested in mouse tumor models. Mouse TEMs with transmembrane domains are particularly preferred for this purpose. Mouse TEMs can also be used as vaccines to raise an immunological response in a human to the human ortholog.
  • the endothelium of human colorectal cancer was chosen to address the issues of tumor angiogenesis, based on the high incidence, relatively slow growth, and resistance to anti-neoplastic agents of these cancers. While certain less common tumor types, such as glioblastomas, are highly vascularized and are regarded as good targets for anti-angiogenic therapy, the importance of angiogenesis for the growth of human colorectal cancers and other common solid tumor types is less well documented.
  • SAGE Serial Analysis of Gene Expression
  • SAGE is not dependent on pre-existing databases of expressed genes, and therefore provides an unbiased view of gene expression profiles. This feature is particularly important in the analysis of cells that constitute only a small fraction of the tissue under study, as transcripts from these cells are unlikely to be well represented in extant EST databases.
  • a library of 100,000 tags from the purified ECs of a colorectal cancer, and a similar library from the ECs of normal colonic mucosa from the same patient were generated. These 193,000 tags corresponded to over 32,500 unique transcripts. Examination of the expression pattern of hematopoietic, epithelial and endothelial markers confirmed the purity of the preparations ( FIG. 2D ).
  • PEMs Pan Endothelial Markers
  • transcripts that were expressed at significantly higher levels in both normal and tumor associated endothelium compared to other tissues.
  • tags expressed at similar levels in both tumor and normal ECs were compared to 1.8 million tags from a variety of cell lines derived from tumors of non-endothelial origin. This simple comparison identified 93 transcripts that were strikingly EC-specific, i.e. expressed at levels at least 20-fold higher in ECs in vivo compared to non-endothelial cells in culture.
  • the 15 tags corresponding to characterized genes which were most highly and specifically expressed in endothelium are shown in Table 1A.
  • VEGFR2 VEGFR2 (KDR), which had previously been reported to be up-regulated in vessels during colon cancer progression, was found to be expressed in both normal and neoplastic colorectal tissue ( FIGS. 3 D and E). The lack of specificity of this gene was in accord with the SAGE data, which indicated that the VEGFR was expressed at 12 copies per cell in both normal and tumor endothelium.
  • the vWF gene expected to be expressed in both normal and tumor endothelium on the basis of the SAGE data as well as previous studies, was expressed at similar levels in normal and tumor ECs from both patients, but was not expressed in purified tumor epithelial cells.
  • PEM2 displayed a pattern similar to vWF.
  • all 9 TEMs chosen for this analysis were prominently expressed in tumor ECs, but were absent or barely detectable in normal ECs (Table 3 and examples in FIG. 4A ).
  • Tumor Endothelium Markers are Expressed in Multiple Tumor Types
  • Tumor Endothelium Markers are Neo-Angiogenic
  • transcripts were expressed in angiogenic states other than that associated with tumorigenesis.

Abstract

To gain a better understanding of tumor angiogenesis, new techniques for isolating endothelial cells (ECs) and evaluating gene expression patterns were developed. When transcripts from ECs derived from normal and malignant colorectal tissues were compared with transcripts from non-endothelial cells, over 170 genes predominantly expressed in the endothelium were identified. Comparison between normal- and tumor-derived endothelium revealed 79 differentially expressed genes, including 46 that were specifically elevated in tumor-associated endothelium. Experiments with representative genes from this group demonstrated that most were similarly expressed in the endothelium of primary lung, breast, brain, and pancreatic cancers as well as in metastatic lesions of the liver. These results demonstrate that neoplastic and normal endothelium in humans are distinct at the molecular level, and have significant implications for the development of anti-angiogenic therapies in the future.

Description

  • This application claims the benefit of provisional application Ser. Nos. 60/222,599 filed Aug. 2, 2000, 60/224,360 filed Aug. 11, 2000, and 60/282,850 filed Apr. 11, 2001, the disclosures of which are expressly incorporated herein.
  • The U.S. government retains certain rights in the invention by virtue of the provisions of National Institutes of Heath grants CA57345 and CA43460, which supported this work.
  • 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 tumor endothelial and normal endothelial cells.
  • BACKGROUND OF THE INVENTION
  • It is now widely recognized that tumors require a blood supply for expansive growth. This recognition has stimulated a profusion of research on tumor angiogenesis, based on the idea that the vasculature in tumors represents a potential therapeutic target. However, several basic questions about tumor endothelium remain unanswered. For example, are vessels of tumors qualitatively different from normal vessels of the same tissue? What is the relationship of tumor endothelium to endothelium of healing wounds or other physiological or pathological forms of angiogenesis? The answers to these questions critically impact on the potential for new therapeutic approaches to inhibit angiogenesis in a specific manner.
  • There is a continuing need in the art to characterize the vasculature of tumors relative to normal vasculature so that any differences can be exploited for therapeutic and diagnostic benefits.
  • One technique which can be used to characterize gene expression, or more precisely gene transcription, is termed serial analysis of gene expression (SAGE). Briefly, the SAGE approach is a method for the rapid quantitative and qualitative analysis of mRNA transcripts based upon the isolation and analysis of short defined sequence tags (SAGE Tags) corresponding to expressed genes. Each Tag is a short nucleotide sequences (9-17 base pairs in length) from a defined position in the transcript. In the SAGE method, the Tags are dimerized to reduce bias inherent in cloning or amplification reactions. (See, U.S. Pat. No. 5,695,937) SAGE is particularly suited to the characterization of genes associated with vasculature stimulation or inhibition because it is capable of detecting rare sequences, evaluating large numbers of sequences at one time, and to provide a basis for the identification of previously unknown genes.
  • SUMMARY OF THE INVENTION
  • One embodiment of the invention provides an isolated molecule comprising an antibody variable region which specifically binds to an extracellular domain of a TEM protein selected from the group consisting of: 1, 3, 9, 17, 19, and 44, as shown in SEQ ID NO: 196, 200, 212, 230, 232, and 271, respectively. The molecule can be, for example, an in tact antibody molecule, a single chain variable region (ScFv), a monoclonal antibody, a humanized antibody, or a human antibody. The molecule can optionally be bound to a cytotoxic moiety, bound to a therapeutic moiety, bound to a detectable moiety, or bound to an anti-tumor agent.
  • According to another embodiment of the invention a method of inhibiting neoangiogenesis is provided. An effective amount of an isolated molecule comprising an antibody variable region which specifically binds to an extracellular domain of a TEM protein selected from the group consisting of: 1, 3, 9, 17, 19, 22, and 44, as shown in SEQ ID NO: 196, 200, 212, 230, 232, 238, and 271, respectively, is administered to a subject in need thereof. Neoangiogenesis is consequently inhibited. The subject may bear a vascularized tumor, may have polycystic kidney disease, may have diabetic retinopathy, may have rheumatoid arthritis, may have psoriasis, for example.
  • Another aspect of the invention is a method of inhibiting tumor growth. An effective amount of an isolated molecule comprising an antibody variable region which specifically binds to an extracellular domain of a TEM protein selected from the group consisting of: 1, 3, 9, 17, 19, 22, and 44, as shown in SEQ ID NO: 196, 200, 212, 230, 232, 238, and 271, respectively, is administered to a human subject bearing a tumor. The growth of the tumor is consequently inhibited.
  • Still another aspect of the invention provides an isolated molecule comprising an antibody variable region which specifically binds to a TEM protein selected from the group consisting of: 3, 9, 17, 19, and 44, as shown in SEQ ID NO: 200, 212, 230, 232, and 271, respectively. The molecule can be, for example, an in tact antibody molecule, a single chain variable region (ScFv), a monoclonal antibody, a humanized antibody, or a human antibody. The molecule can optionally be bound to a cytotoxic moiety, bound to a therapeutic moiety, bound to a detectable moiety, or bound to an anti-tumor agent.
  • According to still another aspect of the invention an isolated and purified human transmembrane protein is provided. The protein is selected from the group consisting of: TEM 3, 9, 17, and 19 as shown in SEQ ID NO: 200, 212, 230, and 232, respectively.
  • Yet another aspect of the invention is an isolated and purified nucleic acid molecule comprising a coding sequence for a transmembrane TEM selected from the group consisting of:: TEM 3, 9, 17, and 19 as shown in SEQ ID NO: 200, 212, 230, and 232, respectively. The isolated and purified nucleic acid molecule may optionally comprise a coding sequence selected from those shown in SEQ ID NO:: 199, 211, 229, and 231.
  • Still another aspect of the invention is a recombinant host cell which comprises a nucleic acid molecule. The nucleic acid molecule comprises a coding sequence for a transmembrane TEM selected from the group consisting of:: TEM 3, 9, 17, and 19 as shown in SEQ ID NO: 200, 212, 230, and 232, respectively. The recombinant host cell optionally comprises a coding sequence selected from those shown in SEQ ID NO: 199, 211, 229, and 231.
  • According to one embodiment of the invention a method is provided for inducing an immune response in a mammal. A nucleic acid molecule comprising a coding sequence for a human transmembrane protein selected from the group consisting of: TEM 1, 3, 9, 13, 17, 19, 22, 30, and 44 as shown in SEQ ID NO: respectively, is administered to the mammal. An immune response to the human transmembrane protein is thereby induced in the mammal. Optionally the coding sequence is shown in SEQ ID NO: 196, 200, 212, 220, 230, 232, 238, 250 and 271.
  • According to yet another embodiment of the invention a method of inducing an immune response in a mammal is provided. A purified human transmembrane protein selected from the group consisting of: TEM 1, 3, 9, 13, 17, 19, 22, 30, and 44 as shown in SEQ ID NO: 196, 200, 212, 220, 230, 232, 238, 250 and 271, respectively, is administered to the mammal. An immune response to the human transmembrane protein is thereby induced in the mammal.
  • Another aspect of the invention is a method for identification of a ligand involved in endothelial cell regulation. A test compound is contacted with an isolated and purified human transmembrane protein selected from the group consisting of 1, 3, 9, 13, 17, 30, 19, and 44 as shown in SEQ ID NO: 196, 200, 212, 220, 230, 232, 250, and 271. The isolated and purified human transmembrane protein is also contacted with a molecule comprising an antibody variable region which specifically binds to an extracellular domain of a TEM protein selected from the group consisting of: 1, 3, 9, 13, 17, 30, 19, and 44 as shown in SEQ ID NO: 196, 200, 212, 220, 230, 232, 250, and 271 respectively. Binding of the molecule comprising an antibody variable region to the human transmembrane protein is determined. A test compound which diminishes the binding of the molecule comprising an antibody variable region to the human transmembrane protein is identified as a ligand involved in endothelial cell regulation.
  • Yet another aspect of the invention is a method for identification of a ligand involved in endothelial cell regulation. A test compound is contacted with a cell comprising a human transmembrane protein selected from the group consisting of 1, 3, 9, 17, and 19 as shown in SEQ ID NO: 196, 200, 212, 230, and 232. The cell is also contacted with a molecule comprising an antibody variable region which specifically binds to an extracellular domain of a TEM protein selected from the group consisting of: 1, 3, 9, 17, and 19 as shown in SEQ ID NO: 196, 200, 212, 230, and 232, respectively. Binding of the molecule comprising an antibody variable region to the cell is determined. A test compound which diminishes the binding of the molecule comprising an antibody variable region to the cell is identified as a ligand involved in endothelial cell regulation.
  • Yet another aspect of the invention is a method for identification of a ligand involved in endothelial cell regulation. A test compound is contacted with a human transmembrane protein selected from the group consisting of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 14, 15, 16, 17, 19, 20, 21, 22, 24, 25, 27, 28, 29, 40, 31, 33, 35, 36, 37, 38, 39, 41, 42, 44, 45, and 46 as shown in SEQ ID NO: 196, 198, 200, 202, 204, 206, 208, 210, 212, 214, 216, 218, 223 & 224, 226, 228, 230, 232, 234, 236, 238, 240, 242, 244, 246, 248, 250, 252, 254, 358, 257, 259, 261, 263, 267, 269, 271, 273, and 275. Binding of a test compound to the human transmembrane protein is determined. A test compound which binds to the protein is identified as a ligand involved in endothelial cell regulation.
  • Another embodiment of the present invention is a soluble form of a human transmembrane protein selected from the group consisting of: TEM 1, 3, 9, 17, 19, 22, 30, and 44 as shown in SEQ ID NO: 196, 200, 212, 230, 232, 238, 250, and 271 respectively. The soluble forms lack transmembrane domains. The soluble form may consist of an extracellular domain of the human transmembrane protein.
  • Also provided by the present invention is a method of inhibiting neoangiogenesis in a patient. A soluble form of a human transmembrane protein is adminstered to the patient. Neoangiogenesis in the patient is consequently inhibited. The patient may bear a vascularized tumor, may have polycystic kidney disease, may have diabetic retinopathy, may have rheumatoid arthritis, or may have psoriasis, for example.
  • Another embodiment of the invention provides a method of inhibiting neoangiogenesis in a patient. A soluble form of a human transmembrane protein is administered to the patient. Neoangiogenesis in the patient is consequently inhibited. The patient may bear a vascularized tumor, may have polycystic kidney disease, may have diabetic retinopathy, may have rheumatoid arthritis, or may have psoriasis, for example.
  • According to still another aspect of the invention a method of identifying regions of neoangiogenesis in a patient is provided. A molecule comprising an antibody variable region which specifically binds to an extracellular domain of a TEM protein selected from the group consisting of: 1, 3, 9, 13, 17, 19, 22, 30, and 44, as shown in SEQ ID NO: 196, 200, 212, 220, 230, 232, 238, 250, and 271, respectively, is administered to a patient. The molecule is bound to a detectable moiety. The detectable moiety is detected in the patient, thereby identifying neoangiogenesis.
  • According to another aspect of the invention a method is provided for inducing an immune response to tumor endothelial cells in a patient. A mouse TEM protein selected from the group consisting of: 1, 2, 3, 9, 13, 17, 19, 22, and 30 as shown in SEQ ID NO: 291, 293, 299, 295, 303, 297, 301, 305, and 307, is administered to a patient in need thereof. An immune response to a human TEM protein is consequently induced.
  • Still another embodiment of the invention is a method of screening for neoangiogenesis in a patient. A body fluid collected from the patient is contacted with a molecule comprising an antibody variable region which specifically binds to an extracellular domain of a TEM protein selected from the group consisting of: 1, 3, 9, 17, 19, and 44, as shown in SEQ ID NO: 196, 200, 212, 230, 232, and 271, respectively. Detection of cross-reactive material in the body fluid with the molecule indicates neo-angiogenesis in the patient.
  • Still another embodiment of the invention provides a method of inhibiting neoangiogenesis in a patient. A molecule comprising an antibody variable region which specifically binds to a TEM protein selected from the group consisting of: 4, 6, 7, 10, 12, 14, 20, 25, 27, 31, 36, 37, 38, 39, and 40 as shown in SEQ ID NO: 202, 206, 208, 214, 218, 223 and 224, 234, 242, 244, 252, 257, 259, 261. 263, and 265, is administered to the patient. Neoangiogenesis in the patient consequently inhibited.
  • Yet another aspect of the invention is a method of screening for neoangiogenesis in a patient. A body fluid collected from the patient is contacted with a molecule comprising an antibody variable region which specifically binds to a TEM protein selected from the group consisting of: 4, 6, 7, 10, 12, 14, 20, 25, 27, 31, 36, 37, 38, 39, and 40, as shown in SEQ ID NO: 202, 206, 208, 214, 218, 223 & 224, 234, 242, 244, 252, 257, 259, 261. 263, and 265, respectively. Detection of cross-reactive material in the body fluid with the molecule indicates neoangiogenesis in the patient.
  • Also provided by the present invention is a method of promoting neoangiogenesis in a patient. A TEM protein selected from the group consisting of: 4, 6, 7, 10, 12, 14, 20, 25, 27, 31, 36, 37, 38, 39, and 40, as shown in SEQ ID NO: 202, 206, 208, 214, 218, 223 & 224, 234, 242, 244, 252, 257, 259, 261. 263, and 265, is administered to a patient in need of neoangiogenesis. Neoangiogenesis in the patient is consequently stimulated.
  • One embodiment of the invention provides a method of promoting neoangiogenesis in a patient. A nucleic acid molecule encoding a TEM protein selected from the group consisting of: 4, 6, 7, 10, 12, 14, 20, 25, 27, 31, 36, 37, 38, 39, and 40, as shown in SEQ ID NO: 201, 205, 207, 213, 217, 221 & 222, 233, 241, 243, 251, 256, 258, 260, 262, and 264, is administered to a patient in need of neoangiogenesis. The TEM protein is consequently expressed and neoangiogenesis in the patient is stimulated.
  • Another embodiment of the invention provides a method of screening for neoangiogenesis in a patient. A TEM protein selected from the group consisting of: 4, 6, 7, 10, 12, 14, 20, 25, 27, 31, 36, 37, 38, 39, and 40, as shown in SEQ ID NO:: 202, 206, 208, 214, 218, 223 & 224, 234, 242, 244, 252, 257, 259, 261. 263, and 265, respectively, is detected in a body fluid collected from the patient. Detection of the TEM protein indicates neoangiogenesis in the patient.
  • Another aspect of the invention is a method of screening for neoangiogenesis in a patient. A nucleic acid encoding a TEM protein selected from the group consisting of: 4, 6, 7, 10, 12, 14, 20, 25, 27, 31, 36, 37, 38, 39, and 40 is detected in a body fluid collected from the patient. The nucleic acid is selected from the group consisting of those shown in SEQ ID NO: 201, 205, 207, 213, 217, 221 & 222, 233, 241, 243, 251, 256, 258, 260, 262, and 264. Detection of the TEM protein indicates neoangiogenesis in the patient.
  • Yet another embodiment of the invention is an isolated and purified nucleic acid molecule which encodes a NEM protein selected from the group consisting of: 14, 22, 23, and 33 as shown in SEQ ID NO: 279, 283, 285, 286, 287, and 289. The nucleic acid molecule optionally comprises a coding sequence as shown in SEQ ID NO: 278, 282, 284, and 288. The nucleic acid may be maintained in a recombinant host cell.
  • The present invention also provides an isolated and purified NEM protein selected from the group consisting of: 14, 22, 23, and 33 as shown in SEQ ID NO: 279, 283, 285, 286, 287, and 289.
  • The present invention further provides an isolated molecule comprising an antibody variable region which specifically binds to a NEM protein selected from the group consisting of: 14, 22, 23, and 33, as shown in SEQ ID NO: 279, 283, 285, 286, 287, and 289.
  • An additional embodiment of the present invention is a method of inhibiting neoangiogenesis. An effective amount of a NEM protein selected from the group consisting of: 14, 22, 23, and 33 as shown in SEQ ID NO: 279, 283, 285, 286, 287, and 289 is administered to a subject in need thereof. Neoangiogenesis is thereby inhibited.
  • A still further embodiment of the invention is a method to identify candidate drugs for treating tumors. Cells which express one or more TEM genes selected from the group consisting of: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 14, 15, 16, 17, 19, 20, 21, 22, 24, 25, 27, 28, 29, 40, 31, 33, 35, 36, 37, 38, 39, 41, 42, 44, 45, and 46 as shown in SEQ ID NO:: 195, 197, 199, 201, 203, 205, 207, 209, 211, 213, 215, 217, 221 & 222, 225, 227, 229, 231, 233, 235, 237, 239, 241, 243, 245, 247, 249, 251, 253, 255, 256, 258, 260, 262, 266, 268, 270, 272, and 274, respectively, are contacted with a test compound. Expression of said one or more TEM genes is determined by hybridization of mRNA of said cells to a nucleic acid probe which is complementary to said mRNA. A test compound is identified as a candidate drug for treating tumors if it decreases expression of said one or more TEM genes. Optionally the cells are endothelial cells. Alternatively or additionally, the cells are recombinant host cells which are transfected with an expression construct which encodes said one or more TEMs. Test compounds which increase expression can be identified as candidates for promoting wound healing.
  • Yet another embodiment of the invention is a method to identify candidate drugs for treating tumors. Cells which express one or more TEM proteins selected from the group consisting of: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 14, 15, 16, 17, 19, 20, 21, 22, 24, 25, 27, 28, 29, 40, 31, 33, 35, 36, 37, 38, 39, 41, 42, 44, 45, and 46 as shown in SEQ ID NO: 196, 198, 200, 202, 204, 206, 208, 210, 212, 214, 216, 218, 223 & 224, 226, 228, 230, 232, 234, 236, 238, 240, 242, 244, 246, 248, 250, 252, 254, 358, 257, 259, 261, 263, 267, 269, 271, 273, and 275, respectively, are contacted with a test compound. The amount of said one or more TEM proteins in said cells is determined. A test compound is identified as a candidate drug for treating tumors if it decreases the amount of one or more TEM proteins in said cells. Optionally the cells are endothelial cells. Alternatively or additionally, the cells are recombinant host cells which are transfected with an expression construct which encodes said one or more TEMs. Alternatively, a test compound which increases the amount of one or more TEM proteins in said cells is identified as a candidate drug for treating wound healing.
  • According to another aspect of the invention a method is provided to identify candidate drugs for treating tumors. Cells which express one or more TEM proteins selected from the group consisting of: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 14, 15, 16, 17, 19, 20, 21, 22, 24, 25, 27, 28, 29, 40, 31, 33, 35, 36, 37, 38, 39, 41, 42, 44, 45, and 46 as shown in SEQ ID NO: 196, 198, 200, 202, 204, 206, 208, 210, 212, 214, 216, 218, 223 & 224, 226, 228, 230, 232, 234, 236, 238, 240, 242, 244, 246, 248, 250, 252, 254, 358, 257, 259, 261, 263, 267, 269, 271, 273, and 275, respectively, are contacted with a test compound. Activity of said one or more TEM proteins in said cells is determined. A test compound is identified as a candidate drug for treating tumors if it decreases the activity of one more TEM proteins in said cells. Optionally the cells are endothelial cells. Alternatively or additionally, the cells are recombinant host cells which are transfected with an expression construct which encodes said one or more TEMs. Optionally the cells are endothelial cells. If a test compound increases the activity of one more TEM proteins in said cells it can be identified as a candidate drug for treating wound healing.
  • An additional aspect of the invention is a method to identify candidate drugs for treating patients bearing tumors. A test compound is contacted with recombinant host cells which are transfected with an expression construct which encodes one or more TEM proteins selected from the group consisting of 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 14, 15, 16, 17, 19, 20, 21, 22, 24, 25, 27, 28, 29, 40, 31, 33, 35, 36, 37, 38, 39, 41, 42, 44, 45, and 46 as shown in SEQ ID NO: 198, 200, 202, 204, 206, 208, 210, 212, 214, 216, 218, 223 & 224, 226, 228, 230, 232, 234, 236, 238, 240, 242, 244, 246, 248, 250, 252, 254, 358, 257, 259, 261, 263, 267, 269, 271, 273, and 275, respectively. Proliferation of said cells is determined. A test compound which inhibits proliferation of said cells is identified as a candidate drug for treating patients bearing tumors. A test compound which stimulates proliferation of said cells is identified as a candidate drug for promoting neoangiogenesis, such as for use in wound healing.
  • Another embodiment of the invention provides a method to identify candidate drugs for treating tumors. Cells which express one or more NEM genes selected from the group consisting of: 14, 22, 23, and 33 as shown in SEQ ID NO: 278, 282, 284, and 288, respectively, are contacted with a test compound. Expression of said one or more NEM genes is determined by hybridization of mRNA of said cells to a nucleic acid probe which is complementary to said mRNA. A test compound is identified as a candidate drug for treating tumors if it increases expression of said one or more NEM genes. Optionally the cells are endothelial cells. Alternatively or additionally, the cells are recombinant host cells which are transfected with an expression construct which encodes said one or more NEMs.
  • According to another aspect of the invention a method is provided to identify candidate drugs for treating tumors. Cells which express one or more NEM proteins selected from the group consisting of: 14, 22, 23, and 33 as shown in SEQ ID NO: 279, 283, 285, 286, 287, and 289, are contacted with a test compound. The amount of said one or more NEM proteins in said cells is determined. A test compound is identified as a candidate drug for treating tumors if it increases the amount of one more NEM proteins in said cells. Optionally the cells are endothelial cells. Alternatively or additionally, the cells are recombinant host cells which are transfected with an expression construct which encodes said one or more NEMs.
  • An additional aspect of the invention is a method to identify candidate drugs for treating tumors. Cells which express one or more NEM proteins selected from the group consisting of: 14, 22, 23, and 33 as shown in SEQ ID NO: 279, 283, 285, 286, 287, and 289, are contacted with a test compound. Activity of said one or more NEM proteins in said cells is determined. A test compound is identified as a candidate drug for treating tumors if it increases the activity of said one or more NEM proteins in said cells. Optionally the cells are endothelial cells. Alternatively or additionally, the cells are recombinant host cells which are transfected with an expression construct which encodes said one or more NEMs.
  • Still another embodiment of the invention provides a method to identify candidate drugs for treating patients bearing tumors. A test compound is contacted with recombinant host cells which are transfected with an expression construct which encodes one or more NEM proteins selected from the group consisting of 14, 22, 23, and 33 as shown in SEQ ID NO: 279, 283, 285, 286, 287, and 289. Proliferation of said cells is determined. A test compound which stimulates proliferation of said cells is identified as a candidate drug for treating patients bearing tumors.
  • Another aspect of the invention is a method for identifying endothelial cells. One or more antibodies which bind specifically to a TEM or NEM protein selected from the group consisting of TEM: 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 14, 15, 16, 17, 19, 20, 21, 22, 24, 25, 27, 28, 29, 30, 31, 33, 35, 36, 37, 38, 39, 41, 42, 44, 45, and 46 as shown in SEQ ID NO: 198, 200, 202, 204, 206, 208, 210, 212, 214, 216, 218, 223 & 224, 226, 228, 230, 232, 234, 236, 238, 240, 242, 244, 246, 248, 250, 252, 254, 358, 257, 259, 261, 263, 267, 269, 271, 273, and 275 and NEM 14, 22, 23, and 33 as shown in SEQ ID NO: 279, 283, 285, 286, 287, and 289, is contacted with a population of cells. Cells in the population which have bound to said antibodies are detected. Cells which are bound to said antibodies are identified as endothelial cells. Optionally cells which have bound to said antibodies are isolated from cells which have not bound.
  • Still another aspect of the invention is a method for identifying endothelial cells. One or more nucleic acid hybridization probes which are complementary to a TEM or NEM gene nucleic acid sequence selected from the group consisting of TEM: 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 14, 15, 16, 17, 19, 20, 21, 22, 24, 25, 27, 28, 29, 30, 31, 33, 35, 36, 37, 38, 39, 41, 42, 44, 45, and 46 as shown in SEQ ID NO: 198, 200, 202, 204, 206, 208, 210, 212, 214, 216, 218, 223 & 224, 226, 228, 230, 232, 234, 236, 238, 240, 242, 244, 246, 248, 250, 252, 254, 358, 257, 259, 261, 263, 267, 269, 271, 273, and 275 and NEM 14, 22, 23, and 33 as shown in SEQ ID NO: 279, 283, 285, 286, 287, and 289, is contacted with nucleic acids of a population of cells. Nucleic acids which have specifically hybridized to said nucleic acid hybridization probes are detected. Cells whose nucleic acids specifically hybridized are identified as endothelial cells.
  • Yet another embodiment of the invention is a method of inhibiting neoangiogenesis. An effective amount of an isolated molecule comprising an antibody variable region which specifically binds to an extracellular domain of a mouse TEM protein selected from the group consisting of: 1, 2, 3, 9, 17, and 19, as shown in SEQ ID NO: 291, 293, 299, 295, 297, and 301, respectively, is administered to a subject in need thereof. Neoangiogenesis is thereby inhibited. The subject may be a mouse, may bear a vascularized tumor, may have polycystic kidney disease, may have diabetic retinopathy may have rheumatoid arthritis, or may have psoriasis, for example.
  • These and other embodiments which will be apparent to those of skill in the art upon reading the specification provide the art with reagents and methods for detection, diagnosis, therapy, and drug screening pertaining to neoangiogenesis and pathological processes involving or requiring neoangiogenesis.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1A-1B. vWF expression in colorectal cancers. vWF (red stain) was detected in vessels by in situ hybridization. At low power magnification (FIG. 1.A) vessels were often surrounded by a perivascular cuff of viable cells (red arrows), with a ring of necrotic cells evident at the periphery (black arrows). At high power magnification (FIG. 1.B) the expression of vWF (red) was clearly localized to the vessels. Sections were counterstained with methyl green.
  • FIG. 2A-2D. Purification of Endothelial Cells (ECs) from human normal and malignant tissue. (FIG. 2A) Vessels (red) of frozen sections were stained by immunofluorescence with the P1H12 monoclonal antibody (Chemicon, Temecula, Calif.) and detected using a biotinylated goat anti-mouse IgG secondary antibody followed by rhodamine-linked strepavidin. The region stained is from within the lamina propria of normal colonic mucosa. Note that the larger vessels (arrowheads) and capillaries (arrows) are positive, and staining of hematopoietic cells was undetectable. E-cadherin positive epithelial cells (green) at the edge of the crypt were simultaneously visualized using a rabbit polyclonal antibody (Santa Cruz, Santa Cruz, Calif.), followed by a goat anti-rabbit IgG secondary antibody labelled with alexa (Molecular Probes, Eugene, Oreg.). Sections were imaged at 60× magnification using confocal microscopy. (FIG. 2.B) To isolate pure populations from collagenase dispersed tissues, the epithelial and hematopoietic cell fractions were sequentially removed by negative selection with magnetic beads. The remaining cells were stained with P1H12 and ECs were isolated by positive selection with magnetic beads. (FIG. 2.C) RT-PCR analysis used to assess the purity of the EC preparations. Semiquantitative PCR analysis was performed on cDNA generated either directly from colorectal cancer tissue (unfractionated tumor) or from purified ECs isolated from normal colonic mucosa (normal EC fraction) or colorectal cancer (tumor EC fraction). PCR amplification of the epithelial specific marker cytokeratin 20 (CK20), demonstrated its expression was limited to the unfractionated tumor. Two endothelial specific markers, vWF and VE-cadherin (VE-Cad) showed robust amplification only in the endothelial fractions, validating the purity and enrichment protocol shown in (FIG. 2.B). The ubiquitous housekeeping enzyme GAPDH was observed in all samples. No signal was detected in the no-template (NT) control. cDNA templates were diluted 1:10, 1:100, 1:1000, 1:4000, and 1:40,000 as indicated by the declining wedge. (FIG. 2.D) The relative expression level of select genes was determined by measuring the tag abundance from several SAGE libraries combined into four groups. The first was composed of ˜193,000 tags from the two in vivo-derived EC preparations (Endothelial Cell Fraction) while the second contained a single library of ˜57,000 tags containing macrophages and other leukocytes derived from the negative selection (Hematopoietic Fraction). The fourth library contained ˜401,000 tags from cultured HUVEC and HMVEC (Endothelial Cells in Culture), and the fourth consisted of 748,000 tags from 6 colon cancer cell lines in culture (Epithelial Cells). After normalization, the library with the highest tag number for each marker was given a value of 100%, and the corresponding relative expression levels of the remaining 3 libraries was plotted on the ordinate. Note the high level of CD31 present on hematopoietic cells, the likely cause of the impurity of the initial endothelial selection, compared with the selectivity of P1H12.
  • FIG. 3A-3E). Expression of Pan-Endothelial Markers (PEMs) is limited to ECs. The endothelial origin of PEMs identified by SAGE was confirmed using a highly sensitive in situ hybridization assay. Localization of novel PEMs to the ECs was demonstrated by examining two representative PEMs, PEM3 (FIG. 3A) and PEM6 (FIG. 3B) in lung cancer and colon cancer, respectively. Hevin expression was readily detected in the ECs of a colon tumor (FIG. 3C) despite its low level of expression in cultured ECs. Expression of VEGFR2 was readily detectable in the ECs of both normal (FIG. 3D) and malignant colon tissue (FIG. 3E).
  • FIG. 4A-4J. Expression of Tumor Endothelial Markers (TEMs). (FIG. 4A) RT-PCR analysis confirmed the tumor specific expression of selected novel TEMs. Semiquantitative PCR analysis was performed on cDNA generated either from purified epithelial cells as a negative control (Control) or from purified ECs isolated from normal colonic mucosa (Normal ECs) or colorectal cancer (Tumor ECs) from two different patients. Two endothelial specific markers, vWF and PEM6 showed robust amplification only in the endothelial fractions whereas the ubiquitous housekeeping enzyme GAPDH was observed in all samples. TEM1 (BSC-TEM1), TEM 17 (BSC-TEM7) and TEM22 (BSC-TEM9) were specifically expressed in tumor compared to normal ECs. The cDNA template was diluted 1:10, 1:100, 1:1000, and 1:10,000 as indicated by the declining wedge. (FIG. 4 B-4J) The endothelial origin of TEMs identified by SAGE was confirmed using in situ hybridization as in FIG. 3. Expression of TEM 1 (BSC-TEM1) (FIG. 4 B) and TEM17 (BSC-TEM7) (FIG. 4 C) was demonstrated to be highly specific to the ECs in colorectal cancers; sections were imaged in the absence of a counterstain to show the complete lack of detectable expression in the non-endothelial cells of the tumor. Expression of TEM17 (BSC-TEM7) in ECs was demonstrated in a metastatic liver lesion from a primary colorectal cancer (FIG. 4 D), a lung (FIG. 4 E), breast (FIG. 4 F), pancreatic (FIG. 4 G) and brain cancer (FIG. 4 H), as well as in a sarcoma (FIG. 4 I). TEM 17 (BSC-TEM7) was also localized to vessels during normal physiological angiogenesis of the corpus luteum (FIG. 4 J).
  • DETAILED DESCRIPTION OF THE INVENTION
  • We identified 46 human genes that were expressed at significantly higher levels (>10-fold) in tumor endothelium than in normal endothelium, and 33 genes that were expressed at significantly lower levels in human tumor versus normal endothelium. See Tables 2 and 4, respectively. Most of these genes were either not expressed or expressed at relatively low levels in Endothelial Cells (ECs) maintained in culture. Moreover, we identified 93 genes which are expressed in both normal and tumor human endothelium. Interestingly, the tumor endothelium genes were expressed in all tumors tested, regardless of its tissue or organ source. Most tumor endothelium genes were also expressed in corpus luteum and wounds.
  • As the work has progressed, we have refined and classified our original 46 tumor endothelial markers. We have named these markers TEMs and renumbered them consecutively by the prevalence of their tags in our SAGE analysis. Originally we had not used a consecutive numbering system. Our non-consecutive numbering system has been renamed as BSC-TEMs. For most of the original 46 SAGE Tags, we now provide full-length nucleic acid and protein sequence. In some cases, the sequences were obtained through the public databases, in others the sequences were obtained by cloning and through the use of gene prediction tools. In some cases, we found SAGE Tags corresponding to genes having different splice varients or with known polymorphisms. For example, in one case the SAGE Tag BSC-TEM3 has been found to hybridize to an alternatively spliced form of the transcript encoding BSC-TEM7. The proteins encoded by the two transcripts are the same; therefore they are cumulatively called TEM7. A highly related sequence was found via homology searches, BSC-TEM7R. This paralog sequence is now called TEM3. See Table 2, which follows, showing tumor endothelial markers by order of prevalence (except for TEM 3). Column 1 indicates the prevalence number. Column 2 indicates the original nomenclature. Column 3 indicates the short tags. Column 4 indicates the long tags. Column 5 indicates the accession number in GenBank. Column 6 indicates the sequence identifiers for the short tag, the long tag, the full nucleic acid, and the protein. Column 7 provides a functional description, which is expanded below in the text.
  • TEM1 BSC-TEM1 GGGGCTGCCC GGGGCTGCCCAGCT NM020404 SEQ ID NO: tumor endothelial marker 1 precursor
    A GA 94, 309,
    195, 196
    TEM 2 BSC-TEM2 GATCTCCGTG SEQ ID NO: sapiens tumor endothelial marker 2
    T 95, 197, (BSC-TEM2) mRNA/mouse Ras, dexa-
    198 methasone-induced 1 (RASD1), mRNA
    TEM 3 BSC-TEM7R SEQ ID NO: human ortholog of mouse paralog of
    199, 200 mouse TEM-7
    TEM 4 CTTTCTTTGA CTTTCTTTGAGTTT AB034203 SEQ ID NO: Homo sapiens dickkopf-3 (DKK-3) mRNA,
    G TAA 97, 311,
    201, 202
    TEM 5 BSC-TEM4 TATTAACTCT TATTAACTCTCTTT SEQ ID NO: Tumor endothelial marker 4
    C GGA 98, 312,
    203, 204
    TEM 6 CAGGAGACCC CAGGAGACCCCAGG X57766 SEQ ID NO: Human stromelysin-3 mRNA.
    C CCC 99, 314,
    205, 206
    TEM 7 GGAAATGTCA GGAAATGTCAGCAA BC002576 SEQ ID NO: matrix metalloproteinase 2
    A GTA 100, 315, (gelatinase A, 72kD gelatinase, 72kD
    207, 208 type IV collagenase)
    TEM 8 CCTGGTTCAG SEQ ID NO: HeyL transcription factor
    T 101, 316,
    209, 210
    TEM 9 BSC-TEM5 TTTTTAAGAA TTTTTAAGAACTCG SEQ ID NO:
    C GGT 102, 317,
    211, 212
    TEM 10 TTTGGTTTTC TTTGGTTTTCCAAA J03464, SEQ ID NO: Human collagen alpha-2 type I mRNA,
    C AGA M18057, 103, 319 complete cds, clone pHCOL2A1.
    X02488 213, 214
    TEM 11 ATTTTGTATG ATTTTGTATGATTT NM_002508 SEQ ID NO: nidogen/entactin
    A TTA 104, 321,
    215, 216
    TEM 12 ACTTTAGATG ACTTTAGATGGGAA X52022 SEQ ID NO: H. sapiens RNA for type VI collagen
    G GCC 105, 322, alpha3 chain.
    217, 218
    TEM 13 GAGTGAGACC GAGTGAGACCCAGG M11749 SEQ ID NO: Human Thy-1 glycoprotein gene,
    C AGC 106, 324, complete cds.
    219, 220
    TEM 14 GTACACACAC GTACACACACCCCC SEQ ID NO: Cystatin SN
    C ACC 107, 325,
    221, 223
    TEM 14 GTACACACAC GTACACACACCCCC X54667 SEQ ID NO: H. sapiens mRNA for cystatin S.
    C ACC 107, 325,
    222, 224
    TEM 15 CCACAGGGGA CCACAGGGGATTCT NM_000090 SEQ ID NO: Human mRNA 3′ region for pro-alpha1
    T CCT 108, 327, (III) collagen.
    225, 226
    TEM 16 BSC-TEM6 TTAAAAGTCA TTAAAAGTCACTGT SEQ ID NO:
    AC GCA 109, 328,
    227, 228
    TEM 17 BSC-TEM7 ACAGACTGTT ACAGACTGTTAGCC AF279144 SEQ ID NO: Human Tumor endothelial marker 7
    A AAG 110, 329,
    229, 230
    TEM 18 CCACTGCAAC SEQ ID NO:
    C 111
    TEM 19 BSC-TEM8 CTATAGGAGA SEQ ID NO:
    C 112, 330,
    231, 232
    TEM 20 GTTCCACAGA NM_000089 SEQ ID NO: collagen, type I, alpha 2 (COL1A2
    A 113, 233,
    234
    TEM 21 TACCACCTCC TACCACCTCCCTTT SEQ ID NO: Homo sapiens mRNA; cDNA DKFZp762B245
    C CCT 114, 331, (from clone DKFZp762B245);
    235, 236
    TEM 22 BSC-TEM9 GCCCTTTCTC GCCCTTTCTCTGTA NM_006039 SEQ ID NO: endocytic receptor (macrophage
    T GTT 115, 334, mannose receptor family) (KIAA0709),
    237, 238
    TEM 23 TTAAATAGCA TTAAATAGCACCTT SEQ ID NO: no match
    C TAG 116, 335
    TEM 24 AGACATACTG AGACATACTGACAG NM_022648 SEQ ID NO: Homo sapiens mRNA; cDNA DKFZp434G162
    A AAT 117, 336, (from clone DKFZp434G162);
    239, 240
    TEM 25 TCCCCCAGGA TCCCCCAGGAGCCA L35279, SEQ ID NO: Homo sapiens (clone KT2) bone morpho-
    G CCG NM_006129 118, 338, genetic protein-1 (BMP-1) mRNA
    241, 242
    TEM 26 AGCCCAAAGT SEQ ID NO: No Match
    G 119
    TEM 27 ACTACCATAA NM_003062 SEQ ID NO: Homo sapiens mRNA for MEGF5, partial
    C 120, 243. cds.
    244
    TEM 28 TACAAATCGT TACAAATCGTTGTC NM_014859 SEQ ID NO: Homo sapiens mRNA for KIAA0672
    T AAA 121, 339, protein, complete cds.
    245, 246
    TEM 29 TTGGGTGAAA SEQ ID NO: ESTs (2 unigene clusters)
    A 122, 247,
    248
    TEM 30 CATTATCCAA CATTATCCAAAAAC THC534029, SEQ ID NO: integrin, alpha 1
    A AAT X68742, 123, 340,
    AI262158, 249, 250
    AI88747,
    AI394565,
    AA679721
    TEM 31 AGAAACCACG AGAAACCACGGAAA NM_001845 SEQ ID NO: hypothetical protein KIAA1164
    G TGG 124, 341,
    251, 252
    TEM 32 ACCAAAACCA SEQ ID NO: no match
    C 125
    TEM 33 TGAAATAAAC NM_000255 SEQ ID NO: methylmalonyl Coenzyme A mutase
    126, 253,
    254
    TEM 34 TTTGGTTTCC SEQ ID NO: no match
    127
    TEM 35 GTGGAGACGG GTGGAGACGGACTC ESTAI186535 SEQ ID NO: est
    A TGT 128, 345,
    255, 358
    TEM 36 TTTGTGTTGT TTTGTGTTGTATAT NM_004370 SEQ ID NO: est
    A TTA 129, 346,
    256, 257
    TEM 37 TTATGTTTAA TTATGTTTAATAGT NM_002345 SEQ ID NO: Human lumican mRNA, complete cds.
    T TGA 130, 347,
    258, 259
    TEM 38 TGGAAATGAC TGGAAATGACCCAA NM_000088 SEQ ID NO: collagen type1 alpha1
    AAA 131, 348,
    260, 261
    TEM 39 TGCCACACAG TGCCACACAGTGAC NM_003239 SEQ ID NO: Human transforming growth factor-beta
    T TTG 132, 350, 3 (TGF-beta3) mRNA, complete
    262, 263
    TEM 40 GATGAGGAGA GATGAGGAGACTGG SEQ ID NO: collagen, type I, alpha 2
    C CAA 133, 351,
    264, 265
    TEM 41 ATCAAAGGTT ATCAAAGGTTTGAT SEQ ID NO: est
    T TTA 134, 352,
    266, 267
    TEM 42 AGTCACTAGT AGTCACATAGTACA NM_025226 SEQ ID NO: ESTs
    AA 135, 353,
    268, 269
    TEM 43 TTCGGTTGGT TTCGGTTGGTCAAA SEQ ID NO: No match
    C GAT 136, 354
    TEM 44 CCCCACACGG CCCCACACGGGCAA NM_018354v SEQ ID NO: Homo sapiens cDNA FLJ11190 fis, clone
    G GCA 137, 355, PLACE1007583.
    270, 271
    TEM 45 GGCTTGCCTT GGCTTGCCTTTTTG NM_000366 SEQ ID NO: est
    T TAT 138, 356,
    272, 273
    TEM 46 ATCCCTTCCC ATCCCTTCCCGCCA NM_002688 SEQ ID NO: Homo sapiens mRNA for peanut-like
    G CAC 139, 357, protein 1, PNUTL1 (hCDCreI-1).
    274, 275
  • The studies described below provide the first definitive molecular characterization of ECs in an unbiased and general manner. They lead to several important conclusions that have direct bearing on long-standing hypotheses about angiogenesis. First, it is clear that normal and tumor endothelium are highly related, sharing many endothelial cell specific markers. Second, it is equally clear that the endothelium derived from tumors is qualitatively different from that derived from normal tissues of the same type and is also different from primary endothelial cultures. Third, these genes are characteristically expressed in tumors derived from several different tissue types, documenting that tumor endothelium, in general, is different from normal endothelium. Fourth, the genes expressed differentially in tumor endothelium are also expressed during other angiogenic processes such as corpus luteum formation and wound healing. It is therefore more appropriate to regard the formation of new vessels in tumors as “neoangiogenesis” rather than “tumor angiogenesis” per se. This distinction is important from a variety of perspectives, and is consistent with the idea that tumors recruit vasculature using much of, or basically the same signals elaborated during other physiologic or pathological processes. That tumors represent “unhealed wounds” is one of the oldest ideas in cancer biology.
  • The nature and precise biological function of many of the Tumor Endothelial Markers (TEMs) identified here are unknown. Of the previously characterized genes shown in Table 2, it is intriguing that several encode proteins involved in extracellular matrix formation or remodelling (TEM 6, TEM 6, TEM 10, TEM 7, TEM 11, TEM 12, TEM 14, TEM 20, TEM 24, TEM 25, TEM 27, TEM 37, TEM 38, and TEM 40,) Deposition of extracellular matrix is likely critical to the growth of new vessels. Finally, it is perhaps not surprising that so many of the endothelial-specific transcripts identified here, whether expressed only in neovasculature or in endothelium in general, have not been previously characterized, and some are not even represented in EST databases. In part, this may be due to the fact that the EST databases are heavily biased toward certain tissues, but moreover, may be due to the fact that even in highly vascularized tissues endothelial cells are still a relatively small proportion of the population. Thus, the sensitivity of the SAGE method is a particularly appropriate tool.
  • Sequence and literature study has permitted the following identifications to be made among the family of TEM proteins. TEM proteins have been identified which contain transmembrane regions. These include TEM 1, TEM 3, TEM 9, TEM 13, TEM 17, TEM 19, TEM 22, TEM 30, and TEM 44. TEM proteins have been identified which are secreted proteins, including TEM 4, TEM 6, TEM 7, TEM 10, TEM 12, TEM 14, TEM 20, TEM 25, TEM 27, TEM 31, TEM 36, TEM 37, TEM 38, and TEM 39. HeyL (TEM 8) is a transcription factor which may be involved in regulating TEMs as one or more groups. The protein corresponding to the tag for TEM44 was found in the public databases, but no biological function has yet been ascribed to it.
  • TEM 1 has been named endosialin in the literature. It has a signal sequence at amino acids 1-17 and a transmembrare domain at amino acids 686-708. Thus it is a cell surface protein. Its extracellular domain is at resiudes 1-685. Endosialin may be involved in endocytosis. The mouse ortholog is predicted to have a signal peptide at residues 1-21.
  • TEM 2 is a dexamethasone induced, ras related protein homolog of 266 amino acids. It has neither a signal sequence nor a transmembrane domain. Thus it is neither a cell surface nor a secreted protein. TEM 2 plays a role in signal transduction. It regulates alterations in cell morphology, proliferation, and cell-extracellular matrix interactions.
  • TEM 3 (originally termed TEM 7R) has both a signal sequence (at residues 1-24 or 1-30) and a transmembrane domain (at residues 456-477). Thus it is a cell surface protein. The portion of the protein which is extracellular is at amino acids 1-455. TEM 3 has domains with homology to integrins, plexin, and adhesion molecules. TEM 3 may regulate GTPases that control signal transduction pathways linking plasma membrane receptors to the actin cytoskeleton. In the mouse ortholog, the signal peptide is predicted to be residues 1-30.
  • TEM 4 is also known as DKK-3. It has a signal sequence (residues1-16), suggesting that is a secreted protein. TEM 4 regulates wnt signaling, and it may be involved in vasculogenesis and wnt-dependent signaling for endothelial growth. TEM 4 is an inhibitor of Wnt oncogene and such inhibition can be determined by assay. Tsuji et al., Biochem. Biophys. Res. Comm. 268:20-4, 2000.
  • TEM 5 appears to be neither secreted nor a cell surface protein. TEM 5 appears to be a component of a G protein—GTPase signaling pathway.
  • TEM 6 is also known as stromelysin-3/Matrix metalloproteinase 11 (MMP-11). It has a signal sequence at residues 1-31, but no transmembrane domain. It has an alternative signal peptide splice site at residues 108-109. Thus it appears to be a secreted protein. TEM 6 belongs to the zinc metalloprotease family, also known as the matrixin subfamily. TEM 6 is expressed in most invasive carcinomas. Alpha 1-protease inhibitor is a natural substrate of MMP 11. TEM 6 degrades extracellular matrix proteins such as collagen and is involved in extracellular matrix remodeling and cell migration. Stromelysin can be assayed using a casein-resorufin substrate, for example. See Tortorella and Amer, Inflammation Research 46 Supp. 2:S 122-3, 1997.
  • TEM 7 is a protein of many names, also being known as matrix metalloproeinase 2, gelatinase A, and 72 KD type IV collagenase. TEM 7 has a signal sequence at residues 1-26 and is a secreted protein. Like TEM 6, TEM 7 belongs to the matrixin subfamily (zinc metalloproteinases). TEM 7 cleaves gelatin type I, collagen type I, IV, V VII and X. TEM 7 associates with integrin on the surface of endothelial cells and promotes vascular invasion. TEM 7 is involved in tissue remodeling. TEM 7 can be assayed using zymography or quenched fluorescent substrate hydrolysis, for example. Garbett, et al., Molecular Pathology 53:99-106, 2000. A fluorogenic matrix metalloproteinase substrate assay can also be used which employs methoxycoumarin containing septapeptide analog of the alpha2(I) collagen cleavage site. See Bhide et al., J. Periodontology 71:690-700, 2000.
  • TEM 8 is HEYL protein. It has neither a signal sequence nor a transmembrane domain. It is related to the hairy/Enhancer of split genes. TEM 8 is likely a nuclear protein, having a role as a transcription factor. TEM 8 belongs to a new class of Notch signal transducers and plays a key role in various developmental processes, such as vascular development, somatogenesis and neurogenesis. SNP's at residues 615 and 2201 have Cytosine bases. Notch 3 mutations underlie the CADASIL vascular disorder. See Mech Dev 2000 November; 98 (1-2):175
  • TEM 9 is a G-protein coupled receptor homolog, having both a signal sequence at residues 1-26 and 7 transmembrane domains. Thus it is a cell surface protein. Its extracellular region resides in amino acids 1-769. Its transmembrane domains are at residues 817-829 (TM2 and TM3), residues 899-929 (TM4 and TM5), and residues 1034-1040 (TM6 and TM7). TEM 9 acts as a G-protein coupled receptor with extracellular domains characteristic of cell adhesion proteins. One of its splice variants may function as a soluble receptor. TEM 9 may regulate cell polarity and cell migration. It may be involved in exocytosis based on latrophilin function. The mouse ortholog has a predicted signal peptide at residues 1-29.
  • TEM 10 is collagen type I, alpha2 (COL1A2), which has a signal sequence at residues 1-22. It is an extracellular matrix (ECM) protein which is secreted subsequent to synthesis. TEM 10 interacts with a number of proteins including other ECM proteins, certain growth factors, and matrix metalloproteases. TEM 10 is required for the induction of endothelial tube formation and is involved in tissue remodeling. A variant at nucleotide 3233 which substitutes an A, is associated with osteogenesis imperfecta type IV. A variant at nucleotide 4321 substituting an A retains a wild type phenotype. Nucleotide 715 is a site of a polymorphism. Nucleotides 695-748 are deleted in Ehlers-Danos syndrome. Other mutations are associated with idiopathic osteoporosis, and atypical Marfan syndrome. Variants are known at nucleotides 226(T,C), 314(A,C), 385(T,C), 868 (G,A), 907(C,T), 965(A,G), 970(T,A), 1784 (G,C), 2017(T,G), 2172(C,A), 2284(T,C), 2308(T,C), 2323(T,G), 2344(T,G), 2604(G,A), 2974(A,T), 2903(A,G), 2995(C,T), 3274(C,T), 3581(A,C), 3991(A,C), 4201(G,T), 4434(C,T), 4551(A,C), 4606(C,A), 4947(T,C), 4978(C,T), 4982(G,T), 5051(G,T). PolyA sites are located at nucleotides 4450, 4550, 4885, and 5082. PolyA signals are located at 4420-4424, 4515-4520, 4529-4534, 4866-4871, 5032-5037, 5053-5058. TEM 10, 20, and 40 derive from the same gene but are different isoforms having different lengths.
  • TEM 11 is Nidogen/Entactin. It is a secreted protein which has a signal sequence at residues 1-28. TEM 11 is an extracellular matrix protein which is a component of a basement membrane. TEM 11 binds to laminin and collagen IV and other extracellular matrix proteins. TEM 11 regulates capillary formation and is involved in tissue remodelling. Variations have been observed at nucleotides 4265(T,C), 4267(G,C,T), and 4738(T,G). Nidogen can be assayed by its effect on the morphology of astrocytes. See Grimpe et al., GLIA 28:138-49, 1999.
  • TEM 12 is the alpha 3 chain of collagen type VI. It has a signal sequence at residues 1-25. A secreted protein, TEM 12 is an extracellular matrix protein. TEM 12 has a splice variant. TEM 12 is a major constituent of vascular subendothelium and is involved in tissue remodeling. It regulates platelet activation and aggregation. Alternatively spliced domains are located at nucleotides 347-964, 965-1567, 2153-3752, and 4541-5041.
  • TEM 13 is also known as Thy-1 glycoprotein. It has both a signal sequence (at residues 1-19) and a transmembrane domain (at residues 143-159). Residues 131-161 are removed in a matured form of the protein. The extracellular region of the protein is residues 1-142 or residues 1-130. TEM 13 has a glycosyl phosphatidylinositol (GPI) anchor at residue 130 anchoring it to the membrane. TEM 13 is detectable in its soluble form in human serum. TEM 13 is reported to be a marker for activated endothelial cells (a marker of adult but not embryonic angiogenesis). TEM 13 on vascular endothelial cells may function as a possible vascular permeability modulator. Antibody to Thy-1 is a mitogenic signal for the CD4+CD45+ and CD8+CD45+ cells, but fails to induce proliferation in the CD45− T cells. Pingel et al., International Immunology 6:169-78, 1994. Thy-1 can be assayed as an inhibitor of such signal.
  • TEM 14 is also known as cystatin S. It is a secreted protein with a signal sequence at residues 1-20 and an extracellular region at residues 1-141. It is a cysteine protease inhibitor. TEM 14 may regulate cysteine protease function involved in angiogenesis and tissue remodeling. TEM14 is an inhibitor of the activity of papain and such inhibition can be assayed. Hiltke et al., J. Dental Research 78:1401-9, 1999.
  • TEM 15 is collagen type III, alpha 1 (COL3A1). It has a signal sequence (residues 1-23) and is secreted. Type III collagen binds to von Willebrand factor. It is involved in cell-cell adhesion, proliferation, and migration activities. Variants at nucloetides 2104(C,A), 2194(G,A), 2346(C,T), 2740(C,T), 3157(T), 3468(G), 3652(T), 3666(C), 3693(C), 3755(G), 3756(T), 3824(C), 4546(A, G), 4661(G), 4591(C,T), 4665(C), 5292(C), 5293(C), and 5451 (A) have been observed.
  • TEM 16 is a tensin homolog which is apparently an intracellular protein. It may have splice variants or isoforms. One form with 1704 amino acids has a region at the N-terminal domain which is similar to a tumor suppressor protein, phosphatase and tensin homolog (PTEN). Tensin is a focal adhesion molecule that binds to actins and phosphorylated proteins. It is involved in cell migration linking signal tranduction pathways to the cytoskeleton. PTEN regulates tumor induced angiogenesis.
  • TEM 17 (BSC-TEM 7) has a signal sequence which includes residues 1-18 and a transmembrane domain at residues 427-445. It is a cell surface marker with an extracellular region comprising residues 1-426. It has homologs in both mouse and C. elegans. Residues 137-244 share weak homology with nidogen; residues 280-344 share homology to PSI domains found in plexin, semaphorins and integrin beta subunits. Variants have been observed at nucleotides 1893(A,G), 1950(C,G), 2042(A,G), and 2220(G,A). In mouse TEM 17 the signal sequence includes residues 1-19.
  • TEM 19 was originally reported to be tumor endothelial marker 8, i.e., BSC-TEM 8. It has a signal sequence at residues 1-27 and a transmembrane domain at residues 322-343. It is a cell surface protein having an extracellular region at residues 1-321. TEM 19 has a von Willebrand Factor (vWF) A domain at residues 44-216; a domain at residues 34-253 which is found in leukointegrin alpha D chain; and a domain at residues 408-560 found in PRAM-1 or adaptor molecule-1 of the vinculin family. TEM 19's function is adhesion related. von Willebrand Factor domains are typically involved in a variety of functions including vascular processes. TEM 19 may play a role in the migration of vascular endothelial cells. The mouse ortholog has a predicted signal peptide at residues 1-27.
  • TEM 20 is collagen type I, alpha 2 (COL1A2). It has a signal sequence at residues 1-22 and is a secreted extracellular matrix protein. TEM 20 induces endothelial tube formation in vitro and is involved in tissue remodeling. Variants have been observed at nucleotides 226(T,C), 314(A,C), 385(T,C), 868 (G,A), 907(C,T), 965(A,G), 970(T,A), 1784(G,C), 2017(T,G), 2172(C,A), 2284(T,C), 2308(T,C), 2323(T,G), 2344(T,G), 2604(G,A), 2794(A,T), 2903(A,G), 2995(C,T), 3274(C,T), 3581(A,C), 3991(A,C), 4201(G,T), 4434(C,T), 4551(A,C), 4606(C,A), 4895-4901 (--, GGACAAC), 4947(T,C), 4978(C,T), 4982(G,T), 5051(G,T).
  • TEM 21 is a Formin-like protein homolog which is an intracellular protein. Formin related proteins interact with Rho family small GTPases, profilin, and other actin associated proteins. Formin-binding proteins bind to FH1 domains with their WW domains. TEM 21 has a proline rich FH1 domain at residues 221-449. Formin related proteins play crucial roles in morphogenesis, cell polarity, cytokinesis and reorganization of the actin cytoskeleton. They may also regulate apoptosis, cell adhesion and migration.
  • TEM 22 is an endocytic receptor in the macrophage mannose receptor family. It has both a signal sequence at residues 1-30 and a transmembrane domain at residues 1415-1435, and resides on the cell surface. Its extracellular domain is amino acids 1-1414. TEM 22 may be present as a soluble (secreted) form and act as an inhibitor. It may bind secreted phopholipase A2 (sPLA2) and mediate biological responses elicited by sPLA2. TEM 22 may have endocytic properties for sPLA2 and mediate endocytosis for endothelial related proteins. It may promote cell adhesion and be involved in cell-cell communication. Variations have been observed at nucleotide 5389 (A, G). TEM 22 mediates uptake of micro-organisms and host-derived glycoproteins. Groger et al., J. Immunology 165:5428-34, 2000.
  • TEM 24 is tensin, an intracellular protein. It is a focal adhesion molecule that binds to actin filaments and interacts with phosphotyrosine containing proteins. It may mediate kinase signaling activities and regulate cellular transformation. Variations have been observed at nucleotides 2502 (A, G), 2622(A, G), 6027(A, G). TEM24 binds to actin filaments and interacts with phosphotyrosine-containing proteins. Chen et al., Biochem. J. 351 Pt 2:403-11, 2000. TEM24 also binds to phosphoinositide3-kinase. Auger et al., J. Bio. Chem. 271:23452-7, 1996 TEM 24 also binds to nuclear protein p130. Lo et al., Bioassays 16:817-23, 1994.
  • TEM 25 is Bone morphogenic protein 1 (BMP-1) which has a signal sequence at residues 1-22. It is a secreted protein. There are at least 6 isoforms of BMP-1 as well as splice variants which add carboxy terminal CUB domains and an additional EGF domain. TEM 25 is a metalloprotease enzyme. It cleaves the C-terminal propeptide of collagen type I, II and III and laminin 5 gamma 2, proteins that are important for vascular processes. It is involved in cartilage formation. Variations have been observed at nucleotides 3106(C,T), 3248(G,A), 3369(G,A). TEM 25 cleave probiglycan at a single site, removing the propeptide and producing a biglycan molecule with an NH(2) terminus identical to that of the mature form found in tissues. Sctt et al., J. Biol. Chem. 275:30504-11, 2000. Laminin alpha 3 and gamma2 short chains are substrates of TEM 25. Amano et al., J. Biol. Chem. 275:22728-35, 2000.
  • TEM 27 is known as Slit homolog 3, a secreted protein with a signal sequence at residues 1-27. TEM 27 is a secreted guide protein involved in migration, repulsion and patterning. It interacts with “round about” receptors (Robo receptors). TEM 27 may interact with extracellular matrix (ECM) proteins and is involved in cell adhesion. Variations have been observed at nucleotides 4772 (C,T)
  • TEM 28 is similar to mouse nadrin (neuron specific GTPase activating protein). TEM 28 is an intracellular protein with a RhoGAP domain. The RhoGAP domain activates RhoA, Rac1, and Cdc42 GTPases. It is involved in the reorganization of actin filaments and enhancing exocytosis. It may also be involved in cell signalling. Variations have been observed at nucleotide 3969 (A,C),
  • TEM 29 is protein tyrosine phosphatase type IVA, member 3, isoform 1, an intracellular protein. It has alternate splice variants. TEM 29 belongs to a small class of prenylated protein tyrosine phosphatases (PTPs). It may be membrane associated by prenylation. PTPs are cell signaling molecules and play regulatory roles in a variety of cellular processes and promote cell proliferation. PTP PRL-3 regulates angiotensin-II induced signaling events.
  • TEM 30 is integrin alpha 1, a cell surface protein having both a signal sequence (residues 1-28) and a transmembrane domain (residues 1142-1164). Its extracellular region includes amino acids 1-1141. TEM 30 is a receptor for laminin and collagen. It mediates a variety of adhesive interactions. TEM 30 is abundantly expressed on microvascular endothelial cells. It stimulates endothelial cell proliferation and vascularization. TEM 30 may regulate angiostatin production. Variations have been observed at nucleotide 418 (C,T). TEM 30 activates the Ras/Shc/mitogen-activated protein kinase pathway promoting fibroblast cell proliferation. It also acts to inhibit collagen and metalloproteinase synthesis. Pozzi et al., Proc. Nat. Acad. Sci. USA 97:2202-7, 2000,
  • TEM 31 is Collagen IV alpha 1 (COL4A1) a secreted protein with a at residues 1-27. TEM 31 is a component of the basement membrane. It binds to alpha3 beta lintegrin and promotes integrin mediated cell adhesion. Non-collagenous domains of type IV subunits are involved in tumoral angiogenesis. TEM 31 is involved in tissue remodeling. Variations have been observed at nucleotide 4470 (C,T)
  • TEM 33 is methylmalonyl Co-A Mutase a protein which is localized in the mitochondrial matrix. It degrades several amino acids, odd-numbered-acid fatty acids, and cholesterol to the tricarboylic acid cycle. A defect in TEM 33 causes a fatal disorder in organic acid metabolism termed methylmalonic acidurea. Variations have been observed at nucleotides 1531(G,A), 1671(G,A), 2028(T,C), 2087(G,A), 2359(A,G), 2437(C,A), 2643(G,C), 2702(G,C). TEM 33 converts L-methylmalonyl CoA to succinyl CoA. This reaction can be assayed as is known in the art. See, e.g., Clin. Chem. 41(8 Pt I): 1164-70, 1995.
  • TEM 36 is collagen type XII, alpha1 (COL12A1), an extracellular matrix protein having a signal sequence at residues 1-23 or 24. TEM 36 has von Willebrand Factor (vWF) type A domains, Fibronectin type III domains, and thrombospondin N-terminal like domain. TEM 36 is expressed in response to stress environment. TEM 36 may organize extracellular matrix architecture and be involved in matrix remodeling. There are two isoforms of the protein, a long form and a short form. The short form is missing amino acids 25-1188, and therefore nucleotides 73 to 3564. Both forms share the signal sequence and are therefore both secreted.
  • TEM 37 is lumican, an extracellular matrix sulfated proteoglycan having a signal sequence at residues 1-18. Lumican interacts with proteins that are involved in matrix assembly such as collagen type I and type VI; it is involved in cell proliferation and tissue morphogenesis. Lumican plays an important role in the regulation of collagen fiber assembly. Variations have been observed at nucleotides 1021(G,T), 1035(A,G), 1209(A,G), 1259(A,C), 1418(C,A), 1519(T,A). TEM 37 is a binding partner of TGF-β. See FASEB J. 15:559-61, 2000. One assay that can be used to determine TEM 37 activity is a collagen fibril formation/sedimentation assay. Svensson et al., FEBS Letters 470:178-82, 2000.
  • TEM 38 is collagen type I, alpha 1 (COL1A1), an extracellular matrix protein having a signal sequence at residues 1-22. Type I collagen promotes endothelial cell migration and vascularization and induces tube formation and is involved in tissue remodelling. Telopeptide derivative is used as a marker for malignancy and invasion for certain cancer types. Variations have been observed at nucleotides 296(T,G), 1810(G,A), 1890(G,A), 2204(T,A), 3175(G,C), 3578(C,T), 4298(C,T), 4394(A,T), 4410(A,C), 4415(C.A), 4419 (A,T), 4528(C,A), 4572(G,T), 4602(T,C), 5529(T,C), 5670(C,T), 5985(C,T), 6012(C,T).
  • TEM 39 is transforming growth factor β-3 (TGF-beta3). It has a signal sequence at residues 1-23. It is a secreted protein. TEM 39 regulates cell growth and differentiation. TGF-beta isoforms play a major role in vascular repair processes and remodeling. Variations have been observed at nucleotide 2020(G,T).
  • TEM 41 is similar to Olfactomedin like protein. It appears to be an intracellular protein, having no obvious predicted signal sequence. Olfactomedin is the major glycoprotein of the extracellular mucous matrix of olfactory neuroepithelium. TEM 41 shares homology with latrophilin (extracellular regions) which has cell-adhesive type domains. TEM 41 may be involved in adhesive function.
  • TEM 42 is MSTP032 protein, a cell surface protein having a transmembrane domain at residues 42-61. Its function is unknown and it shares little homology with other proteins. Variations have been observed at nucleotides 418(A,T), 724(C,A).
  • TEM 44 is a hypothetical protein FLJ1190 (NM018354) which has two predicted transmembrane domains at residues 121-143 and 176-197. Residues 144-175 may form an extracellular region. TEM 44's function is not known and shares no homology to other known proteins.
  • TEM 45 is tropomyosin 1 (alpha), a protein which is intracellular. It forms dimers with a beta subunit. It influences actin function. TEM 45 may be involved in endothelial cell cytoskeletal rearrangement. Variations have been observed at nucleotides 509(A,C), 621(A,C), 635(T,G), 642(C,G), 1059(G,T).
  • TEM 46 is peanut-like 1 protein/septin 5, which belongs to the septin family. Proteins in the septin family bind to GTP and phosphatidylinositol 4,5-bisphosphate. They are involved in the signal tranduction cascades controlling cytokinesis and cell division.
  • NEM 4 is a member of the small inducible cytokine subfamily A (cys-cys), member 14 (SCYA14). NEM4 is a secreted protein characterized by two adjacent cysteine residues. One isoform lacks internal 16 amino acids compared to isoform 2.
  • NEM 22 shares homology with guanylate kinase-interacting protein 1Maguin-1. It is a membrane associated protein.
  • NEM 23 is human signaling lymphocytic activation molecule (SLAM). It has a signal sequence at residues 1-20. The extracellular domain may reside at residues 21-237. There is a secreted isoform of the protein.
  • NEM33 is netrin 4. It induces neurite outgrowth and promotes vascular development. At higher concentration, neurite outgrowth is inhibited.
  • 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 angiogenesis in the future. Genes which have been identified as tumor endothelial markers (TEMs) correspond to tags shown in SEQ ID NOS: 94-139, 173-176, 180-186. Genes which have been identified as normal endothelial markers (NEMs) correspond to tags shown in SEQ ID NOS: 140-172. Genes which have been identified as pan-endothelial markers (PEMs) i.e., expressed in both tumor and normal endothelial cells correspond to tags shown in SEQ ID NOS: 1-93. Genes which have been previously identified as being expressed predominantly in the endothelium correspond to PEM tags shown in SEQ ID NOS: 1-6, 8, 10-15. Markers in each class can be used interchangeably for some purposes.
  • 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. Tags disclosed herein, because of the way that they were made, represent sequences which are 3′ of the 3′ most restriction enzyme recognition site for the tagging enzyme used to generate the SAGE tags. In this case, the tags are 3′ of the most 3′ most NlaIII site in the cDNA molecules corresponding to mRNA. Nucleic acids corresponding to tags may be RNA, cDNA, or genomic DNA, for example. Such corresponding nucleic acids can be determined by comparison to sequence databases to determine sequence identities. Sequence comparisons can be done using any available technique, such as BLAST, available from the National Library of Medicine, National Center for Biotechnology Information. Tags can also be used as hybridization probes to libraries of genomic or cDNA to identify the genes from which they derive. Thus, using sequence comparisons or cloning, or combinations of these methods, one skilled in the art can obtain full-length nucleic acid sequences. Genes corresponding to tags will contain the sequence of the tag at the 3′ end of the coding sequence or of the 3′ untranslated region (UTR), 3′ of the 3′ most recognition site in the cDNA for the restriction endonuclease which was used to make the tags. 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 including 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 JRF, 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 JRF, Wang P, CG 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 stem 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, Gemmill R, Jakobovits A, Klapholz S. Analysis of the structural integrity of YACs comprising human immunoglobulin genes in yeast and in embryonic stem 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 (186 Re), 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 (Bateman 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 (Bateman 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 TEM: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 TEM of interest is administered to a subject. The immunogenic 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 TEM of interest may be delivered in an expressing cell, such as a purified population of tumor endothelial cells or a populations of fused tumor endothelial and dendritic cells. Nucleic acids encoding the TEM of interest may be delivered in a viral or non-viral delivery vector or vehicle. Non-human sequences encoding the human TEM of interest or other mammalian homolog can be used to induce the desired immunologic response in a human subject. For several of the TEMs of the present invention, mouse, rat or other ortholog sequences are described herein or 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. These include the human markers identified by SEQ ID NOS: 1-172, i.e., the normal, pan-endothelial, and the tumor endothelial markers. Homologous mouse markers include tumor endothelial markers of SEQ ID NO: 182-186 and 190-194. 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 tumor endothelial cells for diagnostic purposes, testing cells suspected of containing one or more TEMs. 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 TEMs, as well as for secreted TEMs. Intracellular and/or membrane associated TEMs 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 TEMs.
  • 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 screen 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 of 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 tumor endothelial cell populations, can be contacted with test substances and the expression of tumor endothelial markers and/or normal endothelial markers determined. Test substances which decrease the expression of tumor endothelial markers (TEMs) are candidates for inhibiting angiogenesis and the growth of tumors. Conversely, markers which are only expressed in normal endothelium but not in tumor endothelium (NEMs) can be monitored. Test substances which increase the expression of such NEMs in tumor endothelium and other human cells can be identified as candidate antitumor or anti-angiogenic drugs In cases where the activity of a TEM or NEM is known, agents can be screened for their ability to decrease or increase the activity.
  • For those tumor endothelial markers identified as containing transmembrane regions, it is desirable to identify drug candidates capable of binding to the TEM receptors found at the cell surface. For some applications, the identification of drug candidates capable of blocking the TEM receptor from its native ligand will be desired. For some applications, the identification of a drug candidate capable of binding to the TEM 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 TEM receptor:ligand complex, one may be able to promote or inhibit further development of endothelial cells and hence, vascularization.
  • For those tumor endothelial markers identified as being secreted proteins, it is desirable to identify drug candidates capable of binding to the secreted TEM protein. For some applications, the identification of drug candidates capable of interfering with the binding of the secreted TEM 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 TEM: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 which 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 TEM protein activity can also be used as a drug screen. Human and mouse TEMS can be used for this purpose.
  • 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 which 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. NEMs, can be used to restrict, diminish, reduce, or inhibit proliferation of tumor or other abnormal or undesirable vasculature. TEMs 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 TEMs can also be used to therapeutic benefit. For example, antibodies, T cells specific for a TEM, antisense to a TEM, and ribozymes specific for a TEM 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 TEMS 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 TEMs are disclosed in SEQ ID NO: 182-186 and 190-194. Mouse TEMs can be used as antigens for raising antibodies which can be tested in mouse tumor models. Mouse TEMs with transmembrane domains are particularly preferred for this purpose. Mouse TEMs 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. 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 Visualization of Vasculature of Colorectal Cancers
  • The endothelium of human colorectal cancer was chosen to address the issues of tumor angiogenesis, based on the high incidence, relatively slow growth, and resistance to anti-neoplastic agents of these cancers. While certain less common tumor types, such as glioblastomas, are highly vascularized and are regarded as good targets for anti-angiogenic therapy, the importance of angiogenesis for the growth of human colorectal cancers and other common solid tumor types is less well documented.
  • We began by staining vessels in colorectal cancers using von Willebrand Factor (vWF) as a marker. In each of 6 colorectal tumors, this examination revealed a high density of vessels throughout the tumor parenchyma (Examples in FIGS. 1A and B). Interestingly, these analyses also substantiated the importance of these vessels for tumor growth, as endothelium was often surrounded by a perivascular cuff of viable cells, with a ring of necrotic cells evident at the periphery (Example in FIG. 1A). Although these preliminary studies suggested that colon tumors are angiogenesis-dependent, reliable markers that could distinguish vessels in colon cancers from the vessels in normal colon are currently lacking. One way to determine if such markers exist is by analyzing gene expression profiles in endothelium derived from normal and neoplastic tissue.
  • EXAMPLE 2 Purification of Endothelial Cells
  • Global systematic analysis of gene expression in tumor and normal endothelium has been hampered by at least three experimental obstacles. First, endothelium is enmeshed in a complex tissue consisting of vessel wall components, stromal cells, and neoplastic cells, requiring highly selective means of purifying ECs for analysis. Second, techniques for defining global gene expression profiles were not available until recently. And third, only a small fraction of the cells within a tumor are endothelial, mandating the development of methods that are suitable for the analysis of global expression profiles from relatively few cells.
  • To overcome the first obstacle, we initially attempted to purify ECs from dispersed human colorectal tissue using CD31, an endothelial marker commonly used for this purpose. This resulted in a substantial enrichment of ECs but also resulted in contamination of the preparations by hematopoietic cells, most likely due to expression of CD31 by macrophages. We therefore developed a new method for purifying ECs from human tissues using P1H12, a recently described marker for ECs. Unlike CD31, P1H12 was specifically expressed on the ECs of both colorectal tumors and normal colorectal mucosa. Moreover, immunofluorescence staining of normal and cancerous colon with a panel of known cell surface endothelial markers (e.g. VE-cadherin, CD31 and CD34) revealed that P1H12 was unique in that it stained all vessels including microvessels (see FIG. 2A and data not shown). In addition to selection with P1H12, it was necessary to optimize the detachment of ECs from their neighbors without destroying their cell surface proteins as well as to employ positive and negative affinity purifications using a cocktail of antibodies (FIG. 2B). The ECs purified from normal colorectal mucosa and colorectal cancers were essentially free of epithelial and hematopoietic cells as judged by RT-PCR (FIG. 2C) and subsequent gene expression analysis (see below).
  • EXAMPLE 3 Comparison of Tumor and Normal Endothelial Cell Expression Patterns
  • To overcome the remaining obstacles, a modification of the Serial Analysis of Gene Expression (SAGE) technique was used. SAGE associates individual mRNA transcripts with 14 base pair tags derived from a specific position near their 3′ termini. The abundance of each tag provides a quantitative measure of the transcript level present within the mRNA population studied.
  • SAGE is not dependent on pre-existing databases of expressed genes, and therefore provides an unbiased view of gene expression profiles. This feature is particularly important in the analysis of cells that constitute only a small fraction of the tissue under study, as transcripts from these cells are unlikely to be well represented in extant EST databases. We adapted the SAGE protocol so that it could be used on small numbers of purified ECs obtained from the procedure outlined in FIG. 2B. A library of 100,000 tags from the purified ECs of a colorectal cancer, and a similar library from the ECs of normal colonic mucosa from the same patient were generated. These 193,000 tags corresponded to over 32,500 unique transcripts. Examination of the expression pattern of hematopoietic, epithelial and endothelial markers confirmed the purity of the preparations (FIG. 2D).
  • EXAMPLE 4 Markers of Normal and Tumor Endothelium
  • We next sought to identify Pan Endothelial Markers (PEMs), that is, transcripts that were expressed at significantly higher levels in both normal and tumor associated endothelium compared to other tissues. To identify such PEMs, tags expressed at similar levels in both tumor and normal ECs were compared to 1.8 million tags from a variety of cell lines derived from tumors of non-endothelial origin. This simple comparison identified 93 transcripts that were strikingly EC-specific, i.e. expressed at levels at least 20-fold higher in ECs in vivo compared to non-endothelial cells in culture. The 15 tags corresponding to characterized genes which were most highly and specifically expressed in endothelium are shown in Table 1A. Twelve of these 15 most abundant endothelial transcripts had been previously shown to be preferentially expressed in endothelium, while the other 3 genes had not been associated with endothelium in the past (Table 1A). These data sets also revealed many novel PEMs, which became increasingly prevalent as tag expression levels decreased (Table 1B). For many of the transcripts, their endothelial origin was confirmed by SAGE analysis of 401,000 transcripts derived from primary cultures of human umbilical vein endothelial cells (HUVEC) and human dermal microvascular endothelial cells (HMVEC) (Table 1A and B). To further validate the expression of these PEMs in vivo, we developed a highly sensitive non-radioactive in situ hybridization method that allowed the detection of transcripts expressed at relatively low levels in frozen sections of human tissues. Two uncharacterized markers, PEM3 and PEM6, were chosen for this analysis. In each case, highly specific expression was clearly limited to vascular ECs in both normal and neoplastic tissues (FIGS. 3 A and B and data not shown). These data also suggest that ECs maintained in culture do not completely recapitulate expression patterns observed in vivo. For example, Hevin and several other PEM's were expressed at high levels in both tumor and normal ECs in vivo, but few or no transcripts were detected in cultured HUVEC or HMVEC (Table 1). The source of the Hevin transcripts was confirmed to be endothelium by in situ hybridization in normal and malignant colorectal tissue (FIG. 3C).
  • Many of the markers reported in Table 1 were expressed at significantly higher levels than previously characterized genes commonly associated with ECs. For example, the top 25 markers were all expressed at greater than 200 copies per cell. In contrast, the receptors for VEGF (VEGFR-1 and VEGFR-2) were expressed at less than 20 copies per cell. Interestingly, VEGFR2 (KDR), which had previously been reported to be up-regulated in vessels during colon cancer progression, was found to be expressed in both normal and neoplastic colorectal tissue (FIGS. 3 D and E). The lack of specificity of this gene was in accord with the SAGE data, which indicated that the VEGFR was expressed at 12 copies per cell in both normal and tumor endothelium.
  • EXAMPLE 5 Tumor Versus Normal Endothelium
  • We next attempted to identify transcripts that were differentially expressed in endothelium derived from normal or neoplastic tissues. This comparison revealed 33 tags that were preferentially expressed in normal-derived endothelium at levels at least 10-fold higher than in tumor-derived endothelium. Conversely, 46 tags were expressed at 10-fold or higher levels in tumor vessels. Because those transcripts expressed at higher levels in tumor endothelium are most likely to be useful in the future for diagnostic and therapeutic purposes, our subsequent studies focussed on this class. Of the top 25 tags most differentially expressed, 12 tags corresponded to 11 previously identified genes, one with an alternative polyadenylation site (see Table 2). Of these 10 genes, 6 have been recognized as markers associated with angiogenic vessels. The remaining 14 tags corresponded to uncharacterised genes, most of which have only been deposited as ESTs (Table 2).
  • To validate the expression patterns of these genes, we chose to focus on 9 Tumor Endothelial Markers (BSC-TEM 1-9; TEM 1, 2, 5, 9, 16, 17, 19, and 22) for which EST sequences but no other information was available (Table 2). These tags were chosen simply because they were among the most differentially expressed on the list and because we were able to obtain suitable probes. In many cases, this required obtaining near full-length sequences through multiple rounds of sequencing and cDNA walking (See accession numbers in Table 2). RT-PCR analysis was then used to evaluate the expression of the corresponding transcripts in purified ECs derived from normal and tumor tissues of two patients different from the one used to construct the SAGE libraries. As shown in FIG. 4 A, the vWF gene, expected to be expressed in both normal and tumor endothelium on the basis of the SAGE data as well as previous studies, was expressed at similar levels in normal and tumor ECs from both patients, but was not expressed in purified tumor epithelial cells. As expected, PEM2 displayed a pattern similar to vWF. In contrast, all 9 TEMs chosen for this analysis were prominently expressed in tumor ECs, but were absent or barely detectable in normal ECs (Table 3 and examples in FIG. 4A). It is important to note that these RT-PCR assays were extremely sensitive indicators of expression, and the absence of detectable transcripts in the normal endothelium, combined with their presence in tumor endothelial RNAs even when diluted 100-fold, provides compelling confirmatory evidence for their differential expression. These results also show that these transcripts were not simply expressed differentially in the ECs of the original patient, but were characteristic of colorectal cancer endothelium in general.
  • It could be argued that the results noted above were compromised by the possibility that a small number of non-endothelial cells contaminated the cell populations used for SAGE and RT-PCR analyses, and that these non-endothelial cells were responsible for the striking differences in expression of the noted transcripts. To exclude this possibility, we performed in situ hybridization on normal and neoplastic colon tissue. In every case where transcripts could be detected (BSC-TEM 1, 3, 4, 5, 7, 8, and 9; TEM 1, 5, 9, 17, and 19), they were specifically localized to ECs (Table 3 and examples in FIGS. 4 B and C). Although caution must be used when interpreting negative in situ hybridization results, none of the TEMs were expressed in vascular ECs associated with normal colorectal tissue even though vWF and Hevin were clearly expressed (Table 3).
  • EXAMPLE 6 Tumor Endothelium Markers are Expressed in Multiple Tumor Types
  • Were these transcripts specifically expressed in the endothelium within primary colorectal cancers, or were they characteristic of tumor endothelium in general? To address this question, we studied the expression of a representative TEM (BSC-TEM7; TEM 17) in a liver metastasis from a colorectal cancer, a sarcoma, and in primary cancers of the lung, pancreas, breast and brain. As shown in FIG. 4, the transcript was found to be expressed specifically in the endothelium of each of these cancers, whether metastatic (FIG. 4D) or primary (FIG. 4E-1). Analysis of the other six TEMs, (BSC-TEM 1, 3, 4, 5, 7, 8 and 9; TEM 1, 5, 9, 17, and 19) revealed a similar pattern in lung tumors, brain tumors, and metastatic lesions of the liver (see Table 3).
  • EXAMPLE 7 Tumor Endothelium Markers are Neo-Angiogenic
  • Finally, we asked whether these transcripts were expressed in angiogenic states other than that associated with tumorigenesis. We thus performed in situ hybridizations on corpus luteum tissue as well as healing wounds. Although there were exceptions, we found that these transcripts were generally expressed both in the corpus luteum and in the granulation tissue of healing wounds (Table 3 and example in FIG. 4J). In all tissues studied, expression of the genes was either absent or exclusively confined to the EC compartment.
  • REFERENCES AND NOTES
  • The disclosure of each reference cited is expressly incorporated herein.
    • 1. J. Folkman, in Cancer Medicine J. Holland, Bast Jr, R C, Morton D L, Frei III, E, Kufe, D W, Weichselbaum, R R, Ed. (Williams & Wilkins, Baltimore, 1997) pp. 181.
    • 2. R. S. Kerbel, Carcinogenesis 21, 505 (2000).
    • 3. P. Wesseling, D. J. Ruiter, P. C. Burger, J Neurooncol 32, 253 (1997).
    • 4. Q. G. Dong, et al., Arterioscler Thromb Vasc Biol 17, 1599 (1997).
    • 5. P. W. Hewett, J. C. Murray, In Vitro Cell Dev Biol Anim 32, 462 (1996).
    • 6. M. A. Hull, P. W. Hewett, J. L. Brough, C. J. Hawkey, Gastroenterology 111, 1230 (1996).
    • 7. G. Haraldsen, et al., Gut 37, 225 (1995).
    • 8. The original EC isolation protocol was the same as that shown in FIG. 2B except that dispersed cells were stained with anti-CD31 antibodies instead of anti-P1H12, and magnetic beads against CD64 and CD14 were not included in the negative selection. After generating 120,000 SAGE tags from these two EC preparations, careful analysis of the SAGE data revealed that, in addition to endothelial-specific markers, several macrophage-specific markers were also present.
    • 9. A. Solovey, et al., N Engl J Med 337, 1584 (1997).
    • 10. V. E. Velculescu, L. Zhang, B. Vogelstein, K. W. Kinzler, Science 270, 484-487 (1995).
    • 11. In order to reduce the minimum amount of starting material required from ˜50 million cells to ˜50,000 cells (i.e. ˜1000-fold less) we and others (38) have introduced several modifications to the original SAGE protocol. A detailed version of our modified “MicroSAGE” protocol is available from the authors upon request.
    • 12. 96,694 and 96,588 SAGE tags were analyzed from normal and tumor derived ECs, respectively, and represented 50,298 unique tags. A conservative estimate of 32,703 unique transcripts was derived by considering only those tags observed more than once in the current data set or in the 134,000 transcripts previously identified in human transcriptomes (39).
    • 13. To identify endothelial specific transcripts, we normalized the number of tags analyzed in each group to 100,000, and limited our analysis to transcripts that were expressed at levels at least 20-fold higher in ECs than in non-endothelial cell lines in culture and present at fewer than 5 copies per 100,000 transcripts in non-endothelial cell lines and the hematopoietic fraction (˜57,000 tags)(41). Non-endothelial cell lines consisted of 1.8×106 tags derived from a total of 14 different cancer cell lines including colon, breast, lung, and pancreatic cancers, as well as one non-transformed keratinocyte cell line, two kidney epithelial cell lines, and normal monocytes. A complete list of PEMs is available at www.sagenet.org\angio\table1.htm.
    • 14. M. Tucci, et al., J Endocrinol 157, 13 (1998).
    • 15. T. Oono, et al., J Invest Dermatol 100, 329 (1993).
    • 16. K. Motamed, Int J Biochem Cell Biol 31, 1363 (1999).
    • 17. N. Bardin, et al., Tissue Antigens 48, 531 (1996).
    • 18. D. M. Bradham, A. Igarashi, R. L. Potter, G. R. Grotendorst, J Cell Biol 114, 1285 (1991).
    • 19. K. Akaogi, et al., Proc Natl Acad Sci USA 93,8384 (1996).
    • 20. Y. Muragaki, et al., Proc Natl Acad Sci USA 92, 8763 (1995).
    • 21. M. L. Iruela-Arispe, C. A. Diglio, E. H. Sage, Arterioscler Thromb 11, 805 (1991).
    • 22. J. P. Girard, T. A. Springer, Immunity 2, 113 (1995).
    • 23. E. A. Jaffe, et al., J Immunol 143, 3961 (1989).
    • 24. J. P. Girard, et al., Am J Pathol 155, 2043 (1999).
    • 25. H. Ohtani, N. Sasano, J Electron Microsc 36, 204 (1987).
    • 26. For non-radioactive in situ hybridization, digoxigenin (DIG)-labelled sense and anti-sense riboprobes were generated through PCR by amplifying 500-600 bp products and incorporating a T7 promoter into the anti-sense primer. In vitro transcription was performed using DIG RNA labelling reagents and T7 RNA polymerase (Roche, Indianapolis, Ind.). Frozen tissue sections were fixed with 4% paraformaldehyde, permeabilized with pepsin, and incubated with 200 ng/ml of riboprobe overnight at 55° C. For signal amplification, a horseradish peroxidase (HRP) rabbit anti-DIG antibody (DAKO, Carpinteria, Calif.) was used to catalyse the deposition of Biotin-Tyramide (from GenPoint kit, DAKO). Further amplification was achieved by adding HRP rabbit anti-biotin (DAKO), biotin-tyramide, and then alkaline-phosphatase (AP) rabbit anti-biotin (DAKO). Signal was detected using the AP substrate Fast Red TR/Napthol AS-MX (Sigma, St. Louis, Mo.), and cells were counterstained with hematoxylin unless otherwise indicated. A detailed protocol including the list of primers used to generate the probes can be obtained from the authors upon request.
    • 27. Transcript copies per cell were calculated assuming an average cell contains 300,000 transcripts.
    • 28. R. S. Warren, H. Yuan, M. R. Matli, N. A. Gillett, N. Ferrara, J Clin Invest 95, 1789 (1995).
    • 29. Y. Takahashi, Y. Kitadai, C. D. Bucana, K. R. Cleary, L. M. Ellis, Cancer Res 55, 3964 (1995).
    • 30. L. F. Brown, et al., Cancer Res 53, 4727 (1993).
    • 31. Endothelial-specific transcripts were defined as those expressed at levels at least 5-fold higher in ECs in vivo than in non-endothelial cell lines in culture (13), and present at no more than 5 copies per 100,000 transcripts in non-endothelial cell lines and the hematopoietic cell fraction (41). Transcripts showing statistically different levels of expression (P<0.05) were then identified using Monte Carlo analysis as previously described (40). Transcripts preferentially expressed in normal endothelium were then defined as those expressed at levels at least 10-fold higher in normal endothelium than in tumor endothelium. Conversely, tumor endothelial transcripts were at least 10-fold higher in tumor versus normal endothelium. See www.sagenet.org\angio\table2.htm and www.sagenet.org\angio\table3.htm for a complete list of differentially expressed genes.
    • 32. M. Turlaro, et al., Eur J Clin Invest 29, 793 (1999).
    • 33. W. S. Lee, et al., Circ Res 82, 845 (1998).
    • 34. J. Niquet, A. Represa, Brain Res Dev Brain Res 95, 227 (1996).
    • 35. L. Fouser, L. Iruela-Arispe, P. Bornstein, E. H. Sage, J Biol Chem 266, 18345 (1991).
    • 36. M. L. Iruela-Arispe, P. Hasselaar, H. Sage, Lab Invest 64, 174 (1991).
    • 37. H. F. Dvorak, N Engl J Med 315, 1650 (1986).
    • 38. B. Virlon, et al., Proc Natl Acad Sci USA 96, 15286 (1999).
    • 39. V. E. Velculescu, et al., Nat Genet 23, 387 (1999).
    • 40. L. Zhang, et al., Science 276, 1268 (1997).
    • 41. Human colon tissues were obtained within ½ hour after surgical removal from patients. Sheets of epithelial cells were peeled away from normal tissues with a glass slide following treatment with 5 mM DDT, then 10 mM EDTA, leaving the lamina propria intact. After a 2 h incubation in collagenase at 37° C., cells were filtered sequentially through 400 um, 100 um, 50 um and 25 um mesh, and spun through a 30% pre-formed Percoll gradient to pellet RBCs. Epithelial cells (Epithelial Fraction), which were found to non-specifically bind magnetic beads, were removed using Dynabeads coupled to BerEP4 (Dynal, Lake Success, N.Y.). Subsequently, macrophages and other leukocytes (Hematopoietic Fraction) were removed using a cocktail of beads coupled to anti-CD45, anti-CD 14 and anti-CD64 (Dynal). The remaining cells were stained with P1H12 antibody, purified with anti-mouse IgG-coupled magnetic beads, and lysed in mRNA lysis buffer. A detailed protocol can be obtained from the authors upon request.
    • 42. H. Sheikh, H. Yarwood, A. Ashworth, C. M. Isacke, J Cell Sci 113, 1021-32 (2000).
  • SEQ ID SEQ ID
    Sequence name NO: NO: Sequence name
    PEM 1 1 1 PEM 1
    PEM 2 2 2 PEM 2
    PEM 3 3 3 PEM 3
    PEM 4 4 4 PEM 4
    PEM 5 5 5 PEM 5
    PEM 6 6 6 PEM 6
    PEM 7 7 7 PEM 7
    PEM 8 8 8 PEM 8
    PEM 9 9 9 PEM 9
    PEM 10 10 10 PEM 10
    PEM 11 11 11 PEM 11
    PEM 12 12 12 PEM 12
    PEM 13 13 13 PEM 13
    PEM 14 14 14 PEM 14
    PEM 15 15 15 PEM 15
    PEM 16 16 16 PEM 16
    PEM 17 17 17 PEM 17
    PEM 18 18 18 PEM 18
    PEM 19 19 19 PEM 19
    PEM 20 20 20 PEM 20
    PEM 21 21 21 PEM 21
    PEM 22 22 22 PEM 22
    PEM 23 23 23 PEM 23
    PEM 24 24 24 PEM 24
    PEM 25 25 25 PEM 25
    PEM 26 26 26 PEM 26
    PEM 27 27 27 PEM 27
    PEM 28 28 28 PEM 28
    PEM 29 29 29 PEM 29
    PEM 30 30 30 PEM 30
    PEM 31 31 31 PEM 31
    PEM 32 32 32 PEM 32
    PEM 33 33 33 PEM 33
    PEM 34 34 34 PEM 34
    PEM 35 35 35 PEM 35
    PEM 36 36 36 PEM 36
    PEM 37 37 37 PEM 37
    PEM 38 38 38 PEM 38
    PEM 39 39 39 PEM 39
    PEM 40 40 40 PEM 40
    PEM 41 41 41 PEM 41
    PEM 42 42 42 PEM 42
    PEM 43 43 43 PEM 43
    PEM 44 44 44 PEM 44
    PEM 45 45 45 PEM 45
    PEM 46 46 46 PEM 46
    PEM 47 47 47 PEM 47
    PEM 48 48 48 PEM 48
    PEM 49 49 49 PEM 49
    PEM 50 50 50 PEM 50
    PEM 51 51 51 PEM 51
    PEM 52 52 52 PEM 52
    PEM 53 53 53 PEM 53
    PEM 54 54 54 PEM 54
    PEM 55 55 55 PEM 55
    PEM 56 56 56 PEM 56
    PEM 57 57 57 PEM 57
    PEM 58 58 58 PEM 58
    PEM 59 59 59 PEM 59
    PEM 60 60 60 PEM 60
    PEM 61 61 61 PEM 61
    PEM 62 62 62 PEM 62
    PEM 63 63 63 PEM 63
    PEM 64 64 64 PEM 64
    PEM 65 65 65 PEM 65
    PEM 66 66 66 PEM 66
    PEM 67 67 67 PEM 67
    PEM 68 68 68 PEM 68
    PEM 69 69 69 PEM 69
    PEM 70 70 70 PEM 70
    PEM 71 71 71 PEM 71
    PEM 72 72 72 PEM 72
    PEM 73 73 73 PEM 73
    PEM 74 74 74 PEM 74
    PEM 75 75 75 PEM 75
    PEM 76 76 76 PEM 76
    PEM 77 77 77 PEM 77
    PEM 78 78 78 PEM 78
    PEM 79 79 79 PEM 79
    PEM 80 80 80 PEM 80
    PEM 81 81 81 PEM 81
    PEM 82 82 82 PEM 82
    PEM 83 83 83 PEM 83
    PEM 84 84 84 PEM 84
    PEM 85 85 85 PEM 85
    PEM 86 86 86 PEM 86
    PEM 87 87 87 PEM 87
    PEM 88 88 88 PEM 88
    PEM 89 89 89 PEM 89
    PEM 90 90 90 PEM 90
    PEM 91 91 91 PEM 91
    PEM 92 92 92 PEM 92
    PEM 93 93 93 PEM 93
    TEM 1 94 94 TEM 1
    TEM 2 95 95 TEM 2
    TEM 3 96 96 TEM 3
    TEM 4 97 97 TEM 4
    TEM 5 98 98 TEM 5
    TEM 6 99 99 TEM 6
    TEM 7 100 100 TEM 7
    TEM 8 101 101 TEM 8
    TEM 9 102 102 TEM 9
    TEM 10 103 103 TEM 10
    TEM 11 104 104 TEM 11
    TEM 12 105 105 TEM 12
    TEM 13 106 106 TEM 13
    TEM 14 107 107 TEM 14
    TEM 15 108 108 TEM 15
    TEM 16 109 109 TEM 16
    TEM 17 110 110 TEM 17
    TEM 18 111 111 TEM 18
    TEM 19 112 112 TEM 19
    TEM 20 113 113 TEM 20
    TEM 21 114 114 TEM 21
    TEM 22 115 115 TEM 22
    TEM 23 116 116 TEM 23
    TEM 24 117 117 TEM 24
    TEM 25 118 118 TEM 25
    TEM 26 119 119 TEM 26
    TEM 27 120 120 TEM 27
    TEM 28 121 121 TEM 28
    TEM 29 122 122 TEM 29
    TEM 30 123 123 TEM 30
    TEM 31 124 124 TEM 31
    TEM 32 125 125 TEM 32
    TEM 33 126 126 TEM 33
    TEM 34 127 127 TEM 34
    TEM 35 128 128 TEM 35
    TEM 36 129 129 TEM 36
    TEM 37 130 130 TEM 37
    TEM 38 131 131 TEM 38
    TEM 39 132 132 TEM 39
    TEM 40 133 133 TEM 40
    TEM 41 134 134 TEM 41
    TEM 42 135 135 TEM 42
    TEM 43 136 136 TEM 43
    TEM 44 137 137 TEM 44
    TEM 45 138 138 TEM 45
    TEM 46 139 139 TEM 46
    NEM 1 140 140 NEM 1
    NEM 2 141 141 NEM 2
    NEM 3 142 142 NEM 3
    NEM 4 143 143 NEM 4
    NEM 5 144 144 NEM 5
    NEM 6 145 145 NEM 6
    NEM 7 146 146 NEM 7
    NEM 8 147 147 NEM 8
    NEM 9 148 148 NEM 9
    NEM 10 149 149 NEM 10
    NEM 11 150 150 NEM 11
    NEM 12 151 151 NEM 12
    NEM 13 152 152 NEM 13
    NEM 14 153 153 NEM 14
    NEM 15 154 154 NEM 15
    NEM 16 155 155 NEM 16
    NEM 17 156 156 NEM 17
    NEM 18 157 157 NEM 18
    NEM 19 158 158 NEM 19
    NEM 20 159 159 NEM 20
    NEM 21 160 160 NEM 21
    NEM 22 161 161 NEM 22
    NEM 23 162 162 NEM 23
    NEM 24 163 163 NEM 24
    NEM 25 164 164 NEM 25
    NEM 26 165 165 NEM 26
    NEM 27 166 166 NEM 27
    NEM 28 167 167 NEM 28
    NEM 29 168 168 NEM 29
    NEM 30 169 169 NEM 30
    NEM 31 170 170 NEM 31
    NEM 32 171 171 NEM 32
    NEM 33 172 172 NEM 33
    TEM 1 DNA 173 173 TEM 1 DNA
    TEM 2 DNA 174 174 TEM 2 DNA
    TEM 7 DNA 175 175 TEM 7 DNA
    TEM 8 DNA 176 176 TEM 8 DNA
    TEM 1 Protein 177 177 TEM 1 Protein
    TEM 2 Protein 178 178 TEM 2 Protein
    TEM 8 Protein 179 179 TEM 8 Protein
    TEM 5 DNA 180 180 TEM 5 DNA
    TEM 7B DNA 181 181 TEM 7B DNA
    mTEM 1 DNA 182 182 mTEM 1 DNA
    mTEM 5 DNA 183 183 mTEM 5 DNA
    mTEM 7 DNA 184 184 mTEM 7 DNA
    mTEM 7B DNA 185 185 mTEM 7B DNA
    mTEM 8 DNA 186 186 mTEM 8 DNA
    TEM 8 Protein 187 187 TEM 8 Protein
    TEM 5 Protein 188 188 TEM 5 Protein
    TEM 7B Protein 189 189 TEM 7B Protein
    mTEM 1 Protein 190 190 mTEM 1 Protein
    mTEM 5 Protein 191 191 mTEM 5 Protein
    mTEM 7 Protein 192 192 mTEM 7 Protein
    mTEM 7b Protein 193 193 mTEM 7b Protein
    mTEM 8 Protein 194 194 mTEM 8 Protein
    TEM 1 DNA 195 195 TEM 1 DNA
    TEM 1 Protein 196 196 TEM 1 Protein
    TEM 2 DNA 197 197 TEM 2 DNA
    TEM 2 Protein 198 198 TEM 2 Protein
    TEM 3 DNA 199 199 TEM 3 DNA
    TEM 3 Protein 200 200 TEM 3 Protein
    TEM 4 DNA 201 201 TEM 4 DNA
    TEM 4 Protein 202 202 TEM 4 Protein
    TEM 5 DNA 203 203 TEM 5 DNA
    TEM 5 Protein 204 204 TEM 5 Protein
    TEM 6 DNA 205 205 TEM 6 DNA
    TEM 6 Protein 206 206 TEM 6 Protein
    TEM 7 DNA 207 207 TEM 7 DNA
    TEM 7 Protein 208 208 TEM 7 Protein
    TEM 8 DNA 209 209 TEM 8 DNA
    TEM 8 Protein 210 210 TEM 8 Protein
    TEM 9 DNA 211 211 TEM 9 DNA
    TEM 9 Protein 212 212 TEM 9 Protein
    TEM 10 DNA 213 213 TEM 10 DNA
    TEM 10 Protein 214 214 TEM 10 Protein
    TEM 11 DNA 215 215 TEM 11 DNA
    TEM 11 Protein 216 216 TEM 11 Protein
    TEM 12 DNA 217 217 TEM 12 DNA
    TEM 12 Protein 218 218 TEM 12 Protein
    TEM 13 DNA 219 219 TEM 13 DNA
    TEM 13 Protein 220 220 TEM 13 Protein
    TEM 14a DNA 221 221 TEM 14a DNA
    TEM 14b DNA 222 222 TEM 14b DNA
    TEM 14a Protein 223 223 TEM 14a Protein
    TEM 14b Protein 224 224 TEM 14b Protein
    TEM 15 DNA 225 225 TEM 15 DNA
    TEM 15 Protein 226 226 TEM 15 Protein
    TEM 16 DNA 227 227 TEM 16 DNA
    TEM 16 Protein 228 228 TEM 16 Protein
    TEM 17 DNA 229 229 TEM 17 DNA
    TEM 17 Protein 230 230 TEM 17 Protein
    TEM 19 DNA 231 231 TEM 19 DNA
    TEM 19 Protein 232 232 TEM 19 Protein
    TEM 20 DNA 233 233 TEM 20 DNA
    TEM 20 Protein 234 234 TEM 20 Protein
    TEM 21 DNA 235 235 TEM 21 DNA
    TEM 21 Protein 236 236 TEM 21 Protein
    TEM 22 DNA 237 237 TEM 22 DNA
    TEM 22 Protein 238 238 TEM 22 Protein
    TEM 24 DNA 239 239 TEM 24 DNA
    TEM 24 Protein 240 240 TEM 24 Protein
    TEM 25 DNA 241 241 TEM 25 DNA
    TEM 25 Protein 242 242 TEM 25 Protein
    TEM 27 DNA 243 243 TEM 27 DNA
    TEM 27 Protein 244 244 TEM 27 Protein
    TEM 28 DNA 245 245 TEM 28 DNA
    TEM 28 Protein 246 246 TEM 28 Protein
    TEM 29 DNA 247 247 TEM 29 DNA
    TEM 29 Protein 248 248 TEM 29 Protein
    TEM 30 DNA 249 249 TEM 30 DNA
    TEM 30 Protein 250 250 TEM 30 Protein
    TEM 31 DNA 251 251 TEM 31 DNA
    TEM 31 Protein 252 252 TEM 31 Protein
    TEM 33 DNA 253 253 TEM 33 DNA
    TEM 33 Protein 254 254 TEM 33 Protein
    TEM 35 DNA 255 255 TEM 35 DNA
    TEM 35 Protein 358 256 TEM 36 DNA
    TEM 36 DNA 256 257 TEM 36 Protein
    TEM 36 Protein 257 258 TEM 37 DNA
    TEM 37 DNA 258 259 TEM 37 Protein
    TEM 37 Protein 259 260 TEM 38 DNA
    TEM 38 DNA 260 261 TEM 38 Protein
    TEM 38 Protein 261 262 TEM 39 DNA
    TEM 39 DNA 262 263 TEM 39 Protein
    TEM 39 Protein 263 264 TEM 40 DNA
    TEM 40 DNA 264 265 TEM 40 Protein
    TEM 40 Protein 265 266 TEM 41 DNA
    TEM 41 DNA 266 267 TEM 41 Protein
    TEM 41 Protein 267 268 TEM 42 DNA
    TEM 42 DNA 268 269 TEM 42 Protein
    TEM 42 Protein 269 270 TEM 44 DNA
    TEM 44 DNA 270 271 TEM 44 Protein
    TEM 44 Protein 271 272 TEM 45 DNA
    TEM 45 DNA 272 273 TEM 45 Protein
    TEM 45 Protein 273 274 TEM 46 DNA
    TEM 46 DNA 274 275 TEM 46 Protein
    TEM 46 Protein 275 276 NEM 4 DNA
    NEM 4 DNA 276 277 NEM 4 Protein
    NEM 4 Protein 277 278 NEM 14 DNA
    NEM 14 DNA 278 279 NEM 14 Protein
    NEM 14 Protein 279 280 NEM 17 DNA
    NEM 17 DNA 280 281 NEM 17 Protein
    NEM 17 Protein 281 282 NEM 22 DNA
    NEM 22 DNA 282 283 NEM 22 Protein
    NEM 22 Protein 283 284 NEM 23 DNA
    NEM 23 DNA 284 285 NEM 23 Protein
    NEM 23 Protein 285 286 NEM 23 Secreted
    NEM 23 Secreted 286 287 NEM 23 Short
    NEM 23 Short 287 288 NEM 33 DNA
    NEM 33 DNA 288 289 NEM 33 Protein
    NEM 33 Protein 289 290 mTEM 1 DNA
    mTEM 1 DNA 290 291 mTEM 1 Protein
    mTEM 1 Protein 291 292 mTEM 2 DNA
    mTEM 2 DNA 292 293 mTEM 2 Protein
    mTEM 2 Protein 293 294 mTEM 9 DNA
    mTEM 3 DNA 298 295 mTEM 9 Protein
    mTEM 3 Protein 299 296 mTEM 17 DNA
    mTEM 9 DNA 294 297 mTEM 17 Protein
    mTEM 9 Protein 295 298 mTEM 3 DNA
    mTEM 13 DNA 302 299 mTEM 3 Protein
    mTEM 13 Protein 303 300 mTEM 19 DNA
    mTEM 17 DNA 296 301 mTEM 19 Protein
    mTEM 17 Protein 297 302 mTEM 13 DNA
    mTEM 19 DNA 300 303 mTEM 13 Protein
    mTEM 19 Protein 301 304 mTEM 22 DNA
    mTEM 22 DNA 304 305 mTEM 22 Protein
    mTEM 22 Protein 305 306 mTEM 30 DNA
    mTEM 30 DNA 306 307 mTEM 30 Protein
    mTEM 30 Protein 307 308 TEM 2 tag
    TEM 2 tag 308 309 TEM 1 long tag
    TEM 1 long tag 309 310 TEM 3 long tag
    TEM 3 long tag 310 311 TEM 4 long tag
    TEM 4 long tag 311 312 TEM 5 long tag
    TEM 5 long tag 312 313 TEM 5 long tag
    TEM 5 long tag 313 314 TEM 6 long tag
    TEM 6 long tag 314 315 TEM 7 long tag
    TEM 7 long tag 315 316 TEM 8 long tag
    TEM 8 long tag 316 317 TEM 9 long tag
    TEM 9 long tag 317 318 TEM 10 long tag
    TEM 10 long tag 318 319 TEM 10 long tag
    TEM 10 long tag 319 320 TEM 10 long tag
    TEM 10 long tag 320 321 TEM 11 long tag
    TEM 11 long tag 321 322 TEM 12 long tag
    TEM 12 long tag 322 323 TEM 13 long tag
    TEM 13 long tag 323 324 TEM 13 long tag
    TEM 13 long tag 324 325 TEM 14 long tag
    TEM 14 long tag 325 326 TEM 15 long tag
    TEM 15 long tag 326 327 TEM 15 long tag
    TEM 15 long tag 327 328 TEM 16 long tag
    TEM 16 long tag 328 329 TEM 17 long tag
    TEM 17 long tag 329 330 TEM 19 long tag
    TEM 19 long tag 330 331 TEM 21 long tag
    TEM 21 long tag 331 332 TEM 21 long tag
    TEM 21 long tag 332 333 TEM 22 long tag
    TEM 22 long tag 333 334 TEM 22 long tag
    TEM 22 long tag 334 335 TEM 23 long tag
    TEM 23 long tag 335 336 TEM 24 long tag
    TEM 24 long tag 336 337 TEM 25 long tag
    TEM 25 long tag 337 338 TEM 25 long tag
    TEM 25 long tag 338 339 TEM 28 long tag
    TEM 28 long tag 339 340 TEM 30 long tag
    TEM 30 long tag 340 341 TEM 31 long tag
    TEM 31 long tag 341 342 TEM 32 long tag
    TEM 32 long tag 342 343 TEM 33 long tag
    TEM 33 long tag 343 344 TEM 33 long tag
    TEM 33 long tag 344 345 TEM 35 long tag
    TEM 35 long tag 345 346 TEM 36 long tag
    TEM 36 long tag 346 347 TEM 37 long tag
    TEM 37 long tag 347 348 TEM 38 long tag
    TEM 38 long tag 348 349 TEM 38 long tag
    TEM 38 long tag 349 350 TEM 39 long tag
    TEM 39 long tag 350 351 TEM 40 long tag
    TEM 40 long tag 351 352 TEM 41 long tag
    TEM 41 long tag 352 353 TEM 42 long tag
    TEM 42 long tag 353 354 TEM 43 long tag
    TEM 43 long tag 354 355 TEM 44 long tag
    TEM 44 long tag 355 356 TEM 45 long tag
    TEM 45 long tag 356 357 TEM 46 long tag
    TEM 46 long tag 357 358 TEM 35 Protein

Claims (86)

1. An isolated molecule comprising an antibody variable region which specifically binds to an extracellular domain of a TEM protein selected from the group consisting of: 1, 9, 17, 19, and 44, as shown in SEQ ID NO: 196, 212, 230, 232, and 271, respectively.
2. The isolated molecule of claim 1 which is an in tact antibody molecule.
3. The isolated molecule of claim 1 which is a single chain variable region (ScFv).
4. The isolated molecule of claim 1 which is a monoclonal antibody.
5. The isolated molecule of claim 1 which is a humanized antibody.
6. The isolated molecule of claim 1 which is a human antibody.
7. The isolated molecule of claim 1 which is bound to a cytotoxic moiety.
8. The isolated molecule of claim 1 which is bound to a therapeutic moiety.
9. The isolated molecule of claim 1 which is bound to a detectable moiety.
10. The isolated molecule of claim 1 which is bound to an anti-tumor agent.
11. A method of inhibiting neoangiogenesis, comprising:
administering to a subject in need thereof an effective amount of an isolated molecule comprising an antibody variable region which specifically binds to an extracellular domain of a TEM protein selected from the group consisting of: 1, 9, 17, 19, 22, and 44, as shown in SEQ ID NO: 196, 212, 230, 232, 238, and 271, respectively, whereby neoangiogenesis is inhibited.
12. The method of claim 11 wherein the subject bears a vascularized tumor.
13. The method of claim 11 wherein the subject has polycystic kidney disease.
14. The method of claim 11 wherein the subject has diabetic retinopathy.
15. The method of claim 11 wherein the subject has rheumatoid arthritis.
16. The method of claim 11 wherein the subject has psoriasis.
17. A method of inhibiting tumor growth, comprising:
administering to a human subject bearing a tumor an effective amount of an isolated molecule comprising an antibody variable region which specifically binds to an extracellular domain of a TEM protein selected from the group consisting of: 1, 9, 17, 19, 22, and 44, as shown in SEQ ID NO: 196, 212, 230, 232, 238, and 271, respectively, whereby growth of the tumor is inhibited.
18. An isolated molecule comprising an antibody variable region which specifically binds to a TEM protein selected from the group consisting of: 9, 17, 19, and 44, as shown in SEQ ID NO: 212, 230, 232, and 271, respectively.
19. The isolated molecule of claim 18 which is a single chain variable region (ScFv).
20. The isolated molecule of claim 18 which is a monoclonal antibody.
21. The isolated molecule of claim 18 which is a humanized antibody.
22. The isolated molecule of claim 18 which is a human antibody.
23. The isolated molecule of claim 18 which is bound to a cytotoxic moiety.
24. The isolated molecule of claim 18 which is bound to a therapeutic moiety.
25. The isolated molecule of claim 18 which is bound to a detectable moiety.
26. The isolated molecule of claim 18 which is bound to an anti-tumor agent.
27. The isolated molecule of claim 18 which is an in tact antibody molecule.
28. An isolated and purified human transmembrane protein selected from the group consisting of: TEM 9, 17, and 19 as shown in SEQ ID NO: 212, 230, and 232, respectively.
29. An isolated and purified nucleic acid molecule comprising a coding sequence for a transmembrane TEM selected from the group consisting of:: TEM 9, 17, and 19 as shown in SEQ ID NO: 212, 230, 232, respectively.
30. The isolated and purified nucleic acid molecule of claim 29 which comprises a coding sequence selected from those shown in SEQ ID NO: 211, 229, and 231.
31. A recombinant host cell which comprises a nucleic acid molecule comprising a coding sequence for a transmembrane TEM selected from the group consisting of: TEM 9, 17, and 19 as shown in SEQ ID NO: 212, 230, and 232, respectively.
32. The recombinant host cell of claim 31 which comprises a coding sequence selected from those shown in SEQ ID NO: 211, 229, and 231.
33. A method of inducing an immune response in a mammal, comprising:
administering to the mammal a nucleic acid molecule comprising a coding sequence for a human transmembrane protein selected from the group consisting of: TEM 1, 9, 13, 17, 19, 22, 30, and 44 as shown in SEQ ID NO: 196, 212, 220, 230, 232, 238, 250 and 271, respectively, whereby an immune response to the human transmembrane protein is induced in the mammal.
34. The method of claim 33 wherein the coding sequence is shown in SEQ ID NO: 195, 211, 219, 229, 231, 237, 249, 270.
35. A method of inducing an immune response in a mammal, comprising:
administering to the mammal a purified human transmembrane protein selected from the group consisting of: TEM 1, 9, 13, 17, 19, 22, 30, and 44 as shown in SEQ ID NO: 196, 212, 220, 230, 232, 238, 250 and 271, respectively, whereby an immune response to the human transmembrane protein is induced in the mammal.
36. A method for identification of a ligand involved in endothelial cell regulation, comprising:
contacting a test compound with an isolated and purified human transmembrane protein selected from the group consisting of 1, 9, 13, 17, 19, 30, and 44 as shown in SEQ ID NO: 196, 212, 220, 230, 250, 232 and 271;
contacting the isolated and purified human transmembrane protein with a molecule comprising an antibody variable region which specifically binds to an extracellular domain of a TEM protein selected from the group consisting of: 1, 9, 13, 17, 19, 30, and 44 as shown in SEQ ID NO: 196, 212, 220, 230, 250, 232 and 271, respectively;
determining binding of the molecule comprising an antibody variable region to the human transmembrane protein, wherein a test compound which diminishes the binding of the molecule comprising an antibody variable region to the human transmembrane protein is identified as a ligand involved in endothelial cell regulation.
37. A method for identification of a ligand involved in endothelial cell regulation, comprising:
contacting a test compound with a cell comprising a human transmembrane protein selected from the group consisting of 1, 9, 17, and 19 as shown in SEQ ID NO: 196, 212, 230, and 232;
contacting the cell with a molecule comprising an antibody variable region which specifically binds to an extracellular domain of a TEM protein selected from the group consisting of: 1, 9, 17, and 19 as shown in SEQ ID NO: 196, 212, 230, and 232, respectively;
determining binding of the molecule comprising an antibody variable region to the cell, wherein a test compound which diminishes the binding of the molecule comprising an antibody variable region to the cell is identified as a ligand involved in endothelial cell regulation.
38. A soluble form of a human transmembrane protein selected from the group consisting of: TEM 1, 9, 17, 19, 22, 30 and 44 as shown in SEQ ID NO: 196, 212, 230, 232, 238, 250, and 271, respectively, wherein the soluble forms lack transmembrane domains.
39. The soluble form of claim 38 wherein the soluble form consists of an extracellular domain of the human transmembrane protein.
40. A method of inhibiting neoangiogenesis in a patient, comprising:
administering to the patient a soluble form of a human transmembrane protein according to claim 38, whereby neoangiogenesis in the patient is inhibited.
41. A method of inhibiting neoangiogenesis in a patient, comprising:
administering to the patient a soluble form of a human transmembrane protein according to claim 39, whereby neoangiogenesis in the patient is inhibited.
42. The method of claim 40 wherein the patient bears a vascularized tumor.
43. The method of claim 41 wherein the patient bears a vascularized tumor.
44. The method of claim 40 wherein the patient has polycystic kidney disease.
45. The method of claim 40 wherein the patient has diabetic retinopathy.
46. The method of claim 40 wherein the patient has rheumatoid arthritis.
47. The method of claim 40 wherein the patient has psoriasis.
48. The method of claim 41 wherein the patient has polycystic kidney disease.
49. The method of claim 41 wherein the patient has diabetic retinopathy.
50. The method of claim 41 wherein the patient has rheumatoid arthritis.
51. The method of claim 41 wherein the patient has psoriasis.
52. A method of identifying regions of neoangiogenesis in a patient, comprising:
administering to a patient a molecule comprising an antibody variable region which specifically binds to an extracellular domain of a TEM protein selected from the group consisting of: 1, 9, 13, 17, 19, 22, 30, and 44, as shown in SEQ ID NO: 196, 212, 220, 230, 232, 238, 250, and 271, respectively, wherein the molecule is bound to a detectable moiety; and
detecting the detectable moiety in the patient, thereby identifying neoangiogenesis.
53. A method of screening for neoangiogenesis in a patient, comprising:
contacting a body fluid collected from the patient with a molecule comprising an antibody variable region which specifically binds to an extracellular domain of a TEM protein selected from the group consisting of: 1, 9, 17, 19, and 44, as shown in SEQ ID NO: 196, 212, 230, 232, and 271, respectively, wherein detection of cross-reactive material in the body fluid with the molecule indicates neoangiogenesis in the patient.
54. A method of screening for neoangiogenesis in a patient, comprising:
contacting a body fluid collected from the patient with a molecule comprising an antibody variable region which specifically binds to a TEM protein selected from the group consisting of: 4, 6, 7, 10, 12, 14, 25, 27, 31, 36, 37, 38, 39, as shown in SEQ ID NO: 202, 206, 208, 214, 218, 223 & 224, 242, 244, 252, 257, 259, 261, and 263, respectively, wherein detection of cross-reactive material in the body fluid with the molecule indicates neoangiogenesis in the patient.
55. A method of promoting neoangiogenesis in a patient, comprising:
administering to a patient in need of neoangiogenesis a TEM protein selected from the group consisting of: 4, 6, 7, 10, 12, 14, 20, 25, 27, 31, 36, 37, 38, 39, and 40, as shown in SEQ ID NO: 202, 206, 208, 214, 218, 223 & 224, 234, 242, 244, 252, 257, 259, 261. 263, and 265, whereby neoangiogenesis in the patient is stimulated.
56. A method of promoting neoangiogenesis in a patient, comprising:
administering to a patient in need of neoangiogenesis a nucleic acid molecule encoding a TEM protein selected from the group consisting of: 4, 6, 7, 10, 12, 14, 20, 25, 27, 31, 36, 37, 38, 39, and 40, as shown in SEQ ID NO: 202, 206, 208, 214, 218, 223 & 224, 234, 242, 244, 252, 257, 259, 261. 263, and 265, whereby the TEM protein is expressed and neoangiogenesis in the patient is stimulated.
57. A method of screening for neoangiogenesis in a patient, comprising:
detecting a TEM protein selected from the group consisting of: 4, 6, 7, 10, 12, 14, 20, 25, 27, 31, 36, 37, 38, 39, and 40, as shown in SEQ ID NO: 202, 206, 208, 214, 218, 223 & 224, 234, 242, 244, 252, 257, 259, 261. 263, and 265, respectively, in a body fluid collected from the patient, wherein detection of the TEM protein indicates neoangiogenesis in the patient.
58. A method of screening for neoangiogenesis in a patient, comprising:
detecting in a body fluid collected from the patient a nucleic acid encoding a TEM protein selected from the group consisting of: 4, 6, 7, 10, 12, 14, 20, 25, 27, 31, 36, 37, 38, 39, and 40, wherein the nucleic acid is selected from the group consisting of those shown in SEQ ID NO: 201, 205, 207, 213, 217, 221 & 222, 233, 241, 243, 251, 256, 258, 260, 262, and 264, respectively, wherein detection of the TEM protein indicates neoangiogenesis in the patient.
59. An isolated and purified nucleic acid molecule which encodes a NEM protein selected from the group consisting of: 14, 22, 23, and 33 as shown in SEQ ID NO: 279, 283, 285, 286, 287, and 289.
60. The nucleic acid molecule of claim 60 wherein the nucleic acid molecule comprises a coding sequence as shown in SEQ ID NO: 278, 282, 284, and 288.
61. A recombinant host cell which comprises a nucleic acid according to claim 60.
62. An isolated and purified NEM protein selected from the group consisting of: 14, 22, 23, and 33 as shown in SEQ ID NO: 279, 283, 285, 286, 287, and 289, respectively.
63. An isolated molecule comprising an antibody variable region which specifically binds to a NEM protein selected from the group consisting of: 14, 22, 23, and 33, as shown in SEQ ID NO: 279, 283, 285, 286, 287, and 289.
64. A method of inhibiting neoangiogenesis, comprising:
administering to a subject in need thereof an effective amount of a NEM protein selected from the group consisting of: 14, 22, 23, and 33 as shown in SEQ ID NO: 279, 283, 285, 286, 287, and 289, whereby neoangiogenesis is inhibited.
65. A method to identify candidate drugs for treating tumors, comprising:
contacting cells which express one or more TEM genes selected from the group consisting of: 1, 2, 4, 5, 6, 7, 8, 9, 10, 11, 12, 14, 15, 16, 17, 19, 20, 21, 22, 24, 25, 27, 28, 29, 30, 31, 33, 35, 36, 37, 38, 39, 41, 42, 44, 45, and 46 as shown in SEQ ID NO: 195, 197, 201, 203, 205, 207, 209, 211, 213, 215, 217, 219, 221 & 222, 225, 227, 229, 231, 233, 235, 237, 239, 241, 243, 245, 247, 249, 251, 253, 255, 256, 258, 260, 262, 266, 268, 270, 272, and 274, respectively, with a test compound;
determining expression of said one or more TEM genes by hybridization of mRNA of said cells to a nucleic acid probe which is complementary to said mRNA; and
identifying a test compound as a candidate drug for treating tumors if it decreases expression of said one or more TEM genes.
66. The method of claim 66 wherein the cells are endothelial cells.
67. The method of claim 66 wherein the cells are recombinant host cells which are transfected with an expression construct which encodes said one or more TEMs.
68. A method to identify candidate drugs for treating tumors, comprising:
contacting cells which express one or more TEM proteins selected from the group consisting of: 2, 4, 5, 6, 7, 8, 9, 10, 11, 12, 14, 15, 16, 17, 19, 20, 21, 22, 24, 25, 27, 28, 29, 30, 31, 33, 35, 36, 37, 38, 39, 41, 42, 44, 45, and 46 as shown in SEQ ID NO: 198, 202, 204, 206, 208, 210, 212, 214, 216, 218, 223 & 224, 226, 228, 230, 232, 234, 236, 238, 240, 242, 244, 246, 248, 250, 252, 254, 358, 257, 259, 261, 263, 267, 269, 271, 273, and 275, respectively, with a test compound;
determining amount of said one or more TEM proteins in said cells; and
identifying a test compound as a candidate drug for treating tumors if it decreases the amount of one more TEM proteins in said cells.
69. The method of claim 69 wherein the cells are endothelial cells.
70. The method of claim 69 wherein the cells are recombinant host cells which are transfected with an expression construct which encodes said one or more TEMs.
71. A method to identify candidate drugs for treating tumors, comprising:
contacting cells which express one or more TEM proteins selected from the group consisting of: 2, 4, 5, 6, 7, 8, 9, 10, 11, 12, 14, 15, 16, 17, 19, 20, 21, 22, 24, 25, 27, 28, 29, 40, 31, 33, 35, 36, 37, 38, 39, 41, 42, 44, 45, and 46 as shown in SEQ ID NO: 198, 202, 204, 206, 208, 210, 212, 214, 216, 218, 223 & 224, 226, 228, 230, 232, 234, 236, 238, 240, 242, 244, 246, 248, 250, 252, 254, 358, 257, 259, 261, 263, 267, 269, 271, 273, and 275 respectively, with a test compound;
determining activity of said one or more TEM proteins in said cells; and
identifying a test compound as a candidate drug for treating tumors if it decreases the activity of one more TEM proteins in said cells.
72. The method of claim 72 wherein the cells are endothelial cells.
73. The method of claim 72 wherein the cells are recombinant host cells which are transfected with an expression construct which encodes said one or more TEMs.
74. A method to identify candidate drugs for treating patients bearing tumors, comprising:
contacting a test compound with recombinant host cells which are transfected with an expression construct which encodes one or more TEM proteins selected from the group consisting of 2, 4, 5, 6, 7, 8, 9, 10, 11, 12, 14, 15, 16, 17, 19, 20, 21, 22, 24, 25, 27, 28, 29, 40, 31, 33, 35, 36, 37, 38, 39, 41, 42, 44, 45, and 46 as shown in SEQ ID NO: 198, 202, 204, 206, 208, 210, 212, 214, 216, 218, 223 & 224, 226, 228, 230, 232, 234, 236, 238, 240, 242, 244, 246, 248, 250, 252, 254, 358, 257, 259, 261, 263, 267, 269, 271, 273, and 275, respectively;
determining proliferation of said cells; and
identifying a test compound which inhibits proliferation of said cells as a candidate drug for treating patients bearing tumors.
75. A method to identify candidate drugs for treating tumors, comprising:
contacting cells which express one or more NEM genes selected from the group consisting of: 14, 22, 23, and 33 as shown in SEQ ID NO: 278, 282, 284, and 288, respectively, with a test compound;
determining expression of said one or more NEM genes by hybridization of mRNA of said cells to a nucleic acid probe which is complementary to said mRNA; and
identifying a test compound as a candidate drug for treating tumors if it increases expression of said one or more NEM genes.
76. The method of claim 76 wherein the cells are endothelial cells.
77. The method of claim 76 wherein the cells are recombinant host cells which are transfected with an expression construct which encodes said one or more NEMs.
78. A method to identify candidate drugs for treating tumors, comprising:
contacting cells which express one or more NEM proteins selected from the group consisting of: 14, 22, 23, and 33 as shown in SEQ ID NO: 279, 283, 285, 286, 287, and 289, with a test compound;
determining amount of said one or more NEM proteins in said cells; and
identifying a test compound as a candidate drug for treating tumors if it increases the amount of one more NEM proteins in said cells.
79. The method of claim 79 wherein the cells are endothelial cells.
80. The method of claim 79 wherein the cells are recombinant host cells which are transfected with an expression construct which encodes said one or more NEMs.
81. A method to identify candidate drugs for treating tumors, comprising:
contacting cells which express one or more NEM proteins selected from the group consisting of: 14, 22, 23, and 33 as shown in SEQ ID NO: 279, 283, 285, 286, 287, and 289, with a test compound;
determining activity of said one or more NEM proteins in said cells; and
identifying a test compound as a candidate drug for treating tumors if it increases the activity of one more NEM proteins in said cells.
82. The method of claim 82 wherein the cells are endothelial cells.
83. The method of claim 82 wherein the cells are recombinant host cells which are transfected with an expression construct which encodes said one or more NEMs.
84. A method to identify candidate drugs for treating patients bearing tumors, comprising:
contacting a test compound with recombinant host cells which are transfected with an expression construct which encodes one or more NEM proteins selected from the group consisting of 14, 22, 23, and 33 as shown in SEQ ID NO: 279, 283, 285, 286, 287, and 289;
determining proliferation of said cells; and
identifying a test compound which stimulates proliferation of said cells as a candidate drug for treating patients bearing tumors.
85. A method for identification of a ligand involved in endothelial cell regulation, comprising:
contacting a test compound with a human transmembrane TEM protein selected from the group consisting of 1, 2, 4, 5, 6, 7, 8, 9, 10, 11, 12, 14, 15, 16, 17, 19, 20, 21, 22, 24, 25, 27, 28, 29, 40, 31, 33, 35, 36, 37, 38, 39, 41, 42, 44, 45, and 46 as shown in SEQ ID NO: 196, 198, 202, 204, 206, 208, 210, 212, 214, 216, 218, 223 & 224, 226, 228, 230, 232, 234, 236, 238, 240, 242, 244, 246, 248, 250, 252, 254, 358, 257, 259, 261, 263, 267, 269, 271, 273, and 275;
determining binding of a test compound to the human transmembrane protein, wherein a test compound which binds to the protein is identified as a ligand involved in endothelial cell regulation.
86. A method of inducing an immune response in a mammal, comprising:
administering to the mammal a cell which expresses a transmembrane protein selected from the group consisting of: TEM 1, 9, 13, 17, 19, 22, 30, and 44 as shown in SEQ ID NO: 196, 212, 220, 230, 232, 238, 250 and 271, respectively, wherein the cell is a recombinant cell which comprises a vector encoding said transmembrane protein, or the cell is a of a dendritic cell and a tumor endothelium cell, whereby an immune response to the human transmembrane protein is induced in the mammal.
US11/930,528 2000-08-02 2007-10-31 Endothelial Cell Expression Patterns Abandoned US20090017030A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US11/930,528 US20090017030A1 (en) 2000-08-02 2007-10-31 Endothelial Cell Expression Patterns

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US22259900P 2000-08-02 2000-08-02
US22436000P 2000-08-11 2000-08-11
US28285001P 2001-04-11 2001-04-11
US09/918,715 US7402660B2 (en) 2000-08-02 2001-08-01 Endothelial cell expression patterns
US10/979,159 US7358351B2 (en) 2000-08-02 2004-11-03 Endothelial cell expression patterns
US11/930,528 US20090017030A1 (en) 2000-08-02 2007-10-31 Endothelial Cell Expression Patterns

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US10/979,159 Continuation US7358351B2 (en) 2000-08-02 2004-11-03 Endothelial cell expression patterns

Publications (1)

Publication Number Publication Date
US20090017030A1 true US20090017030A1 (en) 2009-01-15

Family

ID=27397112

Family Applications (3)

Application Number Title Priority Date Filing Date
US09/918,715 Expired - Lifetime US7402660B2 (en) 2000-08-02 2001-08-01 Endothelial cell expression patterns
US10/979,159 Expired - Lifetime US7358351B2 (en) 2000-08-02 2004-11-03 Endothelial cell expression patterns
US11/930,528 Abandoned US20090017030A1 (en) 2000-08-02 2007-10-31 Endothelial Cell Expression Patterns

Family Applications Before (2)

Application Number Title Priority Date Filing Date
US09/918,715 Expired - Lifetime US7402660B2 (en) 2000-08-02 2001-08-01 Endothelial cell expression patterns
US10/979,159 Expired - Lifetime US7358351B2 (en) 2000-08-02 2004-11-03 Endothelial cell expression patterns

Country Status (6)

Country Link
US (3) US7402660B2 (en)
EP (1) EP1307557A2 (en)
JP (3) JP2004527210A (en)
AU (1) AU2001283062A1 (en)
CA (1) CA2416732C (en)
WO (1) WO2002010217A2 (en)

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090176228A1 (en) * 2007-11-19 2009-07-09 Celera Corporation Lung cancer markers, and uses thereof
US20100021454A1 (en) * 2005-04-22 2010-01-28 Morphotek, Inc. Antibodies With Immune Effector Activity And That Internalize In Endosialin-Positive Cells
US20100260769A1 (en) * 2009-04-09 2010-10-14 Morphotek, Inc. Endosialin binding molecules
US20110033455A1 (en) * 2007-04-05 2011-02-10 Morphotek Inc, Methods For Inhibiting The Binding Of Endosialin To Ligands
US20130058935A1 (en) * 2010-05-11 2013-03-07 Fox Chase Cancer Center Antibodies to tumor endothelial marker 7r
WO2015006501A3 (en) * 2013-07-09 2015-03-12 President And Fellows Of Harvard College Microvessel endothelial cell surface markers and uses thereof
US10206941B2 (en) 2013-07-09 2019-02-19 President And Fellows Of Harvard College Venule endothelial cell genes and uses thereof

Families Citing this family (49)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998027932A2 (en) * 1996-12-20 1998-07-02 Human Genome Sciences, Inc. Cerebellum and embryo specific protein
ES2283343T3 (en) * 1999-12-23 2007-11-01 Kirin Beer Kabushiki Kaisha METHODS AND MATERIALS RELATED TO POLYPEPTIDES AND SIMILAR POLYNUCLEOTIDES TO THE MOTHER CELL GROWTH FACTOR.
AU2001239941A1 (en) 2000-02-29 2001-09-12 The General Hospital Corporation Beta netrin and uses thereof
AU2001283062A1 (en) 2000-08-02 2002-02-13 The Johns Hopkins University Endothelial cell expression patterns
WO2004005883A2 (en) * 2002-07-02 2004-01-15 The Johns Hopkins University Secreted and cytoplasmic tumor endothelial markers
EP1395657B1 (en) 2000-12-05 2007-04-18 Wisconsin Alumni Research Foundation Receptor for bacillus anthracis toxin
EP1345965A2 (en) * 2000-12-23 2003-09-24 Kirin Beer Kabushiki Kaisha Methods and materials relating to stem cell growth factor-like polypeptides and polynucleotides
US6969588B2 (en) 2001-02-08 2005-11-29 Applera Corporation Isolated human ras-like proteins, nucleic acid molecules encoding these human ras-like proteins, and uses thereof
JPWO2002064769A1 (en) * 2001-02-15 2004-06-17 塩野義製薬株式会社 New disease markers
CA2443995C (en) * 2001-04-11 2014-06-03 The Johns Hopkins University Endothelial cell expression patterns
JP2005516582A (en) * 2001-05-25 2005-06-09 アムジェン インコーポレーテッド Tumor endothelial marker 7α molecule and use thereof
US7262026B2 (en) 2001-08-23 2007-08-28 Japan Science And Technology Agency Hematopoietic stem cell proliferation regulators and polynucleotides encoding the same
JP2005530856A (en) * 2002-06-21 2005-10-13 ジョンズ ホプキンス ユニバーシティー スクール オブ メディシン Membrane-related tumor endothelial marker
JP2006512051A (en) * 2002-06-27 2006-04-13 セントカー・インコーポレーテツド CNGH0005 polypeptides, antibodies, compositions, methods and uses
US7364727B2 (en) * 2002-07-22 2008-04-29 Cell Genesys, Inc. Metastatic colon cancer specific promoter and uses thereof
EP1576131A4 (en) * 2002-08-15 2008-08-13 Genzyme Corp Brain endothelial cell expression patterns
US20060258560A1 (en) * 2002-09-30 2006-11-16 Chunlin Yang Dry tissue sealant compositions
EP1603587A1 (en) * 2003-03-04 2005-12-14 Genzyme Corporation Use of tumor endothelial markers 1, 9 and 17 to promote angiogenesis
KR20060003860A (en) * 2003-03-04 2006-01-11 기린 비루 가부시키가이샤 Endothelial cell specific antibodies and uses thereof
EP2311468B1 (en) 2003-08-08 2014-01-15 Perseus Proteomics Inc. Gene overexpressed in cancer
AU2005250188A1 (en) * 2004-06-02 2005-12-15 Valtion Teknillinen Tutkimuskeskus Method for activating of T cell protein tyrosine phosphatase and therapeutical methods based thereon
WO2006029045A2 (en) * 2004-09-03 2006-03-16 Kirin Brewery Co., Ltd. Endothelial cell specific antibodies and uses thereof
GB0423974D0 (en) * 2004-10-28 2004-12-01 Ares Trading Sa Proteins
EP2322206A3 (en) * 2004-11-22 2013-06-05 Centre National de la Recherche Scientifique Mutated netrin 1, fragments thereof and uses thereof as drugs
WO2006091326A1 (en) * 2005-01-28 2006-08-31 Attogen Inc. Anti-prl-3 antibodies and methods of use thereof
JP2010512327A (en) 2006-12-11 2010-04-22 ユニヴァーシティー オブ ユタ リサーチ ファウンデーション Compositions and methods for the treatment of pathological angiogenesis and vascular permeability
EP1947114A1 (en) * 2007-01-19 2008-07-23 INSERM (Institut National de la Santé et de la Recherche Médicale) Mutated netrin 4, fragments thereof and uses thereof as drugs
TW201425333A (en) * 2007-04-11 2014-07-01 Oncotherapy Science Inc TEM8 peptides and vaccines comprising the same
EA009327B1 (en) 2007-04-27 2007-12-28 Петр Генриевич ЛОХОВ Method for producing antineoplastic vaccine based on surface antigens of endothelial cells
WO2008142198A2 (en) * 2007-05-23 2008-11-27 Valtion Teknillinen Tutkimuskeskus A method for inhibiting or stimulating angiogenesis in an individual
NZ583851A (en) * 2007-09-14 2012-06-29 Formycon Gmbh Use of slit, nephrin, ephrin or semaphorin for treatment of cartilage diseases
EP2262524A4 (en) * 2008-04-16 2012-07-11 Univ Utah Res Found Compositions and methods for treating pathologic angiogenesis and vascular permeability
US9290556B2 (en) * 2008-09-29 2016-03-22 The Trustees Of The University Of Pennsylvania Tumor vascular marker-targeted vaccines
KR20180044433A (en) 2010-03-24 2018-05-02 알엑스아이 파마슈티칼스 코포레이션 Rna interference in dermal and fibrotic indications
US9095504B2 (en) 2010-03-24 2015-08-04 Rxi Pharmaceuticals Corporation RNA interference in ocular indications
FR2958936A1 (en) * 2010-04-14 2011-10-21 Sanofi Aventis ROBO1-FC FUSION PROTEIN AND ITS USE IN THE TREATMENT OF TUMORS
US9309322B2 (en) * 2010-11-12 2016-04-12 Scott & White Healthcare (Swh) Antibodies to tumor endothelial marker 8
US9181340B2 (en) 2011-06-14 2015-11-10 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services TEM8 antibodies, conjugates thereof, and their use
CN104105791A (en) * 2012-01-09 2014-10-15 苏州工业园区为真生物医药科技有限公司 Colorectal cancer diagnosis and indication marker
CN104136630B (en) * 2012-01-09 2017-04-05 苏州工业园区为真生物医药科技有限公司 Diagnosis and the mark of indication breast carcinoma
WO2014143884A2 (en) * 2013-03-15 2014-09-18 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Synthetic methylmalonyl-coa mutase transgene for the treatment of mut class methylmalonic acidemia (mma)
US9944918B2 (en) 2013-03-15 2018-04-17 The United States Of America, As Represented By The Secretary, Dept. Of Health & Human Services Synthetic methylmalonyl-CoA mutase transgene for the treatment of MUT class methylmalonic acidemia (MMA)
MX2016003744A (en) 2013-10-11 2016-08-11 Us Health Tem8 antibodies and their use.
CN104911168B (en) * 2015-06-30 2018-03-30 中国人民解放军第三军医大学第一附属医院 I-type collagen high-affinity polypeptide and its application
US10471122B2 (en) * 2015-07-31 2019-11-12 Blue Blood Biotech Corp. Composition for use in promoting wound healing
CN109310788A (en) * 2016-05-03 2019-02-05 法国国家健康和医学研究院 CD31SHEDInflammation imaging is used for as molecular target
GB2551591B (en) * 2016-06-23 2019-08-07 Tate & Lyle Tech Ltd Liquid-liquid extraction of DMF
SG11201909153UA (en) * 2017-04-10 2019-10-30 Immatics Biotechnologies Gmbh Peptides and combination thereof for use in the immunotherapy against cancers
KR102014467B1 (en) * 2017-12-14 2019-08-26 (주)녹십자웰빙 Cosmetic composition and pharmaceutical composition for improving atopic dermatis, alopetic, wound, or skin wrinkle

Citations (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5342757A (en) * 1992-11-13 1994-08-30 Ludwig Institute For Cancer Research Monoclonal antibodies which specifically binds to endosialin, a 165 Kd glycoprotein found on tumor vascular endothelium, and uses thereof
US5530101A (en) * 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
US5584458A (en) * 1995-07-18 1996-12-17 Levelite Technology, Inc. Quick-action mount for self-leveling laser
US5644034A (en) * 1989-08-07 1997-07-01 Peptide Technology Ltd. Tumour necrosis factor binding ligands
US5811522A (en) * 1994-03-08 1998-09-22 Ludwig Institute For Cancer Research Recombinant humanized anti-FB5 antibodies
US6117977A (en) * 1996-04-24 2000-09-12 Genentech, Inc. Type C lectins
US6130068A (en) * 1998-10-26 2000-10-10 Immunex Corporation Viral encoded semaphorin protein receptor DNA and polypeptides
US6612190B2 (en) * 1997-09-08 2003-09-02 Ngk Insulators, Ltd. Mass sensor and mass sensing method
US6657103B1 (en) * 1990-01-12 2003-12-02 Abgenix, Inc. Human antibodies derived from immunized xenomice
US7074913B2 (en) * 2000-12-05 2006-07-11 Wisconsin Alumni Research Foundation Receptor for B anthracis toxin

Family Cites Families (49)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPH04166095A (en) 1990-10-25 1992-06-11 Toshiba Corp Monoclonal antibody specifically binding to ascidian entactin, ascidian entactin gene and ascidian entactin
US5587458A (en) * 1991-10-07 1996-12-24 Aronex Pharmaceuticals, Inc. Anti-erbB-2 antibodies, combinations thereof, and therapeutic and diagnostic uses thereof
US5616483A (en) 1992-06-11 1997-04-01 Aktiebolaget Astra Genomic DNA sequences encoding human BSSL/CEL
GB9223707D0 (en) 1992-11-12 1992-12-23 B & H Nottingham Ltd Electrical connector
US5437665A (en) * 1993-10-12 1995-08-01 Munro; Malcolm G. Electrosurgical loop electrode instrument for laparoscopic surgery
US5611522A (en) * 1996-04-01 1997-03-18 Knight Industries, Inc. Hydraulically actuated portable hoist
EP0939805A2 (en) * 1996-10-04 1999-09-08 Genetics Institute, Inc. Secreted proteins and polynucleotides encoding them
WO1998037094A2 (en) 1997-02-24 1998-08-27 Genetics Institute, Inc. Secreted proteins and polynucleotides encoding them
AU6956698A (en) 1997-04-10 1998-10-30 Genetics Institute Inc. Secreted expressed sequence tags (sests)
WO1999021997A1 (en) 1997-10-28 1999-05-06 Immunex Corporation Viral encoded semaphorin protein receptor dna and polypeptides
SE9801424D0 (en) 1998-04-22 1998-04-22 Astra Ab Expression methods
DE19820190A1 (en) 1998-04-28 1999-11-04 Metagen Gesellschaft Fuer Genomforschung Mbh Human pancreatic tumor nucleic acid sequences
US6573095B1 (en) 1998-04-29 2003-06-03 Genesis Research & Development Corporation Limited Polynucleotides isolated from skin cells
WO2001053455A2 (en) 1999-12-23 2001-07-26 Hyseq, Inc. Novel nucleic acids and polypeptides
CA2383592A1 (en) * 1999-03-31 2000-10-05 Curagen Corporation 2384891 acids including open reading frames encoding polypeptides; orfx
JP2003529324A (en) 1999-06-15 2003-10-07 ジェネンテック・インコーポレーテッド Secreted and transmembrane polypeptides and nucleic acids encoding them
WO2001009318A1 (en) 1999-07-29 2001-02-08 Helix Research Institute Liver cancer-associated genes
EP1074617A3 (en) 1999-07-29 2004-04-21 Research Association for Biotechnology Primers for synthesising full-length cDNA and their use
AU7073800A (en) 1999-08-25 2001-03-19 Regents Of The University Of California, The Novel plexins and uses thereof
WO2001018542A2 (en) 1999-09-03 2001-03-15 Millennium Pharmaceuticals, Inc. Identification, assessment, prevention, and therapy of ovarian cancer
AU770157B2 (en) 1999-09-08 2004-02-12 Immunex Corporation Use of semaphorin for modulation of cellular efflux
WO2001017559A1 (en) 1999-09-08 2001-03-15 Immunex Corporation Semaphorin modulation of immune cell migration
WO2001019988A1 (en) 1999-09-17 2001-03-22 Genetics Institute, Inc. Secreted proteins and polynucleotides encoding them
JP4623825B2 (en) 1999-12-16 2011-02-02 協和発酵バイオ株式会社 Novel polynucleotide
AU2596901A (en) 1999-12-21 2001-07-03 Millennium Pharmaceuticals, Inc. Compositions, kits, and methods for identification, assessment, prevention, and therapy of breast cancer
US6380362B1 (en) 1999-12-23 2002-04-30 Genesis Research & Development Corporation Ltd. Polynucleotides, polypeptides expressed by the polynucleotides and methods for their use
WO2001052616A2 (en) 1999-12-23 2001-07-26 Hyseq, Inc. Novel nucleic acids and polypeptides
WO2001053312A1 (en) 1999-12-23 2001-07-26 Hyseq, Inc. Novel nucleic acids and polypeptides
ES2283343T3 (en) 1999-12-23 2007-11-01 Kirin Beer Kabushiki Kaisha METHODS AND MATERIALS RELATED TO POLYPEPTIDES AND SIMILAR POLYNUCLEOTIDES TO THE MOTHER CELL GROWTH FACTOR.
AU2001227385A1 (en) * 2000-01-21 2001-07-31 Hyseq, Inc. Methods and materials relating to stem cell growth factor-like poypeptides and polynucleotides
WO2001055168A1 (en) 2000-01-31 2001-08-02 Human Genome Sciences, Inc. Nucleic acids, proteins and antibodies
CA2395398A1 (en) 2000-01-31 2001-08-02 Human Genome Sciences, Inc. Nucleic acids, proteins, and antibodies
CA2393616A1 (en) 2000-01-31 2001-08-02 Human Genome Sciences, Inc. Nucleic acids, proteins, and antibodies
AU2001234944A1 (en) 2000-02-03 2001-08-14 Hyseq, Inc. Novel nucleic acids and polypeptides
AU3087801A (en) 2000-02-04 2001-08-14 Molecular Dynamics Inc Human genome-derived single exon nucleic acid probes useful for analysis of geneexpression in human breast and hbl 100 cells
AU6802801A (en) 2000-03-01 2001-09-24 Genentech Inc Secreted and transmembrane polypeptides and nucleic acids encoding the same
WO2001071042A2 (en) 2000-03-23 2001-09-27 Pe Corporation (Ny) Detection kits, such as nucleic acid arrays, for detecting the expression of 10,000 or more drosophila genes and uses thereof
US6436703B1 (en) 2000-03-31 2002-08-20 Hyseq, Inc. Nucleic acids and polypeptides
US20030027139A1 (en) 2000-04-06 2003-02-06 Kenneth Jacobs Polynucleotides encoding novel secreted proteins
AU2001274888A1 (en) 2000-05-19 2001-12-03 Human Genome Sciences, Inc. Nucleic acids, proteins, and antibodies
AU2001260847A1 (en) 2000-05-24 2001-12-03 Genesis Research And Development Corporation Limited Compositions isolated from skin cells and methods for their use
WO2001092523A2 (en) 2000-05-30 2001-12-06 Curagen Corporation Human polynucleotides and polypeptides encoded thereby
EP1358349A2 (en) 2000-06-05 2003-11-05 Avalon Pharmaceuticals Cancer gene determination and therapeutic screening using signature gene sets
AU2001273052A1 (en) 2000-06-29 2002-01-14 Anadys Pharmaceuticals, Inc. Antifungal compounds and methods of use
AU2002210791A1 (en) 2000-07-28 2002-02-13 Compugen Inc. Oligonucleotide library for detecting rna transcripts and splice variants that populate a transcriptome
CA2418377A1 (en) 2000-07-28 2002-02-07 The Administrators Of The Tulane Educational Fund Assay methods for identifying compounds which may protect stratified squamous epithelium against damage by noxious substances
AU2001283062A1 (en) 2000-08-02 2002-02-13 The Johns Hopkins University Endothelial cell expression patterns
AU2001290548A1 (en) 2000-09-11 2002-03-26 Hyseq, Inc. Novel nucleic acids and polypeptides
AU2001292802A1 (en) 2000-09-19 2002-04-02 Dana-Farber Cancer Institute Inc. Genetic markers for tumors

Patent Citations (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5530101A (en) * 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
US5644034A (en) * 1989-08-07 1997-07-01 Peptide Technology Ltd. Tumour necrosis factor binding ligands
US6657103B1 (en) * 1990-01-12 2003-12-02 Abgenix, Inc. Human antibodies derived from immunized xenomice
US5437865A (en) * 1992-11-13 1995-08-01 Memorial Sloan-Kettering Cancer Center Isolated sialylated glycoprotein endosialin, which is expressed by tumor associated vascular endothelium
US5342757A (en) * 1992-11-13 1994-08-30 Ludwig Institute For Cancer Research Monoclonal antibodies which specifically binds to endosialin, a 165 Kd glycoprotein found on tumor vascular endothelium, and uses thereof
US6391302B1 (en) * 1994-03-08 2002-05-21 Ludwig Institute For Cancer Research Method for treating cancers which present antigen FB5 with humanized antibodies
US5811522A (en) * 1994-03-08 1998-09-22 Ludwig Institute For Cancer Research Recombinant humanized anti-FB5 antibodies
US6090930A (en) * 1994-03-08 2000-07-18 Ludwig Institute For Cancer Research Recombinant human anti-FB5 antibodies
US6217868B1 (en) * 1994-03-08 2001-04-17 Ludwig Institute For Cancer Research Methods for detecting cancer cells by using humanized antibodies which bind specifically to FB5 antigen
US5584458A (en) * 1995-07-18 1996-12-17 Levelite Technology, Inc. Quick-action mount for self-leveling laser
US6117977A (en) * 1996-04-24 2000-09-12 Genentech, Inc. Type C lectins
US6612190B2 (en) * 1997-09-08 2003-09-02 Ngk Insulators, Ltd. Mass sensor and mass sensing method
US6174689B1 (en) * 1998-10-26 2001-01-16 Immunex Corporation Viral encoded semaphorin protein receptor DNA and polypeptides
US6130068A (en) * 1998-10-26 2000-10-10 Immunex Corporation Viral encoded semaphorin protein receptor DNA and polypeptides
US7074913B2 (en) * 2000-12-05 2006-07-11 Wisconsin Alumni Research Foundation Receptor for B anthracis toxin
US7435418B2 (en) * 2000-12-05 2008-10-14 Wisconsin Alumni Research Foundation Receptor for B. anthracis toxin

Cited By (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100021454A1 (en) * 2005-04-22 2010-01-28 Morphotek, Inc. Antibodies With Immune Effector Activity And That Internalize In Endosialin-Positive Cells
US8389691B2 (en) 2005-04-22 2013-03-05 Morphotek, Inc. Antibodies with immune effector activity and that internalize in endosialin-positive cells
US8524237B2 (en) 2005-04-22 2013-09-03 Morphotek, Inc. Antibodies with immune effector activity and that internalize in endosialin-positive cells
US9505842B2 (en) 2007-04-05 2016-11-29 Morphotek, Inc. Methods for inhibiting the binding of endosialin to ligands
US20110033455A1 (en) * 2007-04-05 2011-02-10 Morphotek Inc, Methods For Inhibiting The Binding Of Endosialin To Ligands
US10053509B2 (en) 2007-04-05 2018-08-21 Eisai, Inc. Methods for inhibiting the binding of endosialin to ligands
US8895000B2 (en) 2007-04-05 2014-11-25 Morphotek, Inc. Methods for inhibiting the binding of endosialin to ligands
US7892760B2 (en) 2007-11-19 2011-02-22 Celera Corporation Lung cancer markers, and uses thereof
US20090176228A1 (en) * 2007-11-19 2009-07-09 Celera Corporation Lung cancer markers, and uses thereof
US20100260769A1 (en) * 2009-04-09 2010-10-14 Morphotek, Inc. Endosialin binding molecules
US8883978B2 (en) * 2010-05-11 2014-11-11 Institute For Cancer Research Antibodies to tumor endothelial marker 7R
US9534050B2 (en) 2010-05-11 2017-01-03 Institute For Cancer Research Antibodies to tumor endothelial marker 7R
US20130058935A1 (en) * 2010-05-11 2013-03-07 Fox Chase Cancer Center Antibodies to tumor endothelial marker 7r
WO2015006501A3 (en) * 2013-07-09 2015-03-12 President And Fellows Of Harvard College Microvessel endothelial cell surface markers and uses thereof
US10206941B2 (en) 2013-07-09 2019-02-19 President And Fellows Of Harvard College Venule endothelial cell genes and uses thereof
US10307487B2 (en) 2013-07-09 2019-06-04 President And Fellows Of Harvard College Microvessel endothelial cell surface markers and uses thereof

Also Published As

Publication number Publication date
WO2002010217A2 (en) 2002-02-07
WO2002010217A3 (en) 2002-09-06
AU2001283062A1 (en) 2002-02-13
JP2013100284A (en) 2013-05-23
US7358351B2 (en) 2008-04-15
US20030017157A1 (en) 2003-01-23
CA2416732C (en) 2016-02-09
EP1307557A2 (en) 2003-05-07
CA2416732A1 (en) 2002-02-07
US20050142138A1 (en) 2005-06-30
JP2015192672A (en) 2015-11-05
US7402660B2 (en) 2008-07-22
JP2004527210A (en) 2004-09-09
WO2002010217A9 (en) 2003-02-06

Similar Documents

Publication Publication Date Title
US7402660B2 (en) Endothelial cell expression patterns
EP1534331B1 (en) Membrane associated tumor endothelium markers
JP6196350B2 (en) Identification of tumor-associated cell surface antigens for diagnosis and therapy
EP1572867B1 (en) Endothelial cell expression patterns
ES2277432T3 (en) COMPOUNDS AND METHODS FOR THERAPY AND DIAGNOSIS OF CANCER DE PULMON.
AU2002258543A1 (en) Endothelial cell expression patterns
JP5380075B2 (en) Cancer-related antigen
WO2004005883A2 (en) Secreted and cytoplasmic tumor endothelial markers
US20090233270A9 (en) Secreted and cytoplasmic tumor endothelial markers
JP2001508052A (en) Diagnosis and treatment method of Hodgkin&#39;s lymphoma

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

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

AS Assignment

Owner name: NATIONAL INSTITUTES OF HEALTH - DIRECTOR DEITR, MA

Free format text: CONFIRMATORY LICENSE;ASSIGNOR:THE JOHNS HOPKINS UNIVERSITY;REEL/FRAME:050928/0109

Effective date: 20191014