US20030118585A1 - Use of protein biomolecular targets in the treatment and visualization of brain tumors - Google Patents

Use of protein biomolecular targets in the treatment and visualization of brain tumors Download PDF

Info

Publication number
US20030118585A1
US20030118585A1 US09/983,000 US98300001A US2003118585A1 US 20030118585 A1 US20030118585 A1 US 20030118585A1 US 98300001 A US98300001 A US 98300001A US 2003118585 A1 US2003118585 A1 US 2003118585A1
Authority
US
United States
Prior art keywords
protein
ptpζ
ser
leu
brain
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
US09/983,000
Inventor
Sabine Muller
Thorsten Melcher
Daniel Chin
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.)
AGY Therapeutics Inc
Original Assignee
AGY Therapeutics Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to US09/983,000 priority Critical patent/US20030118585A1/en
Application filed by AGY Therapeutics Inc filed Critical AGY Therapeutics Inc
Assigned to AGY THERAPEUTICS reassignment AGY THERAPEUTICS ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: MELCHER, THORSTEN, CHIN, DANIEL, MULLER, SABINE
Priority to JP2002575022A priority patent/JP2004537282A/en
Priority to PCT/US2002/008992 priority patent/WO2002076510A1/en
Priority to CA002439630A priority patent/CA2439630A1/en
Priority to EP02726668A priority patent/EP1383543A4/en
Publication of US20030118585A1 publication Critical patent/US20030118585A1/en
Priority to US10/652,981 priority patent/US7060275B2/en
Priority to US11/097,728 priority patent/US20050260132A1/en
Assigned to GENERAL ELECTRIC CAPITAL CORPORATION reassignment GENERAL ELECTRIC CAPITAL CORPORATION SECURITY AGREEMENT Assignors: AGY THERAPEUTICS, INC.
Assigned to AGY THERAPEUTICS, INC. reassignment AGY THERAPEUTICS, INC. RELEASE BY SECURED PARTY (SEE DOCUMENT FOR DETAILS). Assignors: GENERAL ELECTRIC CAPITAL CORPORATION
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • A61K49/0002General or multifunctional contrast agents, e.g. chelated agents

Definitions

  • the present invention relates to the use of proteins which are differentially expressed in primary brain tumor tissues, as compared to normal brain tissues, as biomolecular targets for brain tumor treatment therapies.
  • the present invention relates to the use of immunotherapeutic and immunoimaging agents which specifically bind to one or more of angiopoietin related protein 2 (ARP-2,) secreted protein acidic, rich in cysteine (SPARC,) c-met proto-oncogene (C-MET,) brevican (BEHAB,) CD-44 antigen (CD-44,) tetraspanin 3 (TSPN3,) pleiotrophin (PTN,) osteopontin (OPN,) vasoactive intestinal peptide receptor-2 (VIPR-2,) and receptor protein tyrosine phosphatase zeta (PTP ⁇ ) for the treatment and visualization of brain tumors in patients.
  • ARP-2 angiopoietin related protein 2
  • SPARC secreted protein acidic
  • C-MET
  • Brain tumors are considered to have one of the least favorable prognoses for long term survival: the average life expectancy of an individual diagnosed with a central nervous system (CNS) tumor is just eight to twelve months.
  • CNS central nervous system
  • Several unique characteristics of both the brain and its particular types of neoplastic cells create daunting challenges for the complete treatment and management of brain tumors. Among these are 1) the physical characteristics of the intracranial space, 2) the relative biological isolation of the brain from the rest of the body, 3) the relatively essential and irreplaceable nature of the organ mass, and 4) the unique nature of brain tumor cells.
  • the brain is made of, primarily, astrocytes (which make up the majority of the brain mass, and serve as a scaffold and support for the neurons), neurons (which carry the actual electrical impulses of the nervous system), and a minor contingent of other cells such as insulating oligodendrocytes (which produce myelin).
  • brain tumors e.g., astrocytomas, neuroblastomas, glioblastomas, oligodendrogliomas, etc.
  • astrocytoma and glioblastoma are often used broadly.
  • the brain is encased in the relatively rigid shell of the skull, and is cushioned by the cerebrospinal fluid, much like a fetus in the womb. Because of the relatively small volume of the skull cavity, minor changes in the volume of tissue in the brain can dramatically increase intracranial pressure, causing damage to the entire organ (i.e., “water on the brain”).
  • BBB Blood-Brain-Barrier
  • brain tumors are very heterogeneous, with different cell doubling times, treatment resistances, and other biochemical idiosyncrasies between the various cell populations that make up the tumor. This pervasive and variable nature greatly adds to the difficulty of treating brain tumors while preserving the health and function of normal brain tissue.
  • ARP-2 Angiopoeitin Related Protein-2, Angiopoeitin Like-2 [ANGPTL-2]
  • Angiopoeitin related protein-2 (ARP-2), is related to the angiopoeitin family of proteins, that includes Ang-1 and Ang-2. Like members of the angiopoeitin family, ARP-2 contains a coiled-coil domain in the amino terminal portion and a fibrinogen-like domain in the carboxyl terminal portion. However, ARP-2 has a low homology with Ang-1 and Ang-2 and unlike Ang-1 and Ang-2, ARP-2 does not bind to the Tie-2 receptor, nor does ARP-2 bind to the closely related Tie-1 receptor. Hence, ARP-2 is believed to be part of a newly identified family of proteins termed angiopocitin related proteins. Like the angiopoeitins, ARP-2 is a member of the fibrinogen superfamily, which also includes the fibrinogens and lectins.
  • ARP-2 is a glycosylated, secretory protein that induces sprouting in endothelial cells, most likely through autocrine or paracrine signaling, and it is preferentially expressed in the blood vessels and muscle cells. Hence, ARP-2 mediates the differentiated state of endothelial cells or for vascular remodeling and development. ARP-2 has not heretofore been associated with brain tumors.
  • SPARC Secreted Protein, Aacidic, Cysteine-Rich; Osteonectin; Basement Membrane Protein (bm) 40
  • Secreted protein acidic and rich in cysteine, SPARC or BM-40 is a member of the counter-adhesive family of proteins. It is a developmentally regulated, secreted glycoprotein expressed in fetal astrocytes, particularity during tissue remodeling, vessel morphogenesis, and in response to stress. It has been hypothesized that SPARC may affect cell migration and vascular morphogenesis either by directly interacting with extracellular matrix (ECM) proteins (such as collagens I, III, IV and V) or by initiating a receptor mediated signaling event that induces changes in cytoplasmic components associated with focal adhesions. SPARC has been found to bind directly to vitronectin, a multifunctional adhesive protein that is a component of the brain vascular basement membranes.
  • ECM extracellular matrix
  • SPARC may indirectly affect cell migration and motility by regulating the expression of matrix metallo-proteases and by modulating the expression of other proteolytic enzymes (such as collagenase) that degrade the ECM. Increased SPARC expression has also been observed in two forms of low-grade malignant gliomas, in all grades of human astrocytic tumors, and in tumor cells invading adjacent brain at the tumor/brain interface.
  • SPARC may be an astrocytoma invasion related gene that functions in connection with vitronectin to balance the modulation of cellular adhesion to the ECM and it may promote diffuse tumor cell infiltration into adjacent brain by affecting both tumor and endothelial cell-ECM interactions.
  • SPARC is also found in bone, dentine, and many normal and neoplastic human soft tissues it may also play a regulatory function in the control of such diverse processes as bone mineralization, cell shape, tissue remodeling or repair, cell migration, proliferation, and differentiation. SPARC is also synthesized, stored, and secreted by human blood platelets, binds to plasminogen, and enhances tissue plasminogen activator conversion of plasminogen to plasmin.
  • c-MET Metal Proto-Oncogene Tyrosine Kinase, Hepatocyte Growth Factor Receptor [HGFR]
  • c-MET is a member of the Hepatocyte Growth Factor Receptor (HGFR) family and a heterodimeric cellular receptor for Hepatocyte Growth Factor (HGF).
  • HGFR Hepatocyte Growth Factor Receptor
  • HGF Hepatocyte Growth Factor
  • c-MET contains a disulfide-linked ⁇ -chain of 50-kDa (which is located in the extracellular domain,) a 145-kDa ⁇ -chain (which includes an extracellular region,) a transmembrane spanning domain, and an intracellular tyrosine kinase domain that can be activated by autophosphorylation.
  • HGFR is a subset of the protein tyrosine-kinase family of membrane-spanning, cell surface receptors.
  • the receptor-ligand pair, c-MET and HGF function as a growth factor, regulating cell growth, migration, and morphogenesis, and hence, may play a role in neoplastic formation and metastasis.
  • HGF or macrophage stimulating protein (MSP) binding the c-MET protein receptor goes through a conformational change wherein the intracellular tyrosine residues of the ⁇ subunit become phosphorylated at residue 1235, and a second messenger signal cascade is induced. This change activates c-MET's intracellular receptor kinase activity, which is important to the growth and differentiation of epithelial cells in normal and malignant tissues.
  • c-MET has been identified in both normal brain and on glial tumors, and is thought to be determinant in the pathological processes of various malignancies. For instance, detailed studies have shown that glioblastoma multiforme (GBM), a highly malignant brain tumor of astrocytic origin, expresses c-MET, and this research suggests a role in tumor progression.
  • GBM glioblastoma multiforme
  • BEHAB Brain-Enriched Hyaluronan Binding Protein, Brevican
  • BEHAB is a brain-specific, extracellular matrix protein, that is a member of the chondroitin sulfate proteoglycan (CSPG) family. BEHAB is expressed only in the CNS. Although its function is unclear, BEHAB is reported to bind to HA at the N-terminus, lectins at the C-terminus, and may mediate binding of other ECM components like tenascin. This suggests that BEHAB may play a role in cell-cell and cell-matrix interactions thereby maintaining the extracellular environment of the brain. It has been reported that the highest levels of expression of BEHAB is during brain development and at times and places where glial cells are highly motile, as in cases of brain injury or trauma.
  • CSPG chondroitin sulfate proteoglycan
  • BEHAB expression is also unregulated in primary gliomas of the central nervous system, but not in tumors of non-glial origin. In surgical samples of human gliomas (including astrocytoma, oligodendroglioma, and glioblastoma tumors), BEHAB expression is consistently and dramatically increased over the level of expression in the normal brain. Hence, BEHAB expression correlates with an invasive phenotype that promotes gliogenesis by contributing to cell movement through the ECM.
  • CD-44 is a single-path, type I transmembrane protein with extracellular domains that are flexibly linked to the transmembrane segment.
  • CD-44 is a member of the cartilage link protein family and belongs to the hyaloadherin or link protein superfamily (LPSF). As other members of the LPS family, CD-44 can be extensively glycosylated and is typically decorated with glycosaminoglycans (e.g., chondroitin, heparin, and keratin sulfate).
  • the genomic structure of CD-44 consists of 21 exons, at least 11 of which can be variably spliced (v1 -v10), that are located in the membrane-proximal extracellular region.
  • CD-44 isoforms (at least 30 different isoforms have been characterized to date) that are widely distributed and expressed in a cell-specific manner.
  • CD-44H expressed on hematopoietic cells
  • CD-44E expressed in epithelial cells.
  • CD-44(H) has also been found to be expressed in lymphocytes, macrophages, erythrocytes, fibroblasts, epithelial and endothelial cells, and neurons. It is the predominant isoform in normal brain and neuroectoderm-derived tumors and is expressed on both normal astrocytes and oligodendrocytes as well on neoplastic astrocytes and glioblastomas.
  • CD-44 The family of CD-44 proteins has been implicated in lymphocyte activation and homing, endothelial migration, and tumor cell metastasis.
  • CD-44 is believed to be the major receptor for Hyaluronic acid (HA).
  • HA Hyaluronic acid
  • CD-44 also mediates the attachment of glioma cells to chondroitin sulfate, types I and IV collagen, fibronectin laminin, vitronectin and Martrigel. This suggest that CD-44 may play a role in cell-cell and cell-matrix interactions, affecting the extracellular environment of the brain.
  • HA is a major component of the brain ECM
  • CD-44 is one of the principal cellular receptors of HA
  • CD-44 expression coincides with brain tumor growth and invasiveness.
  • PTN Pleotrophin, Heparin Binding Growth Factor 8, Neurite Growth-Promoting Factor 1
  • Pleiotrophin or PTN is a platelet-derived, growth factor inducible, member of the pleiotrophin family of proteins that includes midkine and retinoic acid-induced heparin-binding protein. It is a developmentally regulated, secreted cytokine that stimulates mitogenesis, angiogenesis, and neurite and glial process outgrowth guidance activities.
  • PTN is expressed in the brain, intestine, muscle, skin, heart, lung and kidney. In the adult, PTN is found primarily in the brain in association with axonal tracts during active mitogenesis and may therefore play an important role in the development and maintenance of the nervous system.
  • PTN has also been found to have oncogenic properties, inducing malignant transformation and tumor growth and progression. It has been described as a proto-oncogene that is expressed in many human tumors and cell lines derived from human tumors. PTN is a mitogen for fibroblasts, epithelial and endothelial cells, stimulates plasminogen-activator production, can induce tube formation, and therefore can serve as a tumor angiogenesis factor.
  • OPN Oleopontin, Secreted Phosphoprotein 1, Bone Sialoprotein-1
  • Osteopontin or OPN is a member of the osteopontin family. It is a glycosylated sialoprotein that is heavily phosphorylated and expressed in a variety of cells including bone, kidney, placenta, nerve cells and macrophages, as well as T lymphocytes, epidermal and bone cells.
  • OPN is a part of the mineralized bone matrix and may play a role in bone resorption, by facilitating the attachment of osteoclasts to the bone surface, and may be functionally important as an adhesive and chemotactic molecule for vascular cells.
  • OPN is a secreted protein that binds tightly to hydroxyapatite, and hence, is important to cell matrix interactions. It has been observed to interact with the CD-44 homing receptor to physiologically induce macrophage chemotaxis, which may be a mechanism utilized by metastatic brain tumors in the process of dissemination.
  • OPN has been observed in the microvasculature of glioblastomas associated with VEGF expression and OPN mRNA has been found to be overexpressed in high grade and metastatic brain tumors. Hence, OPN expression correlates with the malignancy grade of gliomas.
  • VIPR-2 Vasoactive Intestinal Peptide Receptor-2
  • Vasoactive intestinal polypeptide receptor II (VIPR-2), VPAC-2, is a member of the G-protein receptor family, which includes such members as the calcitonin, parathyroid hormone, secretin, glucagon and VIP-1 receptors.
  • VIPR-2 is a seven-transmembrane spanning G protein-coupled receptor that responds to VIP by stimulating cAMP production.
  • VIPR-2 is found in the brain as well as peripheral tissues such as the pancreas, skeletal muscle, heart, lung, kidneys, stomach, adipocytes and the liver, and in various cells of the immune system. In the brain, VIPR-2 functions as a neuroendocrine hormone and neurotransmitter receptor, and is found in the thalamus, hippocampus, suprachiasmatic nucleus and hypothalamus.
  • VIPR-2 is encoded by a nucleotide sequence of approximately 2.8 kb, which codes for a 438 amino acid sequence of approximately 48-64 kDa.
  • the receptor-ligand pair, VIPR-2 and VIP have various functions dependent upon the tissue where in they are located.
  • VIP is a late-developing, 28 amino acid peptide that, along with its receptor, is widely distributed throughout the peripheral body, and plays a role in cardiovascular, reproductive, pulmonary, immune and gastrointestinal systems, to effect vasodilatation, bronchodilation, immunosuppression, hormonal secretion, and increased gastric motility.
  • the cerebral cortex has one of the highest reported concentrations of VIP, localized to intrinsic neurons throughout all neocortical regions.
  • VIP and its receptor have behavioral, electrophysiological, secretory, metabolic, vascular, and mitogenic effects.
  • the receptor-ligand pair play a role in cortical differentiation, the relaying of sensory information to the cortex, and the regulation of morphogenic events by the release of diffusible signals from glial cells.
  • VIPR-2 and VIP also play a role in the growth and differentiation of neuroblastomas.
  • TSPAN3 Tetraspanin 3, Tetraspanin TM-4A
  • the Tetraspanin superfamily is a family of approximately 20 integral membrane proteins that are broadly expressed in most human tissues including neural and bone marrow derived tissues.
  • the family shares a common motif that includes four putative transmembrane domains (TM1-4), a small extracellular domain (EC1) of 20-27 amino acids, and a larger extracellular domain (EC2) between TMS3 and TMS4 of 70-130 amino acids.
  • TM1-4 putative transmembrane domains
  • EC1 small extracellular domain
  • EC2 extracellular domain
  • EC2 extracellular domain
  • EC2 extracellular domain
  • Two conserved features of tetraspanins are critical to their structure and function. First, charged residues are present in or near the TM domains, second, a cluster of cysteine residues is in the putative EC2 domain. Most of the tetraspanins are modified by N-glycosylation.
  • Tetraspanin proteins affect the regulation of cellular proliferation, motility, differentiation, development.
  • Tetraspanins may act as adapters in ric complexes that link plasma membrane proteins, like integrins, into signaling complexes with other signaling molecules (e.g., phosphatidylinositol 4-kinase) at the plasma membrane and play a role in integrin-mediated cell migration, metastasis and tumor cell invasion.
  • signaling molecules e.g., phosphatidylinositol 4-kinase
  • a number of tetraspanins have also been discovered as tumor-associated proteins, including C-029, PETA-3/SFA-1, and SAS, which is amplified in a subset of sarcomas.
  • CD9 is expressed on 90% of non-T cell acute lymphoblastic leukemia cells and on 50% of chronic lymphocytic and acute myeloblastic leukemias.
  • CD63 is also expressed in early stage melanomas.
  • Vital cellular functions are regulated in part by the balance between the activities of protein kinases and protein phosphatases.
  • protein-modifying enzymes add or remove a phosphate group from serine, threonine, or tyrosine residues in specific proteins.
  • Some tyrosine kinases (PTK's) and phosphatases (PTPase's) have been theorized to have a role in some types of oncogenesis, which is thought to result from an imbalance in their activities.
  • PTPase molecules There are two classes of PTPase molecules: low molecular weight proteins with a single conserved phosphatase domain such as T-cell protein-tyrosine phosphatase (PTPT; MIM 176887), and high molecular weight receptor-linked PTPases with two tandemly repeated and conserved phosphatase domains separated by 56 to 57 amino acids. Examples of this latter group of receptor proteins include: leukocyte-common antigen (PTPRC; MIM 151460) and leukocyte antigen related tyrosine phosphatase (PTPRF; MIM 179590).
  • PTPase molecules low molecular weight proteins with a single conserved phosphatase domain such as T-cell protein-tyrosine phosphatase (PTPT; MIM 176887), and high molecular weight receptor-linked PTPases with two tandemly repeated and conserved phosphatase domains separated by 56 to 57 amino acids. Examples of this latter group of receptor proteins include
  • Protein tyrosine phosphatase zeta [also known as PTPRZ, HPTP-ZETA, HPTPZ, RPTP-BETA( ⁇ ), or RPTPB] was isolated as a cDNA sequence by two groups in the early nineties. The complete cDNA sequence of the protein is provided in SEQ ID NO. 1, and the complete deduced amino acid sequence is provided in SEQ ID NO. 2. Splicing variants and features are indicated in the sequences. Levy et al. (“The cloning of a receptor-type protein tyrosine phosphatase expressed in the central nervous system” J. Biol. Chem.
  • PTP- ⁇ the protein, which they designated PTP- ⁇ (PTP ⁇ )
  • PTP- ⁇ is a transmembrane protein with 2 cytoplasmic PTPase domains and a 1,616-amino acid extracellular domain.
  • PTP- ⁇ MIM 176886
  • the 266 N-terminal residues of the extracellular domain are have a high degree of similarity to carbonic anhydrases (see MIM 114880).
  • the human gene encoding PTP ⁇ has been mapped to chromosome 7q31.3-q32 by chromosomal in situ hybridization (Ariyama et al., “Assignment of the human protein tyrosine phosphatase, receptor-type, zeta (PTPRZ) gene to chromosome band 7q31.3 ” Cytogenet. Cell Genet. 70: 52-54 (1995)).
  • Northern blot analysis has shown that showed that PTP-zeta is expressed only in the human central nervous system.
  • PTP ⁇ has only been found to be expressed in the nervous system. By in situ hybridization, it has been localized to different regions of the adult brain, including the Purkinje cell layer of the cerebellum, the dentate gyrus, and the subependymal layer of the anterior horn of the lateral ventricle. High levels of PTP ⁇ have been seen in regions of the brain where there is continued neurogenesis and neurite outgrowth, and it seems to play a role in morphogenesis and plasticity of the nervous system. Phosphacan immunoreactivity has been associated with perineuronal nets around parvalbumin-expressing neurons in adult rat cerebral cortex. Neurons as well as astrocytes have been shown to express phosphacan.
  • the transmembrane forms of PTP ⁇ are expressed on the migrating neurons especially at the lamellipodia along the leading processes.
  • PTP ⁇ is postulated to be involved in the neuronal migration as a neuronal receptor of pleiotrophin distributed along radial glial fibers.
  • PTP ⁇ has been shown to be highly expressed in radial glia and other forms of glial cells that play an important role during development.
  • the anti-PTP ⁇ staining localizes to the radial processes of these cells, which act as guides during neuronal migration and axonal elongation.
  • the pattern of RPTP-zeta expression has also been shown to change with the progression of glial cell differentiation.
  • the three splicing variants of RPTP-zeta have been shown to have different spatial and temporal patterns of expression in the developing brain.
  • the 9.5-kb and 6.4-kb transcripts which encode the ⁇ and ⁇ transmembrane protein tyrosine phosphatases, were predominantly expressed in glial progenitors located in the subventricular zone.
  • the 8.4-kb transcript which encodes the secreted chondroitin sulfate proteoglycan phosphacan, was expressed at high levels by more mature glia that have migrated out of the subventricular zone.
  • the three transcripts have also been shown to be differentially expressed in glial cell cultures.
  • PTP ⁇ -deficient mice were viable, fertile, and showed no gross anatomical alterations in the nervous system or other organs. Therefore, it was deduced that PTP ⁇ is not essential for neurite outgrowth and node formation in mice.
  • the ultrastructure of nerves of the central nervous system in PTP ⁇ -deficient mice suggests a fragility of myelin.
  • conduction velocity was not altered.
  • the normal development of neurons and glia in was thought to indicate that PTP ⁇ function is not necessary for these processes in vivo, or that a loss of PTP ⁇ can be compensated for by other protein tyrosine phosphatases expressed in the nervous system.
  • Phosphacan which is the shortest, secreted form of PTP ⁇ , containing the full extracellular region, previously was designated 3F8 and 6B4 chondroitin sulfate proteoglycan or 3H1 keratin sulfate proteoglycan depending on the glycosylation status.
  • amphoterin and pleiotrophin are heparin-binding proteins that are developmentally regulated in brain and functionally involved in neurite outgrowth.
  • Binding of phosphacan to Ng-CAM/L1, NCAM, and tenascin-C (FNIII domain) is mediated by complex-type N-linked oligosaccharides on the proteoglycan.
  • Phosphacan shows saturable, reversible, high-affinity binding to fibroblast growth factor-2 (FGF-2). The interaction is mediated primarily through the core protein. Immunocytochemical studies have also shown an overlapping localization of FGF-2 and phosphacan in the developing central nervous system. The core protein of phosphacan may also regulate the access of FGF-2 to cell surface signaling receptors in nervous tissue.
  • the carbonic anhydrase (CAH) domain of PTP ⁇ has been shown to bind specifically to contactin.
  • Contactin is a 140 kDa GPI membrane-anchored neuronal cell recognition protein expressed on the surface of neuronal cells.
  • the CAH domain of RPTP zeta was shown to induce cell adhesion and neurite growth of primary tectal neurons, and differentiation of neuroblastoma cells. These responses were blocked by antibodies against contactin, demonstrating that contactin is a neuronal receptor for RPTP zeta.
  • Caspr (p190/Caspr, a contactin-associated transmembrane receptor) and contactin exist as a complex in rat brain and are bound to each other by means of lateral (cis) interactions in the plasma membrane.
  • the extracellular domain of Caspr contains a neurophilin/coagulation factor homology domain, a region related to fibrinogen beta/gamma, epidermal growth factor-like repeats, neurexin motifs as well as unique PGY repeats found in a molluscan adhesive protein.
  • the cytoplasmic domain of Caspr contains a proline-rich sequence capable of binding to a subclass of SH3 domains of signaling molecules.
  • Caspr may function as a signaling component of contactin, enabling recruitment and activation of intracellular signaling pathways in neurons.
  • the role of the extracellular domains in neural adhesion and neurite growth induction was investigated by the use of fusion protein constructs. The results suggested that binding of glial PTP ⁇ to the contactin/Nr-CAM complex is important for neurite growth and neuronal differentiation.
  • PTP ⁇ was shown to bind to a heparin-binding growth factor midkine through the chondroitin sulfate portion of the receptor.
  • the interactions of pleiotrophin (PTN) with the receptor in U373-MG cells was also studied.
  • Pleiotrophin was shown to bind to the spacer domain. Results suggested that PTN signals through “ligand-dependent receptor inactivation” of PTP ⁇ and disrupts its normal roles in the regulation of steady-state tyrosine phosphorylation of downstream signaling molecules.
  • PTN was shown to bind to and functionally inactivate the catalytic activity of PTP ⁇ .
  • PTP ⁇ An active site-containing domain of PTP ⁇ both binds ⁇ -catenin and functionally reduces its levels of tyrosine phosphorylation when added to lysates of pervanadate-treated cells.
  • PTP ⁇ was shown to be intrinsically active, and thought to function as an important regulator in the reciprocal control of the steady-state tyrosine phosphorylation levels of ⁇ -catenin by tyrosine kinases and phosphatases.
  • the present invention provides novel methods and reagents for specifically targeting brain tumor neoplastic cells for both therapeutic and imaging purposes, by targeting brain tumor protein targets (T BT ). These targets have been identified by the applicants as being overexpressed in brain tumors, and thus allow for the selective inhibition of cell function or selective marking for visualization with therapeutic or visualizing compositions which have a specific affinity for thes eprotein targets.
  • T BT brain tumor protein targets
  • ARP-2 angiopoietin related protein 2
  • SPARC secreted protein acidic, rich in cysteine
  • C-MET c-met proto-oncogene
  • BEHAB CD-44 antigen
  • CD-44 CD-44
  • TSPN3 tetraspanin 3
  • PPN pleiotrophin
  • OPN osteopontin
  • VIP-2 vasoactive intestinal peptide receptor-2
  • PTP ⁇ vasoactive intestinal peptide receptor-2
  • PTP ⁇ receptor protein tyrosine phosphatase zeta
  • the present invention provides T BT affinity-based compounds and compositions useful in treating a brain tumor in a patient.
  • the compositions and compounds of this aspect of the invention generally fall into two groups: T BT -binding conjugate compounds, which comprise a cytotoxic moiety (C), which inhibits the growth of tumor cells; and T BT -binding compound compositions in which the T BT binding moiety alters the normal function of the T BT in or around the tumor cell, thus inhibiting cell growth and/or function.
  • C cytotoxic moiety
  • T BT -binding therapeutic conjugate compounds are provided. These compounds have the general formula ⁇ (T BT )C, wherein ⁇ (T BT ) is one or more moieties which specifically binds to a T BT , and C is one or more cytotoxic moieties.
  • ⁇ (T BT ) is an antibody or an antibody fragment.
  • ⁇ (T BT ) is an antibody or an antibody fragment which elicits a reduced immune response when administered to a human patient.
  • Preferred cytotoxic moieties for use in these embodiments of the invention include radioactive moieties, chemotoxic moieties, and toxin proteins.
  • the invention also provides compositions comprising these T BT -binding therapeutic conjugate compounds in a pharmaceutically acceptable carrier.
  • T BT -binding therapeutic compounds which alter the normal function of the T BT in or around brain tumor cells and inhibit brain tumor cell growth.
  • These T BT -binding therapeutic compounds have the general formula ⁇ (T BT ), wherein ⁇ (T BT ) is one or more moieties which specifically binds to a T BT , and wherein the binding of ⁇ (T BT ) alters the function of the T BT .
  • ⁇ (T BT ) is an antibody or an antibody fragment.
  • ⁇ (T BT ) is an antibody or an antibody fragment which elicits a reduced immune response when administered to a human patient. It is preferred that the therapeutic compounds of this second group of embodiments of the first aspect of the invention be formulated into therapeutic compositions comprising the T BT -binding compound in a pharmaceutically acceptable carrier.
  • the present invention provides methods for using these compounds and compositions to treat a brain tumor in a patient.
  • the methods comprise administering an effective amount of a composition, comprising a T BT -binding compound from the first or second group of embodiments of the first aspect and a pharmaceutically acceptable carrier, to a patient in need thereof.
  • Brain tumors treated in this fashion may be glioblastomas, astrocytomas, neuroblastomas, or any type of brain tumor.
  • Administration of the therapeutic composition may be by any acceptable means.
  • One preferred method for administration is by intrathecal administration, although intravascular administration is also preferred.
  • the present invention provides T BT affinity-based compounds and compositions for the visualization of brain tumors in patients.
  • These compounds have the general formula ⁇ (T BT )I, wherein ⁇ (T BT ) is one or more moieties which specifically binds to a T BT , and I is one or more imaging moieties.
  • ⁇ (T BT ) is an antibody or an antibody fragment.
  • ⁇ A(T BT ) is an antibody or an antibody fragment which elicits a reduced immune response when administered to a human patient.
  • Preferred I moieties include radiographic moieties (useful in, e.g., x-ray, scintillation, or other radiation imaging methods,) positron-emitting moieties, magnetic spin contrast moieties, and optically visible moieties (such as visible particles, fluorescent dyes, and visible-spectrum dyes.) It is preferred that the imaging compounds of these embodiments of the third aspect of the invention be formulated into therapeutic compositions comprising the T BT -binding compound in a pharmaceutically acceptable carrier.
  • the present invention provides methods of using the compounds and compositions of the third aspect of the invention to visualize a brain tumor in a patient. These methods generally comprise administering an effective amount of an imaging compound of the general formula ⁇ (T BT )I in a pharmaceutically acceptable carrier to the patient, and then visualizing the imaging moieties of the compound. Administration of the imaging composition may be by any acceptable means. Intravascular administration of the imaging composition is preferred in these methods, although intrathecal administration is also preferred.
  • Preferred methods of visualizing the imaging moieties of the compounds include radiographic imaging techniques (e.g., x-ray imaging and scintillation imaging techniques), positron-emission tomography, magnetic resonance imaging techniques, and direct or indirect (e.g., endoscopic) visual inspection.
  • radiographic imaging techniques e.g., x-ray imaging and scintillation imaging techniques
  • positron-emission tomography positron-emission tomography
  • magnetic resonance imaging techniques e.g., magnetic resonance imaging techniques
  • direct or indirect visual inspection e.g., endoscopic
  • a method to treat a brain tumor by administering a therapeutic amount of a composition comprising a compound of the general formula ⁇ (ARP2)C, wherein ⁇ (ARP2) is one or more moieties which specifically binds to a human angiopoietin related protein-2, and C is one or more cytotoxic moieties, and a pharmaceutically acceptable carrier.
  • ⁇ (ARP2) is one or more moieties which specifically binds to a human angiopoietin related protein-2
  • C is one or more cytotoxic moieties
  • a method to treat a brain tumor by administering a therapeutic amount of a composition comprising a compound of the general formula ⁇ (ARP2), wherein ⁇ (ARP2) is one or more moieties which specifically binds to a human angiopoietin related protein-2, wherein the binding of ⁇ (ARP2) alters the function of the angiopoietin related protein-2, and a pharmaceutically acceptable carrier.
  • ⁇ (ARP2) is one or more moieties which specifically binds to a human angiopoietin related protein-2, wherein the binding of ⁇ (ARP2) alters the function of the angiopoietin related protein-2, and a pharmaceutically acceptable carrier.
  • a method for visualizing a brain tumor in a patient by first administering to a patient an effective amount of a composition comprising: a compound of the general formula ⁇ (ARP2)I, wherein ⁇ (ARP2) is one or more moieties which specifically binds to a human angiopoietin related protein-2, and I is one or more imaging moieties and a pharmaceutically acceptable carrier, and then visualizing the imaging moieties of the compound.
  • a composition for the visualization of a brain tumor comprising a compound of the general formula ⁇ (ARP2)I, wherein ⁇ (ARP2) is one or more moieties which specifically binds to a human angiopoietin related protein-2, and I is one or more imaging moieties, and a pharmaceutically acceptable carrier.
  • a method to treat a brain tumor by administering a therapeutic amount of a composition comprising a compound of the general formula ⁇ (TSPAN3)C, wherein ⁇ (TSPAN3) is one or more moieties which specifically binds to a human tetraspanin 3, and C is one or more cytotoxic moieties, and a pharmaceutically acceptable carrier.
  • a method to treat a brain tumor by administering a therapeutic amount of a composition comprising a compound of the general formula ⁇ (TSPAN3), wherein ⁇ (TSPAN3) is one or more moieties which specifically binds to a human tetraspanin 3, wherein the binding of ⁇ (TSPAN3) alters the function of the tetraspanin 3, and a pharmaceutically acceptable carrier.
  • ⁇ (TSPAN3) is one or more moieties which specifically binds to a human tetraspanin 3, wherein the binding of ⁇ (TSPAN3) alters the function of the tetraspanin 3, and a pharmaceutically acceptable carrier.
  • a composition for the treatment of a brain tumor comprising: a compound of the general formula ⁇ (TSPAN3), wherein ⁇ (TSPAN3) is one or more moieties which specifically binds to a human tetraspanin 3, wherein the binding of ⁇ (TSPAN3) alters the function of the tetraspanin 3, and a pharmaceutically acceptable carrier.
  • a method for visualizing a brain tumor in a patient by first administering to a patient an effective amount of a composition comprising: a compound of the general formula ⁇ (TSPAN3)I, wherein ⁇ (TSPAN3) is one or more moieties which specifically binds to a human tetraspanin 3, and I is one or more imaging moieties, and a pharmaceutically acceptable carrier, and then visualizing the imaging moieties of the compound.
  • a composition for the visualization of a brain tumor comprising a compound of the general formula ⁇ (TSPAN3)I, wherein ⁇ (TSPAN3) is one or more moieties which specifically binds to a human tetraspanin 3, and I is one or more imaging moieties, and a pharmaceutically acceptable carrier.
  • a method to treat a brain tumor by administering a therapeutic amount of a composition comprising a compound of the general formula ⁇ (PTP ⁇ )C, wherein ⁇ (PTP ⁇ ) is one or more moieties which specifically binds to a human PTP ⁇ selected from the group consisting of PTP ⁇ SM1 and PTP ⁇ SM2, further wherein ⁇ (PTP ⁇ ) does not specifically bind to human PTP ⁇ ⁇ , human PTP ⁇ ⁇ , or phosphacan, and C is one or more cytotoxic moieties, and a pharmaceutically acceptable carrier.
  • ⁇ (PTP ⁇ ) is one or more moieties which specifically binds to a human PTP ⁇ selected from the group consisting of PTP ⁇ SM1 and PTP ⁇ SM2, further wherein ⁇ (PTP ⁇ ) does not specifically bind to human PTP ⁇ ⁇ , human PTP ⁇ ⁇ , or phosphacan
  • C is one or more cytotoxic moieties, and a pharmaceutically acceptable carrier.
  • a compound for the treatment of a brain tumor of the general formula ⁇ (PTP ⁇ )C wherein ⁇ (PTP ⁇ ) is one or more moieties which specifically binds to a human PTP ⁇ selected from the group consisting of PTP ⁇ SM1 And PTP ⁇ SM2, further wherein ⁇ (PTP ⁇ ) does not specifically bind to human PTP ⁇ ⁇ , human PTP ⁇ ⁇ ,or human phosphacan, and C is one or more cytotoxic moieties.
  • a method to treat a brain tumor by administering a therapeutic amount of a composition comprising a compound of the general formula ⁇ (PTP ⁇ ), wherein ⁇ (PTP ⁇ ) is one or more moieties which specifically binds to a human PTP ⁇ selected from the group consisting of PTP ⁇ SM1 And PTP ⁇ SM2, further wherein ⁇ (PTP ⁇ ) does not specifically bind to human PTP ⁇ ⁇ , human PTP ⁇ , or phosphacan, wherein the binding of ⁇ (PTP ⁇ ) alters the function of the human PTP ⁇ ,and a pharmaceutically acceptable carrier.
  • ⁇ (PTP ⁇ ) is one or more moieties which specifically binds to a human PTP ⁇ selected from the group consisting of PTP ⁇ SM1 And PTP ⁇ SM2, further wherein ⁇ (PTP ⁇ ) does not specifically bind to human PTP ⁇ ⁇ , human PTP ⁇ , or phosphacan, wherein the binding of ⁇ (PTP ⁇ ) alters the function of the human PTP ⁇
  • a composition for the treatment of a brain tumor comprising a compound of the general formula ⁇ (PTP ⁇ ), wherein ⁇ (PTP ⁇ ) is one or more moieties which specifically binds to PTP ⁇ selected from the group consisting of PTP ⁇ SM1 And PTP ⁇ SM2, further wherein ⁇ (PTP ⁇ ) does not specifically bind to human PTP ⁇ ⁇ , human PTP ⁇ ⁇ , or phosphacan, wherein the binding of ⁇ (PTP ⁇ ) alters the function of the PTP ⁇ , and a pharmaceutically acceptable carrier.
  • ⁇ (PTP ⁇ ) is one or more moieties which specifically binds to PTP ⁇ selected from the group consisting of PTP ⁇ SM1 And PTP ⁇ SM2, further wherein ⁇ (PTP ⁇ ) does not specifically bind to human PTP ⁇ ⁇ , human PTP ⁇ ⁇ , or phosphacan, wherein the binding of ⁇ (PTP ⁇ ) alters the function of the PTP ⁇ , and a pharmaceutically acceptable carrier.
  • a method for visualizing a brain tumor in a patient by first administering to a patient an effective amount of a composition comprising: a compound of the general formula ⁇ (PTP ⁇ )I, wherein ⁇ (PTP ⁇ ) is one or more moieties which specifically binds to a human PTP ⁇ selected from the group consisting of PTP ⁇ SM1 And PTP ⁇ SM2, further wherein ⁇ (PTP ⁇ ) does not specifically bind to human PTP ⁇ ⁇ , human PTP ⁇ ⁇ , or phosphacan, and I is one or more imaging moieties and a pharmaceutically acceptable carrier, and then visualizing the imaging moieties of the compound.
  • a composition comprising: a compound of the general formula ⁇ (PTP ⁇ )I, wherein ⁇ (PTP ⁇ ) is one or more moieties which specifically binds to a human PTP ⁇ selected from the group consisting of PTP ⁇ SM1 And PTP ⁇ SM2, further wherein ⁇ (PTP ⁇ ) does not specifically bind to human PTP
  • a composition for the visualization of a brain tumor comprising a compound of the general formula ⁇ (PTP ⁇ )I, wherein ⁇ (PTP ⁇ ) is one or more moieties which specifically binds to a human PTP ⁇ selected from the group consisting of PTP ⁇ SM1 And PTP ⁇ SM2, further wherein ⁇ (PTP ⁇ ) does not specifically bind to human PTP ⁇ ⁇ , human PTP ⁇ ⁇ , or phosphacan, and I is one or more imaging moieties, and a pharmaceutically acceptable carrier.
  • a method to treat a brain tumor by administering a therapeutic amount of a composition comprising a compound of the general formula ⁇ (SPARC)C, wherein ⁇ (SPARC) is one or more moieties which specifically binds to a human secreted protein, rich in cysteine, and C is one or more cytotoxic moieties, and a pharmaceutically acceptable carrier.
  • ⁇ (SPARC) is one or more moieties which specifically binds to a human secreted protein, rich in cysteine
  • C is one or more cytotoxic moieties
  • a method to treat a brain tumor by administering a therapeutic amount of a composition comprising a compound of the general formula ⁇ (SPARC), wherein ⁇ (SPARC) is one or more moieties which specifically binds to a human secreted protein, rich in cysteine, wherein the binding of ⁇ (SPARC) alters the function of the secreted protein, rich in cysteine, and a pharmaceutically acceptable carrier.
  • ⁇ (SPARC) is one or more moieties which specifically binds to a human secreted protein, rich in cysteine, wherein the binding of ⁇ (SPARC) alters the function of the secreted protein, rich in cysteine, and a pharmaceutically acceptable carrier.
  • a method for visualizing a brain tumor in a patient by first administering to a patient an effective amount of a composition comprising: a compound of the general formula ⁇ (SPARC)I, wherein ⁇ (SPARC) is one or more moieties which specifically binds to a human secreted protein, rich in cysteine, and I is one or more imaging moieties, and a pharmaceutically acceptable carrier, and then visualizing the imaging moieties of the compound.
  • a composition comprising: a compound of the general formula ⁇ (SPARC)I, wherein ⁇ (SPARC) is one or more moieties which specifically binds to a human secreted protein, rich in cysteine, and I is one or more imaging moieties, and a pharmaceutically acceptable carrier, and then visualizing the imaging moieties of the compound.
  • a composition for the visualization of a brain tumor comprising a compound of the general formula ⁇ (SPARC)I, wherein ⁇ (SPARC) is one or more moieties which specifically binds to a human secreted protein, rich in cysteine, and I is one or more imaging moieties, and a pharmaceutically acceptable carrier.
  • a method to treat a brain tumor by administering a therapeutic amount of a composition comprising a compound of the general formula ⁇ (c-MET)C, wherein ⁇ (c-MET) is one or more moieties which specifically binds to a human c-MET oncogene product, and C is one or more cytotoxic moieties and a pharmaceutically acceptable carrier.
  • a method to treat a brain tumor by administering a therapeutic amount of a composition comprising a compound of the general formula ⁇ (c-MET), wherein ⁇ (c-MET) is one or more moieties which specifically binds to a human c-MET oncogene product, wherein the binding of ⁇ (TSPAN3) alters the function of the c-MET oncogene product, and a pharmaceutically acceptable carrier.
  • a method for visualizing a brain tumor in a patient by first administering to a patient an effective amount of a composition comprising: a compound of the general formula ⁇ (c-MET)I, wherein ⁇ (c-MET) is one or more moieties which specifically binds to a human c-MET oncogene product, and I is one or more imaging moieties and a pharmaceutically acceptable carrier, and then visualizing the imaging moieties of the compound.
  • a method to treat a brain tumor by administering a therapeutic amount of a composition comprising a compound of the general formula ⁇ (CD44)C, wherein ⁇ (CD44) is one or more moieties which specifically binds to a human CD44 antigen, and C is one or more cytotoxic moieties, and a pharmaceutically acceptable carrier.
  • a method to treat a brain tumor by administering a therapeutic amount of a composition comprising a compound of the general formula ⁇ (CD44), wherein ⁇ (CD44) is one or more moieties which specifically binds to a human CD44 antigen, wherein the binding of ⁇ (CD44) alters the function of the CD44 antigen, and a pharmaceutically acceptable carrier.
  • a method for visualizing a brain tumor in a patient by first administering to a patient an effective amount of a composition comprising: a compound of the general formula ⁇ (CD44)I, wherein ⁇ (CD44) is one or more moieties which specifically binds to a human CD44 antigen, and I is one or more imaging moieties, and a pharmaceutically acceptable carrier, and then visualizing the imaging moieties of the compound.
  • a composition comprising: a compound of the general formula ⁇ (CD44)I, wherein ⁇ (CD44) is one or more moieties which specifically binds to a human CD44 antigen, and I is one or more imaging moieties, and a pharmaceutically acceptable carrier, and then visualizing the imaging moieties of the compound.
  • a method to treat a brain tumor by administering a therapeutic amount of a composition comprising a compound of the general formula ⁇ (VIPR2)C, wherein ⁇ (VIPR2) is one or more moieties which specifically binds to a human vasoactive intestinal peptide receptor-2, and C is one or more cytotoxic moieties and a pharmaceutically acceptable carrier.
  • ⁇ (VIPR2) is one or more moieties which specifically binds to a human vasoactive intestinal peptide receptor-2
  • C is one or more cytotoxic moieties and a pharmaceutically acceptable carrier.
  • a method to treat a brain tumor by administering a therapeutic amount of a composition comprising a compound of the general formula ⁇ (VIPR2), wherein ⁇ (VIPR2) is one or more moieties which specifically binds to a human vasoactive intestinal peptide receptor-2, wherein the binding of ⁇ (VIPR2) alters the function of the vasoactive intestinal peptide receptor-2, and a pharmaceutically acceptable carrier.
  • a composition comprising a compound of the general formula ⁇ (VIPR2), wherein ⁇ (VIPR2) is one or more moieties which specifically binds to a human vasoactive intestinal peptide receptor-2, wherein the binding of ⁇ (VIPR2) alters the function of the vasoactive intestinal peptide receptor-2, and a pharmaceutically acceptable carrier.
  • a method for visualizing a brain tumor in a patient by first administering to a patient an effective amount of a composition comprising: a compound of the general formula ⁇ (VIPR2)I, wherein ⁇ (VIPR2) is one or more moieties which specifically binds to a human tetraspanin 3, and I is one or more imaging moieties, and a pharmaceutically acceptable carrier, and then visualizing the imaging moieties of the compound.
  • a composition comprising: a compound of the general formula ⁇ (VIPR2)I, wherein ⁇ (VIPR2) is one or more moieties which specifically binds to a human tetraspanin 3, and I is one or more imaging moieties, and a pharmaceutically acceptable carrier, and then visualizing the imaging moieties of the compound.
  • a method to treat a brain tumor by administering a therapeutic amount of a composition comprising a compound of the general formula ⁇ (OPN)C, wherein ⁇ (OPN) is one or more moieties which specifically binds to a human osteopontin, and C is one or more cytotoxic moieties and a pharmaceutically acceptable carrier.
  • a method to treat a brain tumor by administering a therapeutic amount of a composition comprising a compound of the general formula ⁇ (OPN), wherein ⁇ (OPN) is one or more moieties which specifically binds to a human osteopontin, wherein the binding of ⁇ (OPN) alters the function of the osteopontin, and a pharmaceutically acceptable carrier.
  • a method for visualizing a brain tumor in a patient by first administering to a patient an effective amount of a composition comprising: a compound of the general formula ⁇ (OPN)I, wherein ⁇ (OPN) is one or more moieties which specifically binds to a human osteopontin, and I is one or more imaging moieties, and a pharmaceutically acceptable carrier, and then visualizing the imaging moieties of the compound.
  • a composition comprising: a compound of the general formula ⁇ (OPN)I, wherein ⁇ (OPN) is one or more moieties which specifically binds to a human osteopontin, and I is one or more imaging moieties, and a pharmaceutically acceptable carrier, and then visualizing the imaging moieties of the compound.
  • a composition for the visualization of a brain tumor comprising a compound of the general formula ⁇ (OPN)I, wherein ⁇ (OPN) is one or more moieties which specifically binds to a human osteopontin, and I is one or more imaging moieties, and a pharmaceutically acceptable carrier.
  • a method to treat a brain tumor by administering a therapeutic amount of a composition comprising a compound of the general formula ⁇ (PTN)C, wherein ⁇ (PTN) is one or more moieties which specifically binds to a human pleiotrophin, and C is one or more cytotoxic moieties, and a pharmaceutically acceptable carrier.
  • ⁇ (PTN) is one or more moieties which specifically binds to a human pleiotrophin
  • C is one or more cytotoxic moieties
  • a method to treat a brain tumor by administering a therapeutic amount of a composition comprising a compound of the general formula ⁇ (PTN), wherein ⁇ (PTN) is one or more moieties which specifically binds to a human pleiotrophin, wherein the binding of ⁇ (PTN) alters the function of the pleiotrophin, and a pharmaceutically acceptable carrier.
  • a method for visualizing a brain tumor in a patient by first administering to a patient an effective amount of a composition comprising: a compound of the general formula ⁇ (PTN)I, wherein ⁇ (PTN) is one or more moieties which specifically binds to a human pleiotrophin, and I is one or more imaging moieties, and a pharmaceutically acceptable carrier, and then visualizing the imaging moieties of the compound.
  • a composition comprising: a compound of the general formula ⁇ (PTN)I, wherein ⁇ (PTN) is one or more moieties which specifically binds to a human pleiotrophin, and I is one or more imaging moieties, and a pharmaceutically acceptable carrier, and then visualizing the imaging moieties of the compound.
  • a composition for the visualization of a brain tumor comprising a compound of the general formula ⁇ (PTN)I, wherein ⁇ (PTN) is one or more moieties which specifically binds to a human pleiotrophin, and I is one or more imaging moieties, and a pharmaceutically acceptable carrier.
  • Brain tumors are known to be relatively heterogeneous, and thus all patients may not respond the same to a particular protein target treatment.
  • the invention also provides, in yet another aspect, combination therapeutic and/or visualization agents, compositions, and methods.
  • ARP-2 angiopoietin related protein 2
  • SPARC secreted protein acidic, rich in cysteine
  • c-MET c-met proto-oncogene
  • BEHAB cd-44 antigen
  • CD-44 tetraspanin 3
  • PPN3 pleiotrophin
  • PPN osteopontin
  • VIP-2 vasoactive intestinal peptide receptor-2
  • PTP ⁇ receptor protein tyrosine phosphatase zeta
  • the brain tumor protein targets are selected from the group consisting of angiopoietin related protein 2 (ARP-2,) c-met proto-oncogene (c-MET,) cd-44 antigen (CD-44,) tetraspanin 3 (TSPN3,) osteopontin (OPN,) and receptor protein tyrosine phosphatase zeta (PTP ⁇ ).
  • ARP-2 angiopoietin related protein 2
  • c-MET c-met proto-oncogene
  • CD-44 cd-44 antigen
  • TSPN3 tetraspanin 3
  • osteopontin osteopontin
  • PTP ⁇ receptor protein tyrosine phosphatase zeta
  • Embodiments of the combination aspect of the invention may target a group of proteins from the identified targets with similar compartmentalization characteristics.
  • the combination aspects may target two or more secreted proteins from the group angiopoietin related protein 2 (ARP-2,) secreted protein acidic, rich in cysteine (SPARC,) brevican (BEHAB,) pleiotrophin (PTN,) secreted forms of receptor protein tyrosine phosphatase zeta (PTP ⁇ ) [including the novel isoform PTP ⁇ SM1].
  • ARP-2 angiopoietin related protein 2
  • SPARC secreted protein acidic, rich in cysteine
  • BEHAB brevican
  • PPN pleiotrophin
  • PTP ⁇ receptor protein tyrosine phosphatase zeta
  • the combination aspects may target one or more of the extracellular matrix binding proteins secreted protein acidic, rich in cysteine (SPARC,) and/or brevican (BEHAB,) with one or more of the other identified protein targets.
  • the combination aspects may target one or more of the membrane-bound proteins from the group c-met proto-oncogene (c-MET,) cd-44 antigen (CD-44,) tetraspanin 3 (TSPN3,) osteopontin (OPN,) vasoactive intestinal peptide receptor-2 (VIPR-2,) and membrane bound forms of receptor protein tyrosine phosphatase zeta (PTP ⁇ ) [including the novel isoform PTP ⁇ SM2].
  • c-MET c-met proto-oncogene
  • CD-44 CD-44
  • TSPN3 tetraspanin 3
  • osteopontin osteopontin
  • VIP-2 vasoactive intestinal peptide receptor-2
  • PTP ⁇ membrane bound forms of receptor protein tyrosine phosphatase zeta
  • At least one of the protein targets is selected from the proteins angiopoietin related protein 2 (ARP-2,) tetraspanin 3 (TSPN3,), and receptor protein tyrosine phosphatase zeta (PTP ⁇ ) [including the two novel isoforms PTP ⁇ SM1 and SM2].
  • ARP-2 angiopoietin related protein 2
  • TSPN3 tetraspanin 3
  • PTP ⁇ receptor protein tyrosine phosphatase zeta
  • one of these proteins and another protein selected from the secreted group, the extracellular matrix group, or the membrane-bound group.
  • ARP-2 angiopoietin related protein 2
  • TSPN3 tetraspanin 3
  • PTP ⁇ receptor protein tyrosine phosphatase zeta
  • ARP-2 angiopoietin related protein 2
  • SPARC secreted protein acidic, rich in cysteine
  • C-MET c-met proto-oncogene
  • BEHAB brevican
  • CD-44 antigen CD-44
  • TSPN3 tetraspanin 3
  • PPN3 pleiotrophin
  • PPN osteopontin
  • VIPR-2 vasoactive intestinal peptide receptor-2
  • PTP ⁇ receptor protein tyrosine phosphatase zeta
  • TSPN3 target tetraspanin 3
  • SPARC protein acidic, rich in cysteine
  • C-MET c-met proto-oncogene
  • BEHAB brevican
  • CD-44 antigen CD-44
  • ARP-2 angiopoietin related protein 2
  • PPN pleiotrophin
  • OPN osteopontin
  • VIP-2 vasoactive intestinal peptide receptor-2
  • PTP ⁇ receptor protein tyrosine phosphatase zeta
  • PTP ⁇ target receptor protein tyrosine phosphatase zeta
  • ARP-2 angiopoietin related protein 2
  • SPARC secreted protein acidic, rich in cysteine
  • C-MET c-met proto-oncogene
  • BEHAB brevican
  • CD-44 antigen CD-44
  • TSPN3 tetraspanin 3
  • PPN3 pleiotrophin
  • OPN vasoactive intestinal peptide receptor-2
  • VIP-2 vasoactive intestinal peptide receptor-2
  • pleiotrophin (PTN) is a known ligand of PTP ⁇
  • another preferred embodiment of the combination aspects of the invention utilizes these proteins as targets, either alone or in combination with one or more of the other identified targets.
  • the present invention provides two novel splicing isoforms of PTP ⁇ , shown to be expressed in brain tissue.
  • PTP ⁇ SM1 and PTP ⁇ SM2 described in more detail below, differ in structure from the three known isoforms heretofore disclosed.
  • PTP ⁇ SM1 comprises the amino acids encoded by the first nine exons of PTP ⁇ - ⁇ , with three unique additional carboxy terminal amino acids encoded by additional 3′ mRNA sequence from the intron of the gene between exons nine and ten.
  • the mRNA for PTP ⁇ SM2 comprises all exons of PTP ⁇ - ⁇ , with a 116 nucleotide insertion, in the correct reading frame, in the mRNA sequence between exons 23 and 24, from the from the intron of the gene between exons 23 and 24.
  • embodiments of this aspect of the invention include the mature proteins of PTP ⁇ splice variants SM1 or SM2, and nucleic acids encoding these novel spice variants, as well proteins with significant homology to the splice variants.
  • the invention provides nucleic acid polymers comprising the sequence of nucleotides 148 to 1272 of SEQ ID NO. 1, the complement of nucleotides 148 to 1272 of SEQ ID NO. 1, nucleotides 148 to 7209 of SEQ ID NO. 3, or the complement of nucleotides 148 to 7209 of SEQ ID NO. 3.
  • the invention provides polypeptides comprising the amino acid sequence of SEQ ID NO. 2 or the amino acid sequence of SEQ ID NO. 4.
  • the invention provides polypeptides comprising a distinctive portion of the amino acid sequence of SEQ ID NO. 2 or SEQ ID NO. 4.
  • Such peptides are useful for the production of antibodies against the PTP ⁇ SM1 or SM2 splicing variants.
  • these polypeptides comprise a portion of the amino acid sequence of SEQ ID NO. 2 or SEQ ID NO. 4 which is at least 6, more preferably at least 8, more preferably at least 10, more preferably at least 15, and most preferably at lest 20 amino acids in length.
  • the polypeptides comprise the three unique terminal amino acids of PTP ⁇ SM1 after exon 9.
  • the polypeptides comprise a portion of the unique exon 23a of PTP ⁇ SM2, wherein the portion is preferably at least 3 amino acids in length, more preferably at least 6 amino acids in length, more preferably at least 9 amino acids in length, and most preferably at least 15 amino acids in length.
  • the invention also provides affinity reagents which specifically bind to PTP ⁇ splice variants SM1 or SM2, but do not bind to the other known splice variants of PTP ⁇ (e.g., ⁇ , ⁇ , or phosphacan forms).
  • these affinity reagents are antibodies or antibody fragments.
  • the invention also provides nucleic acid sequences encoding the PTP ⁇ splice variants SM1 or SM2.
  • the invention also encompasses nucleic acid probes which hybridize to the mRNA encoding PTP ⁇ splice variants SM1 or SM2, but not mRNA encoding other known splice variants of PTP ⁇ .
  • FIG. 1 A diagram of the three known splicing variant isoforms of PTP ⁇ . The approximate position of the domains of the isoforms is indicated underneath the isoforms, as well as the approximate exon size (for size reference, exon 12 is 3.6 kilobases.)
  • Isoform PTP ⁇ - ⁇ is the full length isoform, which contains the primary amino acid sequence aa 25-2314 of SEQ ID NO. 2 (aa 1-24 are a signal polypeptide). In Isoform PTP ⁇ - ⁇ , aa 755-1614 are missing.
  • Isoform PTP ⁇ -S phosphacan
  • FIG. 2 A diagram of the two newly discovered splicing variant isoforms of PTP ⁇ . The approximate position of the domains of the isoforms is indicated underneath the isoforms, as well as the approximate exon size (for size reference, exon 12 is 3.6 kilobases.)
  • SM 1 fails to splice correctly after the 9 th exon, yielding an mRNA with tow extra codons followed by a stop codon after the normal terminus of exon 9.
  • SM 2 contains a 116 nucleotide insertion from between exons 23 &24.
  • FIG. 3 A diagram comparing the three known PTP ⁇ isoforms with the two novel isoforms.
  • Applicants have identified several brain tumor protein targets and genes which are differentially regulated between brain cancer tissue (glioblastoma) and normal brain tissue. Applicants have performed differential cloning between cancerous and normal brains and have identified the brain tumor protein target genes by DNA sequence analysis. Based on the observation in other diseases, particularly other cancers, in which overexpressed genes can contribute to the pathology of the disease, these overexpressed genes and their protein products mediate the initiation and progression of brain tumors. Thus, the overexpressed brain tumor protein targets, which are presented on the cell surface, provide excellent targets for immunotherapeutic agents which either deliver cytotoxic agents to directly promote tumor cell death, or which alter the function of the brain tumor protein targets to inhibit the normal physiology of the tumor cell.
  • immunotherapeutic agents which either deliver cytotoxic agents to directly promote tumor cell death, or which alter the function of the brain tumor protein targets to inhibit the normal physiology of the tumor cell.
  • immunoimaging agents targeted to the brain tumor protein targets may be utilized to visualize the tumor mass either in diagnostic methods (e.g., magnetic resonance imaging (MRI) or radiography), or in surgery (e.g., by the use of optically visual dye moieties in the immunoimaging agent).
  • diagnostic methods e.g., magnetic resonance imaging (MRI) or radiography
  • surgery e.g., by the use of optically visual dye moieties in the immunoimaging agent.
  • Applicants have identified the brain tumor protein targets by a direct examination of the expression level of genes in actual tumor cells. These samples provide a more accurate and realistic picture of tumor cell biology, especially on the detailed transcriptome level, than animal models or established cell tissue culture cell lines. Several groups have found that cell lines established from astrocytomas and other cell lines do not exhibit expression patterns which reflect the actual expression of the original tumor. For instance, Schreiber, et.
  • N-Oct proteins nervous system-specific transcription factors known as N-Oct proteins are differentially expressed in human neuroblastoma and glioblastoma cell lines in vitro.
  • these results were compared to freshly isolated human primary and metastatic brain tumors, of the five astrocytomas and three glioblastomas analyzed, all but two tumors displayed the complete N-Oct protein profile, irrespective of histopathological tumor grade.
  • angiogenin in tissue samples of different brain tumors and cultured glioma cells. Anticancer Res 20: 1679-84 (2000), could show that angiogenin is detectable in different kinds of intracranial tumor tissue samples. Although angiogenin could be detected in primary cultivated glioma cells, it was not detected in the permanent cell lines.
  • ds-cDNA's from control and disease states were subjected to kinetic re-annealing hybridization during which normalization of transcript abundances and enrichment for differentially expressed transcripts (i.e., subtraction) occurs.
  • Normalized-subtracted ds-cDNAs were cloned into a plasmid vector, a large number of recombinant bacterial clones were picked, and their recombinant inserts were isolated by PCR.
  • High-density cDNA arrays of those PCR products were screened with cDNA probes derived from the original control and disease states. Thus, only clones displaying a significant transcriptional induction and/or repression were sequenced and carried forward for massive expression profiling using a variety of temporal, spatial and disease-related probe sets.
  • each of these proteins is individually useful as a target for the treatment and/or imaging of brain tumors.
  • ARP-2 was selected as a target for selective immuno-therapeutic agents in targeting and/or imaging brain tumors.
  • the mature protein consists of 493 amino acids and contains two potential consensus glycosylation sites.
  • the complete cDNA sequence encoding ARP-2 is provided in SEQ ID NO. 7, and the complete amino acid sequence of ARP-2 is provided in SEQ ID NO. 8.
  • ARP-2 is a 64 kDa, single chain, acidic, angiopoeitin-like protein that includes multiple functional domains, such as a hydrophobic signal sequence from amino acids 1-21 (which is typical of secreted proteins), a coiled-coil domain at the amino terminal end from approximately amino acid sequences 22-274, and a fibrinogen-like domain, from approximately about residues 275 through 493.
  • functional domains such as a hydrophobic signal sequence from amino acids 1-21 (which is typical of secreted proteins), a coiled-coil domain at the amino terminal end from approximately amino acid sequences 22-274, and a fibrinogen-like domain, from approximately about residues 275 through 493.
  • Two major isoforms have been observed, one 2.4 Kb in size and the other about 4 Kb. Both forms are abundant in heart, small intestine, spleen and stomach.
  • a compound that specifically binds to ARP-2 is any compound (such as an antibody) that has a binding affinity for any naturally occurring isoform, splice variant, or polymorphism of ARP-2, explicitly including the isoforms described herein.
  • specific binding compounds e.g., antibodies
  • proteins include truncated forms or domains of ARP-2, and recombinantly engineered alterations of ARP-2.
  • a portion of SEQ ID NO. 8 may be engineered to include a non-naturally occurring cysteine for cross linking to an immunoconjugate protein, as described.
  • the antibodies utilized in the compositions and methods of the invention bind to the fibrinogen domain but need not be restricted to this domain.
  • the antibody may bind to the extracellular region of ARP-2. It is to be noted that antibodies which bind to this secreted protein are useful in the invention as cytotoxic delivery agents, as well as functional inhibition agents, as one of ordinary skill would expect that the concentration of ARP-2 would be increased adjacent the tumor cells which, due to the need for vascularization, over-express the protein.
  • the entire protein (either the unsecreted precursor or the secreted protein), or a portion thereof, may be utilized.
  • the carboxyl-terminal fibrinogen like domain, or any portion of the amino-terminal coiled-coil domain may be utilized.
  • amino acids 22-274 which make up the fibrinogen like domain, may be used.
  • Larger ARP-2 proteins and domains may be produced utilizing any suitable recombinant vector/protein production system, such as the baculovirus transfection system outlined below, after being amplified from a fetal brain cDNA library (as available from, e.g., Clontech, Palo Alto, Calif.) or another suitable source.
  • antibodies may be raised by immunizing the production animal with the protein and a suitable adjuvant (e.g., Fruend's, Fruend's complete, oil-in-water emulsions, etc.).
  • a suitable adjuvant e.g., Fruend's, Fruend's complete, oil-in-water emulsions, etc.
  • the ARP-2 protein (or a portion thereof) can serve as the ARP-2 antigen.
  • BSA bovine serum albumin
  • KLH keyhole limpet hemocyanin
  • peptides derived from the full ARP-2 sequence may be utilized.
  • one or more 8-30 aa peptide portions of the EC domain of ARP-2 are utilized, with peptides in the range of 10-20 being a more economical choice.
  • Custom-synthesized peptides in this range are available from a multitude of vendors, and can be order conjugated to KLH or BSA.
  • peptides in excess of 30 amino acids may be synthesized by solid-phase methods, or may be recombinantly produced in a suitable recombinant protein production system.
  • an animal cell system e.g., Sf9 or other insect cells, CHO or other mammalian cells
  • Other information useful in designing an antigen for the production of antibodies to ARP-2 may be deduced by those of skill in the art by homology analysis of SEQ ID NO. 8.
  • ARP-2 fibrinogen domain of human ARP-2 is hypothesized to interact with one or more an unknown receptor for the purposes of angiogenesis.
  • the interaction of ARP-2 to these molecules may be through either of the aforementioned structural motifs.
  • antibody moieties are utilized which bind to ARP-2 at a site on the protein that alters the binding of an extracellular molecule to ARP-2.
  • ARP-2 activity altering antibodies may be utilized in therapeutic compositions in an unconjugated form (e.g., the antibody in an acceptable pharmaceutical carrier), or may be conjugated to either a therapeutic moiety (creating a double-acting therapeutic agent) or an imaging moiety (creating a duel therapeutic/imaging agent).
  • Selection of antibodies which alter (enhance or inhibit) the binding of a ARP-2 to a receptor may be accomplished by a straightforward binding inhibition/enhancement assay.
  • a labeled (e.g., fluorescently or enzyme-labeled) antibody to ARP-2 which has been immobilized in a microtiter well, is assayed in both the presence and absence of the appropriate ligand.
  • the change in binding is indicative of either an enhancer (increased binding) or competitive inhibitor (decreased binding) relationship between the antibody and the ligand.
  • Such assays may be carried out in high-throughput formats (e.g., 384 well plate formats, in robotic systems) for the automated selection of monoclonal antibody candidates for use as ARP-2 ligand-binding inhibitors or enhancers.
  • antibodies which are useful for altering the function of ARP-2 may be assayed in functional formats, such as endothelial sprouting assays and cell migration assays described in the examples. Thus, antibodies that exhibit the appropriate anti-tumor effect may be selected without direct knowledge of a binding ligand.
  • SPARC was selected as a target for selective immuno-therapeutic agents in targeting and/or imaging brain tumors.
  • the mature protein consists of 286 amino acids (after cleavage of the signal peptide) and contains two potential Asn-X-Thr/Ser N-glycosylation sites, located at positions 71 and 99 of the mature protein.
  • the complete cDNA sequence encoding SPARC is provided in SEQ ID NO. 9, and the complete amino acid sequence of SPARC is provided in SEQ ID NO. 10.
  • SPARC is an abundant 33 kDa, single chain, acidic, extracellular calcium binding protein that contains a flexible N-terminal acidic domain I ( ⁇ 50 amino acids), a follistatin-like (FS) domain ( ⁇ 75 residues), and a C-terminal extracellular calcium-binding (EC) domain with a pair of EF-hand loops ( ⁇ 150 residues).
  • the N-terminal domain shows a low affinity Ca2+ binding site, a transglutaminase cross linking site, and inhibits cell spreading in cell culture assays.
  • Calcium-dependent binding of SPARC to the triple helix of several fibrillar collagen types and basement membrane collagen type IV has been mapped to the EC domain. Two isoforms have been described, bone SPARC with a molecular weight of 31,000 kDa and platelet SPARC with a molecular weight of 33,000 kDa.
  • a compound that specifically binds to SPARC is any compound (such as an antibody) that has a binding affinity for any naturally occurring isoform, splice variant, or polymorphism of SPARC, explicitly including the isoforms described herein.
  • specific binding compounds e.g., antibodies
  • proteins include truncated forms or domains of SPARC, and recombinantly engineered alterations of SPARC.
  • a portion of SEQ ID NO. 10 may be engineered to include a non-naturally occurring cysteine for cross linking to an immunoconjugate protein, as described.
  • the antibodies utilized in the compositions and methods of the invention bind to the extracellular domain (amino acids 130-280). It is preferable that this binding inhibit the activity of SPARC.
  • the antibody may bind to the EF hand which is known to bind Ca2+ with high affinity, but need not be restricted to this domain. It is to be noted that antibodies which bind to SPARC are useful in both cytotoxic and imaging embodiments of the invention, as one of ordinary skill would expect that the concentration of SPARC in the extracellular matrix would be increased around tumor cells which over-express the protein.
  • the entire protein (either the unsecreted precursor or the secreted protein), or a portion thereof, may be utilized.
  • the C terminal extracellular (EC) domain or any portion of the flexible N-terminal domain I, or FS domain may be utilized.
  • amino acids 125-275 which make up the EC domain, may be used.
  • Larger SPARC proteins and domains may be produced utilizing any suitable recombinant vector/protein production system, such as the baculovirus transfection system outlined below, after being amplified from a fetal brain cDNA library (as available from, e.g., Clontech, Palo Alto, Calif.) or another suitable source.
  • antibodies may be raised by immunizing the production animal with the protein and a suitable adjuvant (e.g., Fruend's, Fruend's complete, oil-in-water emulsions, etc.).
  • a suitable adjuvant e.g., Fruend's, Fruend's complete, oil-in-water emulsions, etc.
  • the SPARC protein (or a portion thereof) can serve as the SPARC antigen.
  • Commonly utilized conjugate proteins that are commercially available for such use include bovine serum albumin (BSA) and keyhole limpet hemocyanin (KLH).
  • peptides derived from the full SPARC sequence may be utilized.
  • one or more 8-30 aa peptide portions of the EC domain of SPARC are utilized, with peptides in the range of 10-20 being a more economical choice.
  • Custom-synthesized peptides in this range are available from a multitude of vendors, and can be order conjugated to KLH or BSA.
  • peptides in excess of 30 amino acids may be synthesized by solid-phase methods, or may be recombinantly produced in a suitable recombinant protein production system.
  • an animal cell system e.g., Sf9 or other insect cells, CHO or other mammalian cells
  • Sf9 or other insect cells e.g., Sf9 or other insect cells, CHO or other mammalian cells
  • Other information useful in designing an antigen for the production of antibodies to SPARC, including glycosylation sites, is provided in SEQ ID NO. 10.
  • the EC domain of human SPARC is known to interact with the collagens I, III, IV and V, and to bind to vitronectin, all of which are components of the extracellular matrix surrounding gliomas.
  • the binding of SPARC to these molecules may play a significant role in the oncogenesis and growth of neoplastic cells in the brain.
  • antibody moieties are utilized which bind to SPARC at a site on the protein that alters the binding of an extracellular molecule, such as an ECM molecule, to SPARC.
  • SPARC activity altering antibodies may be utilized in therapeutic compositions in an unconjugated form (e.g., the antibody in an acceptable pharmaceutical carrier), or may be conjugated to either a therapeutic moiety (creating a double-acting therapeutic agent) or an imaging moiety (creating a duel therapeutic/imaging agent).
  • Selection of antibodies which alter (enhance or inhibit) the binding of a ligand to SPARC may be accomplished by a straightforward binding inhibition/enhancement assay.
  • a labeled (e.g., fluorescently or enzyme-labeled) antibody to SPARC which has been immobilized in a microtiter well, is assayed in both the presence and absence of the appropriate ligand.
  • the change in binding is indicative of either an enhancer (increased binding) or competitive inhibitor (decreased binding) relationship between the antibody and the ligand.
  • Such assays may be carried out in high-throughput formats (e.g., 384 well plate formats, in robotic systems) for the automated selection of monoclonal antibody candidates for use as SPARC ligand-binding inhibitors or enhancers.
  • antibodies which are useful for altering the function of SPARC may be assayed in functional formats, such as the HUVEC tube assay and cell migration assay.
  • functional formats such as the HUVEC tube assay and cell migration assay.
  • antibodies that exhibit the appropriate anti-tumor effect may be selected without direct knowledge of a binding ligand or molecular function.
  • c-MET was selected as a target for selective immuno-therapeutic agents in targeting and/or imaging brain tumors.
  • the complete cDNA sequence encoding c-MET is provided in SEQ ID NO. 11, and the complete amino acid sequence of c-MET is provided in SEQ ID NO. 12.
  • c-MET is a type I membrane protein heterodimer.
  • two different receptor variants originate by post-translational processing of a common singe-chain precursor of 170 kDa.
  • Isoform p190MET is formed of a 50 kDa ⁇ -chain and a 145 kDa ⁇ -chain that are disulfide linked
  • isoform p140Met is formed of a 50 kDa ⁇ -chain and an 85 kDa ⁇ -chain, lacking the cytoplasmic kinase domain.
  • This 85 kDa ⁇ chain is likely a trans-membrane glycoprotein that is bound to the cell surface.
  • Truncated forms of c-MET containing the 50 kDa ⁇ -chain and a carboxyl-terminally truncated 75 kDa ⁇ sub-unit have also been described.
  • the 75 kDa form arises by post-translational proteolytic processing, lacks the trans-membrane domain, and is secreted from the cell.
  • a compound that specifically binds to c-MET is any compound (such as an antibody) that has a binding affinity for any naturally occurring isoform, splice variant, or polymorphism of c-MET, explicitly including the three isoforms described herein.
  • specific binding compounds e.g., antibodies
  • proteins include truncated forms or domains of c-MET, and recombinantly engineered alterations of c-MET.
  • a portion of SEQ ID NO. 12 may be engineered to include a non-naturally occurring cysteine for cross-linking to an immunoconjugate protein, as described below.
  • the antibodies utilized in the compositions and methods of the invention bind to the membrane-bound isoforms of the protein, as this will more specifically target the cytotoxic therapeutic agent, or the imaging agent, to the brain tumor cell.
  • embodiments that utilize antibodies that bind to the secreted isoform of the protein are also useful in the invention, as one of ordinary skill would expect that the concentration of the secreted isoform would also be increased adjacent to brain tumor cells which over-express the protein.
  • the amino acid sequence of full length c-MET consists of 1408 amino acids, as the sequence was first deduced by Park et al., (“Sequence of MET proto-oncogene cDNA has features characteristic of the tyrosine kinase family of growth-factor receptors” Proc. nat. Acad. Sci. U.S.A. 84:6379-6383 (1987)) and 1390 amino acids, as later deduced by Prat et al. (“C-terminal truncated forms of Met, the Hepatocyte Growth Factor” Mol Cell. Biol. 11:5954-5962 (1991)).
  • the first N-terminal amino acids 1-24 of SEQ ID NO. B′ [B′] are for the most part hydrophobic, and could serve as a signal sequence for transporting the protein into the lumen of the endoplasmic reticulum.
  • the ⁇ chain makes up the extracellular domain of the mature c-MET protein and spans amino acids 24-306 of SEQ ID NO. 12.
  • the ⁇ chain would consist of 1,084-5 amino acids with the predicted ⁇ chain extracellular domain being amino acids 306 to 932, the single transmembrane hydrophobic segment being amino acids 933 to 955, and the intracellular domain being amino acids 956 to 1390 of SEQ ID NO 12.
  • the entire protein, a dimeric subunit, or a portion thereof may be utilized.
  • the extracellular domain of the ⁇ or ⁇ sub-units or the secreted or extracellular portion of the truncated forms may be utilized.
  • amino acids that constitute the ⁇ sub-unit, amino acids 24-306, may be used.
  • Larger c-MET proteins and domains may be produced utilizing any suitable recombinant vector/protein production system, such as the baculovirus transfection system outlined below, after being amplified from a fetal brain cDNA library (as available from, e.g., Clontech, Palo Alto, Calif.) or another suitable source.
  • antibodies may be raised by immunizing the production animal with the protein and a suitable adjuvant (e.g., Fruend's, Fruend's complete, oil-in-water emulsions, etc.).
  • a suitable adjuvant e.g., Fruend's, Fruend's complete, oil-in-water emulsions, etc.
  • the c-MET protein (or a portion thereof) can serve as the c-MET antigen.
  • a smaller peptide it is advantageous to conjugate the peptide with a larger molecule to make an immunostimulatory conjugate for use as the c-MET antigen.
  • conjugate proteins that are commercially available for such use include bovine serum albumin (BSA) and keyhole limpet hemocyanin (KLH).
  • peptides derived from the full c-MET sequence may be utilized.
  • one or more 8-30 amino acid peptide portions of an extracellular domain of c-MET are utilized, with peptides in the range of 10-20 being a more economical choice.
  • Custom-synthesized peptides in this range are available from a multitude of vendors, and can be order conjugated to KLH or BSA.
  • peptides in excess of 30 amino acids may be synthesized by solid-phase methods, or may be recombinantly produced in a suitable recombinant protein production system.
  • an animal cell system e.g., Sf9 or other insect cells, CHO or other mammalian cells
  • Sf9 or other insect cells e.g., Sf9 or other insect cells, CHO or other mammalian cells
  • Other information useful in designing an antigen for the production of antibodies to c-MET, including glycosylation sites, is provided in SEQ ID NO. 12.
  • HGF hepatocyte growth factor
  • HGF is largely expressed in mesenchymal and neuroectodermal tissues and released to the extracellular compartment
  • paracrine and/or autocrine signaling implicate tumor genesis in mesenchymal and neuroectodermal tumors and other tumor cells that over express the c-MET receptor.
  • Recent studies have shown that the c-MET proto-oncogene is frequently overexpressed in many types of epithelial tumors, in spontaneously transformed NIH/3T3 fibroblasts, and in peripheral nerve sheath tumors.
  • antibody moieties are utilized which bind to c-MET at a site on the protein which alters the binding of an extracellular ligand molecule, such as HGF, to c-MET.
  • c-MET activity altering antibodies may be utilized in therapeutic compositions in an unconjugated form (e.g., the antibody in an acceptable pharmaceutical carrier), or may be conjugated to either a therapeutic moiety (creating a double-acting therapeutic agent) or an imaging moiety (creating a duel therapeutic/imaging agent).
  • Selection of antibodies that alter (enhance or inhibit) the binding of a ligand to c-MET may be accomplished by a straightforward binding inhibition/enhancement assay.
  • a labeled (e.g., fluorescently or enzyme-labeled) antibody to c-MET which has been immobilized in a microtiter well, is assayed in both the presence and absence of the ligand.
  • the change in binding is indicative of either an enhancer (increased binding) or competitive inhibitor (decreased binding) relationship between the antibody and the ligand.
  • Such assays may be carried out in high-throughput formats (e.g., 384 well plate formats, in robotic systems) for the automated selection of monoclonal antibody candidates for use as c-MET ligand-binding inhibitors or enhancers.
  • antibodies that are useful for altering the function of c-MET may be assayed in functional formats, such as the endothelial sprouting assay and cell migration assay.
  • functional formats such as the endothelial sprouting assay and cell migration assay.
  • BEHAB was selected as a target for selective immuno-therapeutic agents in targeting and/or imaging brain tumors.
  • the complete cDNA sequence encoding BEHAB GPI isoform is provided in SEQ ID NO. 13, and the complete amino acid sequence of this BEHAB isoform is provided in SEQ ID NO. 14.
  • Two isoforms have been isolated to date: a full-length isoform that is secreted into the extracellular matrix and a shorter isoform that has a hydrophobic carboxy terminus instead of the typical lectican carboxyl terminus, which predicts a glycophosphatidylinositol (GPI) anchor.
  • GPI glycophosphatidylinositol
  • BEHAB contains an N-terminal hyaluronan (HA)-binding domain, which comprises an immunoglobulin-like loop and two proteoglycan tandem repeats, a C-terminal epidermal growth factor (EGF)-like repeat, a C-type lectin-like domain, and a complement regulatory protein (CRP)-like domain.
  • the central region of the protein contains sites for glycosylation and proteolytic cleavage (between glu395-Ser396 of the mature protein, after signal peptide cleavage) by metallo-protease.
  • the complete cDNA of the secreted isoform is 2878 bp encoding 912 amino acids of 99 kDa.
  • the GPI isoform for which sequences SEQ ID NO. 13 and SEQ ID NO. 14 are given, is 2558 bp encoding 672 amino acids of 72 kDa.
  • the GPI-linked form is generated by a ‘no splice’ event, with the transcript reading through an exon/intron junction thereby extending the open reading frame to a stop codon 74 nucleotides further downstream.
  • a compound that specifically binds to BEHAB is any compound (such as an antibody) that has a binding affinity for any naturally occurring isoform, splice variant, or polymorphism of BEHAB, explicitly including the two splice variants described herein.
  • specific binding compounds e.g., antibodies
  • proteins include truncated forms or domains of BEHAB, and recombinantly engineered alterations of BEHAB.
  • a portion of SEQ ID NO. 14 may be engineered to include a non-naturally occurring cysteine for cross-linking to an immunoconjugate protein, as described below.
  • the antibodies utilized in the compositions and methods of the invention bind to the membrane-bound isoform of the protein, as this will more specifically target the cytotoxic therapeutic agent, or the imaging agent, to the brain tumor cell.
  • embodiments that utilize antibodies that bind to the secreted isoform of the protein are also useful in the invention, as one of ordinary skill would expect that the concentration of the secreted isoform would also be increased adjacent to brain tumor cells which over-express the protein.
  • the entire protein, or a portion thereof may be utilized.
  • any one of the aforementioned domains of the secreted protein or an extracellular portion of the truncated, membrane bound GPI form may be utilized.
  • amino acids that constitute the hyaluronic acid binding domain amino acids 44-247, which includes the Ig like domain at amino acids 44-140, may be used.
  • BEHAB proteins and domains may be produced utilizing any suitable recombinant vector/protein production system, such as the baculovirus transfection system outlined below, after being amplified from a fetal brain cDNA library (as available from, e.g., Clontech, Palo alto, CA) or another suitable source.
  • a fetal brain cDNA library as available from, e.g., Clontech, Palo alto, CA
  • antibodies may be raised by immunizing the production animal with the protein and a suitable adjuvant (e.g., Fruend's, Fruend's complete, oil-in-water emulsions, etc.).
  • a suitable adjuvant e.g., Fruend's, Fruend's complete, oil-in-water emulsions, etc.
  • the Brevican protein (or a portion thereof) can serve as the BEHAB antigen.
  • Commonly utilized conjugate proteins that are commercially available for such use include bovine serum albumin (BSA) and keyhole limpet hemocyanin (KLH).
  • peptides derived from the full Brevican sequence may be utilized.
  • one or more 8-30 amino acid peptide portions of an extracellular domain of BEHAB are utilized, with peptides in the range of 10-20 being a more economical choice.
  • Custom-synthesized peptides in this range are available from a multitude of vendors, and can be order conjugated to KLH or BSA.
  • peptides in excess of 30 amino acids may be synthesized by solid-phase methods, or may be recombinantly produced in a suitable recombinant protein production system.
  • an animal cell system e.g., Sf9 or other insect cells, CHO or other mammalian cells
  • an animal cell system e.g., Sf9 or other insect cells, CHO or other mammalian cells
  • HA hyaluronic acid
  • antibody moieties are utilized which bind to BEHAB at a site on the protein which alters the binding of an extracellular ligand molecule (e.g., HA) to BEHAB.
  • BEHAB activity altering antibodies may be utilized in therapeutic compositions in an unconjugated form (e.g., the antibody in an acceptable pharmaceutical carrier), or may be conjugated to either a therapeutic moiety (creating a double-acting therapeutic agent) or an imaging moiety (creating a duel therapeutic/imaging agent).
  • Selection of antibodies that alter (enhance or inhibit) the binding of a ligand to BEHAB may be accomplished by a straightforward binding inhibition/enhancement assay.
  • a labeled (e.g., fluorescently or enzyme-labeled) antibody to BEHAB which has been immobilized in a microtiter well, is assayed in both the presence and absence of the ligand.
  • the change in binding is indicative of either an enhancer (increased binding) or competitive inhibitor (decreased binding) relationship between the antibody and the ligand.
  • Such assays may be carried out in high-throughput formats (e.g., 384 well plate formats, in robotic systems) for the automated selection of monoclonal antibody candidates for use as BEHAB ligand-binding inhibitors or enhancers.
  • antibodies that are useful for altering the function of BEHAB may be assayed in functional formats, such as the HUVEC tube assay and the cell migration assay described below.
  • functional formats such as the HUVEC tube assay and the cell migration assay described below.
  • CD-44 was selected as a target for selective immuno-therapeutic agents in targeting and/or imaging brain tumors.
  • the complete cDNA sequence encoding CD-44E is provided in SEQ ID NO. 15, and the complete amino acid sequence of CD-44, indicating various splicing variation locations, is provided in SEQ ID NO. 16.
  • CD-44 is a proteoglycan that is expressed as two major splice variants.
  • CD-44E is a 150 kDa protein isolated from epithelial cells.
  • CD-44E has a C-terminal cytoplasmic tail, a hydrophobic transmembrane domain of 23 amino acids, and an N-terminal extracellular region of 248 amino acids.
  • CD-44E The extracellular domain is O-glycosylated and also binds chondroitin sulfate.
  • CD-44E it has two of the three immunodominant epitope clusters of native gp90Hermes.
  • CD-44E contains an additional 132 amino acids in the extracellular region.
  • CD-44H is a 90 kDa protein isolated from hematopoietic cells.
  • CD-44R1 and CD-44R2 are 2 isoforms expressed by hematopoietic cells.
  • the complete cDNA sequence of the 90 kDa CD-44H isoform consist of 1795 bps, encoding a 341 amino acid protein.
  • a compound that specifically binds to CD-44 is any compound (such as an antibody) that has a binding affinity for any naturally occurring isoform, splice variant, or polymorphism of CD-44, explicitly including the isoforms described herein.
  • specific binding compounds e.g., antibodies
  • proteins include truncated forms or domains of CD-44, and recombinantly engineered alterations of CD-44.
  • a portion of SEQ ID NO. 16 may be engineered to include a non-naturally occurring cysteine for cross-linking to an immunoconjugate protein, as described below.
  • CD-44H protein has an overall primary structure of 90 kDa, which consist of 341 amino acids.
  • the N-terminus is located outside of the cell and the extracellular domain consist of 248 amino acids.
  • the C-terminus is located inside of the cell and the intracellular domain consist of 72 amino acids, while the transmembrane region consist of 21 amino acids.
  • the CD-44 gene contains 20 exons, of which exons 1-5, 15-17 and 19 encode the CD44H isoform.
  • the intervening exons 6, 6a, 7-14 also designated v1-v10) are alternatively spliced to generate the variant isoforms with an insertion at the membrane proximal region of the extracellular domain between amino acids 202 and 203. See Bajorath (2000). Proteins: structure, function, and genetic, 39:103-111; and Ilangumaram et al. Leukemia and Lymphoma, 35:455-469.
  • CD-44 When raising antibodies to CD-44, the entire protein, or a portion thereof, may be utilized. For instance, any portion of the extracellular domain may be utilized. For instance, the amino acids between the signal sequence and amino acid 202 may be used. Larger CD-44 proteins and domains may be produced utilizing any suitable recombinant vector/protein production system, such as the baculovirus transfection system outlined below, after being amplified from a fetal brain cDNA library (as available from, e.g., Clontech, Palo Alto, Calif.) or another suitable source.
  • a fetal brain cDNA library as available from, e.g., Clontech, Palo Alto, Calif.
  • antibodies may be raised by immunizing the production animal with the protein and a suitable adjuvant (e.g., Fruend's, Fruend's complete, oil-in-water emulsions, etc.).
  • a suitable adjuvant e.g., Fruend's, Fruend's complete, oil-in-water emulsions, etc.
  • the CD-44 protein (or a portion thereof) can serve as the CD-44 antigen.
  • Commonly utilized conjugate proteins that are commercially available for such use include bovine serum albumin (BSA) and keyhole limpet hemocyanin (KLH).
  • peptides derived from the full CD-44 sequence may be utilized.
  • one or more 8-30 amino acid peptide portions of an extracellular domain of CD-44 are utilized, with peptides in the range of 10-20 being a more economical choice.
  • Custom-synthesized peptides in this range are available from a multitude of vendors, and can be order conjugated to KLH or BSA.
  • peptides in excess of 30 amino acids may be synthesized by solid-phase methods, or may be recombinantly produced in a suitable recombinant protein production system.
  • an animal cell system e.g., Sf9 or other insect cells, CHO or other mammalian cells
  • Sf9 or other insect cells e.g., Sf9 or other insect cells, CHO or other mammalian cells
  • Other information useful in designing an antigen for the production of antibodies to CD-44, including glycosylation sites, is provided in SEQ ID NO. D′.
  • Hyaluronan is a polymeric glycosaminoglycan and a major component of the extracellular matrix.
  • CD-44 is one of the principal receptors for HA. Within the normal CNS, the CD-44 protein has been localized to astrocytes in the white matter. CD-44H has been shown to be the predominant isoform in normal brain and neuroectoderm-derived tumors. Hence, the up-regulation of CD-44 may be a crucial step in brain tumor invasiveness and migration.
  • antibody moieties are utilized which bind to CD-44 at a site on the protein which alters the binding of an extracellular ligand molecule (e.g., HA) to CD-44.
  • CD-44 activity altering antibodies may be utilized in therapeutic compositions in an unconjugated form (e.g., the antibody in an acceptable pharmaceutical carrier), or may be conjugated to either a therapeutic moiety (creating a double-acting therapeutic agent) or an imaging moiety (creating a duel therapeutic/imaging agent).
  • Selection of antibodies that alter (enhance or inhibit) the binding of a ligand to CD-44 may be accomplished by a straightforward binding inhibition/enhancement assay.
  • a labeled (e.g., fluorescently or enzyme-labeled) antibody to CD-44 which has been immobilized in a microtiter well, is assayed in both the presence and absence of the ligand.
  • the change in binding is indicative of either an enhancer (increased binding) or competitive inhibitor (decreased binding) relationship between the antibody and the ligand.
  • Such assays may be carried out in high-throughput formats (e.g., 384 well plate formats, in robotic systems) for the automated selection of monoclonal antibody candidates for use as CD-44 ligand-binding inhibitors or enhancers.
  • antibodies that are useful for altering the function of CD-44 may be assayed in functional formats, such as endothelial sprouting assay and cell migration assay.
  • functional formats such as endothelial sprouting assay and cell migration assay.
  • TSPAN3 was selected as a target for selective immuno-therapeutic agents in targeting and/or imaging brain tumors.
  • the complete cDNA sequence encoding TSPAN3 is provided in SEQ ID NO. 17, and the complete amino acid sequence of TSPAN3 is provided in SEQ ID NO. 18.
  • Tetraspanin is a 253 amino acid membrane bound protein. No isoforms have been isolated to date.
  • TSPAN3, as is characteristic of the tetraspanin family, contains four transmembrane domains, putatively comprising amino acids 12-32, 51-71, 86-106, and 213-233.
  • the protein has two putative extracellular domains, amino acids 33-50 and 107-212, and three putative cytoplasmic domains, amino acids 1-11, 72-85, and 234-235. Putative N-linked glycosylation sites are listed in SEQ ID NO. 18.
  • a compound that specifically binds to TSPAN3 is any compound (such as an antibody) that has a binding affinity for any naturally occurring isoform, splice variant, or polymorphism of TSPAN3.
  • specific binding compounds e.g., antibodies
  • proteins include truncated forms or domains of TSPAN3, and recombinantly engineered alterations of TSPAN3.
  • a portion of SEQ ID NO. 18 may be engineered to include a non-naturally occurring cysteine for cross-linking to an immunoconjugate protein, as described below.
  • the antibodies utilized in the compositions and methods of the invention bind to the membrane-bound isoform of the protein, as this will more specifically target the cytotoxic therapeutic agent, or the imaging agent, to the brain tumor cell.
  • the only currently known form of TSPAN3 is membrane-bound.
  • embodiments that utilize antibodies that bind to any secreted isoform of the protein are also useful in the invention, as one of ordinary skill would expect that the concentration of the secreted isoform would also be increased adjacent to brain tumor cells which over-express the protein.
  • the antibodies utilized in the invention bind to an extracellular domain of the protein, as are described in the SEQ ID NO. 18.
  • cysteine residues at positions 147, 148, and 197 of SEQ ID NO. 18 in the second extracellular domain are highly conserved among the tetraspanin family and are thought to be essential for proper tetraspanin function.
  • the antibodies utilized in the invention bind to an epitope comprising, or alternatively very near to, one of these cysteine residues.
  • TSPAN3 When raising antibodies to TSPAN3, the entire protein, or a portion thereof, may be utilized. For instance, any one of the aforementioned domains of the secreted protein or an extracellular portion of the truncated, membrane bound GPI form may be utilized. For instance, amino acids that constitute one of the extracellular domains, amino acids 33-50 or 107-212, may be used. Larger TSPAN3 proteins and domains may be produced utilizing any suitable recombinant vector/protein production system, such as the baculovirus transfection system outlined below, after being amplified from a fetal brain cDNA library (as available from, e.g., Clontech, Palo Alto, Calif.) or another suitable source.
  • a fetal brain cDNA library as available from, e.g., Clontech, Palo Alto, Calif.
  • antibodies may be raised by immunizing the production animal with the protein and a suitable adjuvant (e.g., Fruend's, Fruend's complete, oil-in-water emulsions, etc.).
  • a suitable adjuvant e.g., Fruend's, Fruend's complete, oil-in-water emulsions, etc.
  • the tetraspanin 3 protein (or a portion thereof) can serve as the TSPAN3 antigen.
  • Commonly utilized conjugate proteins that are commercially available for such use include bovine serum albumin (BSA) and keyhole limpet hemocyanin (KLH).
  • peptides derived from the full Brevican sequence may be utilized.
  • one or more 8-30 amino acid peptide portions of an extracellular domain of TSPAN3 are utilized, with peptides in the range of 10-20 being a more economical choice.
  • Custom-synthesized peptides in this range are available from a multitude of vendors, and can be order conjugated to KLH or BSA.
  • peptides in excess of 30 amino acids may be synthesized by solid-phase methods, or may be recombinantly produced in a suitable recombinant protein production system.
  • an animal cell system e.g., Sf9 or other insect cells, CHO or other mammalian cells
  • Sf9 or other insect cells e.g., Sf9 or other insect cells, CHO or other mammalian cells
  • Other information useful in designing an antigen for the production of antibodies to TSPAN3, including glycosylation sites, is provided in SEQ ID NO. 18.
  • TSPAN3 activity altering antibodies may be utilized in therapeutic compositions in an unconjugated form (e.g., the antibody in an acceptable pharmaceutical carrier), or may be conjugated to either a therapeutic moiety (creating a double-acting therapeutic agent) or an imaging moiety (creating a duel therapeutic/imaging agent).
  • Selection of antibodies that alter (enhance or inhibit) the binding of a ligand to TSPAN3 may be accomplished by a straightforward binding inhibition/enhancement assay.
  • a labeled (e.g., fluorescently or enzyme-labeled) antibody to TSPAN3 which has been immobilized in a microtiter well, is assayed in both the presence and absence of the ligand.
  • the change in binding is indicative of either an enhancer (increased binding) or competitive inhibitor (decreased binding) relationship between the antibody and the ligand.
  • Such assays may be carried out in high-throughput formats (e.g., 384 well plate formats, in robotic systems) for the automated selection of monoclonal antibody candidates for use as TSPAN3 ligand-binding inhibitors or enhancers.
  • antibodies that are useful for altering the function of TSPAN3 may be assayed in functional formats, such as the HUVEC tube assay and the cell migration assay described below.
  • functional formats such as the HUVEC tube assay and the cell migration assay described below.
  • VIPR-2 was selected as a prime target for selective immuno-therapeutic agents in targeting and/or imaging brain tumors.
  • the complete cDNA sequence encoding VIPR-2 is provided in SEQ ID NO. 19, and the complete amino acid sequence of VIPR-2 is provided in SEQ ID NO. 20.
  • VIPR-2 is a seven transmembrane spanning G-protein receptor.
  • the complete VIPR-2 protein is encoded by 13 exons.
  • the initiator codon of the approximated 438 amino acid-encoding open reading frame is located in exon 1 and the termination signal is located in exon 13.
  • the 5′ untranslated region extends 187 bp upstream of the initiator codon and is extremely GC-rich (80%).
  • the polyadenylation signal is located 2416 bp downstream of the stop codon. Intron sizes range from 68 bp (intron 11) to 45 bp (intron 4), the entire human gene spans 117 kb, while the cDNA sequence spans 1317 bp. Recent studies have also isolated two VIP-2 receptor mRNAs of 4.6 kb and 2.3 kb in size.
  • a compound that specifically binds to VIPR-2 is any compound (such as an antibody) that has a binding affinity for any naturally occurring isoform, splice variant, or polymorphism of VIPR-2, explicitly including any isoforms described herein.
  • specific binding compounds e.g., antibodies
  • proteins include truncated forms or domains of VIPR-2, and recombinantly engineered alterations of VIPR-2.
  • a portion of SEQ ID NO. 20 may be engineered to include a non-naturally occurring cysteine for cross-linking to an immunoconjugate protein, as described below.
  • the antibodies utilized in the compositions and methods of the invention bind to the membrane-bound isoforms of the protein, as this will more specifically target the cytotoxic therapeutic agent, or the imaging agent, to the brain tumor cell.
  • embodiments that utilize antibodies that bind to the secreted isoform of the protein are also useful in the invention, as one of ordinary skill would expect that the concentration of the secreted isoform would also be increased adjacent to brain tumor cells which over-express the protein.
  • the amino acid sequence of full length VIPR-2 consists of 437 amino acids with a predicted molecular mass is 49 kDa, as the sequence was first deduced by Lutz et al. FEBS. 334:3-8, 1993. Lutz et al.
  • the receptor is a seven membrane spanning protein where in the first 22 amino acids constitute a typical hydrophobic signal sequence, and the remaining amino acids constitute two membrane spanning regions between amino acids 127 to 148 and 158 to 178, two more membrane spanning domains between amino acids 202 to 227 and 238 to 261, another between 278 to 303, and two final membrane spanning regions between 327 to 347 and 359 to 380, with three potential N-linked glycosylation sites found in the amino terminal extracellular domain at residues 57, 87 and 91.
  • Sreedharan et al. describes the VIPR-2 receptor as being a 457 amino-acid protein encoded by a 2.8 kb cDNA of 52 kDa.
  • the entire protein or a portion thereof may be utilized.
  • the extracellular domains of any of the seven transmembrane spanning portions of the protein may be utilized.
  • amino acids 179 to 201 may be used.
  • Larger VIPR-2 proteins and domains may be produced utilizing any suitable recombinant vector/protein production system, such as the baculovirus transfection system outlined below, after being amplified from a fetal brain cDNA library (as available from, e.g., Clontech, Palo Alto, Calif.) or another suitable source.
  • antibodies may be raised by immunizing the production animal with the protein and a suitable adjuvant (e.g., Fruend's, Fruend's complete, oil-in-water emulsions, etc.).
  • a suitable adjuvant e.g., Fruend's, Fruend's complete, oil-in-water emulsions, etc.
  • the VIPR-2 protein (or a portion thereof) can serve as the VIPR-2 antigen.
  • Commonly utilized conjugate proteins that are commercially available for such use include bovine serum albumin (BSA) and keyhole limpet hemocyanin (KLH).
  • peptides derived from the full VIPR-2 sequence may be utilized.
  • one or more 8-30 amino acid peptide portions of an extracellular domain of VIPR-2 are utilized, with peptides in the range of 10-20 being a more economical choice.
  • Custom-synthesized peptides in this range are available from a multitude of vendors, and can be order conjugated to KLH or BSA.
  • peptides in excess of 30 amino acids may be synthesized by solid-phase methods, or may be recombinantly produced in a suitable recombinant protein production system.
  • an animal cell system e.g., Sf9 or other insect cells, CHO or other mammalian cells
  • Sf9 or other insect cells e.g., Sf9 or other insect cells, CHO or other mammalian cells
  • Other information useful in designing an antigen for the production of antibodies to VIPR-2, including glycosylation sites, is provided in SEQ ID NO. 20.
  • the extracellular domain of human VIPR-2 binds PACAP-27, PACAP-38, VIP and secretin. Because these factors have been found to affect tumor cell growth, and due to the recent discovery that the VIPR-2 receptor is overexpressed in glioblastomas (Astrocytoma grade IV), the binding of these factors to the VIPR-2 receptor may play a significant role in the oncogenesis and growth of astrocytoma cells in the brain.
  • antibody moieties are utilized which bind to VIPR-2 at a site on the protein which alters the binding of extracellular ligand molecules, such as VIP, to VIPR-2.
  • VIPR-2 activity altering antibodies may be utilized in therapeutic compositions in an unconjugated form (e.g., the antibody in an acceptable pharmaceutical carrier), or may be conjugated to either a therapeutic moiety (creating a double-acting therapeutic agent) or an imaging moiety (creating a duel therapeutic/imaging agent).
  • Selection of antibodies that alter (enhance or inhibit) the binding of a ligand to VIPR-2 may be accomplished by a straightforward binding inhibition/enhancement assay.
  • a labeled (e.g., fluorescently or enzyme-labeled) antibody to VIPR-2 which has been immobilized in a microtiter well, is assayed in both the presence and absence of the ligand (e.g., vasoactive intestinal peptide.)
  • the change in binding is indicative of either an enhancer (increased binding) or competitive inhibitor (decreased binding) relationship between the antibody and the ligand.
  • Such assays may be carried out in high-throughput formats (e.g., 384 well plate formats, in robotic systems) for the automated selection of monoclonal antibody candidates for use as VIPR-2 ligand-binding inhibitors or enhancers.
  • antibodies that are useful for altering the function of VIPR-2 may be assayed in functional formats, such as the HUVEC tube assay and cell migration assay.
  • functional formats such as the HUVEC tube assay and cell migration assay.
  • PTN was selected as a target for selective immuno-therapeutic agents in targeting and/or imaging brain tumors.
  • the entire PTN gene spans 65 kb and 7 exons, while the mature protein is approximately 136 amino acids (after cleavage of a 32 amino acid signal peptide) with distinctive lysine and arginine-rich clusters within both N- and C-terminal domains.
  • the complete cDNA sequence encoding PTN is provided in SEQ ID NO. 21, and the complete amino acid sequence of PTN is provided in SEQ ID NO. 22.
  • PTN is a 18 kDa, single chain, secreted protein with 10 conserved disulfide linked cysteine residues.
  • a compound that specifically binds to PTN is any compound (such as an antibody) that has a binding affinity for any naturally occurring isoform, splice variant, or polymorphism of PTN, explicitly including the isoforms described herein.
  • specific binding compounds e.g., antibodies
  • proteins include truncated forms or domains of PTN, and recombinantly engineered alterations of PTN.
  • a portion of SEQ ID NO. 22 may be engineered to include a non-naturally occurring cysteine for cross linking to an immunoconjugate protein, as described.
  • the gene sequence of PTN isolated from human genomic DNA consists of five exons and four introns. While exon 1 does not encode an amino acid sequence, exon 2 encodes the hydrophobic signal sequence of 32 amino acids, exons 3 and 4 code for the amino terminal and the ten cysteine residues, and exon 5 codes for the highly basic C-terminal domains.
  • the human cDNA starts toward the end of exon 1, while the coded for protein begins at exon 2.
  • the mature protein consist of 136 amino acids encoded by exons 2 to 5.
  • the minimum size of the gene is 42 kb, with a mRNA of 1650 nucleotides, spanning five exons, the majority of the protein being coded for by exon 3 (174 base pairs in length) and exon 4 (162 base pairs in length).
  • exon 3 174 base pairs in length
  • exon 4 162 base pairs in length
  • the entire protein or a portion thereof may be utilized.
  • amino acid domains encoded for by exons 3 and 4 i.e. amino acids 7 to 64 or 65 to 118, respectfully.
  • residues 41 to 64 may be used to abolish the transformation potential of PTN.
  • Larger PTN proteins and domains may be produced utilizing any suitable recombinant vector/protein production system, such as the baculovirus transfection system outlined below, after being amplified from a fetal brain cDNA library (as available from, e.g., Clontech, Palo Alto, Calif.) or another suitable source. It is to be noted that antibodies which bind to this secreted protein are useful in cytotoxic and imaging embodiments of the invention, as one of ordinary skill would expect that the concentration of the PTN would be increased adjacent to tumor cells which over-express the protein.
  • antibodies may be raised by immunizing the production animal with the protein and a suitable adjuvant (e.g., Fruend's, Fruend's complete, oil-in-water emulsions, etc.).
  • a suitable adjuvant e.g., Fruend's, Fruend's complete, oil-in-water emulsions, etc.
  • the PTN protein (or a portion thereof) can serve as the PTN antigen.
  • a smaller peptide it is advantageous to conjugate the peptide with a larger molecule to make an immunostimulatory conjugate for use as the PTN antigen.
  • conjugate proteins that are commercially available for such use include bovine serum albumin (BSA) and keyhole limpet hemocyanin (KLH).
  • peptides derived from the full PTN sequence may be utilized.
  • one or more 8-30 aa peptide portions of the protein are utilized, with peptides in the range of 10-20 being a more economical choice.
  • Custom-synthesized peptides in this range are available from a multitude of vendors, and can be order conjugated to KLH or BSA.
  • peptides in excess of 30 amino acids may be synthesized by solid-phase methods, or may be recombinantly produced in a suitable recombinant protein production system.
  • an animal cell system e.g., Sf9 or other insect cells, CHO or other mammalian cells
  • Sf9 or other insect cells e.g., Sf9 or other insect cells, CHO or other mammalian cells
  • Other information useful in designing an antigen for the production of antibodies to PTN, including glycosylation sites, is provided in SEQ ID NO. 22.
  • PTN has been shown to bind to extracellular domain of RPTP beta and zeta. This binding inactivates the catalytic activity of RPTP, and PTN binds all the three major isoforms pf RPTP beta and zeta. PTN has also been shown to interact with syndecan-3.
  • antibody moieties are utilized which bind to PTN at a site on the protein that alters the binding of a cell surface molecule, such as the ones listed above, to PTN.
  • Such PTN activity altering antibodies may be utilized in therapeutic compositions in an unconjugated form (e.g., the antibody in an acceptable pharmaceutical carrier), or may be conjugated to either a therapeutic moiety (creating a double-acting therapeutic agent) or an imaging moiety (creating a duel therapeutic/imaging agent).
  • Selection of antibodies which alter (enhance or inhibit) the binding of a ligand to PTN may be accomplished by a straightforward binding inhibition/enhancement assay.
  • a labeled (e.g., fluorescently or enzyme-labeled) antibody to PTN which has been immobilized in a microtiter well, is assayed in both the presence and absence of the appropriate ligand.
  • the change in binding is indicative of either an enhancer (increased binding) or competitive inhibitor (decreased binding) relationship between the antibody and the ligand.
  • Such assays may be carried out in high-throughput formats (e.g., 384 well plate formats, in robotic systems) for the automated selection of monoclonal antibody candidates for use as PTN ligand-binding inhibitors or enhancers.
  • antibodies which are useful for altering the function of PTN may be assayed in functional formats, such as the HUVEC tube assay and the cell migration assay described below.
  • antibodies that exhibit the appropriate anti-PTN activity may be selected without direct knowledge of a binding ligand or the particular biomolecular interactions of PTN.
  • OPN was selected as a target for selective immuno-therapeutic agents in targeting and/or imaging brain tumors.
  • the mature protein consists of approximately 298 amino acids (after cleavage of a 16 amino acid signal peptide) and contains two potential Asn-Xaa-Ser N-glycosylation site, located at positions 65 and 92 of the mature protein.
  • the complete cDNA sequence encoding OPN is provided in SEQ ID NO. 23, and the complete amino acid sequence of OPN is provided in SEQ ID NO. 24.
  • OPN is an abundant 34 kDa, single chain, phosphorylated glycoprotein, with a presumed site for cell attachment at residues 144-148.
  • OPN-A and OPN-B differ by the addition of 14 amino acids at residue 58 of the protein. Amino acids 58-71 are absent in OPN-B, and amino acids 31-57 are absent in OPN-C.
  • OPN is a negatively charged, highly hydrophilic secreted protein.
  • a compound that specifically binds to OPN is any compound (such as an antibody) that has a binding affinity for any naturally occurring isoform, splice variant, or polymorphism of OPN, explicitly including the three isoforms described herein.
  • specific binding compounds e.g., antibodies
  • proteins include truncated forms or domains of OPN, and recombinantly engineered alterations of OPN.
  • a portion of SEQ ID NO. 24 may be engineered to include a non-naturally occurring cysteine for cross linking to an immunoconjugate protein, as described.
  • the cDNA sequence of OPN isolated from human bone cells has an overall structure of approximately 34 kDA that consist of 298 amino acids, which is 14 amino acids less than the cDNA sequence of OPN isolated from human osteosarcoma by Keifer et al. (OPN-B).
  • the cDNA transcript for OPN-A is 1.5 kb with an open reading frame of 900 nucleotides, of which the first 16 amino acids are hydrophobic in nature and probably constitute a signal sequence for the secreted protein.
  • the OPN gene contains 7 exons that are alternatively spliced to generate the variant isoforms, the most common variant being the addition of a 42 bp (14 amino acid) sequence located at base 280 of OPN-A. See Young et al. (1990). Genomics, 7:491-502 and Keifer et al. Nucleic Acids Res. 17:3306.
  • the entire protein or a portion thereof may be utilized. For instance, amino acid domains 4 to 12 (from the N-terminus) or 29 to 37 (from the N-terminus) may be utilized. Larger OPN proteins and domains may be produced utilizing any suitable recombinant vector/protein production system, such as the baculovirus transfection system outlined below, after being amplified from a fetal brain cDNA library (as available from, e.g., Clontech, Palo Alto, Calif.) or another suitable source. It is to be noted that antibodies which bind to this secreted protein are useful in cytotoxic and imaging embodiments of the invention, as one of ordinary skill would expect that the concentration of OPN would be increased adjacent to tumor cells which over-express the protein.
  • antibodies may be raised by immunizing the production animal with the protein and a suitable adjuvant (e.g., Fruend's, Fruend's complete, oil-in-water emulsions, etc.).
  • a suitable adjuvant e.g., Fruend's, Fruend's complete, oil-in-water emulsions, etc.
  • the OPN protein (or a portion thereof) can serve as the OPN antigen.
  • Commonly utilized conjugate proteins that are commercially available for such use include bovine serum albumin (BSA) and keyhole limpet hemocyanin (KLH).
  • peptides derived from the full OPN sequence may be utilized.
  • one or more 8-30 aa peptide portions of the protein are utilized, with peptides in the range of 10-20 being a more economical choice.
  • Custom-synthesized peptides in this range are available from a multitude of vendors, and can be order conjugated to KLH or BSA.
  • peptides in excess of 30 amino acids may be synthesized by solid-phase methods, or may be recombinantly produced in a suitable recombinant protein production system.
  • an animal cell system e.g., Sf9 or other insect cells, CHO or other mammalian cells
  • Sf9 or other insect cells e.g., Sf9 or other insect cells, CHO or other mammalian cells
  • Other information useful in designing an antigen for the production of antibodies to OPN, including glycosylation sites, is provided in SEQ ID NO. 22.
  • the cell attachment sequence of human OPN (amino acids 144 to 148) is believed to interact with various cell surface proteins (such as CD-44) to affect cell adhesion, and a highly acidic stretch composed almost exclusively of aspartic acid residues (amino acids 72 to 81) is believed to be the mineral binding site within the protein. Because CD-44 is frequently over expressed on primary brain tumors and metastases the binding of OPN to these various cell-surface adhesion protein molecules may play a significant role in the senescence and growth of tumor cells in the brain.
  • antibody moieties are utilized which bind to OPN at a site on the protein that alters the binding of a cell surface molecule, e.g., CD-44, to OPN.
  • OPN activity altering antibodies may be utilized in therapeutic compositions in an unconjugated form (e.g., the antibody in an acceptable pharmaceutical carrier), or may be conjugated to either a therapeutic moiety (creating a double-acting therapeutic agent) or an imaging moiety (creating a duel therapeutic/imaging agent).
  • Selection of antibodies which alter (enhance or inhibit) the binding of a ligand to OPN may be accomplished by a straightforward binding inhibition/enhancement assay.
  • a labeled (e.g., fluorescently or enzyme-labeled) antibody to OPN which has been immobilized in a microtiter well, is assayed in both the presence and absence of the appropriate ligand.
  • the change in binding is indicative of either an enhancer (increased binding) or competitive inhibitor (decreased binding) relationship between the antibody and the ligand.
  • Such assays may be carried out in high-throughput formats (e.g., 384 well plate formats, in robotic systems) for the automated selection of monoclonal antibody candidates for use as OPN ligand-binding inhibitors or enhancers.
  • antibodies which are useful for altering the function of OPN may be assayed in functional formats, such as the HUVEC tube assay and the cell migration assay described below.
  • antibodies that exhibit the appropriate anti-OPN activity may be selected without direct knowledge of a the biomolecular role of OPN.
  • PTP ⁇ was also selected as a prime target for selective immuno-therapeutic agents in treating or imaging brain tumors.
  • the complete cDNA sequence encoding PTP ⁇ is provided in SEQ ID NO. 5, and the complete amino acid sequence of PTP ⁇ is provided in SEQ ID NO. 6.
  • Three different splice variants have been described, which include two membrane bound variants (full length: PTP ⁇ - ⁇ , and shorter version PTP ⁇ - ⁇ ) and one secreted form (Phosphacan). See FIG. 1.
  • Isoform PTP ⁇ - ⁇ is the full length isoform, which contains the primary amino acid sequence aa 25-2314 of SEQ ID NO. 6 (aa 1-24 are a signal polypeptide).
  • PTP ⁇ This full length long form of PTP ⁇ is a type I membrane protein. After the signal peptide it contains a carbonic anhydrase like (CAH) and a fibronectin type III like (FN3) domain, followed by a long cysteine free spacer (S) domain. This follows a 860 amino acid long insert domain, which can be glycosylated. After a single transmembrane segment, in the intracellular region it has 2 phosphatase domains, but only the membrane-proximal PTPase domain is catalytically active (Krueger 1992).
  • CAH carbonic anhydrase like
  • FN3 fibronectin type III like
  • S cysteine free spacer
  • Isoform PTP ⁇ - ⁇ 755-1614 are missing.
  • Isoform PTP ⁇ -S phosphacan
  • Northern Blot analysis have shown that the PTP zeta is exclusively expressed in the human central nervous system. In mouse embryos, the PTP ⁇ transcript was mainly detected in the ventricular and subventricular zone of the brain and the spinal cord. The same pattern was detected in adult mice.
  • a compound which specifically binds to human protein tyrosine phosphatase-zeta is any compound (such as an antibody) which has a binding affinity for any naturally occurring isoform, spice variant, or polymorphism of PTP ⁇ , explicitly including the three splice variants describe herein.
  • PTP ⁇ SM1 and PTP ⁇ SM2 are subsets of compounds which specifically bind to PTP ⁇ .
  • such “specific” binding compounds may also bind to other closely related proteins which exhibit significant homology (such as greater than 90% identity, more preferably greater than 95% identity, and most preferably greater than 99% identity) with the amino acid sequence of PTP ⁇ .
  • proteins include truncated forms or domains of PTP ⁇ , and recombinantly engineered alterations of PTP ⁇ .
  • an portion of SEQ ID NO. 6 may be engineered to include a non-naturally occurring cysteine for cross-linking to an immunoconjugate protein, as described below.
  • the antibodies utilized in the compositions and methods of the invention bind to the membrane-bound isoforms of the protein, as this will more specifically target the cytotoxic therapeutic agent, or the imaging agent, to the brain tumor cell.
  • embodiments which utilize antibodies which bind to the secreted isoform of the protein are also useful in the invention, as one of ordinary skill would expect that the concentration of the secreted isoform would also be increased adjacent to brain tumor cells which over-express the protein.
  • the amino acid sequence of full length PTP ⁇ consists of 2307 amino acids, as the sequence was deduced by Levy (in which aa 1722-1728 of SEQ ID NO. 2 were missing) (See also U.S. Pat. Nos. 5,604,094, and 6,160,090, fully incorporated herein by reference), or 2314 amino acids as the sequence was deduced by Krueger, et al., (“A human transmembrane protein-tyrosine phosphatase, PTP zeta, is expressed in brain and has an N-terminal receptor domain homologous to carbonic anhydrases” Proc. Nat. Acad. Sci. U.S.A. 89:7417-7421 (1992)).
  • Amino acids 1-24 of SEQ ID NO. 6 are a signal sequence which directs the proper placement of the transmembrane protein.
  • the extracellular domain of the mature PTP ⁇ protein spans amino acids 25-1635 of SEQ ID NO. 6 in the long and secreted forms (this forms the entire secreted form), and amino acids 25-754,1615-1635 in the short isoform.
  • the transmembrane region of the protein spans amino acids 1636-1661 of SEQ ID NO. 6, and the balance of the protein forms the cytoplasmic domain, amino acids 1662-2314.
  • the entire protein (any of the three isoforms) or a portion thereof may be utilized.
  • the extracellular domain of the long or short form, the entire secreted form, or a portion of extracellular domain may be utilized.
  • amino acids 25-754 which are common to both ⁇ and ⁇ isoforms, may be used.
  • Such larger PTP ⁇ proteins and domains may be produced utilizing any suitable recombinant vector/protein production system, such as the baculovirus transfection system outlined below, after being amplified from a fetal brain cDNA library (as available from, e.g., Clontech, Palo Alto, Calif.) or another suitable source.
  • antibodies may be raised by immunizing the production animal with the protein and a suitable adjuvant (e.g., Fruend's, Fruend's complete, oil-in-water emulsions, etc.).
  • a suitable adjuvant e.g., Fruend's, Fruend's complete, oil-in-water emulsions, etc.
  • the PTP ⁇ protein (or a portion thereof) can serve as the PTP ⁇ antigen.
  • conjugate proteins which are commercially available for such use include bovine serum albumin (BSA) and keyhole limpet hemocyanin (KLH).
  • peptides derived from the full PTP ⁇ sequence may be utilized.
  • one or more 8-30 aa peptide portions of an extracellular domain of PTP ⁇ are utilized, with peptides in the range of 10-20 being a more economical choice.
  • Custom-synthesized peptides in this range are available from a multitude of vendors, and can be order conjugated to KLH or BSA.
  • peptides in excess of 30 amino acids may be synthesized by solid-phase methods, or may be recombinantly produced in a suitable recombinant protein production system.
  • an animal cell system e.g., Sf9 or other insect cells, CHO or other mammalian cells
  • Sf9 or other insect cells e.g., Sf9 or other insect cells, CHO or other mammalian cells
  • Other information useful in designing an antigen for the production of antibodies to PTP ⁇ , including glycosylation sites, is provided in SEQ ID NO. 6.
  • the extracellular domain of human PTP ⁇ is known to bind to tenascin-C, tenascin-R, pleiotrophin (NM — 002825), midkine (NM — 002391), FGF-2 (XM — 00366), Nr-CAM 1NM — 005010), L1/Ng-CAM, contactin (NM — 001843), N-CAM (XM — 006332), and axonin-1NM — 005076.)
  • the first 5 of these molecules are either components of the extracellular matrix in gliomas or are soluble factors known to be present in gliomas, and the latter 4 are neuronal surface molecules.
  • PTP ⁇ activity altering antibodies may be utilized in therapeutic compositions in an unconjugated form (e.g., the antibody in an acceptable pharmaceutical carrier), or may be conjugated to either a therapeutic moiety (creating a double-acting therapeutic agent) or an imaging moiety (creating a duel therapeutic/imaging agent).
  • Selection of antibodies which alter (enhance or inhibit) the binding of a ligand to PTP ⁇ may be accomplished by a straightforward binding inhibition/enhancement assay.
  • a labeled (e.g., fluorescently or enzyme-labeled) antibody to PTP ⁇ which has been immobilized in a microtiter well, is assayed in both the presence and absence of the ligand.
  • the change in binding is indicative of either an enhancer (increased binding) or competitive inhibitor (decreased binding) relationship between the antibody and the ligand.
  • Such assays may be carried out in high-throughput formats (e.g., 384 well plate formats. in robotic systems) for the automated selection of monoclonal antibody candidates for use as PTP ⁇ ligand-binding inhibitors or enhancers.
  • antibodies which are useful for altering the function of PTP ⁇ may be assayed in functional formats, such as the HUVEC tube assay and the cell migration assay described below.
  • antibodies that exhibit the appropriate anti-PTP ⁇ activity may be selected without direct knowledge of a the biomolecular role of PTP ⁇ .
  • the protein PTP ⁇ SM1 (amino acid sequence SEQ ID NO. 2, cDNA sequence SEQ ID NO. 1) comprises the amino acids encoded by the first nine exons of PTP ⁇ - ⁇ , with three unique additional carboxy terminal amino acids, see FIG. 2. These are encoded by additional 3′ mRNA sequence (nucleotides 1262-1272 of SEQ ID NO. 1) from the intron of the gene between exons nine and ten.
  • the PTP ⁇ SM1 clone was isolated from a human fetal brain cDNA library, an has been shown to be expressed in several human glioblastoma cell lines. Expression of the SM1 splice variant has also been confirmed in primary brain tumor samples.
  • the protein comprises only extracellular domains of PTP ⁇ , and is expected to be secreted by the cell.
  • PTP ⁇ SM1 may serve a cell signaling or messenger function, and may have bind to a receptor on the surface of cells which are associated with or part of central nervous system tissues.
  • antibodies specific for PTP ⁇ SM1, and not specific for the other splicing isoforms of PTP ⁇ may be especially efficacious in the brain tumor therapeutic or imaging compositions of the invention.
  • the PTP ⁇ SM1 protein mainly comprises the carbonic anhydrase-like domain which has been identified in PTP ⁇ ⁇ .
  • CA domain could function as a carbonic anhydrase enzyme.
  • Two of the three histidines implicated in binding of the catalytic zinc are missing from the CA domain of the receptor.
  • active enzymes there is a conserved HxHWG ⁇ 18,20 ⁇ ELH motif (the three histidines bind zinc), however, in the receptor this is modified to TFHWG ⁇ 18,20 ⁇ EMQ; i.e. two of the three critical zinc atoms would be missing.
  • CAH 8 carbonic anhydrase related protein
  • the protein PTP ⁇ SM2 (amino acid sequence SEQ ID NO. 4) comprises the amino acids encoded by all exons of PTP ⁇ - ⁇ , plus a 116 nucleotide “extra” exon, in the correct reading frame, between exons 23 and 24 (nucleotides 6229-6345 of SEQ ID NO. 3). This extra exon, designated exon 23a, contains a portion of the intron sequence between exons 23 and 24 of the PTP ⁇ gene.
  • PTP ⁇ SM2 expression has been verified in several human glioblastoma cell lines, and has also been confirmed in primary brain tumor samples. As PTP ⁇ SM2 comprises all the domains of PTP ⁇ ⁇ , the protein is expected to be membrane-bound. The extra exon lies within the cytoplasmic domain of the protein, and thus may alter the protein tyrosine phosphatase function of PTP ⁇ SM2.
  • a novel splicing variant PTP ⁇ protein having an amino acid sequence which includes the amino acid sequence of PTP ⁇ SM1 (SEQ ID NO. 2) or PTP ⁇ SM2 (SEQ. ID NO. 4) may be produced by recombinant techniques known in the art utilizing any suitable vector, in any suitable host cell.
  • the term “vector” is intended to include any physical or biochemical vehicle containing nucleic acid polymers of interest, by which those nucleic acid polymers are transferred into a host cell, thereby transfecting that cell with the introduced nucleic acid polymers.
  • the transfected nucleic acid sequence preferably contains a control sequence, such as a promoter sequence, suitable for transcription of the nucleic acid sequence in the host cell.
  • vectors include DNA plasmids, viruses, liposomes, particle gun pellets, and transfection vectors known to those of skill in the molecular biology arts.
  • the term “host cell” is intended to mean the target cell for vector transformation, in which the transferred nucleic acid polymer will be replicated and/or expressed.
  • bacterial cells may be suitable for production of the proteins for antibody production or structural study purposes, eukaryotic cell hosts are preferred for production of the protein for functional assays or therapeutic purposes.
  • Preferred eukaryotic cell hosts include insect cell lines (e.g., Sf9, Sf21, or High FiveTM cell lines), and mammalian cell lines (e.g., HeLa, CHO-K1, COS-7, COS-1, HEK293, HEPG2, Jurkat, MDCK, PAE, PC-12, and other acceptable mammalian cell lines).
  • insect cell lines e.g., Sf9, Sf21, or High FiveTM cell lines
  • mammalian cell lines e.g., HeLa, CHO-K1, COS-7, COS-1, HEK293, HEPG2, Jurkat, MDCK, PAE, PC-12, and other acceptable mammalian cell lines.
  • mammalian cell lines e.g., HeLa, CHO-K1, COS-7, COS-1, HEK293, HEPG2, Jurkat, MDCK, PAE, PC-12, and other acceptable mammalian cell lines.
  • the invention also provides vectors incorporating a nu
  • additional functionalities include, for example, polyhistidine domains, c-myc domains (specifically comprising amino acids 410-419 of the human c-myc oncogene product), ⁇ -galactosidase, ⁇ -glucuronidase, glutathione-S-transferase, maltose binding protein, human influenza virus hemagglutanin, green fluorescent protein, chloramphenicol acetyltransferase, luciferase, thioredoxin, and others.
  • the PTP ⁇ SM1 and SM2 proteins of the invention also encompass proteins comprising the amino acid sequence of SEQ ID NO. 2 or SEQ ID NO. 4 and such additional amino acid functionalities.
  • the invention also provides polypeptides which have a unique activity of PTP ⁇ SM1 or PTP ⁇ SM2 which is not shared by the other PTP ⁇ splice variant (e.g., an antigenic epitope) and which include a portion of the amino acid sequence of PTP ⁇ SM1 or PTP ⁇ SM2 which is at least about 8 to 12 amino acid residues in length, more preferably at least about 20 amino acids in length.
  • These polypeptides preferably comprise an amino acid sequence which is not found in PTP ⁇ ⁇ , PTP ⁇ ⁇ , or phosphacan, wherein the included portion of the sequence confers the unique activity on the polypeptide.
  • Such polypeptides may be utilized as described above to produce affinity reagents which specifically bind to PTP ⁇ splice variants SM1 or SM2, but do not bind to the other known splice variants of PTP ⁇ .
  • the invention thus provides such specific affinity reagents, which may be produces from such polypeptides, or from an entire PTP ⁇ SM1or PTP ⁇ SM2 protein.
  • these affinity reagents are antibodies or antibody fragments.
  • the invention also provides nucleic acid polymers encoding the PTP ⁇ splice variants SM1 or SM2.
  • These nucleic acid polymers most preferably comprises a nucleic acid sequence of SEQ. ID NO. 1 or SEQ ID NO. 3, or the predictable variants thereof which one of ordinary skill of the art could derive using the degeneracy of the genetic code.
  • Such nucleic acid polymers are useful for the production of PTP ⁇ SM1 or PTP ⁇ SM2 by recombinant methods, as described above.
  • the invention also encompasses nucleic acid probes or primers which hybridize to the mRNA encoding PTP ⁇ splice variants SM1 or SM2, but not mRNA encoding other known splice variants of PTP ⁇ .
  • probes or primers provided by the invention are preferably able to hybridize with SEQ. ID NO. 1 or SEQ. ID NO. 3 (or their complements) under stringent conditions (e.g., 0.5 ⁇ to 2 ⁇ SSC buffer, 0.1% SDS, and a temperature of 55-65° C.), but do not hybridize to SEQ ID NO. 5 (or its complement) under the same conditions.
  • PTP ⁇ SM1 or PTP ⁇ SM2 coding sequence specific probes are preferably from about 16 to about 40 nucleotides in length, more preferably from about 18 nucleotides to about 30 nucleotides in length. However, probes may be of a smaller size, preferably from about 8 to about 15 nucleotides in length, if two ore more probes are hybridized to adjacent sequences, so that terminal nucleic acid base-stacking interactions may stabilize their hybridization. In preferred embodiments of PTP ⁇ SM1 specific nucleic acid probes, the probes hybridize at or near the novel splice site at the 3′ end of exon 9, or its complement.
  • the probes hybridize at or adjacent to a location selected from: the novel splice site at the 3′ end of exon 23, at least a portion of the novel exon 23a, the novel splice site at the 5′ end of exon 24, or the complement of any one of these.
  • the level of the expression of these splice variants may be useful for staging or characterizing glioblastoma cells. Such cells may be extracted, for instance, from a primary tumor.
  • the invention provides for the monitoring of the relative expression level of PTP ⁇ SM1 or PTP ⁇ SM2, or both, in relation to each other or to one or more of the known PTP ⁇ splice variants.
  • the level of expression of PTP ⁇ SM1 is compare to at least one other splice variant selected from PTP ⁇ SM2, PTP ⁇ ⁇ , PTP ⁇ ⁇ , and phosphacan.
  • the level of expression of PTP ⁇ SM2 is compare to at least one other splice variant selected from PTP ⁇ SM1, PTP ⁇ ⁇ , PTP ⁇ ⁇ , and phosphacan.
  • Such comparison may be made in either a qualitative or quantitative manner.
  • One means for comparison is by hybridizing splice-variant specific nucleic acid probes to a sample of nucleic acids (which may be amplified) obtained from brain tumor cells.
  • the expression level of the splice variants may be deduced by the amplification of splice variant nucleic acid sequences, and the analysis of the size of those amplified products using methods known in the art.
  • protein levels may be studied utilizing splice-variant specific antibodies in either sandwich immunoassay or in-situ staining formats.
  • Various expression level assay techniques are known to those of skill in the molecular biological arts, and thus the specific techniques mentioned above should be considered merely exemplary.
  • antibody or “antibody moiety” is intended to include any polypeptide chain-containing molecular structure that has a specific shape which fits to and recognizes an epitope, where one or more non-covalent binding interactions stabilize the complex between the molecular structure and the epitope.
  • Antibodies which bind specifically to one of the brain tumor protein targets are referred to as anti-brain tumor protein target antibodies, or ⁇ (T BT ), or more specifically ⁇ (ARP2), ⁇ (SPARC), ⁇ (CMET), ⁇ (CD44), ⁇ (BEHAB), ⁇ (TSPAN3), ⁇ (VIPR2), ⁇ (OPN), ⁇ (PTN), and ⁇ (PTP ⁇ ).
  • the specific or selective fit of a given structure and its specific epitope is sometimes referred to as a “lock and key” fit.
  • the archetypal antibody molecule is the immunoglobulin, and all types of immunoglobulins (IgG, IgM, IgA, IgE, IgD, etc.), from all sources (e.g., human, rodent, rabbit, cow, sheep, pig, dog, other mammal, chicken, turkey, emu, other avians, etc.) are considered to be “antibodies.”
  • Antibodies utilized in the present invention may be polyclonal antibodies, although monoclonal antibodies are preferred because they may be reproduced by cell culture or recombinantly, and may be modified to reduce their antigenicity.
  • Polyclonal antibodies may be raised by a standard protocol by injecting a production animal with an antigenic composition, formulated as described above. See, e.g., Harlow and Lane, Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory, 1988.
  • an T BT antigen comprising an antigenic portion of the brain tumor protein targets' polypeptide is initially injected into any of a wide variety of mammals (e.g., mice, rats, rabbits, sheep or goats).
  • mammals e.g., mice, rats, rabbits, sheep or goats.
  • a superior immune response may be elicited if the polypeptide is joined to a carrier protein, such as ovalbumin, BSA or KLH.
  • the peptide-conjugate is injected into the animal host, preferably according to a predetermined schedule incorporating one or more booster immunizations, and the animals are bled periodically.
  • Polyclonal antibodies specific for the polypeptide may then be purified from such antisera by, for example, affinity chromatography using the polypeptide coupled to a suitable solid support.
  • hybridomas may be formed by isolating the stimulated immune cells, such as those from the spleen of the inoculated animal. These cells are then fused to immortalized cells, such as myeloma cells or transformed cells, which are capable of replicating indefinitely in cell culture, thereby producing an immortal, immunoglobulin-secreting cell line.
  • immortalized cells such as myeloma cells or transformed cells, which are capable of replicating indefinitely in cell culture, thereby producing an immortal, immunoglobulin-secreting cell line.
  • the immortal cell line utilized is preferably selected to be deficient in enzymes necessary for the utilization of certain nutrients.
  • Many such cell lines (such as myelomas) are known to those skilled in the art, and include, for example: thymidine kinase (TK) or hypoxanthine-guanine phosphoriboxyl transferase (HGPRT). These deficiencies allow selection for fused cells according to their ability to grow on, for example, hypoxanthine aminopter
  • the immortal fusion partners utilized are derived from a line that does not secrete immunoglobulin.
  • the resulting fused cells, or hybridomas are cultured under conditions that allow for the survival of fused, but not unfused, cells and the resulting colonies screened for the production of the desired monoclonal antibodies. Colonies producing such antibodies are cloned, expanded, and grown so as to produce large quantities of antibody, see Kohler and Milstein, 1975 Nature 256:495 (the disclosures of which are hereby incorporated by reference).
  • mice Large quantities of monoclonal antibodies from the secreting hybridomas may then be produced by injecting the clones into the peritoneal cavity of mice and harvesting the ascites fluid therefrom.
  • the mice preferably primed with pristine, or some other tumor-promoter, and immunosuppressed chemically or by irradiation, may be any of various suitable strains known to those in the art.
  • the ascites fluid is harvested from the mice and the monoclonal antibody purified therefrom, for example, by CM Sepharose column or other chromatographic means.
  • the hybridomas may be cultured in vitro or as suspension cultures. Batch, continuous culture, or other suitable culture processes may be utilized. Monoclonal antibodies are then recovered from the culture medium or supernatant.
  • ⁇ -exclusive list of available commercial antibodies includes: for SPARC/Osteonectin, from Zymed, mouse anti-bovine MAb (cross-reactivity with human), suitable for ELISA, WB, IH (paraffin), Cat# 33-5500; for c-MET, from Zymed, rabbit anti-human polyclonal, suitable for ELISA, WB, IH. Cat# 71-8000, and from RDI, rabbit anti-human MAb, suitable for WB, IP, IH.
  • mice anti-human MAb known to block binding of hyaluronic acid to its receptor CD44, “CD44/H-CAM Ab-2”; for Brevican/BEHAB, from BD Transduction Lab., a mouse anti-human MAb, WB, IF, Cat# B68820; for VIP 2 receptor, from Exalpha, mouse anti-rat (possible human cross-specificity, which is easily assayed) MAb, WB, IH.
  • Laminin receptor 67 kDa from Lab vision, mouse anti-human MAb, IH, ELISA, not for WB.
  • laminin receptor Ab-1 for Osteopontin, from Chemicon, rat anti-human MAb, raised against rh-Osteopontin—recognizes native protein well, WB, IH, ELISA.
  • MAB3057 for Pleiotrophin, from R&D goat anti-human polyclonal, WB, recognizes rh-Pleiotrophin.
  • BAF252 and from Oncogene goat anti-human polyclonal, WB, ELISA, detects rh-Pleiotrophin.
  • the antibodies or antigen binding fragments may be produced by genetic engineering.
  • antibody-producing cells are sensitized to the desired antigen or immunogen.
  • the messenger RNA isolated from the immune spleen cells or hybridomas is used as a template to make cDNA using PCR amplification.
  • a library of vectors, each containing one heavy chain gene and one light chain gene retaining the initial antigen specificity, is produced by insertion of appropriate sections of the amplified immunoglobulin cDNA into the expression vectors.
  • a combinatorial library is constructed by combining the heavy chain gene library with the light chain gene library.
  • the vectors that carry these genes are co-transfected into a host (e.g. bacteria, insect cells, mammalian cells, or other suitable protein production host cell.).
  • a host e.g. bacteria, insect cells, mammalian cells, or other suitable protein production host cell.
  • antibody gene synthesis is induced in the transfected host, the heavy and light chain proteins self-assemble to produce active antibodies that can be detected by screening with the antigen or immunogen.
  • recombinant antibodies are produced in a recombinant protein production system which correctly glycosylates and processes the immunoglobulin chains, such as insect or mammalian cells.
  • a recombinant protein production system which correctly glycosylates and processes the immunoglobulin chains, such as insect or mammalian cells.
  • An advantage to using insect cells which utilize recombinant baculoviruses for the production of antibodies for use in the present invention is that the baculovirus system allows production of mutant antibodies much more rapidly than stably transfected mammalian cell lines.
  • insect cells have been shown to correctly process and glycosylate eukaryotic proteins, which prokaryotic cells do not.
  • the baculovirus expression of foreign protein has been shown to constitute as much as 50-75% of the total cellular protein late in viral infection, making this system an excellent means of producing milligram quantities of the recombinant antibodies.
  • baculovirus Autographia californica nuclear polyhedrosis virus (AcNPV) and recombinant viral stocks in Spodoptera frugiperda (Sf9) cells to prepare large quantities of protein has been described by Smith et al. (1985), Summers and Smith (1987).
  • a preferred method of preparing recombinant antibodies is through the expression of DNA encoding recombinant antibody (produced by screening, as above, or by protein engineering to include more human-like domains, as discussed below) via the baculoviral expression system in Sf9 insect cells. Production of recombinant proteins in Sf9 cells is well known in the art, and one of ordinary skill would be able to select from a number of acceptable protocols (e.g., that described in U.S. Pat. No. 6,603,905).
  • antibodies which have a reduced propensity to induce a violent or detrimental immune response in humans such as anaphylactic shock
  • HAMA human-anti-murine-antibody “HAMA” response
  • These antibodies are preferred for all administrative routes, including intrathecal administration.
  • an immune response still can occur in the form of increased leukocyte infiltration, and inflammation.
  • the concurrent binding and inactivation of the therapeutic or imaging agent generally outweighs this benefit.
  • humanized, chimeric, or xenogenic human antibodies which produce less of an immune response when administered to humans, are preferred for use in the present invention.
  • Chimeric antibodies may be made by recombinant means by combining the murine variable light and heavy chain regions (VK and VH), obtained from a murine (or other animal-derived) hybridoma clone, with the human constant light and heavy chain regions, in order to produce an antibody with predominantly human domains.
  • VK and VH murine variable light and heavy chain regions
  • the production of such chimeric antibodies is well known in the art, and may be achieved by standard means (as described, e.g., in U.S. Pat. No. 5,624,659, incorporated fully herein by reference).
  • Humanized antibodies are engineered to contain even more human-like immunoglobulin domains, and incorporate only the complementarity-determining regions of the animal-derived antibody.
  • polyclonal or monoclonal antibodies may be produced from animals which have been genetically altered to produce human immunoglobulins, such as the Abgenix XenoMouse or the Medarex HuMAb® technology.
  • the transgenic animal may be produced by initially producing a “knock-out” animal which does not produce the animal's natural antibodies, and stably transforming the animal with a human antibody locus (e.g., by the use of a human artificial chromosome). Only human antibodies are then made by the animal. Techniques for generating such animals, and deriving antibodies therefrom, are described in U.S. Pat. Nos. 6,162,963 and 6,150,584, incorporated fully herein by reference. Such fully human xenogenic antibodies are a preferred antibody for use in the methods and compositions of the present invention.
  • single chain antibodies can be produced from phage libraries containing human variable regions. See U.S. Pat. No. 6,174,708, incorporated fully herein by reference. Also see Kuan, C. T., Reist, C. J., Foulon, C. F., Lorimer, I. A., Archer, G., Pegram, C. N., Pastan, I., Zalutsky, M. R., and Bigner, D. D. (1999). 125I-labeled anti-epidermal growth factor receptor-viii single-chain Fv exhibits specific and high-level targeting of glioma xenografts. Clin Cancer Res.
  • LMB-7 single-chain immunotoxin
  • immunoglobulin fragments comprising the epitope binding site (e.g., Fab′, F(ab′) 2 , or other fragments) are useful as antibody moieties in the present invention.
  • Such antibody fragments may be generated from whole immunoglobulins by ficin, pepsin, papain, or other protease cleavage.
  • “Fragment,” or minimal immunoglobulins may be designed utilizing recombinant immunoglobulin techniques.
  • Fv immunoglobulins for use in the present invention may be produced by linking a variable light chain region to a variable heavy chain region via a peptide linker (e.g., poly-glycine or another sequence which does not form an alpha helix or beta sheet motif).
  • a peptide linker e.g., poly-glycine or another sequence which does not form an alpha helix or beta sheet motif.
  • Fv fragments are heterodimers of the variable heavy chain domain (V H ) and the variable light chain domain (V L ).
  • the heterodimers of heavy and light chain domains that occur in whole IgG, for example, are connected by a disulfide bond.
  • Recombinant Fvs in which V H and V L are connected by a peptide linker are typically stable, see, for example, Huston et al., Proc. Natl. Acad, Sci. USA 85:5879-5883 (1988) and Bird et al., Science 242:423-426 (1988), both fully incorporated herein, by reference.
  • immunoglobulins with added chemical linkers, detectable moieties [fluorescent dyes, enzymes, substrates, chemiluminescent moieties], or specific binding moieties [such as streptavidin, avidin, or biotin] may be utilized in the methods and compositions of the present invention.
  • detectable moieties fluorescent dyes, enzymes, substrates, chemiluminescent moieties
  • specific binding moieties such as streptavidin, avidin, or biotin
  • antibody or “antibody moiety” will be used throughout to generally refer to molecules which specifically bind to an epitope of the brain tumor protein targets, although the term will encompass all immunoglobulins, derivatives, fragments, recombinant or engineered immunoglobulins, and modified immunoglobulins, as described above.
  • Candidate anti-T BT antibodies can be tested for anti-T BT activity by any suitable standard means. As a first screen, the antibodies may be tested for binding against the brain tumor protein target antigen utilized to produce them, or against the entire brain tumor protein target extracellular domain or protein. As a second screen, anti-T BT candidates may be tested for binding to an appropriate glioblastoma cell line (i.e., one which approximates primary tumor brain tumor protein target expression), or to primary tumor tissue samples. For these screens, the anti-T BT candidate antibody may be labeled for detection (e.g., with fluorescein or another fluorescent moiety, or with an enzyme such as horseradish peroxidase).
  • an enzyme such as horseradish peroxidase
  • the candidate antibody, or an antibody conjugate produced as described below may be tested for appropriate activity (i.e., the ability to decrease tumor cell growth and/or to aid in visualizing tumor cells) in an in vivo model, such as an appropriate glioblastoma cell line, or in a mouse or rat human brain tumor model, as described below.
  • appropriate activity i.e., the ability to decrease tumor cell growth and/or to aid in visualizing tumor cells
  • an in vivo model such as an appropriate glioblastoma cell line, or in a mouse or rat human brain tumor model, as described below.
  • antibodies which are useful for altering the function of ARP-2, SPARC, c-MET, BEHAB, CD-44, TSPN3, PTN, OPN, VIPR-2, or PTP ⁇ may be assayed in functional formats, such as glioblastoma cell culture or mouse/rat CNS tumor model studies. In glioblastoma cell models of activity, expression of the protein is first verified in the particular cell strain to be used. If necessary, the cell line may be stably transfected with a coding sequence of the protein under the control of an appropriate constituent promoter, in order to express the protein at a level comparable to that found in primary tumors.
  • glioblastoma cells The ability of the glioblastoma cells to survive in the presence of the candidate function-altering anti-protein antibody is then determined.
  • cell migration assays as described below in Example 1, may be utilized to determine the effect of the candidate antibody therapeutic agent on the tumor-like behavior of the cells.
  • endothelial cell sprouting assays such as described in Example 2 may be utilized to determine the ability of the candidate antibody therapeutic to inhibit vascular neogenesis, an important function in tumor biology.
  • the anti-T BT antibodies for use in the present invention may have utility without conjugation when the native activity of the brain tumor protein target is altered in the tumor cell.
  • Such antibodies which may be selected as described above, may be utilized without further modification to include a cytotoxic or imaging moiety.
  • These types of compositions have the advantage of reduced toxicity (in that only the toxicity of the antibody moieties themselves must be taken into account when dosing), and are simpler to manufacture.
  • non-conjugated activity altering anti-T BT antibody therapeutics are a preferred embodiment of the invention.
  • the conjugation of cytotoxic or imaging agents is yet another preferred embodiment when utilizing these antibodies because the added moieties add functionality to the therapeutic.
  • the anti-T BT antibodies may be coupled or conjugated to one or more therapeutic cytotoxic or imaging moieties.
  • cytotoxic moiety simply means a moiety which inhibits cell growth or promotes cell death when proximate to or absorbed by the cell.
  • Suitable cytotoxic moieties in this regard include radioactive isotopes (radionuclides), chemotoxic agents such as differentiation inducers and small chemotoxic drugs, toxin proteins, and derivatives thereof.
  • imaging moiety means a moiety which can be utilized to increase contrast between a tumor and the surrounding healthy tissue in a visualization technique (e.g., radiography, positron-emission tomography, magnetic resonance imaging, direct or indirect visual inspection).
  • suitable imaging moieties include radiography moieties (e.g. heavy metals and radiation emitting moieties), positron emitting moieties, magnetic resonance contrast moieties, and optically visible moieties (e.g., fluorescent or visible-spectrum dyes, visible particles, etc.).
  • radiography moieties e.g. heavy metals and radiation emitting moieties
  • positron emitting moieties positron emitting moieties
  • magnetic resonance contrast moieties e.g., and optically visible moieties (e.g., fluorescent or visible-spectrum dyes, visible particles, etc.).
  • optically visible moieties e.g., fluorescent or visible-spectrum dyes, visible particles, etc.
  • therapeutic or imaging agents may be conjugated to the anti-PTP ⁇ moiety by any suitable technique, with appropriate consideration of the need for pharmokinetic stability and reduced overall toxicity to the patient.
  • a therapeutic agent may be coupled to a suitable antibody moiety either directly or indirectly (e.g. via a linker group).
  • a direct reaction between an agent and an antibody is possible when each possesses a functional group capable of reacting with the other.
  • a nucleophilic group such as an amino or sulfhydryl group, may be capable of reacting with a carbonyl-containing group, such as an anhydride or an acid halide, or with an alkyl group containing a good leaving group (e.g., a halide).
  • a linker group can function as a spacer to distance an antibody from an agent in order to avoid interference with binding capabilities.
  • a linker group can also serve to increase the chemical reactivity of a substituent on a moiety or an antibody, and thus increase the coupling efficiency. An increase in chemical reactivity may also facilitate the use of moieties, or functional groups on moieties, which otherwise would not be possible.
  • Suitable linkage chemistries include maleimidyl linkers and alkyl halide linkers (which react with a sulfhydryl on the antibody moiety) and succinimidyl linkers (which react with a primary amine on the antibody moiety).
  • Several primary amine and sulfhydryl groups are present on immunoglobulins, and additional groups may be designed into recombinant immunoglobulin molecules. It will be evident to those skilled in the art that a variety of bifunctional or polyfunctional reagents, both homo- and hetero-functional (such as those described in the catalog of the Pierce Chemical Co., Rockford, Ill.), may be employed as a linker group.
  • Coupling may be effected, for example, through amino groups, carboxyl groups, sulfhydryl groups or oxidized carbohydrate residues.
  • cytotoxic or imaging moieties may be coupled to the anti-T BT antibody moiety through a an oxidized carbohydrate group at a glycosylation site, as described in U.S. Pat. Nos. 5,057,313 and 5,156,840.
  • Yet another alternative method of coupling the antibody moiety to the cytotoxic or imaging moiety is by the use of a non-covalent binding pair, such as streptavidin/biotin, or avidin/biotin.
  • a non-covalent binding pair such as streptavidin/biotin, or avidin/biotin.
  • one member of the pair is covalently coupled to the antibody moiety and the other member of the binding pair is covalently coupled to the cytotoxic or imaging moiety.
  • a cytotoxic moiety is more potent when free from the antibody portion of the immunoconjugates of the present invention, it may be desirable to use a linker group which is cleavable during or upon internalization into a cell, or which is gradually cleavable over time in the extracellular environment.
  • linker group which is cleavable during or upon internalization into a cell, or which is gradually cleavable over time in the extracellular environment.
  • the mechanisms for the intracellular release of a cytotoxic moiety agent from these linker groups include cleavage by reduction of a disulfide bond (e.g., U.S. Pat. No. 4,489,710), by irradiation of a photolabile bond (e.g., U.S. Pat. No.
  • cytotoxic and/or imaging moiety it may be desirable to couple more than one cytotoxic and/or imaging moiety to an antibody.
  • cytotoxic strategies may be simultaneously implemented, an antibody may be made useful as a contrasting agent for several visualization techniques, or a therapeutic antibody may be labeled for tracking by a visualization technique.
  • multiple molecules of an imaging or cytotoxic moiety are coupled to one antibody molecule.
  • more than one type of moiety may be coupled to one antibody.
  • immunoconjugates with more than one moiety may be prepared in a variety of ways.
  • more than one moiety may be coupled directly to an antibody molecule, or linkers which provide multiple sites for attachment (e.g., dendrimers) can be used.
  • linkers which provide multiple sites for attachment
  • a carrier with the capacity to hold more than one cytotoxic or imaging moiety can be used.
  • a carrier may bear the agents in a variety of ways, including covalent bonding either directly or via a linker group, and non-covalent associations.
  • Suitable covalent-bond carriers include proteins such as albumins (e.g., U.S. Pat. No. 4,507,234), peptides, and polysaccharides such as aminodextran (e.g., U.S. Pat. No. 4,699,784), each of which have multiple sites for the attachment of moieties.
  • a carrier may also bear an agent by non-covalent associations, such as non-covalent bonding or by encapsulation, such as within a liposome vesicle (e.g., U.S. Pat. Nos.
  • Encapsulation carriers are especially useful for imaging moiety conjugation to anti-T BT antibody moieties for use in the invention, as a sufficient amount of the imaging moiety (dye, magnetic resonance contrast reagent, etc.) for detection may be more easily associated with the antibody moiety.
  • encapsulation carriers are also useful in chemotoxic therapeutic embodiments, as they can allow the therapeutic compositions to gradually release a chemotoxic moiety over time while concentrating it in the vicinity of the tumor cells.
  • Carriers and linkers specific for radionuclide agents include radiohalogenated small molecules and chelating compounds.
  • U.S. Pat. No. 4,735,792 discloses representative radiohalogenated small molecules and their synthesis.
  • a radionuclide chelate may be formed from chelating compounds that include those containing nitrogen and sulfur atoms as the donor atoms for binding the metal, or metal oxide, radionuclide.
  • U.S. Pat. No. 4,673,562 to Davison et al. discloses representative chelating compounds and their synthesis.
  • Such chelation carriers are also useful for magnetic spin contrast ions for use in magnetic resonance imaging tumor visualization methods, and for the chelation of heavy metal ions for use in radiographic visualization methods.
  • radionuclides for use as cytotoxic moieties are radionuclides which are suitable for pharmacological administration.
  • radionuclides include 123 I, 125 I, 131 I, 90 Y, 211 At, 67 Cu, 186 Re, 188 Re, 212 Pb, and 212 Bi.
  • Iodine and astatine isotopes are more preferred radionuclides for use in the therapeutic compositions of the present invention, as a large body of literature has been accumulated regarding their use.
  • 131 I is particularly preferred, as are other ⁇ -radiation emitting nuclides, which have an effective range of several millimeters.
  • 123 I, 125 I, 131 I, or 211 At may be conjugated to antibody moieties for use in the compositions and methods utilizing any of several known conjugation reagents, including Iodogen, N-succinimidyl 3-[ 211 At]astatobenzoate, N-succinimidyl 3-[ 133 I]iodobenzoate (SIB), and, N-succinimidyl 5-[ 131 I]iodob-3-pyridinecarboxylate (SIPC). Any iodine isotope may be utilized in the recited iodo-reagents.
  • a suitable antibody for use in the present invention may be easily made by coupling an Fab fragment of the BD Transduction Labs R20720 anti-PTP ⁇ MAb with 131 I Iodogen according to the manufacturer's instructions.
  • Other radionuclides may be conjugated to anti-T BT antibody moieties by suitable chelation agents known to those of skill in the nuclear medicine arts.
  • Preferred chemotoxic agents include small-molecule drugs such as carboplatin, cisplatin, vincristine, taxanes such as paclitaxel and doceltaxel, hydroxyurea, gemcitabine, vinorelbine, irinotecan, tirapazamine, matrilysin, methotrexate, pyrimidine and purine analogs, and other suitable small toxins known in the art.
  • Preferred chemotoxin differentiation inducers include phorbol esters and butyric acid. Chemotoxic moieties may be directly conjugated to the anti-T BT antibody moiety via a chemical linker, or may encapsulated in a carrier, which is in turn coupled to the anti-T BT antibody moiety.
  • Preferred toxin proteins for use as cytotoxic moieties include ricins A and B, abrin, diphtheria toxin, bryodin 1 and 2, momordin, trichokirin, cholera toxin, gelonin, Pseudomonas exotoxin, Shigella toxin, pokeweed antiviral protein, and other toxin proteins known in the medicinal biochemistry arts.
  • ricins A and B abrin, diphtheria toxin, bryodin 1 and 2, momordin, trichokirin, cholera toxin, gelonin, Pseudomonas exotoxin, Shigella toxin, pokeweed antiviral protein, and other toxin proteins known in the medicinal biochemistry arts.
  • these toxin agents may elicit undesirable immune responses in the patient, especially if injected intravascularly, it is preferred that they be encapsulated in a carrier for coupling to the anti-T BT antibody moiety.
  • Preferred radiographic moieties for use as imaging moieties in the present invention include compounds and chelates with relatively large atoms, such as gold, iridium, technetium, barium, thallium, iodine, and their isotopes. It is preferred that less toxic radiographic imaging moieties, such as iodine or iodine isotopes, be utilized in the compositions and methods of the invention. Examples of such compositions which may be utilized for x-ray radiography are described in U.S. Pat. No. 5,709,846, incorporated fully herein by reference. Such moieties may be conjugated to the anti-T BT antibody moiety through an acceptable chemical linker or chelation carrier.
  • radionuclides which emit radiation capable of penetrating the scull may be useful for scintillation imaging techniques.
  • Suitable radionuclides for conjugation include 99 Tc, 111 In, and 67 Ga.
  • Positron emitting moieties for use in the present invention include 18 F, which can be easily conjugated by a fluorination reaction with the anti-T BT antibody moiety according to the method described in U.S. Pat. No. 6,187,284.
  • Preferred magnetic resonance contrast moieties include chelates of chromium(III), manganese(II), iron(II), nickel(II), copper(II), praseodymium(III), neodymium(III), samarium(III) and ytterbium(III) ion. Because of their very strong magnetic moment, the gadolinium(III), terbium(III), dysprosium(III), holmium(III), erbium(III), and iron(III) ions are especially preferred. Examples of such chelates, suitable for magnetic resonance spin imaging, are described in U.S. Pat. No. 5,733,522, incorporated fully herein by reference. Nuclear spin contrast chelates may be conjugated to the anti-T BT antibody moieties through a suitable chemical linker.
  • Optically visible moieties for use as imaging moieties include fluorescent dyes, or visible-spectrum dyes, visible particles, and other visible labeling moieties.
  • Fluorescent dyes such as fluorescein, coumarin, rhodamine, bodipy Texas red, and cyanine dyes, are useful when sufficient excitation energy can be provided to the site to be inspected visually. Endoscopic visualization procedures may be more compatible with the use of such labels.
  • visible spectrum dyes are preferred.
  • Acceptable dyes include FDA-approved food dyes and colors, which are non-toxic, although pharmaceutically acceptable dyes which have been approved for internal administration are preferred.
  • such dyes are encapsulated in carrier moieties, which are in turn conjugated to the anti-T BT antibody.
  • visible particles such as colloidal gold particles or latex particles, may be coupled to the anti-T BT antibody moiety via a suitable chemical linker.
  • the BBB was not considered to present a problem in the diagnosis and treatment of brain tumors, because early scans of human brain tumors suggested that the BTB (blood tumor barrier) was “leaky.” However, as the size of the molecule increases, the rate of movement across the barrier decreases.
  • the BBB has been demonstrated to be heterogeneous in experimental human tumor xenograft animal models and in human patients.
  • a first strategy for drug delivery through the BBB entails disruption of the BBB, either by osmotic means such as mannitol or leukotrienes, or biochemically by the use of vasoactive substances such as bradykinin.
  • osmotic means such as mannitol or leukotrienes
  • vasoactive substances such as bradykinin.
  • a BBB disrupting agent is co-administered with the therapeutic or imaging compositions of the invention when the compositions are administered by intravascular injection.
  • the antibody-therapeutic or antibody-imaging agent will generally be mixed, prior to administration, with a non-toxic, pharmaceutically acceptable carrier substance.
  • a non-toxic, pharmaceutically acceptable carrier substance usually, this will be an aqueous solution, such as normal saline or phosphate-buffered saline (PBS), Ringer's solution, lactate-Ringer's solution, or any isotonic physiologically acceptable solution for administration by the chosen means.
  • PBS normal saline or phosphate-buffered saline
  • Ringer's solution such as phosphate-buffered saline (PBS), Ringer's solution, lactate-Ringer's solution, or any isotonic physiologically acceptable solution for administration by the chosen means.
  • PBS normal saline or phosphate-buffered saline
  • Ringer's solution such as normal saline or phosphate-buffered saline (PBS), Ringer's solution, lactate-Ringer's
  • the clinician of ordinary skill is familiar with appropriate ranges for pH, tonicity, and additives or preservatives when formulating pharmaceutical compositions for administration by intravascular injection, intrathecal injection, injection into the cerebro-spinal fluid, direct injection into the tumor, or by other routes.
  • the antibody-therapeutics and antibody-imaging agents may be stabilized against aggregation and polymerization with amino acids and non-ionic detergents, polysorbate, and polyethylene glycol.
  • additional stabilizers may include various physiologically-acceptable carbohydrates and salts.
  • polyvinylpyrrolidone may be added in addition to the amino acid. Suitable therapeutic immunoglobulin solutions which are stabilized for storage and administration to humans are described in U.S. Pat. No. 5,945,098, incorporated fully herein by reference.
  • Other agents such as human serum albumin (HSA), may be added to the therapeutic or imaging composition to stabilize the antibody conjugates.
  • HSA human serum albumin
  • compositions of the invention may be administered using any medically appropriate procedure, e.g., intravascular (intravenous, intraarterial, intracapillary) administration, injection into the cerebrospinal fluid, intracavity or direct injection in the tumor.
  • Intrathecal administration maybe carried out through the use of an Ommaya reservoir, in accordance with known techniques. (F. Balis et al., Am J. Pediatr. Hematol. Oncol. 11, 74, 76 (1989).
  • administration via intravascular injection is preferred for pre-operative visualization of the tumor.
  • Post-operative visualization or visualization concurrent with an operation may be through intrathecal or intracavity administration, as through an Ommaya reservoir, or also by intravascular administration.
  • Intravascular injection may be by intravenous or intraarterial injection: carotid artery injection is thought to assist in administration to the brain, and is thus preferred.
  • Antibody-agents injected into the blood stream have been shown to cross the blood-brain barrier and to infiltrate the cranial cavity to some extent, usually in the range of 10 ⁇ 4 to 10 ⁇ 3 % [?UNITS?] injected dose per gram. This rate of uptake may be sufficient for imaging reagents, and also may be useful for tumor cell specific cytotoxic agents (e.g, those specifically directed to the inhibition of the function of tumor-cell overexpressed proteins).
  • tumor cell specific cytotoxic agents e.g, those specifically directed to the inhibition of the function of tumor-cell overexpressed proteins.
  • the therapeutics compositions be administered by intrathecal injection, direct injection, or injection into the cerebro-spinal fluid.
  • a preferred method for administration of the therapeutic compositions of the invention is by depositing it into the inner cavity of a cystic tumor by any suitable technique, such as by direct injection (aided by stereotaxic positioning of an injection syringe, if necessary) or by placing the tip of an Ommaya reservoir into a cavity, or cyst, for administration.
  • the tumor is a solid tumor
  • the antibody may be administered by first creating a resection cavity in the location of the tumor. This procedure differs from an ordinary craniotomy and tumor resection only in a few minor respects.
  • tumor resection is a common treatment procedure, and is often indicated to relieve pressure
  • administration of the therapeutic compositions of the invention following tumor resection is a preferred embodiment of the treatment methods of the invention.
  • the cavity is preferable rinsed with saline until all bleeding is stopped by cauterization.
  • the pia-arachnoid membrane, surrounding the tumor cavity at the surface is cauterized to enhance the formation of fibroblastic reaction and scarring in the pia-arachnoid area.
  • the result is the formation of an enclosed, fluid-filled cavity within the brain tissue at the location from where the tumor was removed.
  • either the tip of an Ommaya reservoir or a micro catheter which is connected to a pump device and allows the continues infusion of an antibody solution into the cavity, can be placed into the cavity. See, e.g., U.S. Pat. No. 5,558,852, incorporated fully herein by reference.
  • a convection-enhanced delivery catheter may be implanted directly into the tumor mass, into a natural or surgically created cyst, or into the normal brain mass.
  • Such convection-enhanced pharmaceutical composition delivery devices greatly improve the diffusion of the composition throughout the brain mass.
  • the implanted catheters of these delivery devices utilize high-flow microinfusion (with flow rates in the range of about 0.5 to 15.0 ⁇ l/minute), rather than diffusive flow, to deliver the therapeutic or imaging composition to the brain and/or tumor mass.
  • high-flow microinfusion with flow rates in the range of about 0.5 to 15.0 ⁇ l/minute
  • the effective amount of the therapeutic antibody-conjugate composition or of the imaging antibody-conjugate compositions to be given to a particular patient will depend on a variety of factors, several of which will be different from patient to patient.
  • a competent clinician will be able to determine an effective amount of a therapeutic antibody-conjugate composition to administer to a patient to retard the growth and promote the death of tumor cells, or an effective amount of an imaging composition to administer to a patient to facilitate the visualization of a tumor.
  • Dosage of the antibody-conjugate will depend on the treatment of the tumor, route of administration, the nature of the therapeutics, sensitivity of the tumor to the therapeutics, etc.
  • a clinician can determine the maximum safe dose for an individual, depending on the route of administration. For instance, an intravenously administered dose may be more than an intrathecally administered dose, given the greater body of fluid into which the therapeutic composition is being administered. Similarly, compositions which are rapidly cleared from the body may be administered at higher doses, or in repeated doses, in order to maintain a therapeutic concentration. Imaging moieties are typically less toxic than cytotoxic moieties and may be administered in higher doses in some embodiments. Utilizing ordinary skill, the competent clinician will be able to optimize the dosage of a particular therapeutic or imaging composition in the course of routine clinical trials.
  • the dosage will be 0.001 to 100 milligrams of conjugate per kilogram subject body weight. Doses in the range of 0.01 to 1 mg per kilogram of patient body weight may be utilized for a radionuclide therapeutic composition which is administered intrathecally. Relatively large doses, in the range of 0.1 to 10 mg per kilogram of patient body weight, may used for imaging conjugates with a relatively non-toxic imaging moiety. The amount utilized will depend on the sensitivity of the imaging method, and the relative toxicity of the imaging moiety. In a therapeutic example, where the therapeutic composition comprises a 131 I cytotoxic moiety, the dosage to the patient will typically start at a lower range of 10 mCi, and go up to 100, 300 or even 500 mCi.
  • the dosage to the patient will typically be from 5,000 Rads to 100,000 Rads (preferably at least 13,000 Rads, or even at least 50,000 Rads). Doses for other radionuclides are typically selected so that the tumoricidal dose will be equivalent to the foregoing range for 131 I. Similarly, chemotoxic or toxin protein doses may be scaled accordingly.
  • the antibody conjugate can be administered to the subject in a series of more than one administration.
  • regular periodic administration e.g., every 2-3 days
  • antibody moieties which do not provoke HAMA or other immune responses are preferred.
  • the imaging antibody conjugate compositions may be administered at an appropriate time before the visualization technique. For example, administration within an hour before direct visual inspection may be appropriate, or administration within twelve hours before an MRI scan may be appropriate. Care should be taken, however, to not allow too much time to pass between administration and visualization, as the imaging compound may eventually be cleared from the patient's system.
  • compositions may be utilized as a “dry run” for more toxic cytotoxic antibody conjugates.
  • the physician may first use a visualization technique to determine precisely where in the brain the cytotoxic conjugate will concentrate. If a sufficient degree of tissue selectivity is not achieved (e.g, if the tumor cells are too disperse in the normal tissue, or if the particular brain tumor protein target chosen is not sufficiently overexpressed in the particular patient's tumor cells), then the physician may choose another brain tumor protein target.
  • tissue selectivity e.g, if the tumor cells are too disperse in the normal tissue, or if the particular brain tumor protein target chosen is not sufficiently overexpressed in the particular patient's tumor cells.
  • brain tumors tend to be heterogeneous in character, and pervasive throughout the brain tissue. This combination often makes them difficult to treat, as individual portions of the tumor cells in any particular patient may have differing biological characteristic.
  • various combinations of therapeutic or imaging agents as described above in the Summary of Invention, in order to more fully target all of the cells exhibiting tumorigenic characteristics.
  • Such combination treatments may be by administering blended antibody therapeutic or imaging compositions, individually prepared as described above, and administering the blended therapeutic to the patient as described.
  • the skilled administering physician will be able to take such factors as combined toxicity, and individual antibody agent efficacy, into account when administering such combined agents. Additionally, those of skill in the art will be able to screen the antibodies to avoid potential cross-reaction with each other, in order to assure full efficacy of each antibody therapeutic or imaging agent.
  • each individual brain tumor protein target compositions may be administered simultaneously or in succession for a combined therapy. This may be desirable to avoid accumulated toxicity from several antibody conjugate reagents, or to more closely monitor potential adverse reactions to the individual antibody reagents. Thus, cycles such as where a first antibody therapeutic agent is administered on day one, followed by a second on day two, then a period with out administration, followed by re-administration of the antibody therapeutics on different successive days, is comprehended within the present invention.
  • the mRNA nucleotide sequence for PTP ⁇ SM1 was identified in a human fetal brain phage cDNA library by sequencing.
  • the mRNA nucleotide sequence for PTP ⁇ SM2 was identified by PCR amplification of adult human brain cDNA, and sequencing of the resulting nucleic acids.
  • cDNA was generated from total RNA using the 1 st Strand synthesis kit from Gibco Life Technologies, Inc., and an oligo dT 30 anchored primer.
  • 1 st Strand synthesis kit from Gibco Life Technologies, Inc.
  • an oligo dT 30 anchored primer For each RT-PCR reaction, 1 ⁇ l of cDNA was utilized. The PCR reaction was carried out using an Advantage 2 kit (Clontech) under standard conditions. The products of the PCR reactions were confirmed via sequencing.
  • Both clones were verified by RT-PCR analysis of glioblastoma cell lines and primary tumors.
  • PTP ⁇ SM1 primers CAGCAGTTGGATGGAAGAGGAC [SEQ ID NO. 28] and CACTGAGATTCTGGCACTATTC [SEQ ID NO. 29] were used, producing an identifiable 1116 bp product.
  • RT-PCR analysis was performed, confirming expression of the SM1 splice variant in 11 of 17 different glioblastoma cell lines tested, fetal brain, and adult brain, using the unique 3 end and portion of the 3′ untranslated region as the hybridization target for the probe.
  • RT-PCR analysis was performed on 28 primary brain tumor samples, confirming expression of the PTP ⁇ SM1 variant in 16 of the 28 tumors.
  • PTP ⁇ SM2 For PTP ⁇ SM2, primers AACAATTCCAGGGTCTCACTC [SEQ ID NO. 30] and TTGACTGGCTCAGGAGTATAG [SEQ ID NO. 31] were used, which produce a 130 bp product when the extra exon 23a is present, and a no product when the exon 23a is absent.
  • RT-PCR analysis was performed, confirming expression in 6 of 17 different glioblastoma cell lines tested.
  • RT-PCR analysis was performed on 28 primary brain tumor samples, confirming expression of the PTP ⁇ SM1 variant in 19 of the 28 tumors.
  • RT-PCR analysis was also done for the expression of PTP ⁇ - ⁇ (primers CTGATAATGAGGGCTCCCAAC [SEQ ID NO. 32] and CTCTGCACTTCCTGGTAAAACTCT [SEQ ID NO. 33]) and PTP ⁇ - ⁇ (primers CAGCAGTTGGATGGAAGAGGAC [SEQ ID NO. 34] and CTCTGCACTTCCTGGTAAAACTCT [SEQ ID NO. 35]) in the 28 brain tumor tissue samples.
  • PTP ⁇ - ⁇ was shown to be expressed in 16 of the 28 samples, and the short form PTP ⁇ - ⁇ was shown to be expressed in 19 of the 28 samples.
  • Tumor cells are known to migrate more rapidly towards chemoattractants.
  • the cell migration assay measures the ability of a cell to migrate. The ability to migrate is taken as a measure of tumorigenicity.
  • Chemoattractants generally used are fetal bovine serum, pleiotrophin, bFGF, and VEGF. Thus, this assay can be used to determine migration capability of a cell in which the gene has been knocked down or the gene of interest is being overexpressed.
  • the ChemoTx® disposable chemotaxis system (Neuroprobe, Inc., Gaithersburg, Md.) is used according to the manufacturer's instructions, with a few modifications. Briefly, glioblastoma cultured cells from cell line G55T2 are prepared by splitting the cells the day before the assay is performed. A ChemoTx® chamber with the following specifications is used: Pore size 8 ⁇ m, exposed filter area 8 mm, exposed filter area diameter 3.2 mm 2 . The plate configuration is: 30 ⁇ l per well, 96 well plate. The membrane type is: Track-etched polycarbonate.
  • the filter membrane is coated in 100 ml PBS containing 0.1% acetic acid and 3.5 ml Vitrogen 100 (from Cohesion) at 37° C. overnight. About 30 minutes before starting the assay the coated membrane is washed and rinsed with PBS containing 0.1% BSA. Cells are harvested by using the standard technique (trypsin-EDTA). The cells are washed once with DMEM 10% FBS, and then spun at 1000 RPM, for 5 minutes at room temperature. The pellet is resuspended in DMEM without serum, containing 0.1% BSA (serum free medium).
  • the cells are spun and resuspended again in serum free medium, and then spun and resuspended in the amount of serum free medium needed to provide a concentration of 1 mio. cells/ml, or 25,000 cells per 25 ul. Just prior to the assay, a suitable amount of the antibody to be tested for anti-target function activity is added to the cell suspension.
  • a standard chemoattractant is used to measure the mobility of the cells.
  • the chemoattractants are diluted in serum free medium.
  • a suitable unspecific chemoattractant is DMEM with 5% FBS.
  • the chemoattractant solutions and control solutions without chemoattractant are pipetted (29 ⁇ l) into the lower plate wells. After placing and securing the filter plate over the lower wells, ensuring contact with the solution in the bottom wells, serial dilutions of the cell suspension are pipetted onto each site on the filter top. The plates are them covered and incubated at 37° C., 5% CO 2 , for 3-4 hours.
  • the upper filter side is rinsed with PBS and exposed upper filter areas are cleaned with wet cotton swabs.
  • the filter is stained using Diff-QuikTM (VWR) dye kit, according to the manufacturer's instructions.
  • VWR Diff-QuikTM
  • the migrated cells are counted on the lower filter side using a microscope (Magnification 200 ⁇ ), by counting of 5 high power field sections per well.
  • HUVEC Human Umbilical Vein Endothelial Cells
  • Cell-sprouting morphology can be utilized as an easily visualized assay to determine the inhibitory effect of a candidate antibody on the protein target function for protein targets which stimulate endothelial cell sprouting, such as ARP2.
  • assays have been described extensively in the literature (Nehls, V., et al., Histochem. Cell Biol. 104: 459-466 (1995); Koblizek, T. I., et al., Curr. Biol. 8: 529-532 (1988); and Kwak, H. J., et al., FEBS Lett. 448: 249-253).
  • a endothelial cells from a suitable source such as HUVECs or PPAECs (porcine pulmonary artery endothelial cells) are grown to confluence on microcarrier (MC) beads (diameter 175 ⁇ m, available from Sigma) and placed into a 2.5 mg/ml fibrinogen gel containing the protein target at an appropriate effective concentration (200 ng/ml is an suitable starting concentration, which the skilled practitioner may optimize) and the antibody in an appropriate range of concentrations (this will depend on antibody titer and affinity for the target), and 200 units/ml Trasylol (available from Bayer).
  • MC microcarrier
  • Fibrin gels are incubated in M-199 with a daily supplement of the same amount of recombinant protein and antibody, 2.0% heat-inactivated fetal bovine serum, and 200 units/ml Trasylol. After three days, the extent of sprouting is determined using a phase-contrast microscope (such as those available from Zeiss). A decrease in cell sprouting as compared to controls without antibody indicates a reduction in protein target activity by the antibody.

Abstract

The present invention relates to the use of proteins which are differentially expressed in primary brain tumor tissues, as compared to normal brain tissues, as biomolecular targets for brain tumor treatment therapies. Specifically, the present invention relates to the use of immunotherapeutic and immunoimaging agents which specifically bind to one or more of human proteins angiopoietin related protein 2 (ARP-2,) secreted protein acidic, rich in cysteine (SPARC,) c-met proto-oncogene (C-MET,) brevican (BEHAB,) CD-44 antigen (CD-44,) tetraspanin 3 (TSPN3,) pleiotrophin (PTN,) osteopontin (OPN,) vasoactive intestinal peptide receptor-2 (VIPR-2,) and receptor protein tyrosine phosphatase zeta (PTPζ) for the treatment and visualization of brain tumors in patients. The present invention also provides compounds and pharmaceutically acceptable compositions for administration in the methods of the invention. The present invention also provides novel splice variants of protein PTPζ, PTPζ SM1 and PTPζ SM2. Nucleic acid probes specific for the spliced mRNA encoding these variants and affinity reagents specific for the novel proteins are also provided.

Description

    FIELD OF USE
  • The present invention relates to the use of proteins which are differentially expressed in primary brain tumor tissues, as compared to normal brain tissues, as biomolecular targets for brain tumor treatment therapies. Specifically, the present invention relates to the use of immunotherapeutic and immunoimaging agents which specifically bind to one or more of angiopoietin related protein 2 (ARP-2,) secreted protein acidic, rich in cysteine (SPARC,) c-met proto-oncogene (C-MET,) brevican (BEHAB,) CD-44 antigen (CD-44,) tetraspanin 3 (TSPN3,) pleiotrophin (PTN,) osteopontin (OPN,) vasoactive intestinal peptide receptor-2 (VIPR-2,) and receptor protein tyrosine phosphatase zeta (PTPζ) for the treatment and visualization of brain tumors in patients. The present invention also provides compounds and pharmaceutically acceptable compositions for administration. [0001]
  • BACKGROUND OF THE INVENTION
  • Brain Tumor Biology and Etiology [0002]
  • Brain tumors are considered to have one of the least favorable prognoses for long term survival: the average life expectancy of an individual diagnosed with a central nervous system (CNS) tumor is just eight to twelve months. Several unique characteristics of both the brain and its particular types of neoplastic cells create daunting challenges for the complete treatment and management of brain tumors. Among these are 1) the physical characteristics of the intracranial space, 2) the relative biological isolation of the brain from the rest of the body, 3) the relatively essential and irreplaceable nature of the organ mass, and 4) the unique nature of brain tumor cells. [0003]
  • First and foremost, the intracranial space and physical layout of the brain create significant obstacles to treatment and recovery. The brain is made of, primarily, astrocytes (which make up the majority of the brain mass, and serve as a scaffold and support for the neurons), neurons (which carry the actual electrical impulses of the nervous system), and a minor contingent of other cells such as insulating oligodendrocytes (which produce myelin). These cell types give rise to primary brain tumors (e.g., astrocytomas, neuroblastomas, glioblastomas, oligodendrogliomas, etc.) Although the World Health Organization has recently established standard guidelines, the nomenclature for brain tumors is somewhat imprecise, and the terms astrocytoma and glioblastoma are often used broadly. The brain is encased in the relatively rigid shell of the skull, and is cushioned by the cerebrospinal fluid, much like a fetus in the womb. Because of the relatively small volume of the skull cavity, minor changes in the volume of tissue in the brain can dramatically increase intracranial pressure, causing damage to the entire organ (i.e., “water on the brain”). Thus, even small tumors can have a profound and adverse affect on the brain's function. In contrast, tumors in the relatively distensible abdomen may reach several pounds in size before the patient experiences adverse symptoms. The cramped physical location of the cranium also makes surgery and treatment of the brain a difficult and delicate procedure. However, because of the dangers of increased intracranial pressure from the tumor, surgery is often the first strategy of attack in treating brain tumors. [0004]
  • In addition to its physical isolation, the brain is chemically and biologically isolated from the rest of the body by the so-called “Blood-Brain-Barrier” (or BBB). This physiological phenomenon arises because of the “tightness” of the epithelial cell junctions in the lining of the blood vessels in the brain. Although nutrients, which are actively transported across the cell lining, may reach the brain, other molecules from the bloodstream are excluded. This prevents toxins, viruses, and other potentially dangerous molecules from entering the brain cavity. However, it also prevents therapeutic molecules, including many chemotherapeutic agents that are useful in other types of tumors, from crossing into the brain. Thus, many therapies directed at the brain must be delivered directly into the brain cavity (e.g., by an Ommaya reservoir), or administered in elevated dosages to ensure the diffusion of an effective amount across the BBB. [0005]
  • With the difficulties of administering chemotherapies to the brain, radiotherapy approaches have also been attempted. However, the amount of radiation necessary to completely destroy potential tumor-producing cells also produce unacceptable losses of healthy brain tissue. The retention of patient cognitive function while eliminating the tumor mass is another challenge to brain tumor treatment. Neoplastic brain cells are often pervasive, and travel throughout the entire brain mass. Thus, it is impossible to define a true “tumor margin,” unlike, for example, in lung or bladder cancers. Unlike reproductive (ovarian, uterine, testicular, prostate, etc.), breast, kidney, or lung cancers, the entire organ, or even significant portions, cannot be removed to prevent the growth of new tumors. In addition, brain tumors are very heterogeneous, with different cell doubling times, treatment resistances, and other biochemical idiosyncrasies between the various cell populations that make up the tumor. This pervasive and variable nature greatly adds to the difficulty of treating brain tumors while preserving the health and function of normal brain tissue. [0006]
  • Although current surgical methods offer considerably better post-operative life for patients, the current combination therapy methods (surgery, low-dosage radiation, and chemotherapy) have only improved the life expectancy of patients by one month, as compared to the methods of 30 years ago. Without effective agents to prevent the growth of brain tumor cells that are present outside the main tumor mass, the prognosis for these patients cannot be significantly improved. Although some immuno-affinity agents have been proposed and tested for the treatment of brain tumors, see, e.g., the tenascin-targeting agents described in U.S. Pat. No. 5,624,659, these agents have not proven sufficient for the treatment of brain tumors. Thus, therapeutic agents which are directed towards new molecular targets, and are capable of specifically targeting and killing brain tumor cells, are urgently needed for the treatment of brain tumors. [0007]
  • ARP-2 (Angiopoeitin Related Protein-2, Angiopoeitin Like-2 [ANGPTL-2]) [0008]
  • Angiopoeitin related protein-2 (ARP-2), is related to the angiopoeitin family of proteins, that includes Ang-1 and Ang-2. Like members of the angiopoeitin family, ARP-2 contains a coiled-coil domain in the amino terminal portion and a fibrinogen-like domain in the carboxyl terminal portion. However, ARP-2 has a low homology with Ang-1 and Ang-2 and unlike Ang-1 and Ang-2, ARP-2 does not bind to the Tie-2 receptor, nor does ARP-2 bind to the closely related Tie-1 receptor. Hence, ARP-2 is believed to be part of a newly identified family of proteins termed angiopocitin related proteins. Like the angiopoeitins, ARP-2 is a member of the fibrinogen superfamily, which also includes the fibrinogens and lectins. [0009]
  • ARP-2 is a glycosylated, secretory protein that induces sprouting in endothelial cells, most likely through autocrine or paracrine signaling, and it is preferentially expressed in the blood vessels and muscle cells. Hence, ARP-2 mediates the differentiated state of endothelial cells or for vascular remodeling and development. ARP-2 has not heretofore been associated with brain tumors. [0010]
  • SPARC (Secreted Protein, Aacidic, Cysteine-Rich; Osteonectin; Basement Membrane Protein (bm) 40) [0011]
  • Secreted protein acidic and rich in cysteine, SPARC or BM-40, is a member of the counter-adhesive family of proteins. It is a developmentally regulated, secreted glycoprotein expressed in fetal astrocytes, particularity during tissue remodeling, vessel morphogenesis, and in response to stress. It has been hypothesized that SPARC may affect cell migration and vascular morphogenesis either by directly interacting with extracellular matrix (ECM) proteins (such as collagens I, III, IV and V) or by initiating a receptor mediated signaling event that induces changes in cytoplasmic components associated with focal adhesions. SPARC has been found to bind directly to vitronectin, a multifunctional adhesive protein that is a component of the brain vascular basement membranes. [0012]
  • SPARC may indirectly affect cell migration and motility by regulating the expression of matrix metallo-proteases and by modulating the expression of other proteolytic enzymes (such as collagenase) that degrade the ECM. Increased SPARC expression has also been observed in two forms of low-grade malignant gliomas, in all grades of human astrocytic tumors, and in tumor cells invading adjacent brain at the tumor/brain interface. Hence, SPARC may be an astrocytoma invasion related gene that functions in connection with vitronectin to balance the modulation of cellular adhesion to the ECM and it may promote diffuse tumor cell infiltration into adjacent brain by affecting both tumor and endothelial cell-ECM interactions. [0013]
  • Because SPARC is also found in bone, dentine, and many normal and neoplastic human soft tissues it may also play a regulatory function in the control of such diverse processes as bone mineralization, cell shape, tissue remodeling or repair, cell migration, proliferation, and differentiation. SPARC is also synthesized, stored, and secreted by human blood platelets, binds to plasminogen, and enhances tissue plasminogen activator conversion of plasminogen to plasmin. [0014]
  • c-MET (Met Proto-Oncogene Tyrosine Kinase, Hepatocyte Growth Factor Receptor [HGFR]) [0015]
  • c-MET is a member of the Hepatocyte Growth Factor Receptor (HGFR) family and a heterodimeric cellular receptor for Hepatocyte Growth Factor (HGF). c-MET contains a disulfide-linked α-chain of 50-kDa (which is located in the extracellular domain,) a 145-kDa β-chain (which includes an extracellular region,) a transmembrane spanning domain, and an intracellular tyrosine kinase domain that can be activated by autophosphorylation. Hence, HGFR is a subset of the protein tyrosine-kinase family of membrane-spanning, cell surface receptors. [0016]
  • The receptor-ligand pair, c-MET and HGF, function as a growth factor, regulating cell growth, migration, and morphogenesis, and hence, may play a role in neoplastic formation and metastasis. Upon HGF or macrophage stimulating protein (MSP) binding, the c-MET protein receptor goes through a conformational change wherein the intracellular tyrosine residues of the β subunit become phosphorylated at residue 1235, and a second messenger signal cascade is induced. This change activates c-MET's intracellular receptor kinase activity, which is important to the growth and differentiation of epithelial cells in normal and malignant tissues. c-MET has been identified in both normal brain and on glial tumors, and is thought to be determinant in the pathological processes of various malignancies. For instance, detailed studies have shown that glioblastoma multiforme (GBM), a highly malignant brain tumor of astrocytic origin, expresses c-MET, and this research suggests a role in tumor progression. [0017]
  • BEHAB (Brain-Enriched Hyaluronan Binding Protein, Brevican) [0018]
  • BEHAB is a brain-specific, extracellular matrix protein, that is a member of the chondroitin sulfate proteoglycan (CSPG) family. BEHAB is expressed only in the CNS. Although its function is unclear, BEHAB is reported to bind to HA at the N-terminus, lectins at the C-terminus, and may mediate binding of other ECM components like tenascin. This suggests that BEHAB may play a role in cell-cell and cell-matrix interactions thereby maintaining the extracellular environment of the brain. It has been reported that the highest levels of expression of BEHAB is during brain development and at times and places where glial cells are highly motile, as in cases of brain injury or trauma. BEHAB expression is also unregulated in primary gliomas of the central nervous system, but not in tumors of non-glial origin. In surgical samples of human gliomas (including astrocytoma, oligodendroglioma, and glioblastoma tumors), BEHAB expression is consistently and dramatically increased over the level of expression in the normal brain. Hence, BEHAB expression correlates with an invasive phenotype that promotes gliogenesis by contributing to cell movement through the ECM. [0019]
  • CD-44 Antigen [0020]
  • CD-44 is a single-path, type I transmembrane protein with extracellular domains that are flexibly linked to the transmembrane segment. CD-44 is a member of the cartilage link protein family and belongs to the hyaloadherin or link protein superfamily (LPSF). As other members of the LPS family, CD-44 can be extensively glycosylated and is typically decorated with glycosaminoglycans (e.g., chondroitin, heparin, and keratin sulfate). The genomic structure of CD-44 consists of 21 exons, at least 11 of which can be variably spliced (v1 -v10), that are located in the membrane-proximal extracellular region. Alternative splicing of these exons give rise to a variety of CD-44 isoforms (at least 30 different isoforms have been characterized to date) that are widely distributed and expressed in a cell-specific manner. Among the most frequently occurring isoforms are CD-44H, expressed on hematopoietic cells, and CD-44E, expressed in epithelial cells. CD-44(H) has also been found to be expressed in lymphocytes, macrophages, erythrocytes, fibroblasts, epithelial and endothelial cells, and neurons. It is the predominant isoform in normal brain and neuroectoderm-derived tumors and is expressed on both normal astrocytes and oligodendrocytes as well on neoplastic astrocytes and glioblastomas. [0021]
  • The family of CD-44 proteins has been implicated in lymphocyte activation and homing, endothelial migration, and tumor cell metastasis. CD-44 is believed to be the major receptor for Hyaluronic acid (HA). CD-44/HA interactions underlie a wide spectrum of functions in embryonic morphogenesis and organogenesis, hematopoeisis, lymphocyte homing. CD-44 also mediates the attachment of glioma cells to chondroitin sulfate, types I and IV collagen, fibronectin laminin, vitronectin and Martrigel. This suggest that CD-44 may play a role in cell-cell and cell-matrix interactions, affecting the extracellular environment of the brain. Because HA is a major component of the brain ECM, and CD-44 is one of the principal cellular receptors of HA, CD-44 expression coincides with brain tumor growth and invasiveness. [0022]
  • PTN (Pleiotrophin, Heparin Binding [0023] Growth Factor 8, Neurite Growth-Promoting Factor 1)
  • Pleiotrophin or PTN, is a platelet-derived, growth factor inducible, member of the pleiotrophin family of proteins that includes midkine and retinoic acid-induced heparin-binding protein. It is a developmentally regulated, secreted cytokine that stimulates mitogenesis, angiogenesis, and neurite and glial process outgrowth guidance activities. During development PTN is expressed in the brain, intestine, muscle, skin, heart, lung and kidney. In the adult, PTN is found primarily in the brain in association with axonal tracts during active mitogenesis and may therefore play an important role in the development and maintenance of the nervous system. It has been found to bind heparin, heparin sulfate proteoglycans, the extracellular matrix, and is also a natural ligand for receptor protein tyrosine phosphatase (RPTP), signaling through ligand dependant receptor inactivation of RPTP. Receptor mediated endocytosis occurs following PTN binding and may be disrupted by heparin. [0024]
  • PTN has also been found to have oncogenic properties, inducing malignant transformation and tumor growth and progression. It has been described as a proto-oncogene that is expressed in many human tumors and cell lines derived from human tumors. PTN is a mitogen for fibroblasts, epithelial and endothelial cells, stimulates plasminogen-activator production, can induce tube formation, and therefore can serve as a tumor angiogenesis factor. [0025]
  • OPN (Osteopontin, Secreted Phosphoprotein 1, Bone Sialoprotein-1) [0026]
  • Osteopontin or OPN, is a member of the osteopontin family. It is a glycosylated sialoprotein that is heavily phosphorylated and expressed in a variety of cells including bone, kidney, placenta, nerve cells and macrophages, as well as T lymphocytes, epidermal and bone cells. OPN is a part of the mineralized bone matrix and may play a role in bone resorption, by facilitating the attachment of osteoclasts to the bone surface, and may be functionally important as an adhesive and chemotactic molecule for vascular cells. OPN is a secreted protein that binds tightly to hydroxyapatite, and hence, is important to cell matrix interactions. It has been observed to interact with the CD-44 homing receptor to physiologically induce macrophage chemotaxis, which may be a mechanism utilized by metastatic brain tumors in the process of dissemination. [0027]
  • OPN has been observed in the microvasculature of glioblastomas associated with VEGF expression and OPN mRNA has been found to be overexpressed in high grade and metastatic brain tumors. Hence, OPN expression correlates with the malignancy grade of gliomas. [0028]
  • VIPR-2 (Vasoactive Intestinal Peptide Receptor-2) [0029]
  • Vasoactive intestinal polypeptide receptor II (VIPR-2), VPAC-2, is a member of the G-protein receptor family, which includes such members as the calcitonin, parathyroid hormone, secretin, glucagon and VIP-1 receptors. VIPR-2 is a seven-transmembrane spanning G protein-coupled receptor that responds to VIP by stimulating cAMP production. VIPR-2 is found in the brain as well as peripheral tissues such as the pancreas, skeletal muscle, heart, lung, kidneys, stomach, adipocytes and the liver, and in various cells of the immune system. In the brain, VIPR-2 functions as a neuroendocrine hormone and neurotransmitter receptor, and is found in the thalamus, hippocampus, suprachiasmatic nucleus and hypothalamus. [0030]
  • VIPR-2 is encoded by a nucleotide sequence of approximately 2.8 kb, which codes for a 438 amino acid sequence of approximately 48-64 kDa. The receptor-ligand pair, VIPR-2 and VIP, have various functions dependent upon the tissue where in they are located. VIP is a late-developing, 28 amino acid peptide that, along with its receptor, is widely distributed throughout the peripheral body, and plays a role in cardiovascular, reproductive, pulmonary, immune and gastrointestinal systems, to effect vasodilatation, bronchodilation, immunosuppression, hormonal secretion, and increased gastric motility. However, the cerebral cortex has one of the highest reported concentrations of VIP, localized to intrinsic neurons throughout all neocortical regions. In the brain, VIP and its receptor, have behavioral, electrophysiological, secretory, metabolic, vascular, and mitogenic effects. For instance, the receptor-ligand pair play a role in cortical differentiation, the relaying of sensory information to the cortex, and the regulation of morphogenic events by the release of diffusible signals from glial cells. VIPR-2 and VIP also play a role in the growth and differentiation of neuroblastomas. [0031]
  • TSPAN3 ([0032] Tetraspanin 3, Tetraspanin TM-4A)
  • The Tetraspanin superfamily, is a family of approximately 20 integral membrane proteins that are broadly expressed in most human tissues including neural and bone marrow derived tissues. The family shares a common motif that includes four putative transmembrane domains (TM1-4), a small extracellular domain (EC1) of 20-27 amino acids, and a larger extracellular domain (EC2) between TMS3 and TMS4 of 70-130 amino acids. Two conserved features of tetraspanins are critical to their structure and function. First, charged residues are present in or near the TM domains, second, a cluster of cysteine residues is in the putative EC2 domain. Most of the tetraspanins are modified by N-glycosylation. [0033]
  • Many Tetraspanin proteins affect the regulation of cellular proliferation, motility, differentiation, development. In some cells, Tetraspanins may act as adapters in ultimeric complexes that link plasma membrane proteins, like integrins, into signaling complexes with other signaling molecules (e.g., phosphatidylinositol 4-kinase) at the plasma membrane and play a role in integrin-mediated cell migration, metastasis and tumor cell invasion. A number of tetraspanins have also been discovered as tumor-associated proteins, including C-029, PETA-3/SFA-1, and SAS, which is amplified in a subset of sarcomas. Of the various TM4SF proteins, CD9, CD63, CD81, CD82, and CD151 are the most widely distributed. CD9 is expressed on 90% of non-T cell acute lymphoblastic leukemia cells and on 50% of chronic lymphocytic and acute myeloblastic leukemias. CD63 is also expressed in early stage melanomas. [0034]
  • Protein Tyrosine Phosphatase Receptor Zeta (PTPζ) [0035]
  • Vital cellular functions, such as cell proliferation and signal transduction, are regulated in part by the balance between the activities of protein kinases and protein phosphatases. These protein-modifying enzymes add or remove a phosphate group from serine, threonine, or tyrosine residues in specific proteins. Some tyrosine kinases (PTK's) and phosphatases (PTPase's) have been theorized to have a role in some types of oncogenesis, which is thought to result from an imbalance in their activities. There are two classes of PTPase molecules: low molecular weight proteins with a single conserved phosphatase domain such as T-cell protein-tyrosine phosphatase (PTPT; MIM 176887), and high molecular weight receptor-linked PTPases with two tandemly repeated and conserved phosphatase domains separated by 56 to 57 amino acids. Examples of this latter group of receptor proteins include: leukocyte-common antigen (PTPRC; MIM 151460) and leukocyte antigen related tyrosine phosphatase (PTPRF; MIM 179590). [0036]
  • Protein tyrosine phosphatase zeta (PTPζ) [also known as PTPRZ, HPTP-ZETA, HPTPZ, RPTP-BETA(β), or RPTPB] was isolated as a cDNA sequence by two groups in the early nineties. The complete cDNA sequence of the protein is provided in SEQ ID NO. 1, and the complete deduced amino acid sequence is provided in SEQ ID NO. 2. Splicing variants and features are indicated in the sequences. Levy et al. (“The cloning of a receptor-type protein tyrosine phosphatase expressed in the central nervous system” [0037] J. Biol. Chem. 268: 10573-10581, (1993)) isolated cDNA clones from a human infant brain step mRNA expression library, and deduced the complete amino acid sequence of a large receptor-type protein tyrosine phosphatase containing 2,307 amino acids.
  • Levy found that the protein, which they designated PTP-β (PTPζ), is a transmembrane protein with 2 cytoplasmic PTPase domains and a 1,616-amino acid extracellular domain. As in PTP-γ (MIM 176886), the 266 N-terminal residues of the extracellular domain are have a high degree of similarity to carbonic anhydrases (see MIM 114880). The human gene encoding PTPζ has been mapped to chromosome 7q31.3-q32 by chromosomal in situ hybridization (Ariyama et al., “Assignment of the human protein tyrosine phosphatase, receptor-type, zeta (PTPRZ) gene to chromosome band 7q31.3[0038] ” Cytogenet. Cell Genet. 70: 52-54 (1995)). Northern blot analysis has shown that showed that PTP-zeta is expressed only in the human central nervous system. By in situ hybridization, Levy et al. (1993) localized the expression to different regions of the adult human brain, including the Purkinje cell layer of the cerebellum, the dentate gyrus, and the subependymal layer of the anterior horn of the lateral ventricle. Levy stated that this was the first mammalian tyrosine phosphatase whose expression is restricted to the nervous system. In addition, high levels of expression in the murine embryonic brain suggest an important role in CNS development.
  • Northern analysis has shown three splice variants: the extracellular proteoglycan phosphacan, which contains the full extracellular region of the protein, and the long (α) and short (β) forms of the transmembrane phosphatase. The β form lacks the extracellular 860 aa long insert domain of the protein, therefore it is not glycosylated. PCR studies of the gene in rat genomic DNA indicated that there are no introns at the putative 5′ and 3′ splice sites or in the 2.6 kb segment which is deleted in the short transmembrane protein. The phosphatases and the extracellular proteoglycan have different 3′-untranslated regions. Additional alternative mRNA splicing is likely to result in the deletion of a 7 amino acid insert from the intracellular juxtamembrane region of both long and short phosphatase isoforms. Simultaneous quantitation of the three major isoforms indicated that the mRNA encoding phosphacan had the highest relative abundance in the CNS while that encoding the short phosphatase isoform was most abundant relative to the other PTPζ variants in the PNS. [0039]
  • PTPζ has only been found to be expressed in the nervous system. By in situ hybridization, it has been localized to different regions of the adult brain, including the Purkinje cell layer of the cerebellum, the dentate gyrus, and the subependymal layer of the anterior horn of the lateral ventricle. High levels of PTPζ have been seen in regions of the brain where there is continued neurogenesis and neurite outgrowth, and it seems to play a role in morphogenesis and plasticity of the nervous system. Phosphacan immunoreactivity has been associated with perineuronal nets around parvalbumin-expressing neurons in adult rat cerebral cortex. Neurons as well as astrocytes have been shown to express phosphacan. [0040]
  • The transmembrane forms of PTPζ are expressed on the migrating neurons especially at the lamellipodia along the leading processes. PTPζ is postulated to be involved in the neuronal migration as a neuronal receptor of pleiotrophin distributed along radial glial fibers. PTPζ has been shown to be highly expressed in radial glia and other forms of glial cells that play an important role during development. The anti-PTPζ staining localizes to the radial processes of these cells, which act as guides during neuronal migration and axonal elongation. The pattern of RPTP-zeta expression has also been shown to change with the progression of glial cell differentiation. [0041]
  • The three splicing variants of RPTP-zeta have been shown to have different spatial and temporal patterns of expression in the developing brain. The 9.5-kb and 6.4-kb transcripts, which encode the α and β transmembrane protein tyrosine phosphatases, were predominantly expressed in glial progenitors located in the subventricular zone. The 8.4-kb transcript, which encodes the secreted chondroitin sulfate proteoglycan phosphacan, was expressed at high levels by more mature glia that have migrated out of the subventricular zone. The three transcripts have also been shown to be differentially expressed in glial cell cultures. [0042]
  • In knockout studies, PTPζ-deficient mice were viable, fertile, and showed no gross anatomical alterations in the nervous system or other organs. Therefore, it was deduced that PTPζ is not essential for neurite outgrowth and node formation in mice. The ultrastructure of nerves of the central nervous system in PTPζ-deficient mice suggests a fragility of myelin. However, conduction velocity was not altered. The normal development of neurons and glia in was thought to indicate that PTPζ function is not necessary for these processes in vivo, or that a loss of PTPζ can be compensated for by other protein tyrosine phosphatases expressed in the nervous system. [0043]
  • Following CNS injury, robust induction of phosphatase forms of PTPζ mRNA has been observed in areas of axonal sprouting, and of both phosphatases and phosphacan mRNAs in areas of glial scarring. This is thought to imply that the encoded proteins and the cell adhesion molecules and extracellular matrix proteins to which they bind may contribute to recovery from injury and perhaps also to the regulation of axonal regrowth in the nervous system. Following peripheral nerve crush, all PTPζ mRNAs, including phosphacan and the phosphatase variants with and without the 21 base insert, were observed to be significantly induced in the distal segments of the sciatic nerve with a time course that correlated well with the response of Schwann cells to this injury. [0044]
  • The extracellular domains of PTPζ have been shown to be capable of binding to several cell adhesion molecules. Phosphacan, which is the shortest, secreted form of PTPζ, containing the full extracellular region, previously was designated 3F8 and 6B4 chondroitin sulfate proteoglycan or 3H1 keratin sulfate proteoglycan depending on the glycosylation status. It is synthesized mainly by glia and binds to neurons and to the neural cell adhesion molecules Ng-CAM/L1, NCAM, TAG-1/axonin-1, to tenascin-C and R, to amphoterin and pleiotrophin/heparin-binding growth-associated molecule (HB-GAM) (amphoterin and pleiotrophin are heparin-binding proteins that are developmentally regulated in brain and functionally involved in neurite outgrowth). Binding of phosphacan to Ng-CAM/L1, NCAM, and tenascin-C (FNIII domain) is mediated by complex-type N-linked oligosaccharides on the proteoglycan. Phosphacan, shows saturable, reversible, high-affinity binding to fibroblast growth factor-2 (FGF-2). The interaction is mediated primarily through the core protein. Immunocytochemical studies have also shown an overlapping localization of FGF-2 and phosphacan in the developing central nervous system. The core protein of phosphacan may also regulate the access of FGF-2 to cell surface signaling receptors in nervous tissue. [0045]
  • The carbonic anhydrase (CAH) domain of PTPζ has been shown to bind specifically to contactin. Contactin is a 140 kDa GPI membrane-anchored neuronal cell recognition protein expressed on the surface of neuronal cells. The CAH domain of RPTP zeta was shown to induce cell adhesion and neurite growth of primary tectal neurons, and differentiation of neuroblastoma cells. These responses were blocked by antibodies against contactin, demonstrating that contactin is a neuronal receptor for RPTP zeta. Caspr ((p190/Caspr, a contactin-associated transmembrane receptor) and contactin exist as a complex in rat brain and are bound to each other by means of lateral (cis) interactions in the plasma membrane. The extracellular domain of Caspr contains a neurophilin/coagulation factor homology domain, a region related to fibrinogen beta/gamma, epidermal growth factor-like repeats, neurexin motifs as well as unique PGY repeats found in a molluscan adhesive protein. The cytoplasmic domain of Caspr contains a proline-rich sequence capable of binding to a subclass of SH3 domains of signaling molecules. Caspr may function as a signaling component of contactin, enabling recruitment and activation of intracellular signaling pathways in neurons. The role of the extracellular domains in neural adhesion and neurite growth induction was investigated by the use of fusion protein constructs. The results suggested that binding of glial PTPζ to the contactin/Nr-CAM complex is important for neurite growth and neuronal differentiation. [0046]
  • PTPζ was shown to bind to a heparin-binding growth factor midkine through the chondroitin sulfate portion of the receptor. The interactions of pleiotrophin (PTN) with the receptor in U373-MG cells was also studied. Pleiotrophin was shown to bind to the spacer domain. Results suggested that PTN signals through “ligand-dependent receptor inactivation” of PTPζ and disrupts its normal roles in the regulation of steady-state tyrosine phosphorylation of downstream signaling molecules. PTN was shown to bind to and functionally inactivate the catalytic activity of PTPζ. An active site-containing domain of PTPζ both binds β-catenin and functionally reduces its levels of tyrosine phosphorylation when added to lysates of pervanadate-treated cells. In unstimulated cells, PTPζ was shown to be intrinsically active, and thought to function as an important regulator in the reciprocal control of the steady-state tyrosine phosphorylation levels of β-catenin by tyrosine kinases and phosphatases. [0047]
  • Using the yeast substrate-trapping system, several substrate candidates for PTPζ were isolated. The results indicated that GIT1/Cat-1 is a substrate molecule of PTPζ. In addition, PTPζ was shown to bind to the PSD-95/SAP90 family through the second phosphatase domain. Immunohistochemical analysis revealed that PTPζ and PSD-95/SAP90 are similarly distributed in the dendrites of pyramidal neurons of the hippocampus and neocortex. Subcellular fractionation experiments indicated that PTPζ is concentrated in the postsynaptic density fraction. These results suggested that PTPζ is involved in the regulation of synaptic function as postsynaptic macromolecular complexes with PSD-95/SAP90. [0048]
  • Voltage-gated sodium channels in brain neurons were also found to associate with the membrane bound forms of PTPζ and phosphacan. Both the extracellular domain and the intracellular catalytic domain of PTPζ interacted with sodium channels. Sodium channels were tyrosine phosphorylated and were modulated by the associated catalytic domains of PTPζ. [0049]
  • SUMMARY OF THE INVENTION
  • The present invention provides novel methods and reagents for specifically targeting brain tumor neoplastic cells for both therapeutic and imaging purposes, by targeting brain tumor protein targets (T[0050] BT). These targets have been identified by the applicants as being overexpressed in brain tumors, and thus allow for the selective inhibition of cell function or selective marking for visualization with therapeutic or visualizing compositions which have a specific affinity for thes eprotein targets. Each of angiopoietin related protein 2 (ARP-2,) secreted protein acidic, rich in cysteine (SPARC,) c-met proto-oncogene (C-MET,) brevican (BEHAB,) CD-44 antigen (CD-44,) tetraspanin 3 (TSPN3,) pleiotrophin (PTN,) osteopontin (OPN,) vasoactive intestinal peptide receptor-2 (VIPR-2,) and receptor protein tyrosine phosphatase zeta (PTPζ) [including the two novel isoforms PTPζ SM1 and SM2], as the proteins are described below, have been identified as an independently useful protein target TBT. In some preferred embodiments of the invention, either novel isofom PTPζ SM1 or PTPζ SM2 is the protein target TBT. Thus, the aspects of the invention with relation to each of these TBT are described generally as follows:
  • In a first aspect, the present invention provides T[0051] BT affinity-based compounds and compositions useful in treating a brain tumor in a patient. The compositions and compounds of this aspect of the invention generally fall into two groups: TBT-binding conjugate compounds, which comprise a cytotoxic moiety (C), which inhibits the growth of tumor cells; and TBT-binding compound compositions in which the TBT binding moiety alters the normal function of the TBT in or around the tumor cell, thus inhibiting cell growth and/or function.
  • In a first group of embodiments of this aspect of the invention, T[0052] BT-binding therapeutic conjugate compounds are provided. These compounds have the general formula α(TBT)C, wherein α(TBT) is one or more moieties which specifically binds to a TBT, and C is one or more cytotoxic moieties. In preferred embodiments, α(TBT) is an antibody or an antibody fragment. In particularly preferred embodiments, α(TBT) is an antibody or an antibody fragment which elicits a reduced immune response when administered to a human patient. Preferred cytotoxic moieties for use in these embodiments of the invention include radioactive moieties, chemotoxic moieties, and toxin proteins. The invention also provides compositions comprising these TBT-binding therapeutic conjugate compounds in a pharmaceutically acceptable carrier.
  • In a second group of embodiments of this first aspect of the invention, T[0053] BT-binding therapeutic compounds are provided which alter the normal function of the TBT in or around brain tumor cells and inhibit brain tumor cell growth. These TBT -binding therapeutic compounds have the general formula α(TBT), wherein α(TBT) is one or more moieties which specifically binds to a TBT, and wherein the binding of α(TBT) alters the function of the TBT. In preferred embodiments, α(TBT) is an antibody or an antibody fragment. In particularly preferred embodiments, α(TBT) is an antibody or an antibody fragment which elicits a reduced immune response when administered to a human patient. It is preferred that the therapeutic compounds of this second group of embodiments of the first aspect of the invention be formulated into therapeutic compositions comprising the TBT-binding compound in a pharmaceutically acceptable carrier.
  • In a second aspect, the present invention provides methods for using these compounds and compositions to treat a brain tumor in a patient. The methods comprise administering an effective amount of a composition, comprising a T[0054] BT-binding compound from the first or second group of embodiments of the first aspect and a pharmaceutically acceptable carrier, to a patient in need thereof. Brain tumors treated in this fashion may be glioblastomas, astrocytomas, neuroblastomas, or any type of brain tumor. Administration of the therapeutic composition may be by any acceptable means. One preferred method for administration is by intrathecal administration, although intravascular administration is also preferred.
  • In a third aspect, the present invention provides T[0055] BT affinity-based compounds and compositions for the visualization of brain tumors in patients. These compounds have the general formula α(TBT)I, wherein α(TBT) is one or more moieties which specifically binds to a TBT, and I is one or more imaging moieties. In preferred embodiments, α(TBT) is an antibody or an antibody fragment. In particularly preferred embodiments, αA(TBT) is an antibody or an antibody fragment which elicits a reduced immune response when administered to a human patient. Preferred I moieties include radiographic moieties (useful in, e.g., x-ray, scintillation, or other radiation imaging methods,) positron-emitting moieties, magnetic spin contrast moieties, and optically visible moieties (such as visible particles, fluorescent dyes, and visible-spectrum dyes.) It is preferred that the imaging compounds of these embodiments of the third aspect of the invention be formulated into therapeutic compositions comprising the TBT-binding compound in a pharmaceutically acceptable carrier.
  • In a fourth aspect, the present invention provides methods of using the compounds and compositions of the third aspect of the invention to visualize a brain tumor in a patient. These methods generally comprise administering an effective amount of an imaging compound of the general formula α(T[0056] BT)I in a pharmaceutically acceptable carrier to the patient, and then visualizing the imaging moieties of the compound. Administration of the imaging composition may be by any acceptable means. Intravascular administration of the imaging composition is preferred in these methods, although intrathecal administration is also preferred. Preferred methods of visualizing the imaging moieties of the compounds include radiographic imaging techniques (e.g., x-ray imaging and scintillation imaging techniques), positron-emission tomography, magnetic resonance imaging techniques, and direct or indirect (e.g., endoscopic) visual inspection.
  • Various particular embodiments of these aspects of the invention include: [0057]
  • A method to treat a brain tumor by administering a therapeutic amount of a composition comprising a compound of the general formula α(ARP2)C, wherein α(ARP2) is one or more moieties which specifically binds to a human angiopoietin related protein-2, and C is one or more cytotoxic moieties, and a pharmaceutically acceptable carrier. [0058]
  • A compound for the treatment of a brain tumor of the general formula α(ARP2)C, wherein α(ARP2) is one or more moieties which specifically binds to a human angiopoietin related protein-2, and C is one or more cytotoxic moieties. [0059]
  • A method to treat a brain tumor by administering a therapeutic amount of a composition comprising a compound of the general formula α(ARP2), wherein α(ARP2) is one or more moieties which specifically binds to a human angiopoietin related protein-2, wherein the binding of α(ARP2) alters the function of the angiopoietin related protein-2, and a pharmaceutically acceptable carrier. [0060]
  • A method for visualizing a brain tumor in a patient by first administering to a patient an effective amount of a composition comprising: a compound of the general formula α(ARP2)I, wherein α(ARP2) is one or more moieties which specifically binds to a human angiopoietin related protein-2, and I is one or more imaging moieties and a pharmaceutically acceptable carrier, and then visualizing the imaging moieties of the compound. [0061]
  • A composition for the visualization of a brain tumor comprising a compound of the general formula α(ARP2)I, wherein α(ARP2) is one or more moieties which specifically binds to a human angiopoietin related protein-2, and I is one or more imaging moieties, and a pharmaceutically acceptable carrier. [0062]
  • A method to treat a brain tumor by administering a therapeutic amount of a composition comprising a compound of the general formula α(TSPAN3)C, wherein α(TSPAN3) is one or more moieties which specifically binds to a [0063] human tetraspanin 3, and C is one or more cytotoxic moieties, and a pharmaceutically acceptable carrier.
  • A compound for the treatment of a brain tumor of the general formula α(TSPAN3)C, wherein α(TSPAN3) is one or more moieties which specifically binds to a [0064] human tetraspanin 3, and C is one or more cytotoxic moieties.
  • A method to treat a brain tumor by administering a therapeutic amount of a composition comprising a compound of the general formula α(TSPAN3), wherein α(TSPAN3) is one or more moieties which specifically binds to a [0065] human tetraspanin 3, wherein the binding of α(TSPAN3) alters the function of the tetraspanin 3, and a pharmaceutically acceptable carrier.
  • A composition for the treatment of a brain tumor comprising: a compound of the general formula α(TSPAN3), wherein α(TSPAN3) is one or more moieties which specifically binds to a [0066] human tetraspanin 3, wherein the binding of α(TSPAN3) alters the function of the tetraspanin 3, and a pharmaceutically acceptable carrier.
  • A method for visualizing a brain tumor in a patient by first administering to a patient an effective amount of a composition comprising: a compound of the general formula α(TSPAN3)I, wherein α(TSPAN3) is one or more moieties which specifically binds to a [0067] human tetraspanin 3, and I is one or more imaging moieties, and a pharmaceutically acceptable carrier, and then visualizing the imaging moieties of the compound.
  • A composition for the visualization of a brain tumor comprising a compound of the general formula α(TSPAN3)I, wherein α(TSPAN3) is one or more moieties which specifically binds to a [0068] human tetraspanin 3, and I is one or more imaging moieties, and a pharmaceutically acceptable carrier.
  • A method to treat a brain tumor by administering a therapeutic amount of a composition comprising a compound of the general formula α(PTPζ)C, wherein α(PTPζ) is one or more moieties which specifically binds to a human PTPζ selected from the group consisting of PTPζ SM1 and PTPζ SM2, further wherein α(PTPζ) does not specifically bind to human PTPζ α, human PTPζ β, or phosphacan, and C is one or more cytotoxic moieties, and a pharmaceutically acceptable carrier. [0069]
  • A compound for the treatment of a brain tumor of the general formula α(PTPζ)C, wherein α(PTPζ) is one or more moieties which specifically binds to a human PTPζ selected from the group consisting of PTPζ SM1 And PTPζ SM2, further wherein α(PTPζ) does not specifically bind to human PTPζ α, human PTPζ β,or human phosphacan, and C is one or more cytotoxic moieties. [0070]
  • A method to treat a brain tumor by administering a therapeutic amount of a composition comprising a compound of the general formula α(PTPζ), wherein α(PTPζ) is one or more moieties which specifically binds to a human PTPζ selected from the group consisting of PTPζ SM1 And PTPζ SM2, further wherein α(PTPζ) does not specifically bind to human PTPζ α, human PTPζ, or phosphacan, wherein the binding of α(PTPζ) alters the function of the human PTPζ,and a pharmaceutically acceptable carrier. [0071]
  • A composition for the treatment of a brain tumor comprising a compound of the general formula α(PTPζ), wherein α(PTPζ) is one or more moieties which specifically binds to PTPζ selected from the group consisting of PTPζ SM1 And PTPζ SM2, further wherein α(PTPζ) does not specifically bind to human PTPζ α, human PTPζ α, or phosphacan, wherein the binding of α(PTPζ) alters the function of the PTPζ, and a pharmaceutically acceptable carrier. [0072]
  • A method for visualizing a brain tumor in a patient by first administering to a patient an effective amount of a composition comprising: a compound of the general formula α(PTPζ)I, wherein α(PTPζ) is one or more moieties which specifically binds to a human PTPζ selected from the group consisting of PTPζ SM1 And PTPζ SM2, further wherein α(PTPζ) does not specifically bind to human PTPζ α, human PTPζ β, or phosphacan, and I is one or more imaging moieties and a pharmaceutically acceptable carrier, and then visualizing the imaging moieties of the compound. [0073]
  • A composition for the visualization of a brain tumor comprising a compound of the general formula α(PTPζ)I, wherein α(PTPζ) is one or more moieties which specifically binds to a human PTPζ selected from the group consisting of PTPζ SM1 And PTPζ SM2, further wherein α(PTPζ) does not specifically bind to human PTPζ α, human PTPζ β, or phosphacan, and I is one or more imaging moieties, and a pharmaceutically acceptable carrier. [0074]
  • A method to treat a brain tumor by administering a therapeutic amount of a composition comprising a compound of the general formula α(SPARC)C, wherein α(SPARC) is one or more moieties which specifically binds to a human secreted protein, rich in cysteine, and C is one or more cytotoxic moieties, and a pharmaceutically acceptable carrier. [0075]
  • A compound for the treatment of a brain tumor of the general formula α(SPARC)C, wherein α(SPARC) is one or more moieties which specifically binds to a human secreted protein, rich in cysteine, and C is one or more cytotoxic moieties. [0076]
  • A method to treat a brain tumor by administering a therapeutic amount of a composition comprising a compound of the general formula α(SPARC), wherein α(SPARC) is one or more moieties which specifically binds to a human secreted protein, rich in cysteine, wherein the binding of α(SPARC) alters the function of the secreted protein, rich in cysteine, and a pharmaceutically acceptable carrier. [0077]
  • A method for visualizing a brain tumor in a patient by first administering to a patient an effective amount of a composition comprising: a compound of the general formula α(SPARC)I, wherein α(SPARC) is one or more moieties which specifically binds to a human secreted protein, rich in cysteine, and I is one or more imaging moieties, and a pharmaceutically acceptable carrier, and then visualizing the imaging moieties of the compound. [0078]
  • A composition for the visualization of a brain tumor comprising a compound of the general formula α(SPARC)I, wherein α(SPARC) is one or more moieties which specifically binds to a human secreted protein, rich in cysteine, and I is one or more imaging moieties, and a pharmaceutically acceptable carrier. [0079]
  • A method to treat a brain tumor by administering a therapeutic amount of a composition comprising a compound of the general formula α(c-MET)C, wherein α(c-MET) is one or more moieties which specifically binds to a human c-MET oncogene product, and C is one or more cytotoxic moieties and a pharmaceutically acceptable carrier. [0080]
  • A method to treat a brain tumor by administering a therapeutic amount of a composition comprising a compound of the general formula α(c-MET), wherein α(c-MET) is one or more moieties which specifically binds to a human c-MET oncogene product, wherein the binding of α(TSPAN3) alters the function of the c-MET oncogene product, and a pharmaceutically acceptable carrier. [0081]
  • A method for visualizing a brain tumor in a patient by first administering to a patient an effective amount of a composition comprising: a compound of the general formula α(c-MET)I, wherein α(c-MET) is one or more moieties which specifically binds to a human c-MET oncogene product, and I is one or more imaging moieties and a pharmaceutically acceptable carrier, and then visualizing the imaging moieties of the compound. [0082]
  • A method to treat a brain tumor by administering a therapeutic amount of a composition comprising a compound of the general formula α(CD44)C, wherein α(CD44) is one or more moieties which specifically binds to a human CD44 antigen, and C is one or more cytotoxic moieties, and a pharmaceutically acceptable carrier. [0083]
  • A method to treat a brain tumor by administering a therapeutic amount of a composition comprising a compound of the general formula α(CD44), wherein α(CD44) is one or more moieties which specifically binds to a human CD44 antigen, wherein the binding of α(CD44) alters the function of the CD44 antigen, and a pharmaceutically acceptable carrier. [0084]
  • A method for visualizing a brain tumor in a patient by first administering to a patient an effective amount of a composition comprising: a compound of the general formula α(CD44)I, wherein α(CD44) is one or more moieties which specifically binds to a human CD44 antigen, and I is one or more imaging moieties, and a pharmaceutically acceptable carrier, and then visualizing the imaging moieties of the compound. [0085]
  • A method to treat a brain tumor by administering a therapeutic amount of a composition comprising a compound of the general formula α(VIPR2)C, wherein α(VIPR2) is one or more moieties which specifically binds to a human vasoactive intestinal peptide receptor-2, and C is one or more cytotoxic moieties and a pharmaceutically acceptable carrier. [0086]
  • A method to treat a brain tumor by administering a therapeutic amount of a composition comprising a compound of the general formula α(VIPR2), wherein α(VIPR2) is one or more moieties which specifically binds to a human vasoactive intestinal peptide receptor-2, wherein the binding of α(VIPR2) alters the function of the vasoactive intestinal peptide receptor-2, and a pharmaceutically acceptable carrier. [0087]
  • A method for visualizing a brain tumor in a patient by first administering to a patient an effective amount of a composition comprising: a compound of the general formula α(VIPR2)I, wherein α(VIPR2) is one or more moieties which specifically binds to a [0088] human tetraspanin 3, and I is one or more imaging moieties, and a pharmaceutically acceptable carrier, and then visualizing the imaging moieties of the compound.
  • A method to treat a brain tumor by administering a therapeutic amount of a composition comprising a compound of the general formula α(OPN)C, wherein α(OPN) is one or more moieties which specifically binds to a human osteopontin, and C is one or more cytotoxic moieties and a pharmaceutically acceptable carrier. [0089]
  • A compound for the treatment of a brain tumor of the general formula α(OPN)C, wherein (OPN) is one or more moieties which specifically binds to a [0090] human tetraspanin 3, and C is one or more cytotoxic moieties.
  • A method to treat a brain tumor by administering a therapeutic amount of a composition comprising a compound of the general formula α(OPN), wherein α(OPN) is one or more moieties which specifically binds to a human osteopontin, wherein the binding of α(OPN) alters the function of the osteopontin, and a pharmaceutically acceptable carrier. [0091]
  • A method for visualizing a brain tumor in a patient by first administering to a patient an effective amount of a composition comprising: a compound of the general formula α(OPN)I, wherein α(OPN) is one or more moieties which specifically binds to a human osteopontin, and I is one or more imaging moieties, and a pharmaceutically acceptable carrier, and then visualizing the imaging moieties of the compound. [0092]
  • A composition for the visualization of a brain tumor comprising a compound of the general formula α(OPN)I, wherein α(OPN) is one or more moieties which specifically binds to a human osteopontin, and I is one or more imaging moieties, and a pharmaceutically acceptable carrier. [0093]
  • A method to treat a brain tumor by administering a therapeutic amount of a composition comprising a compound of the general formula α(PTN)C, wherein α(PTN) is one or more moieties which specifically binds to a human pleiotrophin, and C is one or more cytotoxic moieties, and a pharmaceutically acceptable carrier. [0094]
  • A compound for the treatment of a brain tumor of the general formula α(PTN)C, wherein α(PTN) is one or more moieties which specifically binds to a human pleiotrophin, and C is one or more cytotoxic moieties. [0095]
  • A method to treat a brain tumor by administering a therapeutic amount of a composition comprising a compound of the general formula α(PTN), wherein α(PTN) is one or more moieties which specifically binds to a human pleiotrophin, wherein the binding of α(PTN) alters the function of the pleiotrophin, and a pharmaceutically acceptable carrier. [0096]
  • A method for visualizing a brain tumor in a patient by first administering to a patient an effective amount of a composition comprising: a compound of the general formula α(PTN)I, wherein α(PTN) is one or more moieties which specifically binds to a human pleiotrophin, and I is one or more imaging moieties, and a pharmaceutically acceptable carrier, and then visualizing the imaging moieties of the compound. [0097]
  • A composition for the visualization of a brain tumor comprising a compound of the general formula α(PTN)I, wherein α(PTN) is one or more moieties which specifically binds to a human pleiotrophin, and I is one or more imaging moieties, and a pharmaceutically acceptable carrier. [0098]
  • Brain tumors are known to be relatively heterogeneous, and thus all patients may not respond the same to a particular protein target treatment. Thus, in addition to the independent uses of each of the T[0099] BT protein targets as described above, the invention also provides, in yet another aspect, combination therapeutic and/or visualization agents, compositions, and methods. These combination embodiments of the invention may utilize as brain tumor protein targets any two or more of the identified targets angiopoietin related protein 2 (ARP-2,) secreted protein acidic, rich in cysteine (SPARC,) c-met proto-oncogene (c-MET,) brevican (BEHAB,) cd-44 antigen (CD-44,) tetraspanin 3 (TSPN3,) pleiotrophin (PTN,) osteopontin (OPN,) vasoactive intestinal peptide receptor-2 (VIPR-2,) and receptor protein tyrosine phosphatase zeta (PTPζ) [including the two novel isoforms PTPζ SM1 and SM2]. In some preferred embodiments, the brain tumor protein targets are selected from the group consisting of angiopoietin related protein 2 (ARP-2,) c-met proto-oncogene (c-MET,) cd-44 antigen (CD-44,) tetraspanin 3 (TSPN3,) osteopontin (OPN,) and receptor protein tyrosine phosphatase zeta (PTPζ). Embodiments of the combination aspect of the invention may target a group of proteins from the identified targets with similar compartmentalization characteristics. Thus, the combination aspects may target two or more secreted proteins from the group angiopoietin related protein 2 (ARP-2,) secreted protein acidic, rich in cysteine (SPARC,) brevican (BEHAB,) pleiotrophin (PTN,) secreted forms of receptor protein tyrosine phosphatase zeta (PTPζ) [including the novel isoform PTPζ SM1]. Or, the combination aspects may target one or more of the extracellular matrix binding proteins secreted protein acidic, rich in cysteine (SPARC,) and/or brevican (BEHAB,) with one or more of the other identified protein targets. Or, the combination aspects may target one or more of the membrane-bound proteins from the group c-met proto-oncogene (c-MET,) cd-44 antigen (CD-44,) tetraspanin 3 (TSPN3,) osteopontin (OPN,) vasoactive intestinal peptide receptor-2 (VIPR-2,) and membrane bound forms of receptor protein tyrosine phosphatase zeta (PTPζ) [including the novel isoform PTPζ SM2].
  • In preferred embodiments, at least one of the protein targets is selected from the proteins angiopoietin related protein 2 (ARP-2,) tetraspanin 3 (TSPN3,), and receptor protein tyrosine phosphatase zeta (PTPζ) [including the two novel isoforms PTPζ SM1 and SM2]. In other preferred embodiments, one of these proteins and another protein selected from the secreted group, the extracellular matrix group, or the membrane-bound group. In another group of preferred embodiments are combination aspects targeting two or more of angiopoietin related protein 2 (ARP-2,) tetraspanin 3 (TSPN3,), and receptor protein tyrosine phosphatase zeta (PTPζ) [including the two novel isoforms PTPζ SM1 and SM2]. Another group of preferred embodiments are combination aspects which target angiopoietin related protein 2 (ARP-2), and one or more proteins selected from the group secreted protein acidic, rich in cysteine (SPARC,) c-met proto-oncogene (C-MET,) brevican (BEHAB,) CD-44 antigen (CD-44,) tetraspanin 3 (TSPN3,) pleiotrophin (PTN,) osteopontin (OPN,) vasoactive intestinal peptide receptor-2 (VIPR-2,) and receptor protein tyrosine phosphatase zeta (PTPζ) [including the two novel isoforms PTPζ SM1 and SM2]. Another group of preferred embodiments are combination aspects which target tetraspanin 3 (TSPN3), and one or more proteins selected from the group secreted protein acidic, rich in cysteine (SPARC,) c-met proto-oncogene (C-MET,) brevican (BEHAB,) CD-44 antigen (CD-44,) angiopoietin related protein 2 (ARP-2,) pleiotrophin (PTN,) osteopontin (OPN,) vasoactive intestinal peptide receptor-2 (VIPR-2,) and receptor protein tyrosine phosphatase zeta (PTPζ) [including the two novel isoforms PTPζ SM1 and SM2]. Another group of preferred embodiments are combination aspects which target receptor protein tyrosine phosphatase zeta (PTPζ) [including the two novel isoforms PTPζ SM1 and SM2] and one or more proteins selected from the group angiopoietin related protein 2 (ARP-2,) secreted protein acidic, rich in cysteine (SPARC,) c-met proto-oncogene (C-MET,) brevican (BEHAB,) CD-44 antigen (CD-44,) tetraspanin 3 (TSPN3,) pleiotrophin (PTN,) osteopontin (OPN,) and vasoactive intestinal peptide receptor-2 (VIPR-2.) As pleiotrophin (PTN) is a known ligand of PTPζ, another preferred embodiment of the combination aspects of the invention utilizes these proteins as targets, either alone or in combination with one or more of the other identified targets. [0100]
  • In yet another aspect, the present invention provides two novel splicing isoforms of PTPζ, shown to be expressed in brain tissue. These novel isoforms, PTPζ SM1 and PTPζ SM2, described in more detail below, differ in structure from the three known isoforms heretofore disclosed. PTPζ SM1 comprises the amino acids encoded by the first nine exons of PTPζ-α, with three unique additional carboxy terminal amino acids encoded by additional 3′ mRNA sequence from the intron of the gene between exons nine and ten. The mRNA for PTPζ SM2 comprises all exons of PTPζ-α, with a 116 nucleotide insertion, in the correct reading frame, in the mRNA sequence between [0101] exons 23 and 24, from the from the intron of the gene between exons 23 and 24. Thus, embodiments of this aspect of the invention include the mature proteins of PTPζ splice variants SM1 or SM2, and nucleic acids encoding these novel spice variants, as well proteins with significant homology to the splice variants.
  • Thus, in one group of embodiments of this aspect, the invention provides nucleic acid polymers comprising the sequence of nucleotides 148 to 1272 of SEQ ID NO. 1, the complement of nucleotides 148 to 1272 of SEQ ID NO. 1, nucleotides 148 to 7209 of SEQ ID NO. 3, or the complement of nucleotides 148 to 7209 of SEQ ID NO. 3. In another group of embodiments of this aspect, the invention provides polypeptides comprising the amino acid sequence of SEQ ID NO. 2 or the amino acid sequence of SEQ ID NO. 4. [0102]
  • In an additional related aspect, the invention provides polypeptides comprising a distinctive portion of the amino acid sequence of SEQ ID NO. 2 or SEQ ID NO. 4. Such peptides are useful for the production of antibodies against the PTPζ SM1 or SM2 splicing variants. Preferably, these polypeptides comprise a portion of the amino acid sequence of SEQ ID NO. 2 or SEQ ID NO. 4 which is at least 6, more preferably at least 8, more preferably at least 10, more preferably at least 15, and most preferably at lest 20 amino acids in length. In some preferred embodiments of this aspect of the invention, the polypeptides comprise the three unique terminal amino acids of PTPζ SM1 after [0103] exon 9. In other preferred embodiments, the polypeptides comprise a portion of the unique exon 23a of PTPζ SM2, wherein the portion is preferably at least 3 amino acids in length, more preferably at least 6 amino acids in length, more preferably at least 9 amino acids in length, and most preferably at least 15 amino acids in length.
  • In an additional related aspect, the invention also provides affinity reagents which specifically bind to PTPζ splice variants SM1 or SM2, but do not bind to the other known splice variants of PTPζ (e.g., α, β, or phosphacan forms). In preferred embodiments these affinity reagents are antibodies or antibody fragments. [0104]
  • In an additional related aspect, the invention also provides nucleic acid sequences encoding the PTPζ splice variants SM1 or SM2. The invention also encompasses nucleic acid probes which hybridize to the mRNA encoding PTPζ splice variants SM1 or SM2, but not mRNA encoding other known splice variants of PTPζ.[0105]
  • BRIEF DESCRIPTION OF THE FIGURES
  • FIG. 1: A diagram of the three known splicing variant isoforms of PTPζ. The approximate position of the domains of the isoforms is indicated underneath the isoforms, as well as the approximate exon size (for size reference, [0106] exon 12 is 3.6 kilobases.) Isoform PTPζ-α is the full length isoform, which contains the primary amino acid sequence aa 25-2314 of SEQ ID NO. 2 (aa 1-24 are a signal polypeptide). In Isoform PTPζ-β, aa 755-1614 are missing. Isoform PTPζ-S (phosphacan), is a secreted isoform which comprise the extracellular domains of PTPζ-α, in which the transmembrane and cytosol domains are missing.
  • FIG. 2: A diagram of the two newly discovered splicing variant isoforms of PTPζ. The approximate position of the domains of the isoforms is indicated underneath the isoforms, as well as the approximate exon size (for size reference, [0107] exon 12 is 3.6 kilobases.) SM 1 fails to splice correctly after the 9th exon, yielding an mRNA with tow extra codons followed by a stop codon after the normal terminus of exon 9. SM 2 contains a 116 nucleotide insertion from between exons 23 &24.
  • FIG. 3: A diagram comparing the three known PTPζ isoforms with the two novel isoforms.[0108]
  • DETAILED DESCRIPTION OF THE INVENTION
  • Applicants have identified several brain tumor protein targets and genes which are differentially regulated between brain cancer tissue (glioblastoma) and normal brain tissue. Applicants have performed differential cloning between cancerous and normal brains and have identified the brain tumor protein target genes by DNA sequence analysis. Based on the observation in other diseases, particularly other cancers, in which overexpressed genes can contribute to the pathology of the disease, these overexpressed genes and their protein products mediate the initiation and progression of brain tumors. Thus, the overexpressed brain tumor protein targets, which are presented on the cell surface, provide excellent targets for immunotherapeutic agents which either deliver cytotoxic agents to directly promote tumor cell death, or which alter the function of the brain tumor protein targets to inhibit the normal physiology of the tumor cell. In addition, immunoimaging agents targeted to the brain tumor protein targets may be utilized to visualize the tumor mass either in diagnostic methods (e.g., magnetic resonance imaging (MRI) or radiography), or in surgery (e.g., by the use of optically visual dye moieties in the immunoimaging agent). [0109]
  • Applicants have identified the brain tumor protein targets by a direct examination of the expression level of genes in actual tumor cells. These samples provide a more accurate and realistic picture of tumor cell biology, especially on the detailed transcriptome level, than animal models or established cell tissue culture cell lines. Several groups have found that cell lines established from astrocytomas and other cell lines do not exhibit expression patterns which reflect the actual expression of the original tumor. For instance, Schreiber, et. al., “Primary brain tumors differ in their expression of octamer deoxyribonucleic acid-binding transcription factors from long-term cultured glioma cell lines.” Neurosurgery 34: 129-35 (1994), showed that nervous system-specific transcription factors known as N-Oct proteins are differentially expressed in human neuroblastoma and glioblastoma cell lines in vitro. However, when these results were compared to freshly isolated human primary and metastatic brain tumors, of the five astrocytomas and three glioblastomas analyzed, all but two tumors displayed the complete N-Oct protein profile, irrespective of histopathological tumor grade. Similarly, Eberle, et al., “The expression of angiogenin in tissue samples of different brain tumors and cultured glioma cells.”[0110] Anticancer Res 20: 1679-84 (2000), could show that angiogenin is detectable in different kinds of intracranial tumor tissue samples. Although angiogenin could be detected in primary cultivated glioma cells, it was not detected in the permanent cell lines. Finally, Hartmann, et al., “The rate of homozygous CDKN2A/p16 deletions in glioma cell lines and in primary tumors.”Int J Oncol 15: 975-82 (1999), showed that the rate of homozygous deletions of CDKN2A/p16 is variable between different tumor entities, but the rate of deletions is higher in established cell lines in comparison with primary tumors. Hartmann hypothesized that such incongruity may reflect statistical sampling errors, true differences depending on tissue derivatization and CDKN2A/p16 loss under selective pressure in tissue culture. After comparing established cell lines derived from human glioblastomas and their corresponding primary tumors by multiplex PCR methodology, they found that in 2 of 11 cases (18%) the primary tumor had no p16 alteration whereas the corresponding cell lines had a homozygous p16 deletion, and that CDKN2A/p16 was lost already in the earliest passages of the cell lines. Thus, Hartmann concluded that the deletion was the result of selective cell-culture pressures in many cases.
  • These inconsistent results arise because the tumor tissue samples are obtained from their native milieu, without allowing them the opportunity to alter their gene expression levels in response to artificial environmental stimuli. As recently reported by the Brain Tumor Progress Review group of the National Cancer Institute in November, 2000, conventionally used glioblastoma cell lines contain genetic and gene expression alterations that are well defined and do not necessarily reflect the primary tumors from which they were derived. In addition, these cell lines are highly homogenous, unlike a primary brain tumor. Therefore, data derived soley from a cell line cannot reliably reflect the biology, heterogeneity, or therapeutic response of a primary brain tumor. [0111]
  • Applicants obtained tumor tissue, snap frozen in the operation hall from unknown patients, which was confirmed as glioblastoma grade IV by neuropathology. These tissues served as the experimental sample. Human whole brain tissue (Clontech Laboratories, Palo Alto, USA) served as control sample. Poly-A[0112] + RNA prepared from the cells was converted into double-stranded cDNA (dscDNA).
  • Briefly, the ds-cDNA's from control and disease states were subjected to kinetic re-annealing hybridization during which normalization of transcript abundances and enrichment for differentially expressed transcripts (i.e., subtraction) occurs. Normalized-subtracted ds-cDNAs were cloned into a plasmid vector, a large number of recombinant bacterial clones were picked, and their recombinant inserts were isolated by PCR. High-density cDNA arrays of those PCR products were screened with cDNA probes derived from the original control and disease states. Thus, only clones displaying a significant transcriptional induction and/or repression were sequenced and carried forward for massive expression profiling using a variety of temporal, spatial and disease-related probe sets. [0113]
  • The selected PCR products (fragments of 200-2000 bp in size) from clones showing a significant transcriptional induction and/or repression were sequenced and functionally annotated in AGY's proprietary database structure (See WO01/13105). Because large sequence fragments were utilized in the sequencing step, the data generated has a much higher fidelity and specificity than other approaches, such as SAGE. The resulting sequence information was compared to public databases using the BLAST (blastn) and tblastx algorithm. The results are listed in Table 1, below: [0114]
    TABLE 1
    RELATIVE EXPRESSION NUMBER OF CLONES
    PROTEIN LEVEL ISOLATED (out of 20,000)
    ARP2 ˜2 times 13
    SPARC ˜2-5.6 times 100
    CMET ˜1.2-2.5 times 30
    CD 44 ˜2.3-3.0 times 6
    BEHAB ˜2-6 times 180
    TSPAN3 ˜2.0-3.0 times 7
    VIPR2 ˜3.0 times 3
    OPN ˜2.0-3.0 times 19
    PTN ˜˜1.8-2.6 times 26
    PTPζ ˜2.0-4.0 times 20
  • As one of skill in the art will appreciate from this data, each of these proteins is individually useful as a target for the treatment and/or imaging of brain tumors. [0115]
  • Characteristics of Protein Targets Utilized in the Invention [0116]
  • ARP2 [0117]
  • Given the experiments described above, and the results of Table 1, ARP-2 was selected as a target for selective immuno-therapeutic agents in targeting and/or imaging brain tumors. The mature protein consists of 493 amino acids and contains two potential consensus glycosylation sites. The complete cDNA sequence encoding ARP-2 is provided in SEQ ID NO. 7, and the complete amino acid sequence of ARP-2 is provided in SEQ ID NO. 8. ARP-2 is a 64 kDa, single chain, acidic, angiopoeitin-like protein that includes multiple functional domains, such as a hydrophobic signal sequence from amino acids 1-21 (which is typical of secreted proteins), a coiled-coil domain at the amino terminal end from approximately amino acid sequences 22-274, and a fibrinogen-like domain, from approximately about residues 275 through 493. Two major isoforms have been observed, one 2.4 Kb in size and the other about 4 Kb. Both forms are abundant in heart, small intestine, spleen and stomach. [0118]
  • As used herein, a compound that specifically binds to ARP-2 is any compound (such as an antibody) that has a binding affinity for any naturally occurring isoform, splice variant, or polymorphism of ARP-2, explicitly including the isoforms described herein. As one of ordinary skill in the art will appreciate, such “specific” binding compounds (e.g., antibodies) may also bind to other closely related proteins that exhibit significant homology (such as greater than 90% identity, more preferably greater than 95% identity, and most preferably greater than 99% identity) with the amino acid sequence of ARP-2. Such proteins include truncated forms or domains of ARP-2, and recombinantly engineered alterations of ARP-2. For example, a portion of SEQ ID NO. 8 may be engineered to include a non-naturally occurring cysteine for cross linking to an immunoconjugate protein, as described. [0119]
  • In general, it is preferred that the antibodies utilized in the compositions and methods of the invention bind to the fibrinogen domain but need not be restricted to this domain. The antibody may bind to the extracellular region of ARP-2. It is to be noted that antibodies which bind to this secreted protein are useful in the invention as cytotoxic delivery agents, as well as functional inhibition agents, as one of ordinary skill would expect that the concentration of ARP-2 would be increased adjacent the tumor cells which, due to the need for vascularization, over-express the protein. [0120]
  • When raising antibodies to ARP-2, the entire protein (either the unsecreted precursor or the secreted protein), or a portion thereof, may be utilized. For instance, the carboxyl-terminal fibrinogen like domain, or any portion of the amino-terminal coiled-coil domain may be utilized. For instance, amino acids 22-274, which make up the fibrinogen like domain, may be used. Larger ARP-2 proteins and domains may be produced utilizing any suitable recombinant vector/protein production system, such as the baculovirus transfection system outlined below, after being amplified from a fetal brain cDNA library (as available from, e.g., Clontech, Palo Alto, Calif.) or another suitable source. [0121]
  • When utilizing an entire protein, or a larger section of the protein, antibodies may be raised by immunizing the production animal with the protein and a suitable adjuvant (e.g., Fruend's, Fruend's complete, oil-in-water emulsions, etc.). In these cases, the ARP-2 protein (or a portion thereof) can serve as the ARP-2 antigen. When a smaller peptide is utilized, it is advantageous to conjugate the peptide with a larger molecule to make an immunostimulatory conjugate for use as the ARP-2 antigen. Commonly utilized conjugate proteins that are commercially available for such use include bovine serum albumin (BSA) and keyhole limpet hemocyanin (KLH). In order to raise antibodies to particular epitopes, peptides derived from the full ARP-2 sequence may be utilized. Preferably, one or more 8-30 aa peptide portions of the EC domain of ARP-2 are utilized, with peptides in the range of 10-20 being a more economical choice. Custom-synthesized peptides in this range are available from a multitude of vendors, and can be order conjugated to KLH or BSA. Alternatively, peptides in excess of 30 amino acids may be synthesized by solid-phase methods, or may be recombinantly produced in a suitable recombinant protein production system. In order to ensure proper protein glycosylation and processing, an animal cell system (e.g., Sf9 or other insect cells, CHO or other mammalian cells) is preferred. Other information useful in designing an antigen for the production of antibodies to ARP-2 may be deduced by those of skill in the art by homology analysis of SEQ ID NO. 8. [0122]
  • The fibrinogen domain of human ARP-2 is hypothesized to interact with one or more an unknown receptor for the purposes of angiogenesis. The interaction of ARP-2 to these molecules may be through either of the aforementioned structural motifs. Thus, in alternative embodiments of the compositions and methods of the invention, antibody moieties are utilized which bind to ARP-2 at a site on the protein that alters the binding of an extracellular molecule to ARP-2. Such ARP-2 activity altering antibodies may be utilized in therapeutic compositions in an unconjugated form (e.g., the antibody in an acceptable pharmaceutical carrier), or may be conjugated to either a therapeutic moiety (creating a double-acting therapeutic agent) or an imaging moiety (creating a duel therapeutic/imaging agent). [0123]
  • Selection of antibodies which alter (enhance or inhibit) the binding of a ARP-2 to a receptor may be accomplished by a straightforward binding inhibition/enhancement assay. According to standard techniques, the binding of a labeled (e.g., fluorescently or enzyme-labeled) antibody to ARP-2, which has been immobilized in a microtiter well, is assayed in both the presence and absence of the appropriate ligand. The change in binding is indicative of either an enhancer (increased binding) or competitive inhibitor (decreased binding) relationship between the antibody and the ligand. Such assays may be carried out in high-throughput formats (e.g., 384 well plate formats, in robotic systems) for the automated selection of monoclonal antibody candidates for use as ARP-2 ligand-binding inhibitors or enhancers. [0124]
  • In addition, antibodies which are useful for altering the function of ARP-2 may be assayed in functional formats, such as endothelial sprouting assays and cell migration assays described in the examples. Thus, antibodies that exhibit the appropriate anti-tumor effect may be selected without direct knowledge of a binding ligand. [0125]
  • SPARC [0126]
  • Given the experiments described above, and the results of Table 1, SPARC was selected as a target for selective immuno-therapeutic agents in targeting and/or imaging brain tumors. The mature protein consists of 286 amino acids (after cleavage of the signal peptide) and contains two potential Asn-X-Thr/Ser N-glycosylation sites, located at positions 71 and 99 of the mature protein. The complete cDNA sequence encoding SPARC is provided in SEQ ID NO. 9, and the complete amino acid sequence of SPARC is provided in SEQ ID NO. 10. SPARC is an abundant 33 kDa, single chain, acidic, extracellular calcium binding protein that contains a flexible N-terminal acidic domain I (˜50 amino acids), a follistatin-like (FS) domain (˜75 residues), and a C-terminal extracellular calcium-binding (EC) domain with a pair of EF-hand loops (˜150 residues). The N-terminal domain shows a low affinity Ca2+ binding site, a transglutaminase cross linking site, and inhibits cell spreading in cell culture assays. Calcium-dependent binding of SPARC to the triple helix of several fibrillar collagen types and basement membrane collagen type IV has been mapped to the EC domain. Two isoforms have been described, bone SPARC with a molecular weight of 31,000 kDa and platelet SPARC with a molecular weight of 33,000 kDa. [0127]
  • As used herein, a compound that specifically binds to SPARC is any compound (such as an antibody) that has a binding affinity for any naturally occurring isoform, splice variant, or polymorphism of SPARC, explicitly including the isoforms described herein. As one of ordinary skill in the art will appreciate, such “specific” binding compounds (e.g., antibodies) may also bind to other closely related proteins that exhibit significant homology (such as greater than 90% identity, more preferably greater than 95% identity, and most preferably greater than 99% identity) with the amino acid sequence of SPARC. Such proteins include truncated forms or domains of SPARC, and recombinantly engineered alterations of SPARC. For example, a portion of SEQ ID NO. 10 may be engineered to include a non-naturally occurring cysteine for cross linking to an immunoconjugate protein, as described. [0128]
  • In general, it is preferred that the antibodies utilized in the compositions and methods of the invention bind to the extracellular domain (amino acids 130-280). It is preferable that this binding inhibit the activity of SPARC. The antibody may bind to the EF hand which is known to bind Ca2+ with high affinity, but need not be restricted to this domain. It is to be noted that antibodies which bind to SPARC are useful in both cytotoxic and imaging embodiments of the invention, as one of ordinary skill would expect that the concentration of SPARC in the extracellular matrix would be increased around tumor cells which over-express the protein. [0129]
  • When raising antibodies to SPARC the entire protein (either the unsecreted precursor or the secreted protein), or a portion thereof, may be utilized. For instance, the C terminal extracellular (EC) domain, or any portion of the flexible N-terminal domain I, or FS domain may be utilized. For instance, amino acids 125-275, which make up the EC domain, may be used. Larger SPARC proteins and domains may be produced utilizing any suitable recombinant vector/protein production system, such as the baculovirus transfection system outlined below, after being amplified from a fetal brain cDNA library (as available from, e.g., Clontech, Palo Alto, Calif.) or another suitable source. [0130]
  • When utilizing an entire protein, or a larger section of the protein, antibodies may be raised by immunizing the production animal with the protein and a suitable adjuvant (e.g., Fruend's, Fruend's complete, oil-in-water emulsions, etc.). In these cases, the SPARC protein (or a portion thereof) can serve as the SPARC antigen. When a smaller peptide is utilized, it is advantageous to conjugate the peptide with a larger molecule to make an immunostimulatory conjugate for use as the SPARC antigen. Commonly utilized conjugate proteins that are commercially available for such use include bovine serum albumin (BSA) and keyhole limpet hemocyanin (KLH). In order to raise antibodies to particular epitopes, peptides derived from the full SPARC sequence may be utilized. Preferably, one or more 8-30 aa peptide portions of the EC domain of SPARC are utilized, with peptides in the range of 10-20 being a more economical choice. Custom-synthesized peptides in this range are available from a multitude of vendors, and can be order conjugated to KLH or BSA. Alternatively, peptides in excess of 30 amino acids may be synthesized by solid-phase methods, or may be recombinantly produced in a suitable recombinant protein production system. In order to ensure proper protein glycosylation and processing, an animal cell system (e.g., Sf9 or other insect cells, CHO or other mammalian cells) is preferred. Other information useful in designing an antigen for the production of antibodies to SPARC, including glycosylation sites, is provided in SEQ ID NO. 10. [0131]
  • The EC domain of human SPARC is known to interact with the collagens I, III, IV and V, and to bind to vitronectin, all of which are components of the extracellular matrix surrounding gliomas. The binding of SPARC to these molecules may play a significant role in the oncogenesis and growth of neoplastic cells in the brain. Thus, in alternative embodiments of the compositions and methods of the invention, antibody moieties are utilized which bind to SPARC at a site on the protein that alters the binding of an extracellular molecule, such as an ECM molecule, to SPARC. Such SPARC activity altering antibodies may be utilized in therapeutic compositions in an unconjugated form (e.g., the antibody in an acceptable pharmaceutical carrier), or may be conjugated to either a therapeutic moiety (creating a double-acting therapeutic agent) or an imaging moiety (creating a duel therapeutic/imaging agent). [0132]
  • Selection of antibodies which alter (enhance or inhibit) the binding of a ligand to SPARC may be accomplished by a straightforward binding inhibition/enhancement assay. According to standard techniques, the binding of a labeled (e.g., fluorescently or enzyme-labeled) antibody to SPARC, which has been immobilized in a microtiter well, is assayed in both the presence and absence of the appropriate ligand. The change in binding is indicative of either an enhancer (increased binding) or competitive inhibitor (decreased binding) relationship between the antibody and the ligand. Such assays may be carried out in high-throughput formats (e.g., 384 well plate formats, in robotic systems) for the automated selection of monoclonal antibody candidates for use as SPARC ligand-binding inhibitors or enhancers. [0133]
  • In addition, antibodies which are useful for altering the function of SPARC may be assayed in functional formats, such as the HUVEC tube assay and cell migration assay. Thus, antibodies that exhibit the appropriate anti-tumor effect may be selected without direct knowledge of a binding ligand or molecular function. [0134]
  • c-MET [0135]
  • Given the experiments described above, and the results of Table 1, c-MET was selected as a target for selective immuno-therapeutic agents in targeting and/or imaging brain tumors. The complete cDNA sequence encoding c-MET is provided in SEQ ID NO. 11, and the complete amino acid sequence of c-MET is provided in SEQ ID NO. 12. c-MET is a type I membrane protein heterodimer. Generally, two different receptor variants originate by post-translational processing of a common singe-chain precursor of 170 kDa. Isoform p190MET is formed of a 50 kDa α-chain and a 145 kDa α-chain that are disulfide linked, and isoform p140Met is formed of a 50 kDa α-chain and an 85 kDa β-chain, lacking the cytoplasmic kinase domain. This 85 kDa β chain is likely a trans-membrane glycoprotein that is bound to the cell surface. Truncated forms of c-MET containing the 50 kDa α-chain and a carboxyl-terminally truncated 75 kDa β sub-unit have also been described. The 75 kDa form arises by post-translational proteolytic processing, lacks the trans-membrane domain, and is secreted from the cell. [0136]
  • As used herein, a compound that specifically binds to c-MET is any compound (such as an antibody) that has a binding affinity for any naturally occurring isoform, splice variant, or polymorphism of c-MET, explicitly including the three isoforms described herein. As one of ordinary skill in the art will appreciate, such “specific” binding compounds (e.g., antibodies) may also bind to other closely related proteins that exhibit significant homology (such as greater than 90% identity, more preferably greater than 95% identity, and most preferably greater then 99% identity) with the amino acid sequence of c-MET. Such proteins include truncated forms or domains of c-MET, and recombinantly engineered alterations of c-MET. For example, a portion of SEQ ID NO. 12 may be engineered to include a non-naturally occurring cysteine for cross-linking to an immunoconjugate protein, as described below. [0137]
  • In general, it is preferred that the antibodies utilized in the compositions and methods of the invention bind to the membrane-bound isoforms of the protein, as this will more specifically target the cytotoxic therapeutic agent, or the imaging agent, to the brain tumor cell. However, embodiments that utilize antibodies that bind to the secreted isoform of the protein are also useful in the invention, as one of ordinary skill would expect that the concentration of the secreted isoform would also be increased adjacent to brain tumor cells which over-express the protein. [0138]
  • The amino acid sequence of full length c-MET consists of 1408 amino acids, as the sequence was first deduced by Park et al., (“Sequence of MET proto-oncogene cDNA has features characteristic of the tyrosine kinase family of growth-factor receptors” [0139] Proc. nat. Acad. Sci. U.S.A. 84:6379-6383 (1987)) and 1390 amino acids, as later deduced by Prat et al. (“C-terminal truncated forms of Met, the Hepatocyte Growth Factor” Mol Cell. Biol. 11:5954-5962 (1991)). According to Prat et al., the first N-terminal amino acids 1-24 of SEQ ID NO. B′ [B′] are for the most part hydrophobic, and could serve as a signal sequence for transporting the protein into the lumen of the endoplasmic reticulum. The α chain makes up the extracellular domain of the mature c-MET protein and spans amino acids 24-306 of SEQ ID NO. 12. The β chain would consist of 1,084-5 amino acids with the predicted β chain extracellular domain being amino acids 306 to 932, the single transmembrane hydrophobic segment being amino acids 933 to 955, and the intracellular domain being amino acids 956 to 1390 of SEQ ID NO 12.
  • When raising antibodies to c-MET, the entire protein, a dimeric subunit, or a portion thereof may be utilized. For instance, the extracellular domain of the α or β sub-units or the secreted or extracellular portion of the truncated forms may be utilized. For instance, amino acids that constitute the α sub-unit, amino acids 24-306, may be used. Larger c-MET proteins and domains may be produced utilizing any suitable recombinant vector/protein production system, such as the baculovirus transfection system outlined below, after being amplified from a fetal brain cDNA library (as available from, e.g., Clontech, Palo Alto, Calif.) or another suitable source. [0140]
  • When utilizing an entire protein, or a larger section of the protein, antibodies may be raised by immunizing the production animal with the protein and a suitable adjuvant (e.g., Fruend's, Fruend's complete, oil-in-water emulsions, etc.). In these cases, the c-MET protein (or a portion thereof) can serve as the c-MET antigen. When a smaller peptide is utilized, it is advantageous to conjugate the peptide with a larger molecule to make an immunostimulatory conjugate for use as the c-MET antigen. Commonly utilized conjugate proteins that are commercially available for such use include bovine serum albumin (BSA) and keyhole limpet hemocyanin (KLH). In order to raise antibodies to particular epitopes, peptides derived from the full c-MET sequence may be utilized. Preferably, one or more 8-30 amino acid peptide portions of an extracellular domain of c-MET are utilized, with peptides in the range of 10-20 being a more economical choice. Custom-synthesized peptides in this range are available from a multitude of vendors, and can be order conjugated to KLH or BSA. Alternatively, peptides in excess of 30 amino acids may be synthesized by solid-phase methods, or may be recombinantly produced in a suitable recombinant protein production system. In order to ensure proper protein glycosylation and processing, an animal cell system (e.g., Sf9 or other insect cells, CHO or other mammalian cells) is preferred. Other information useful in designing an antigen for the production of antibodies to c-MET, including glycosylation sites, is provided in SEQ ID NO. 12. [0141]
  • The extracellular domain of human c-MET binds hepatocyte growth factor (HGF). [0142]
  • Because HGF is largely expressed in mesenchymal and neuroectodermal tissues and released to the extracellular compartment, paracrine and/or autocrine signaling implicate tumor genesis in mesenchymal and neuroectodermal tumors and other tumor cells that over express the c-MET receptor. Recent studies have shown that the c-MET proto-oncogene is frequently overexpressed in many types of epithelial tumors, in spontaneously transformed NIH/3T3 fibroblasts, and in peripheral nerve sheath tumors. In alternative embodiments of the compositions and methods of the invention, antibody moieties are utilized which bind to c-MET at a site on the protein which alters the binding of an extracellular ligand molecule, such as HGF, to c-MET. Such c-MET activity altering antibodies may be utilized in therapeutic compositions in an unconjugated form (e.g., the antibody in an acceptable pharmaceutical carrier), or may be conjugated to either a therapeutic moiety (creating a double-acting therapeutic agent) or an imaging moiety (creating a duel therapeutic/imaging agent). [0143]
  • Selection of antibodies that alter (enhance or inhibit) the binding of a ligand to c-MET may be accomplished by a straightforward binding inhibition/enhancement assay. According to standard techniques, the binding of a labeled (e.g., fluorescently or enzyme-labeled) antibody to c-MET, which has been immobilized in a microtiter well, is assayed in both the presence and absence of the ligand. The change in binding is indicative of either an enhancer (increased binding) or competitive inhibitor (decreased binding) relationship between the antibody and the ligand. Such assays may be carried out in high-throughput formats (e.g., 384 well plate formats, in robotic systems) for the automated selection of monoclonal antibody candidates for use as c-MET ligand-binding inhibitors or enhancers. [0144]
  • In addition, antibodies that are useful for altering the function of c-MET may be assayed in functional formats, such as the endothelial sprouting assay and cell migration assay. Thus, antibodies which exhibit the appropriate anti-tumor effect may be selected without direct knowledge of a molecular function. [0145]
  • BEHAB [0146]
  • Given the experiments described above, and the results of Table 1, BEHAB was selected as a target for selective immuno-therapeutic agents in targeting and/or imaging brain tumors. The complete cDNA sequence encoding BEHAB GPI isoform is provided in SEQ ID NO. 13, and the complete amino acid sequence of this BEHAB isoform is provided in SEQ ID NO. 14. Two isoforms have been isolated to date: a full-length isoform that is secreted into the extracellular matrix and a shorter isoform that has a hydrophobic carboxy terminus instead of the typical lectican carboxyl terminus, which predicts a glycophosphatidylinositol (GPI) anchor. BEHAB contains an N-terminal hyaluronan (HA)-binding domain, which comprises an immunoglobulin-like loop and two proteoglycan tandem repeats, a C-terminal epidermal growth factor (EGF)-like repeat, a C-type lectin-like domain, and a complement regulatory protein (CRP)-like domain. The central region of the protein contains sites for glycosylation and proteolytic cleavage (between glu395-Ser396 of the mature protein, after signal peptide cleavage) by metallo-protease. The complete cDNA of the secreted isoform is 2878 bp encoding 912 amino acids of 99 kDa. The GPI isoform, for which sequences SEQ ID NO. 13 and SEQ ID NO. 14 are given, is 2558 bp encoding 672 amino acids of 72 kDa. The GPI-linked form is generated by a ‘no splice’ event, with the transcript reading through an exon/intron junction thereby extending the open reading frame to a stop codon 74 nucleotides further downstream. [0147]
  • As used herein, a compound that specifically binds to BEHAB is any compound (such as an antibody) that has a binding affinity for any naturally occurring isoform, splice variant, or polymorphism of BEHAB, explicitly including the two splice variants described herein. As one of ordinary skill in the art will appreciate, such “specific” binding compounds (e.g., antibodies) may also bind to other closely related proteins that exhibit significant homology (such as greater than 90% identity, more preferably greater than 95% identity, and most preferably greater then 99% identity) with the amino acid sequence of BEHAB. Such proteins include truncated forms or domains of BEHAB, and recombinantly engineered alterations of BEHAB. For example, a portion of SEQ ID NO. 14 may be engineered to include a non-naturally occurring cysteine for cross-linking to an immunoconjugate protein, as described below. [0148]
  • In general, it is preferred that the antibodies utilized in the compositions and methods of the invention bind to the membrane-bound isoform of the protein, as this will more specifically target the cytotoxic therapeutic agent, or the imaging agent, to the brain tumor cell. However, embodiments that utilize antibodies that bind to the secreted isoform of the protein are also useful in the invention, as one of ordinary skill would expect that the concentration of the secreted isoform would also be increased adjacent to brain tumor cells which over-express the protein. [0149]
  • When raising antibodies to BEHAB, the entire protein, or a portion thereof, may be utilized. For instance, any one of the aforementioned domains of the secreted protein or an extracellular portion of the truncated, membrane bound GPI form may be utilized. For instance, amino acids that constitute the hyaluronic acid binding domain, amino acids 44-247, which includes the Ig like domain at amino acids 44-140, may be used. Larger BEHAB proteins and domains may be produced utilizing any suitable recombinant vector/protein production system, such as the baculovirus transfection system outlined below, after being amplified from a fetal brain cDNA library (as available from, e.g., Clontech, Palo alto, CA) or another suitable source. [0150]
  • When utilizing an entire protein, or a larger section of the protein, antibodies may be raised by immunizing the production animal with the protein and a suitable adjuvant (e.g., Fruend's, Fruend's complete, oil-in-water emulsions, etc.). In these cases, the Brevican protein (or a portion thereof) can serve as the BEHAB antigen. When a smaller peptide is utilized, it is advantageous to conjugate the peptide with a larger molecule to make an immunostimulatory conjugate for use as the BEHAB antigen. Commonly utilized conjugate proteins that are commercially available for such use include bovine serum albumin (BSA) and keyhole limpet hemocyanin (KLH). In order to raise antibodies to particular epitopes, peptides derived from the full Brevican sequence may be utilized. Preferably, one or more 8-30 amino acid peptide portions of an extracellular domain of BEHAB are utilized, with peptides in the range of 10-20 being a more economical choice. Custom-synthesized peptides in this range are available from a multitude of vendors, and can be order conjugated to KLH or BSA. Alternatively, peptides in excess of 30 amino acids may be synthesized by solid-phase methods, or may be recombinantly produced in a suitable recombinant protein production system. In order to ensure proper protein glycosylation and processing, an animal cell system (e.g., Sf9 or other insect cells, CHO or other mammalian cells) is preferred. [0151]
  • The hyaluronic acid binding domain of human BEHAB binds to hyaluronic acid (HA). Because HA is largely expressed in the ECM surrounding gliomas and because recent studies have shown that the BEHAB protein is frequently overexpressed in primary brain tumors, it is suggested that the up-regulation of BEHAB may be a crucial step in returning the unmalleable mature extracellular matrix to a more immature matrix, permissive for cell growth, thereby promoting the progression of primary brain tumors. Thus, in alternative embodiments of the compositions and methods of the invention, antibody moieties are utilized which bind to BEHAB at a site on the protein which alters the binding of an extracellular ligand molecule (e.g., HA) to BEHAB. Such BEHAB activity altering antibodies may be utilized in therapeutic compositions in an unconjugated form (e.g., the antibody in an acceptable pharmaceutical carrier), or may be conjugated to either a therapeutic moiety (creating a double-acting therapeutic agent) or an imaging moiety (creating a duel therapeutic/imaging agent). [0152]
  • Selection of antibodies that alter (enhance or inhibit) the binding of a ligand to BEHAB may be accomplished by a straightforward binding inhibition/enhancement assay. According to standard techniques, the binding of a labeled (e.g., fluorescently or enzyme-labeled) antibody to BEHAB, which has been immobilized in a microtiter well, is assayed in both the presence and absence of the ligand. The change in binding is indicative of either an enhancer (increased binding) or competitive inhibitor (decreased binding) relationship between the antibody and the ligand. Such assays may be carried out in high-throughput formats (e.g., 384 well plate formats, in robotic systems) for the automated selection of monoclonal antibody candidates for use as BEHAB ligand-binding inhibitors or enhancers. [0153]
  • In addition, antibodies that are useful for altering the function of BEHAB may be assayed in functional formats, such as the HUVEC tube assay and the cell migration assay described below. Thus, antibodies which exhibit the appropriate anti-tumor effect may be selected without direct knowledge of molecular function. [0154]
  • CD-44 [0155]
  • Given the experiments described above, and the results of Table 1, CD-44 was selected as a target for selective immuno-therapeutic agents in targeting and/or imaging brain tumors. The complete cDNA sequence encoding CD-44E is provided in SEQ ID NO. 15, and the complete amino acid sequence of CD-44, indicating various splicing variation locations, is provided in SEQ ID NO. 16. CD-44 is a proteoglycan that is expressed as two major splice variants. CD-44E is a 150 kDa protein isolated from epithelial cells. CD-44E has a C-terminal cytoplasmic tail, a hydrophobic transmembrane domain of 23 amino acids, and an N-terminal extracellular region of 248 amino acids. The extracellular domain is O-glycosylated and also binds chondroitin sulfate. In addition, CD-44E it has two of the three immunodominant epitope clusters of native gp90Hermes. CD-44E contains an additional 132 amino acids in the extracellular region. and CD-44H is a 90 kDa protein isolated from hematopoietic cells. In addition, CD-44R1 and CD-44R2 are 2 isoforms expressed by hematopoietic cells. The complete cDNA sequence of the 90 kDa CD-44H isoform consist of 1795 bps, encoding a 341 amino acid protein. [0156]
  • As used herein, a compound that specifically binds to CD-44 is any compound (such as an antibody) that has a binding affinity for any naturally occurring isoform, splice variant, or polymorphism of CD-44, explicitly including the isoforms described herein. As one of ordinary skill in the art will appreciate, such “specific” binding compounds (e.g., antibodies) may also bind to other closely related proteins that exhibit significant homology (such as greater than 90% identity, more preferably greater than 95% identity, and most preferably greater then 99% identity) with the amino acid sequence of CD-44. Such proteins include truncated forms or domains of CD-44, and recombinantly engineered alterations of CD-44. For example, a portion of SEQ ID NO. 16 may be engineered to include a non-naturally occurring cysteine for cross-linking to an immunoconjugate protein, as described below. [0157]
  • According to the human full length, CD-44H protein has an overall primary structure of 90 kDa, which consist of 341 amino acids. The N-terminus is located outside of the cell and the extracellular domain consist of 248 amino acids. The C-terminus is located inside of the cell and the intracellular domain consist of 72 amino acids, while the transmembrane region consist of 21 amino acids. The CD-44 gene contains 20 exons, of which exons 1-5, 15-17 and 19 encode the CD44H isoform. The intervening [0158] exons 6, 6a, 7-14 (also designated v1-v10) are alternatively spliced to generate the variant isoforms with an insertion at the membrane proximal region of the extracellular domain between amino acids 202 and 203. See Bajorath (2000). Proteins: structure, function, and genetic, 39:103-111; and Ilangumaram et al. Leukemia and Lymphoma, 35:455-469.
  • When raising antibodies to CD-44, the entire protein, or a portion thereof, may be utilized. For instance, any portion of the extracellular domain may be utilized. For instance, the amino acids between the signal sequence and amino acid 202 may be used. Larger CD-44 proteins and domains may be produced utilizing any suitable recombinant vector/protein production system, such as the baculovirus transfection system outlined below, after being amplified from a fetal brain cDNA library (as available from, e.g., Clontech, Palo Alto, Calif.) or another suitable source. [0159]
  • When utilizing an entire protein, or a larger section of the protein, antibodies may be raised by immunizing the production animal with the protein and a suitable adjuvant (e.g., Fruend's, Fruend's complete, oil-in-water emulsions, etc.). In these cases, the CD-44 protein (or a portion thereof) can serve as the CD-44 antigen. When a smaller peptide is utilized, it is advantageous to conjugate the peptide with a larger molecule to make an immunostimulatory conjugate for use as the CD-44 antigen. Commonly utilized conjugate proteins that are commercially available for such use include bovine serum albumin (BSA) and keyhole limpet hemocyanin (KLH). In order to raise antibodies to particular epitopes, peptides derived from the full CD-44 sequence may be utilized. Preferably, one or more 8-30 amino acid peptide portions of an extracellular domain of CD-44 are utilized, with peptides in the range of 10-20 being a more economical choice. Custom-synthesized peptides in this range are available from a multitude of vendors, and can be order conjugated to KLH or BSA. Alternatively, peptides in excess of 30 amino acids may be synthesized by solid-phase methods, or may be recombinantly produced in a suitable recombinant protein production system. In order to ensure proper protein glycosylation and processing, an animal cell system (e.g., Sf9 or other insect cells, CHO or other mammalian cells) is preferred. Other information useful in designing an antigen for the production of antibodies to CD-44, including glycosylation sites, is provided in SEQ ID NO. D′. [0160]
  • Hyaluronan (HA) is a polymeric glycosaminoglycan and a major component of the extracellular matrix. CD-44 is one of the principal receptors for HA. Within the normal CNS, the CD-44 protein has been localized to astrocytes in the white matter. CD-44H has been shown to be the predominant isoform in normal brain and neuroectoderm-derived tumors. Hence, the up-regulation of CD-44 may be a crucial step in brain tumor invasiveness and migration. Thus, in alternative embodiments of the compositions and methods of the invention, antibody moieties are utilized which bind to CD-44 at a site on the protein which alters the binding of an extracellular ligand molecule (e.g., HA) to CD-44. Such CD-44 activity altering antibodies may be utilized in therapeutic compositions in an unconjugated form (e.g., the antibody in an acceptable pharmaceutical carrier), or may be conjugated to either a therapeutic moiety (creating a double-acting therapeutic agent) or an imaging moiety (creating a duel therapeutic/imaging agent). [0161]
  • Selection of antibodies that alter (enhance or inhibit) the binding of a ligand to CD-44 may be accomplished by a straightforward binding inhibition/enhancement assay. According to standard techniques, the binding of a labeled (e.g., fluorescently or enzyme-labeled) antibody to CD-44, which has been immobilized in a microtiter well, is assayed in both the presence and absence of the ligand. The change in binding is indicative of either an enhancer (increased binding) or competitive inhibitor (decreased binding) relationship between the antibody and the ligand. Such assays may be carried out in high-throughput formats (e.g., 384 well plate formats, in robotic systems) for the automated selection of monoclonal antibody candidates for use as CD-44 ligand-binding inhibitors or enhancers. [0162]
  • In addition, antibodies that are useful for altering the function of CD-44 may be assayed in functional formats, such as endothelial sprouting assay and cell migration assay. Thus, antibodies which exhibit the appropriate anti-tumor effect may be selected without direct knowledge of molecular function. [0163]
  • TSPAN3 [0164]
  • Given the experiments described above, and the results of Table 1, TSPAN3 was selected as a target for selective immuno-therapeutic agents in targeting and/or imaging brain tumors. The complete cDNA sequence encoding TSPAN3 is provided in SEQ ID NO. 17, and the complete amino acid sequence of TSPAN3 is provided in SEQ ID NO. 18. Tetraspanin is a 253 amino acid membrane bound protein. No isoforms have been isolated to date. TSPAN3, as is characteristic of the tetraspanin family, contains four transmembrane domains, putatively comprising amino acids 12-32, 51-71, 86-106, and 213-233. The protein has two putative extracellular domains, amino acids 33-50 and 107-212, and three putative cytoplasmic domains, amino acids 1-11, 72-85, and 234-235. Putative N-linked glycosylation sites are listed in SEQ ID NO. 18. [0165]
  • As used herein, a compound that specifically binds to TSPAN3 is any compound (such as an antibody) that has a binding affinity for any naturally occurring isoform, splice variant, or polymorphism of TSPAN3. As one of ordinary skill in the art will appreciate, such “specific” binding compounds (e.g., antibodies) may also bind to other closely related proteins that exhibit significant homology (such as greater than 90% identity, more preferably greater than 95% identity, and most preferably greater then 99% identity) with the amino acid sequence of TSPAN3. Such proteins include truncated forms or domains of TSPAN3, and recombinantly engineered alterations of TSPAN3. For example, a portion of SEQ ID NO. 18 may be engineered to include a non-naturally occurring cysteine for cross-linking to an immunoconjugate protein, as described below. [0166]
  • In general, it is preferred that the antibodies utilized in the compositions and methods of the invention bind to the membrane-bound isoform of the protein, as this will more specifically target the cytotoxic therapeutic agent, or the imaging agent, to the brain tumor cell. The only currently known form of TSPAN3 is membrane-bound. However, embodiments that utilize antibodies that bind to any secreted isoform of the protein are also useful in the invention, as one of ordinary skill would expect that the concentration of the secreted isoform would also be increased adjacent to brain tumor cells which over-express the protein. Likewise, it is preferred that the antibodies utilized in the invention bind to an extracellular domain of the protein, as are described in the SEQ ID NO. 18. The cysteine residues at positions 147, 148, and 197 of SEQ ID NO. 18 in the second extracellular domain are highly conserved among the tetraspanin family and are thought to be essential for proper tetraspanin function. Thus, in some preferred embodiments of the invention, the antibodies utilized in the invention bind to an epitope comprising, or alternatively very near to, one of these cysteine residues. [0167]
  • When raising antibodies to TSPAN3, the entire protein, or a portion thereof, may be utilized. For instance, any one of the aforementioned domains of the secreted protein or an extracellular portion of the truncated, membrane bound GPI form may be utilized. For instance, amino acids that constitute one of the extracellular domains, amino acids 33-50 or 107-212, may be used. Larger TSPAN3 proteins and domains may be produced utilizing any suitable recombinant vector/protein production system, such as the baculovirus transfection system outlined below, after being amplified from a fetal brain cDNA library (as available from, e.g., Clontech, Palo Alto, Calif.) or another suitable source. [0168]
  • When utilizing an entire protein, or a larger section of the protein, antibodies may be raised by immunizing the production animal with the protein and a suitable adjuvant (e.g., Fruend's, Fruend's complete, oil-in-water emulsions, etc.). In these cases, the [0169] tetraspanin 3 protein (or a portion thereof) can serve as the TSPAN3 antigen. When a smaller peptide is utilized, it is advantageous to conjugate the peptide with a larger molecule to make an immunostimulatory conjugate for use as the TSPAN3 antigen. Commonly utilized conjugate proteins that are commercially available for such use include bovine serum albumin (BSA) and keyhole limpet hemocyanin (KLH). In order to raise antibodies to particular epitopes, peptides derived from the full Brevican sequence may be utilized. Preferably, one or more 8-30 amino acid peptide portions of an extracellular domain of TSPAN3 are utilized, with peptides in the range of 10-20 being a more economical choice. Custom-synthesized peptides in this range are available from a multitude of vendors, and can be order conjugated to KLH or BSA. Alternatively, peptides in excess of 30 amino acids may be synthesized by solid-phase methods, or may be recombinantly produced in a suitable recombinant protein production system. In order to ensure proper protein glycosylation and processing, an animal cell system (e.g., Sf9 or other insect cells, CHO or other mammalian cells) is preferred. Other information useful in designing an antigen for the production of antibodies to TSPAN3, including glycosylation sites, is provided in SEQ ID NO. 18.
  • In alternative embodiments of the compositions and methods of the invention, antibody moieties are utilized which bind to TSPAN3 at a site on the protein which alters the binding of an extracellular ligand molecule to TSPAN3. Such TSPAN3 activity altering antibodies may be utilized in therapeutic compositions in an unconjugated form (e.g., the antibody in an acceptable pharmaceutical carrier), or may be conjugated to either a therapeutic moiety (creating a double-acting therapeutic agent) or an imaging moiety (creating a duel therapeutic/imaging agent). [0170]
  • Selection of antibodies that alter (enhance or inhibit) the binding of a ligand to TSPAN3 may be accomplished by a straightforward binding inhibition/enhancement assay. According to standard techniques, the binding of a labeled (e.g., fluorescently or enzyme-labeled) antibody to TSPAN3, which has been immobilized in a microtiter well, is assayed in both the presence and absence of the ligand. The change in binding is indicative of either an enhancer (increased binding) or competitive inhibitor (decreased binding) relationship between the antibody and the ligand. Such assays may be carried out in high-throughput formats (e.g., 384 well plate formats, in robotic systems) for the automated selection of monoclonal antibody candidates for use as TSPAN3 ligand-binding inhibitors or enhancers. [0171]
  • In addition, antibodies that are useful for altering the function of TSPAN3 may be assayed in functional formats, such as the HUVEC tube assay and the cell migration assay described below. Thus, antibodies which exhibit the appropriate anti-tumor effect may be selected without direct knowledge of molecular function. [0172]
  • VIPR-2 [0173]
  • Given the experiments described above, and the results of Table 1, VIPR-2 was selected as a prime target for selective immuno-therapeutic agents in targeting and/or imaging brain tumors. The complete cDNA sequence encoding VIPR-2 is provided in SEQ ID NO. 19, and the complete amino acid sequence of VIPR-2 is provided in SEQ ID NO. 20. VIPR-2 is a seven transmembrane spanning G-protein receptor. The complete VIPR-2 protein is encoded by 13 exons. The initiator codon of the approximated 438 amino acid-encoding open reading frame is located in exon 1 and the termination signal is located in exon 13. The 5′ untranslated region extends 187 bp upstream of the initiator codon and is extremely GC-rich (80%). The polyadenylation signal is located 2416 bp downstream of the stop codon. Intron sizes range from 68 bp (intron 11) to 45 bp (intron 4), the entire human gene spans 117 kb, while the cDNA sequence spans 1317 bp. Recent studies have also isolated two VIP-2 receptor mRNAs of 4.6 kb and 2.3 kb in size. [0174]
  • As used herein, a compound that specifically binds to VIPR-2 is any compound (such as an antibody) that has a binding affinity for any naturally occurring isoform, splice variant, or polymorphism of VIPR-2, explicitly including any isoforms described herein. As one of ordinary skill in the art will appreciate, such “specific” binding compounds (e.g., antibodies) may also bind to other closely related proteins that exhibit significant homology (such as greater than 90% identity, more preferably greater than 95% identity, and most preferably greater then 99% identity) with the amino acid sequence of VIPR-2. Such proteins include truncated forms or domains of VIPR-2, and recombinantly engineered alterations of VIPR-2. For example, a portion of SEQ ID NO. 20 may be engineered to include a non-naturally occurring cysteine for cross-linking to an immunoconjugate protein, as described below. [0175]
  • In general, it is preferred that the antibodies utilized in the compositions and methods of the invention bind to the membrane-bound isoforms of the protein, as this will more specifically target the cytotoxic therapeutic agent, or the imaging agent, to the brain tumor cell. However, embodiments that utilize antibodies that bind to the secreted isoform of the protein are also useful in the invention, as one of ordinary skill would expect that the concentration of the secreted isoform would also be increased adjacent to brain tumor cells which over-express the protein. [0176]
  • The amino acid sequence of full length VIPR-2 consists of 437 amino acids with a predicted molecular mass is 49 kDa, as the sequence was first deduced by Lutz et al. [0177] FEBS. 334:3-8, 1993. Lutz et al. predicted that the receptor is a seven membrane spanning protein where in the first 22 amino acids constitute a typical hydrophobic signal sequence, and the remaining amino acids constitute two membrane spanning regions between amino acids 127 to 148 and 158 to 178, two more membrane spanning domains between amino acids 202 to 227 and 238 to 261, another between 278 to 303, and two final membrane spanning regions between 327 to 347 and 359 to 380, with three potential N-linked glycosylation sites found in the amino terminal extracellular domain at residues 57, 87 and 91. Sreedharan et al. describes the VIPR-2 receptor as being a 457 amino-acid protein encoded by a 2.8 kb cDNA of 52 kDa. Sreedharan et al. Biochem. Biophys. Res. Commun. 203:141-148, 1994.
  • When raising antibodies to VIPR-2, the entire protein or a portion thereof may be utilized. For instance, the extracellular domains of any of the seven transmembrane spanning portions of the protein may be utilized. For instance, amino acids 179 to 201 may be used. Larger VIPR-2 proteins and domains may be produced utilizing any suitable recombinant vector/protein production system, such as the baculovirus transfection system outlined below, after being amplified from a fetal brain cDNA library (as available from, e.g., Clontech, Palo Alto, Calif.) or another suitable source. [0178]
  • When utilizing an entire protein, or a larger section of the protein, antibodies may be raised by immunizing the production animal with the protein and a suitable adjuvant (e.g., Fruend's, Fruend's complete, oil-in-water emulsions, etc.). In these cases, the VIPR-2 protein (or a portion thereof) can serve as the VIPR-2 antigen. When a smaller peptide is utilized, it is advantageous to conjugate the peptide with a larger molecule to make an immunostimulatory conjugate for use as the VIPR-2 antigen. Commonly utilized conjugate proteins that are commercially available for such use include bovine serum albumin (BSA) and keyhole limpet hemocyanin (KLH). In order to raise antibodies to particular epitopes, peptides derived from the full VIPR-2 sequence may be utilized. Preferably, one or more 8-30 amino acid peptide portions of an extracellular domain of VIPR-2 are utilized, with peptides in the range of 10-20 being a more economical choice. Custom-synthesized peptides in this range are available from a multitude of vendors, and can be order conjugated to KLH or BSA. Alternatively, peptides in excess of 30 amino acids may be synthesized by solid-phase methods, or may be recombinantly produced in a suitable recombinant protein production system. In order to ensure proper protein glycosylation and processing, an animal cell system (e.g., Sf9 or other insect cells, CHO or other mammalian cells) is preferred. Other information useful in designing an antigen for the production of antibodies to VIPR-2, including glycosylation sites, is provided in SEQ ID NO. 20. [0179]
  • The extracellular domain of human VIPR-2 binds PACAP-27, PACAP-38, VIP and secretin. Because these factors have been found to affect tumor cell growth, and due to the recent discovery that the VIPR-2 receptor is overexpressed in glioblastomas (Astrocytoma grade IV), the binding of these factors to the VIPR-2 receptor may play a significant role in the oncogenesis and growth of astrocytoma cells in the brain. Thus, in alternative embodiments of the of the invention, antibody moieties are utilized which bind to VIPR-2 at a site on the protein which alters the binding of extracellular ligand molecules, such as VIP, to VIPR-2. Such VIPR-2 activity altering antibodies may be utilized in therapeutic compositions in an unconjugated form (e.g., the antibody in an acceptable pharmaceutical carrier), or may be conjugated to either a therapeutic moiety (creating a double-acting therapeutic agent) or an imaging moiety (creating a duel therapeutic/imaging agent). [0180]
  • Selection of antibodies that alter (enhance or inhibit) the binding of a ligand to VIPR-2 may be accomplished by a straightforward binding inhibition/enhancement assay. According to standard techniques, the binding of a labeled (e.g., fluorescently or enzyme-labeled) antibody to VIPR-2, which has been immobilized in a microtiter well, is assayed in both the presence and absence of the ligand (e.g., vasoactive intestinal peptide.) The change in binding is indicative of either an enhancer (increased binding) or competitive inhibitor (decreased binding) relationship between the antibody and the ligand. Such assays may be carried out in high-throughput formats (e.g., 384 well plate formats, in robotic systems) for the automated selection of monoclonal antibody candidates for use as VIPR-2 ligand-binding inhibitors or enhancers. [0181]
  • In addition, antibodies that are useful for altering the function of VIPR-2 may be assayed in functional formats, such as the HUVEC tube assay and cell migration assay. Thus, antibodies which exhibit the appropriate anti-tumor effect may be selected without direct knowledge of a binding ligand. [0182]
  • PTN [0183]
  • Given the experiments described above, and the results of Table 1, PTN was selected as a target for selective immuno-therapeutic agents in targeting and/or imaging brain tumors. The entire PTN gene spans 65 kb and 7 exons, while the mature protein is approximately 136 amino acids (after cleavage of a 32 amino acid signal peptide) with distinctive lysine and arginine-rich clusters within both N- and C-terminal domains. The complete cDNA sequence encoding PTN is provided in SEQ ID NO. 21, and the complete amino acid sequence of PTN is provided in SEQ ID NO. 22. PTN is a 18 kDa, single chain, secreted protein with 10 conserved disulfide linked cysteine residues. [0184]
  • As used herein, a compound that specifically binds to PTN is any compound (such as an antibody) that has a binding affinity for any naturally occurring isoform, splice variant, or polymorphism of PTN, explicitly including the isoforms described herein. As one of ordinary skill in the art will appreciate, such “specific” binding compounds (e.g., antibodies) may also bind to other closely related proteins that exhibit significant homology (such as greater than 90% identity, more preferably greater than 95% identity, and most preferably greater than 99% identity) with the amino acid sequence of PTN. Such proteins include truncated forms or domains of PTN, and recombinantly engineered alterations of PTN. For example, a portion of SEQ ID NO. 22 may be engineered to include a non-naturally occurring cysteine for cross linking to an immunoconjugate protein, as described. [0185]
  • According to Milner et al. the gene sequence of PTN isolated from human genomic DNA consists of five exons and four introns. While exon 1 does not encode an amino acid sequence, exon 2 encodes the hydrophobic signal sequence of 32 amino acids, [0186] exons 3 and 4 code for the amino terminal and the ten cysteine residues, and exon 5 codes for the highly basic C-terminal domains. Interestingly, the human cDNA starts toward the end of exon 1, while the coded for protein begins at exon 2. Thus, the mature protein consist of 136 amino acids encoded by exons 2 to 5. As reported by Kretschmer et al. the minimum size of the gene is 42 kb, with a mRNA of 1650 nucleotides, spanning five exons, the majority of the protein being coded for by exon 3 (174 base pairs in length) and exon 4 (162 base pairs in length). See Kretschmer et al. (1993). Biochem. Biophys. Res. Commun. 192:420-429.
  • When raising antibodies to PTN, the entire protein or a portion thereof may be utilized. For instance, amino acid domains encoded for by [0187] exons 3 and 4 (i.e. amino acids 7 to 64 or 65 to 118, respectfully). Specifically, residues 41 to 64 may be used to abolish the transformation potential of PTN. Larger PTN proteins and domains may be produced utilizing any suitable recombinant vector/protein production system, such as the baculovirus transfection system outlined below, after being amplified from a fetal brain cDNA library (as available from, e.g., Clontech, Palo Alto, Calif.) or another suitable source. It is to be noted that antibodies which bind to this secreted protein are useful in cytotoxic and imaging embodiments of the invention, as one of ordinary skill would expect that the concentration of the PTN would be increased adjacent to tumor cells which over-express the protein.
  • When utilizing an entire protein, or a larger section of the protein, antibodies may be raised by immunizing the production animal with the protein and a suitable adjuvant (e.g., Fruend's, Fruend's complete, oil-in-water emulsions, etc.). In these cases, the PTN protein (or a portion thereof) can serve as the PTN antigen. When a smaller peptide is utilized, it is advantageous to conjugate the peptide with a larger molecule to make an immunostimulatory conjugate for use as the PTN antigen. Commonly utilized conjugate proteins that are commercially available for such use include bovine serum albumin (BSA) and keyhole limpet hemocyanin (KLH). In order to raise antibodies to particular epitopes, peptides derived from the full PTN sequence may be utilized. Preferably, one or more 8-30 aa peptide portions of the protein are utilized, with peptides in the range of 10-20 being a more economical choice. Custom-synthesized peptides in this range are available from a multitude of vendors, and can be order conjugated to KLH or BSA. Alternatively, peptides in excess of 30 amino acids may be synthesized by solid-phase methods, or may be recombinantly produced in a suitable recombinant protein production system. In order to ensure proper protein glycosylation and processing, an animal cell system (e.g., Sf9 or other insect cells, CHO or other mammalian cells) is preferred. Other information useful in designing an antigen for the production of antibodies to PTN, including glycosylation sites, is provided in SEQ ID NO. 22. [0188]
  • PTN has been shown to bind to extracellular domain of RPTP beta and zeta. This binding inactivates the catalytic activity of RPTP, and PTN binds all the three major isoforms pf RPTP beta and zeta. PTN has also been shown to interact with syndecan-3. Thus, in alternative embodiments of the compositions and methods of the invention, antibody moieties are utilized which bind to PTN at a site on the protein that alters the binding of a cell surface molecule, such as the ones listed above, to PTN. Such PTN activity altering antibodies may be utilized in therapeutic compositions in an unconjugated form (e.g., the antibody in an acceptable pharmaceutical carrier), or may be conjugated to either a therapeutic moiety (creating a double-acting therapeutic agent) or an imaging moiety (creating a duel therapeutic/imaging agent). [0189]
  • Selection of antibodies which alter (enhance or inhibit) the binding of a ligand to PTN may be accomplished by a straightforward binding inhibition/enhancement assay. According to standard techniques, the binding of a labeled (e.g., fluorescently or enzyme-labeled) antibody to PTN, which has been immobilized in a microtiter well, is assayed in both the presence and absence of the appropriate ligand. The change in binding is indicative of either an enhancer (increased binding) or competitive inhibitor (decreased binding) relationship between the antibody and the ligand. Such assays may be carried out in high-throughput formats (e.g., 384 well plate formats, in robotic systems) for the automated selection of monoclonal antibody candidates for use as PTN ligand-binding inhibitors or enhancers. [0190]
  • In addition, antibodies which are useful for altering the function of PTN may be assayed in functional formats, such as the HUVEC tube assay and the cell migration assay described below. Thus, antibodies that exhibit the appropriate anti-PTN activity may be selected without direct knowledge of a binding ligand or the particular biomolecular interactions of PTN. [0191]
  • OPN [0192]
  • Given the experiments described above, and the results of Table 1, OPN was selected as a target for selective immuno-therapeutic agents in targeting and/or imaging brain tumors. The mature protein consists of approximately 298 amino acids (after cleavage of a 16 amino acid signal peptide) and contains two potential Asn-Xaa-Ser N-glycosylation site, located at positions 65 and 92 of the mature protein. The complete cDNA sequence encoding OPN is provided in SEQ ID NO. 23, and the complete amino acid sequence of OPN is provided in SEQ ID NO. 24. OPN is an abundant 34 kDa, single chain, phosphorylated glycoprotein, with a presumed site for cell attachment at residues 144-148. Three isoforms have been identified to be generated by post transcriptional modification, such as alternative splicing, OPN-A, OPN-B, and OPN-C. OPN-A and OPN-B differ by the addition of 14 amino acids at residue 58 of the protein. Amino acids 58-71 are absent in OPN-B, and amino acids 31-57 are absent in OPN-C. OPN is a negatively charged, highly hydrophilic secreted protein. [0193]
  • As used herein, a compound that specifically binds to OPN is any compound (such as an antibody) that has a binding affinity for any naturally occurring isoform, splice variant, or polymorphism of OPN, explicitly including the three isoforms described herein. As one of ordinary skill in the art will appreciate, such “specific” binding compounds (e.g., antibodies) may also bind to other closely related proteins that exhibit significant homology (such as greater than 90% identity, more preferably greater than 95% identity, and most preferably greater than 99% identity) with the amino acid sequence of OPN. Such proteins include truncated forms or domains of OPN, and recombinantly engineered alterations of OPN. For example, a portion of SEQ ID NO. 24 may be engineered to include a non-naturally occurring cysteine for cross linking to an immunoconjugate protein, as described. [0194]
  • According to Young et al. the cDNA sequence of OPN isolated from human bone cells (OPN-A) has an overall structure of approximately 34 kDA that consist of 298 amino acids, which is 14 amino acids less than the cDNA sequence of OPN isolated from human osteosarcoma by Keifer et al. (OPN-B). The cDNA transcript for OPN-A is 1.5 kb with an open reading frame of 900 nucleotides, of which the first 16 amino acids are hydrophobic in nature and probably constitute a signal sequence for the secreted protein. The OPN gene contains 7 exons that are alternatively spliced to generate the variant isoforms, the most common variant being the addition of a 42 bp (14 amino acid) sequence located at base 280 of OPN-A. See Young et al. (1990). [0195] Genomics, 7:491-502 and Keifer et al. Nucleic Acids Res. 17:3306.
  • When raising antibodies to OPN, the entire protein or a portion thereof may be utilized. For instance, [0196] amino acid domains 4 to 12 (from the N-terminus) or 29 to 37 (from the N-terminus) may be utilized. Larger OPN proteins and domains may be produced utilizing any suitable recombinant vector/protein production system, such as the baculovirus transfection system outlined below, after being amplified from a fetal brain cDNA library (as available from, e.g., Clontech, Palo Alto, Calif.) or another suitable source. It is to be noted that antibodies which bind to this secreted protein are useful in cytotoxic and imaging embodiments of the invention, as one of ordinary skill would expect that the concentration of OPN would be increased adjacent to tumor cells which over-express the protein.
  • When utilizing an entire protein, or a larger section of the protein, antibodies may be raised by immunizing the production animal with the protein and a suitable adjuvant (e.g., Fruend's, Fruend's complete, oil-in-water emulsions, etc.). In these cases, the OPN protein (or a portion thereof) can serve as the OPN antigen. When a smaller peptide is utilized, it is advantageous to conjugate the peptide with a larger molecule to make an immunostimulatory conjugate for use as the OPN antigen. Commonly utilized conjugate proteins that are commercially available for such use include bovine serum albumin (BSA) and keyhole limpet hemocyanin (KLH). In order to raise antibodies to particular epitopes, peptides derived from the full OPN sequence may be utilized. Preferably, one or more 8-30 aa peptide portions of the protein are utilized, with peptides in the range of 10-20 being a more economical choice. Custom-synthesized peptides in this range are available from a multitude of vendors, and can be order conjugated to KLH or BSA. Alternatively, peptides in excess of 30 amino acids may be synthesized by solid-phase methods, or may be recombinantly produced in a suitable recombinant protein production system. In order to ensure proper protein glycosylation and processing, an animal cell system (e.g., Sf9 or other insect cells, CHO or other mammalian cells) is preferred. Other information useful in designing an antigen for the production of antibodies to OPN, including glycosylation sites, is provided in SEQ ID NO. 22. [0197]
  • The cell attachment sequence of human OPN (amino acids 144 to 148) is believed to interact with various cell surface proteins (such as CD-44) to affect cell adhesion, and a highly acidic stretch composed almost exclusively of aspartic acid residues (amino acids 72 to 81) is believed to be the mineral binding site within the protein. Because CD-44 is frequently over expressed on primary brain tumors and metastases the binding of OPN to these various cell-surface adhesion protein molecules may play a significant role in the senescence and growth of tumor cells in the brain. Thus, in alternative embodiments of the compositions and methods of the invention, antibody moieties are utilized which bind to OPN at a site on the protein that alters the binding of a cell surface molecule, e.g., CD-44, to OPN. Such OPN activity altering antibodies may be utilized in therapeutic compositions in an unconjugated form (e.g., the antibody in an acceptable pharmaceutical carrier), or may be conjugated to either a therapeutic moiety (creating a double-acting therapeutic agent) or an imaging moiety (creating a duel therapeutic/imaging agent). [0198]
  • Selection of antibodies which alter (enhance or inhibit) the binding of a ligand to OPN may be accomplished by a straightforward binding inhibition/enhancement assay. According to standard techniques, the binding of a labeled (e.g., fluorescently or enzyme-labeled) antibody to OPN, which has been immobilized in a microtiter well, is assayed in both the presence and absence of the appropriate ligand. The change in binding is indicative of either an enhancer (increased binding) or competitive inhibitor (decreased binding) relationship between the antibody and the ligand. Such assays may be carried out in high-throughput formats (e.g., 384 well plate formats, in robotic systems) for the automated selection of monoclonal antibody candidates for use as OPN ligand-binding inhibitors or enhancers. [0199]
  • In addition, antibodies which are useful for altering the function of OPN may be assayed in functional formats, such as the HUVEC tube assay and the cell migration assay described below. Thus, antibodies that exhibit the appropriate anti-OPN activity may be selected without direct knowledge of a the biomolecular role of OPN. [0200]
  • PTPζ[0201]
  • PTPζ was also selected as a prime target for selective immuno-therapeutic agents in treating or imaging brain tumors. The complete cDNA sequence encoding PTPζ is provided in SEQ ID NO. 5, and the complete amino acid sequence of PTPζ is provided in SEQ ID NO. 6. Three different splice variants have been described, which include two membrane bound variants (full length: PTPζ-α, and shorter version PTPζ-β) and one secreted form (Phosphacan). See FIG. 1. Isoform PTPζ-α is the full length isoform, which contains the primary amino acid sequence aa 25-2314 of SEQ ID NO. 6 (aa 1-24 are a signal polypeptide). This full length long form of PTPζ is a type I membrane protein. After the signal peptide it contains a carbonic anhydrase like (CAH) and a fibronectin type III like (FN3) domain, followed by a long cysteine free spacer (S) domain. This follows a 860 amino acid long insert domain, which can be glycosylated. After a single transmembrane segment, in the intracellular region it has 2 phosphatase domains, but only the membrane-proximal PTPase domain is catalytically active (Krueger 1992). [0202]
  • In Isoform PTPζ-β, 755-1614 are missing. Isoform PTPζ-S (phosphacan), is a secreted isoform, which is comprises the extracellular domains of PTPζ-α. Northern Blot analysis have shown that the PTP zeta is exclusively expressed in the human central nervous system. In mouse embryos, the PTPζ transcript was mainly detected in the ventricular and subventricular zone of the brain and the spinal cord. The same pattern was detected in adult mice. Detailed studies have shown that during rat embryogenesis the two transmembrane splice variants of PTPζ are mainly expressed in glial precursor cells and that the secretory version (Phosphacan) is more abundant in mature astrocytes which have already migrated in the ventricle zone. Applicants have characterized two additional novel slice variants, PTPζ SM1 and PTPζ SM2, which are described in detail below. [0203]
  • As used herein, a compound which specifically binds to human protein tyrosine phosphatase-zeta (PTPζ) is any compound (such as an antibody) which has a binding affinity for any naturally occurring isoform, spice variant, or polymorphism of PTPζ, explicitly including the three splice variants describe herein. For example, the compounds which specifically bind to novel isoforms PTPζ SM1 and PTPζ SM2, described below, are subsets of compounds which specifically bind to PTPζ. As one of ordinary skill in the art will appreciate, such “specific” binding compounds (e.g., antibodies) may also bind to other closely related proteins which exhibit significant homology (such as greater than 90% identity, more preferably greater than 95% identity, and most preferably greater than 99% identity) with the amino acid sequence of PTPζ. Such proteins include truncated forms or domains of PTPζ, and recombinantly engineered alterations of PTPζ. For example, an portion of SEQ ID NO. 6 may be engineered to include a non-naturally occurring cysteine for cross-linking to an immunoconjugate protein, as described below. [0204]
  • In general, it is preferred that the antibodies utilized in the compositions and methods of the invention bind to the membrane-bound isoforms of the protein, as this will more specifically target the cytotoxic therapeutic agent, or the imaging agent, to the brain tumor cell. However, embodiments which utilize antibodies which bind to the secreted isoform of the protein are also useful in the invention, as one of ordinary skill would expect that the concentration of the secreted isoform would also be increased adjacent to brain tumor cells which over-express the protein. [0205]
  • The amino acid sequence of full length PTPζ consists of 2307 amino acids, as the sequence was deduced by Levy (in which aa 1722-1728 of SEQ ID NO. 2 were missing) (See also U.S. Pat. Nos. 5,604,094, and 6,160,090, fully incorporated herein by reference), or 2314 amino acids as the sequence was deduced by Krueger, et al., (“A human transmembrane protein-tyrosine phosphatase, PTP zeta, is expressed in brain and has an N-terminal receptor domain homologous to carbonic anhydrases” [0206] Proc. Nat. Acad. Sci. U.S.A. 89:7417-7421 (1992)). Amino acids 1-24 of SEQ ID NO. 6 are a signal sequence which directs the proper placement of the transmembrane protein. The extracellular domain of the mature PTPζ protein spans amino acids 25-1635 of SEQ ID NO. 6 in the long and secreted forms (this forms the entire secreted form), and amino acids 25-754,1615-1635 in the short isoform. The transmembrane region of the protein spans amino acids 1636-1661 of SEQ ID NO. 6, and the balance of the protein forms the cytoplasmic domain, amino acids 1662-2314.
  • When raising antibodies to PTPζ, the entire protein (any of the three isoforms) or a portion thereof may be utilized. For instance, the extracellular domain of the long or short form, the entire secreted form, or a portion of extracellular domain may be utilized. For instance, amino acids 25-754, which are common to both α and β isoforms, may be used. Such larger PTPζ proteins and domains may be produced utilizing any suitable recombinant vector/protein production system, such as the baculovirus transfection system outlined below, after being amplified from a fetal brain cDNA library (as available from, e.g., Clontech, Palo Alto, Calif.) or another suitable source. When utilizing an entire protein, or a larger section of the protein, antibodies may be raised by immunizing the production animal with the protein and a suitable adjuvant (e.g., Fruend's, Fruend's complete, oil-in-water emulsions, etc.). In these cases, the PTPζ protein (or a portion thereof) can serve as the PTPζ antigen. When a smaller peptide is utilized, it is advantageous to conjugate the peptide with a larger molecule to make an immunostimulatory conjugate for use as the PTPζ antigen. Commonly utilized conjugate proteins which are commercially available for such use include bovine serum albumin (BSA) and keyhole limpet hemocyanin (KLH). In order to raise antibodies to particular epitopes, peptides derived from the full PTPζ sequence may be utilized. Preferably, one or more 8-30 aa peptide portions of an extracellular domain of PTPζ are utilized, with peptides in the range of 10-20 being a more economical choice. Custom-synthesized peptides in this range are available from a multitude of vendors, and can be order conjugated to KLH or BSA. Alternatively, peptides in excess of 30 amino acids may be synthesized by solid-phase methods, or may be recombinantly produced in a suitable recombinant protein production system. In order to ensure proper protein glycosylation and processing, an animal cell system (e.g., Sf9 or other insect cells, CHO or other mammalian cells) is preferred. Other information useful in designing an antigen for the production of antibodies to PTPζ, including glycosylation sites, is provided in SEQ ID NO. 6. [0207]
  • The extracellular domain of human PTPζ is known to bind to tenascin-C, tenascin-R, pleiotrophin (NM[0208] 002825), midkine (NM002391), FGF-2 (XM00366), Nr-CAM 1NM005010), L1/Ng-CAM, contactin (NM001843), N-CAM (XM006332), and axonin-1NM005076.) The first 5 of these molecules are either components of the extracellular matrix in gliomas or are soluble factors known to be present in gliomas, and the latter 4 are neuronal surface molecules. The binding of PTPζ to these molecules may play a significant role in the oncogenesis and growth of neoplastic cells in the brain. Thus, in alternative embodiments of the compositions and methods of the invention, antibody moieties are utilized which bind to PTPζ at a site on the protein which alters the binding of an extracellular ligand molecule to PTPζ. Such PTPζ activity altering antibodies may be utilized in therapeutic compositions in an unconjugated form (e.g., the antibody in an acceptable pharmaceutical carrier), or may be conjugated to either a therapeutic moiety (creating a double-acting therapeutic agent) or an imaging moiety (creating a duel therapeutic/imaging agent).
  • Selection of antibodies which alter (enhance or inhibit) the binding of a ligand to PTPζ may be accomplished by a straightforward binding inhibition/enhancement assay. According to standard techniques, the binding of a labeled (e.g., fluorescently or enzyme-labeled) antibody to PTPζ, which has been immobilized in a microtiter well, is assayed in both the presence and absence of the ligand. The change in binding is indicative of either an enhancer (increased binding) or competitive inhibitor (decreased binding) relationship between the antibody and the ligand. Such assays may be carried out in high-throughput formats (e.g., 384 well plate formats. in robotic systems) for the automated selection of monoclonal antibody candidates for use as PTPζ ligand-binding inhibitors or enhancers. [0209]
  • In addition, antibodies which are useful for altering the function of PTPζ may be assayed in functional formats, such as the HUVEC tube assay and the cell migration assay described below. Thus, antibodies that exhibit the appropriate anti-PTPζ activity may be selected without direct knowledge of a the biomolecular role of PTPζ. [0210]
  • Novel PTPζ Splice Variants PTPζ SM1 and PTPζ SM2 [0211]
  • In addition to the known variants of PTPζ for use in the invention, applicants have identified two novel splice variant isoforms of PTPζ, SM1 and SM2, from their clone libraries, see FIG. 2. These novel isoforms, PTPζ SM1 and PTPζ SM2, differ in structure from the three known isoforms heretofore disclosed, as is illustrated in FIG. 3. As only cDNA sequences for the known splice variants had been previously disclosed, rather than the full gene sequence, applicants verified the location of the novel sequences by comparison of the known splice variant sequences and the novel sequences with a publicly available genomic sequence database. [0212]
  • The protein PTPζ SM1 (amino acid sequence SEQ ID NO. 2, cDNA sequence SEQ ID NO. 1) comprises the amino acids encoded by the first nine exons of PTPζ-α, with three unique additional carboxy terminal amino acids, see FIG. 2. These are encoded by additional 3′ mRNA sequence (nucleotides 1262-1272 of SEQ ID NO. 1) from the intron of the gene between exons nine and ten. The PTPζ SM1 clone was isolated from a human fetal brain cDNA library, an has been shown to be expressed in several human glioblastoma cell lines. Expression of the SM1 splice variant has also been confirmed in primary brain tumor samples. The protein comprises only extracellular domains of PTPζ, and is expected to be secreted by the cell. Thus, PTPζ SM1 may serve a cell signaling or messenger function, and may have bind to a receptor on the surface of cells which are associated with or part of central nervous system tissues. Thus, antibodies specific for PTPζ SM1, and not specific for the other splicing isoforms of PTPζ, may be especially efficacious in the brain tumor therapeutic or imaging compositions of the invention. The PTPζ SM1 protein mainly comprises the carbonic anhydrase-like domain which has been identified in PTPζ α. [0213]
  • Applicants have explored the relationship between the putative carbonic anhydrase domain of PTPζ SM1 (SEQ ID NO. 2) and other human carbonic anhydrase domains from carbonic anhydrase III (SEQ ID NO. 25), carbonic anhydrase I (SEQ ID NO. 26), and carbonic anhydrase VIX [e] (SEQ ID NO. 27), shown below: [0214]
    1                                                   50
             cah3 human ˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜AKEW GYASHNGPDH
             cah1 human ˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜ASPDW GYDDKNGPEQ
             cahe human ˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜ML FSALLLEVIW ILAADGGQHW TYEGPHGQDH
    rptpzetaexon9_frame1 MRILKRFLAC IQLLCVCRLD WANGYYRQQR KLVEEIG..W SYTGALNQKN
    51                                                 100
              cah3_human WHELFPNAKG ENQSPIELHT KDIRHD...P SLQPWSVSYD GGSAKTILNN
              cah1_human WSKLYPIANG NNQSPVDIKT SETKHD...T SLKPISVSYN PATAKEIINV
              cahe_human WPASYPECGN NAQSPIDIQT DSVTFDPDLP ALQPHGYDQP GTEPLDLHNN
    rptpzetaexon9_frame1 WGKKYPTCNS PKQSPINIDE DLTQVNVNLK KLKFQGWDKT SLENTFIHNT
    101                                                150
              cah3_human GKTCRVVFDD TYDRSMLRGG PLPGPYRLRQ FHLHWGS.S. DDHGSEHTVD
              cah1_human GHSFHVNFED NDNRSVLKGG PFSDSYRLFQ FHFHWGS.T. NEHGSEHTVD
              cahe_human GHTVQLSLP. ....STLYLG GLPRKYVAAQ LHLHWGQ.KG SPGGSEHQIN
    rptpzetaexon9_frame1 GKTVEINLTN DYRVS...GG VSEMVFKASK ITFHWGKCNM SSDGSEHSLE
    151                                                200
              cah3_human GVKYAAELHL VHWN.PKYNT FKEALKQRDG IAVIGIFLKI GH.ENGEFQI
              cah1_human GVKYSAELHV AHWNSAKYSS LAEAASKADG LAVIGVLMKV GE.ANPKLQK
              cahe_human SEATFAELHI VHYDSDSYDS LSEAAERPQG LAVLGILIEV GETKNIAYEH
    rptpzetaexon9_frame1 GQKFPLEMQI YCFDADRFSS FEEAVKGKGK LRALSILFEV GTEENLDFKA
    201                                                250
              cah3_human FLDALDKIKT KGKEAPFTKF DPSCLFPACR .DYWTYQGSF TTPPCEECIV
              cah1_human VLDALQAIKT KGKRAPFTNF DPSTLLPSSL .DFWTYPGSL THPPLYESVT
              cahe_human ILSHLHEVRH KDQKTSVPPF NLRELLPKQL GQYFRYNGSL TTPPCYQSVL
    rptpzetaexon9_frame1 IIDGVESVSR FGKQAALDPF ILLNLLPNST DKYYIYNGSL TSPPCTDTVD
    251                                                300
              cah3_human WLLLKEPMTV SSDQMAKLRS LLSSAENEPP VP...LVSNW RPPQPINNRV
              cah1_human WIICKESISV SSEQLAQFRS LLSNVEGDNA VP...MQHNN RPTQPLKGRT
              cahe_human WTVFYRRSQI SMEQLEKLQG TLFSTEEEPS KL...LVQNY RALQPLNQRM
    rptpzetaexon9_frame1 WIVFKDTVSI SESQLAVFCE VLTMQQSGYV MLMDYLQNNF REQQYKFSRQ
    301                                                350
              cah3_human VRASFK˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜
              cah1_human VRASF˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜
              cahe_human VFASFIQAGS SYTTGEMLSL GVGILVGCLC LLLAVYFIAR KIRKKRLENR
    rptpzetaexon9_frame1 VFSSYTGKEE IHEAVCSSEP ENVQADPENY TSLLVTWERP RVVYDTMIEK
    351                          380
              cah3_human ˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜
              cah1_human ˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜
              cahe_human KSVVFTSAQA TTEA˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜˜
    rptpzetaexon9_frame1 FAVLYQQLDG EDQTKHEFLT DGYQDLVTI*
  • Based on alignment with these catalytically active carbonic anhydrases, it seems unlikely that the CA domain could function as a carbonic anhydrase enzyme. Two of the three histidines implicated in binding of the catalytic zinc are missing from the CA domain of the receptor. In active enzymes there is a conserved HxHWG{18,20}ELH motif (the three histidines bind zinc), however, in the receptor this is modified to TFHWG {18,20}EMQ; i.e. two of the three critical zinc atoms would be missing. For comparison, it has been found that a carbonic anhydrase related protein (CAH 8) that lacks just one of these histidines also lacks catalytic activity. [0215]
  • The protein PTPζ SM2 (amino acid sequence SEQ ID NO. 4) comprises the amino acids encoded by all exons of PTPζ-α, plus a 116 nucleotide “extra” exon, in the correct reading frame, between [0216] exons 23 and 24 (nucleotides 6229-6345 of SEQ ID NO. 3). This extra exon, designated exon 23a, contains a portion of the intron sequence between exons 23 and 24 of the PTPζ gene. PTPζ SM2 expression has been verified in several human glioblastoma cell lines, and has also been confirmed in primary brain tumor samples. As PTPζ SM2 comprises all the domains of PTPζ α, the protein is expected to be membrane-bound. The extra exon lies within the cytoplasmic domain of the protein, and thus may alter the protein tyrosine phosphatase function of PTPζ SM2.
  • A novel splicing variant PTPζ protein having an amino acid sequence which includes the amino acid sequence of PTPζ SM1 (SEQ ID NO. 2) or PTPζ SM2 (SEQ. ID NO. 4) may be produced by recombinant techniques known in the art utilizing any suitable vector, in any suitable host cell. The term “vector” is intended to include any physical or biochemical vehicle containing nucleic acid polymers of interest, by which those nucleic acid polymers are transferred into a host cell, thereby transfecting that cell with the introduced nucleic acid polymers. The transfected nucleic acid sequence preferably contains a control sequence, such as a promoter sequence, suitable for transcription of the nucleic acid sequence in the host cell. Examples of vectors include DNA plasmids, viruses, liposomes, particle gun pellets, and transfection vectors known to those of skill in the molecular biology arts. The term “host cell” is intended to mean the target cell for vector transformation, in which the transferred nucleic acid polymer will be replicated and/or expressed. Although bacterial cells may be suitable for production of the proteins for antibody production or structural study purposes, eukaryotic cell hosts are preferred for production of the protein for functional assays or therapeutic purposes. Preferred eukaryotic cell hosts include insect cell lines (e.g., Sf9, Sf21, or High Five™ cell lines), and mammalian cell lines (e.g., HeLa, CHO-K1, COS-7, COS-1, HEK293, HEPG2, Jurkat, MDCK, PAE, PC-12, and other acceptable mammalian cell lines). Thus, the invention also provides vectors incorporating a nucleic acid sequence encoding PTPζ SM1 or PTPζ SM2, as well as host cells which express the proteins. [0217]
  • It is common in the molecular biology arts to utilize additional functional amino acid domains or proteins fused with a protein sequence of interest for purification or detection purposes. Such additional functionalities include, for example, polyhistidine domains, c-myc domains (specifically comprising amino acids 410-419 of the human c-myc oncogene product), β-galactosidase, β-glucuronidase, glutathione-S-transferase, maltose binding protein, human influenza virus hemagglutanin, green fluorescent protein, chloramphenicol acetyltransferase, luciferase, thioredoxin, and others. After purification (e.g., by antibody-affinity chromatography) or detection, these extra amino acid sequences may be cleaved (e.g., by thrombin, enterokinase, Factor Xa, or other protease) to yield a functional mature protein. Thus, the PTPζ SM1 and SM2 proteins of the invention also encompass proteins comprising the amino acid sequence of SEQ ID NO. 2 or SEQ ID NO. 4 and such additional amino acid functionalities. [0218]
  • The invention also provides polypeptides which have a unique activity of PTPζ SM1 or PTPζ SM2 which is not shared by the other PTPζ splice variant (e.g., an antigenic epitope) and which include a portion of the amino acid sequence of PTPζ SM1 or PTPζ SM2 which is at least about 8 to 12 amino acid residues in length, more preferably at least about 20 amino acids in length. These polypeptides preferably comprise an amino acid sequence which is not found in PTPζ α, PTPζ β, or phosphacan, wherein the included portion of the sequence confers the unique activity on the polypeptide. Such polypeptides may be utilized as described above to produce affinity reagents which specifically bind to PTPζ splice variants SM1 or SM2, but do not bind to the other known splice variants of PTPζ. The invention thus provides such specific affinity reagents, which may be produces from such polypeptides, or from an entire PTPζ SM1or PTPζ SM2 protein. In preferred embodiments these affinity reagents are antibodies or antibody fragments. [0219]
  • In addition, although the understanding of the field of protein biochemistry is not as complete as that of molecular genetics, the person or ordinary skill in the art of biochemistry is capable of predicting, with reasonable certainty, when certain substitutions to the primary amino acid sequence structure of a protein will not result in any appreciable modification of a protein's structure or function. Such conservative substitutions are made by replacing an amino acid in the sequence with another containing a side chain with like charge, size, and other characteristics. Conservative substitutions in a protein sequence which would be expected to have minimal to no impact on protein structure or function can be readily devised by a person of ordinary skill in the biochemical arts. To the extent that such conservative substitutions can be made while retaining 90%, preferably 95%, and more preferably 99% or more identity to SEQ. ID NO. 2 or SEQ ID NO. 4, and maintain the activity of the native PTPζ SM1 or PTPζ SM2 protein, such altered proteins are within the scope of the present invention. [0220]
  • The invention also provides nucleic acid polymers encoding the PTPζ splice variants SM1 or SM2. These nucleic acid polymers most preferably comprises a nucleic acid sequence of SEQ. ID NO. 1 or SEQ ID NO. 3, or the predictable variants thereof which one of ordinary skill of the art could derive using the degeneracy of the genetic code. Such nucleic acid polymers are useful for the production of PTPζ SM1 or PTPζ SM2 by recombinant methods, as described above. [0221]
  • The invention also encompasses nucleic acid probes or primers which hybridize to the mRNA encoding PTPζ splice variants SM1 or SM2, but not mRNA encoding other known splice variants of PTPζ. Such probes or primers provided by the invention are preferably able to hybridize with SEQ. ID NO. 1 or SEQ. ID NO. 3 (or their complements) under stringent conditions (e.g., 0.5× to 2×SSC buffer, 0.1% SDS, and a temperature of 55-65° C.), but do not hybridize to SEQ ID NO. 5 (or its complement) under the same conditions. These PTPζ SM1 or PTPζ SM2 coding sequence specific probes are preferably from about 16 to about 40 nucleotides in length, more preferably from about 18 nucleotides to about 30 nucleotides in length. However, probes may be of a smaller size, preferably from about 8 to about 15 nucleotides in length, if two ore more probes are hybridized to adjacent sequences, so that terminal nucleic acid base-stacking interactions may stabilize their hybridization. In preferred embodiments of PTPζ SM1 specific nucleic acid probes, the probes hybridize at or near the novel splice site at the 3′ end of [0222] exon 9, or its complement. In preferred embodiments of PTPζ SM2 specific probes, the probes hybridize at or adjacent to a location selected from: the novel splice site at the 3′ end of exon 23, at least a portion of the novel exon 23a, the novel splice site at the 5′ end of exon 24, or the complement of any one of these.
  • Because PTPζ SM1 and PTPζ SM2 have been shown to be expressed in glioblastoma cell lines and primary tumors, the level of the expression of these splice variants may be useful for staging or characterizing glioblastoma cells. Such cells may be extracted, for instance, from a primary tumor. Thus, the invention provides for the monitoring of the relative expression level of PTPζ SM1 or PTPζ SM2, or both, in relation to each other or to one or more of the known PTPζ splice variants. In one preferred embodiment, the level of expression of PTPζ SM1 is compare to at least one other splice variant selected from PTPζ SM2, PTPζ α, PTPζ β, and phosphacan. In another preferred embodiment, the level of expression of PTPζ SM2 is compare to at least one other splice variant selected from PTPζ SM1, PTPζ α, PTPζ β, and phosphacan. Such comparison may be made in either a qualitative or quantitative manner. One means for comparison is by hybridizing splice-variant specific nucleic acid probes to a sample of nucleic acids (which may be amplified) obtained from brain tumor cells. Alternatively, the expression level of the splice variants may be deduced by the amplification of splice variant nucleic acid sequences, and the analysis of the size of those amplified products using methods known in the art. In another alternative embodiment, protein levels may be studied utilizing splice-variant specific antibodies in either sandwich immunoassay or in-situ staining formats. Various expression level assay techniques are known to those of skill in the molecular biological arts, and thus the specific techniques mentioned above should be considered merely exemplary. [0223]
  • Antibodies for Use in the Antibody-Therapeutics Methods of the Invention [0224]
  • Generally, as the term is utilized in the specification, “antibody” or “antibody moiety” is intended to include any polypeptide chain-containing molecular structure that has a specific shape which fits to and recognizes an epitope, where one or more non-covalent binding interactions stabilize the complex between the molecular structure and the epitope. Antibodies which bind specifically to one of the brain tumor protein targets are referred to as anti-brain tumor protein target antibodies, or α(T[0225] BT), or more specifically α(ARP2), α(SPARC), α(CMET), α(CD44), α(BEHAB), α(TSPAN3), α(VIPR2), α(OPN), α(PTN), and α(PTPζ). The specific or selective fit of a given structure and its specific epitope is sometimes referred to as a “lock and key” fit. The archetypal antibody molecule is the immunoglobulin, and all types of immunoglobulins (IgG, IgM, IgA, IgE, IgD, etc.), from all sources (e.g., human, rodent, rabbit, cow, sheep, pig, dog, other mammal, chicken, turkey, emu, other avians, etc.) are considered to be “antibodies.” Antibodies utilized in the present invention may be polyclonal antibodies, although monoclonal antibodies are preferred because they may be reproduced by cell culture or recombinantly, and may be modified to reduce their antigenicity.
  • Polyclonal antibodies may be raised by a standard protocol by injecting a production animal with an antigenic composition, formulated as described above. See, e.g., Harlow and Lane, Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory, 1988. In one such technique, an T[0226] BT antigen comprising an antigenic portion of the brain tumor protein targets' polypeptide is initially injected into any of a wide variety of mammals (e.g., mice, rats, rabbits, sheep or goats). Alternatively, in order to generate antibodies to relatively short peptide portions of the brain tumor protein target (see discussion above), a superior immune response may be elicited if the polypeptide is joined to a carrier protein, such as ovalbumin, BSA or KLH. The peptide-conjugate is injected into the animal host, preferably according to a predetermined schedule incorporating one or more booster immunizations, and the animals are bled periodically. Polyclonal antibodies specific for the polypeptide may then be purified from such antisera by, for example, affinity chromatography using the polypeptide coupled to a suitable solid support.
  • Alternatively, for monoclonal antibodies, hybridomas may be formed by isolating the stimulated immune cells, such as those from the spleen of the inoculated animal. These cells are then fused to immortalized cells, such as myeloma cells or transformed cells, which are capable of replicating indefinitely in cell culture, thereby producing an immortal, immunoglobulin-secreting cell line. The immortal cell line utilized is preferably selected to be deficient in enzymes necessary for the utilization of certain nutrients. Many such cell lines (such as myelomas) are known to those skilled in the art, and include, for example: thymidine kinase (TK) or hypoxanthine-guanine phosphoriboxyl transferase (HGPRT). These deficiencies allow selection for fused cells according to their ability to grow on, for example, hypoxanthine aminopterinthymidine medium (HAT). [0227]
  • Preferably, the immortal fusion partners utilized are derived from a line that does not secrete immunoglobulin. The resulting fused cells, or hybridomas, are cultured under conditions that allow for the survival of fused, but not unfused, cells and the resulting colonies screened for the production of the desired monoclonal antibodies. Colonies producing such antibodies are cloned, expanded, and grown so as to produce large quantities of antibody, see Kohler and Milstein, 1975 Nature 256:495 (the disclosures of which are hereby incorporated by reference). [0228]
  • Large quantities of monoclonal antibodies from the secreting hybridomas may then be produced by injecting the clones into the peritoneal cavity of mice and harvesting the ascites fluid therefrom. The mice, preferably primed with pristine, or some other tumor-promoter, and immunosuppressed chemically or by irradiation, may be any of various suitable strains known to those in the art. The ascites fluid is harvested from the mice and the monoclonal antibody purified therefrom, for example, by CM Sepharose column or other chromatographic means. Alternatively, the hybridomas may be cultured in vitro or as suspension cultures. Batch, continuous culture, or other suitable culture processes may be utilized. Monoclonal antibodies are then recovered from the culture medium or supernatant. [0229]
  • Several monoclonal antibodies against various isoforms of the brain tumor protein targets are currently available from commercial sources. For instance, a non-exclusive list of available commercial antibodies includes: for SPARC/Osteonectin, from Zymed, mouse anti-bovine MAb (cross-reactivity with human), suitable for ELISA, WB, IH (paraffin), Cat# 33-5500; for c-MET, from Zymed, rabbit anti-human polyclonal, suitable for ELISA, WB, IH. Cat# 71-8000, and from RDI, rabbit anti-human MAb, suitable for WB, IP, IH. Cat# RDI-MET Cabr.; for CD44, from RDI, mouse anti-human MAb, only for IH and FACS, Cat# RD1-M1676clb., and from Lab vision, mouse anti-human MAb, known to block binding of hyaluronic acid to its receptor CD44, “CD44/H-CAM Ab-2”; for Brevican/BEHAB, from BD Transduction Lab., a mouse anti-human MAb, WB, IF, Cat# B68820; for VIP 2 receptor, from Exalpha, mouse anti-rat (possible human cross-specificity, which is easily assayed) MAb, WB, IH. Cat#2140M; for Laminin receptor 67 kDa, from Lab vision, mouse anti-human MAb, IH, ELISA, not for WB. “laminin receptor Ab-1”; for Osteopontin, from Chemicon, rat anti-human MAb, raised against rh-Osteopontin—recognizes native protein well, WB, IH, ELISA. “MAB3057”; for Pleiotrophin, from R&D goat anti-human polyclonal, WB, recognizes rh-Pleiotrophin. “BAF252”, and from Oncogene goat anti-human polyclonal, WB, ELISA, detects rh-Pleiotrophin. “PC87L”.; for PTPζ-α and PTPζ-β, from BD Transduction Labs, mouse anti-human MAb (WB, IH, IF), denominated “R20720” and from Chemicon, mouse anti-human MAb (WB, IH, IP), denominated “MAB5210”, which recognizes both of the transmembrane isoforms, and also recognizes the soluble isoform (phosphacan, PTPζ-S). These antibodies are suitable for use in the compositions of the present invention, especially in Fab fragment form (which eliminates significant portions of the antigenic mouse constant heavy and light chain regions). However, it is preferred that such antibodies be humanized or chimerized according to one of the procedures outlined below. [0230]
  • In addition, the antibodies or antigen binding fragments may be produced by genetic engineering. In this technique, as with the standard hybridoma procedure, antibody-producing cells are sensitized to the desired antigen or immunogen. The messenger RNA isolated from the immune spleen cells or hybridomas is used as a template to make cDNA using PCR amplification. A library of vectors, each containing one heavy chain gene and one light chain gene retaining the initial antigen specificity, is produced by insertion of appropriate sections of the amplified immunoglobulin cDNA into the expression vectors. A combinatorial library is constructed by combining the heavy chain gene library with the light chain gene library. This results in a library of clones which co-express a heavy and light chain (resembling the Fab fragment or antigen binding fragment of an antibody molecule). The vectors that carry these genes are co-transfected into a host (e.g. bacteria, insect cells, mammalian cells, or other suitable protein production host cell.). When antibody gene synthesis is induced in the transfected host, the heavy and light chain proteins self-assemble to produce active antibodies that can be detected by screening with the antigen or immunogen. [0231]
  • Preferably, recombinant antibodies are produced in a recombinant protein production system which correctly glycosylates and processes the immunoglobulin chains, such as insect or mammalian cells. An advantage to using insect cells which utilize recombinant baculoviruses for the production of antibodies for use in the present invention is that the baculovirus system allows production of mutant antibodies much more rapidly than stably transfected mammalian cell lines. In addition, insect cells have been shown to correctly process and glycosylate eukaryotic proteins, which prokaryotic cells do not. Finally, the baculovirus expression of foreign protein has been shown to constitute as much as 50-75% of the total cellular protein late in viral infection, making this system an excellent means of producing milligram quantities of the recombinant antibodies. [0232]
  • The use of the baculovirus [0233] Autographia californica nuclear polyhedrosis virus (AcNPV) and recombinant viral stocks in Spodoptera frugiperda (Sf9) cells to prepare large quantities of protein has been described by Smith et al. (1985), Summers and Smith (1987). A preferred method of preparing recombinant antibodies is through the expression of DNA encoding recombinant antibody (produced by screening, as above, or by protein engineering to include more human-like domains, as discussed below) via the baculoviral expression system in Sf9 insect cells. Production of recombinant proteins in Sf9 cells is well known in the art, and one of ordinary skill would be able to select from a number of acceptable protocols (e.g., that described in U.S. Pat. No. 6,603,905).
  • It should be noted that antibodies which have a reduced propensity to induce a violent or detrimental immune response in humans (such as anaphylactic shock), and which also exhibit a reduced propensity for priming an immune response which would prevent repeated dosage with the antibody therapeutic or imaging agent (e.g., the human-anti-murine-antibody “HAMA” response), are preferred for use in the invention. These antibodies are preferred for all administrative routes, including intrathecal administration. Even through the brain is relatively isolated in the cranial cavity, behind the blood brain barrier, an immune response still can occur in the form of increased leukocyte infiltration, and inflammation. Although some increased immune response against the tumor is desirable, the concurrent binding and inactivation of the therapeutic or imaging agent generally outweighs this benefit. Thus, humanized, chimeric, or xenogenic human antibodies, which produce less of an immune response when administered to humans, are preferred for use in the present invention. [0234]
  • Chimeric antibodies may be made by recombinant means by combining the murine variable light and heavy chain regions (VK and VH), obtained from a murine (or other animal-derived) hybridoma clone, with the human constant light and heavy chain regions, in order to produce an antibody with predominantly human domains. The production of such chimeric antibodies is well known in the art, and may be achieved by standard means (as described, e.g., in U.S. Pat. No. 5,624,659, incorporated fully herein by reference). Humanized antibodies are engineered to contain even more human-like immunoglobulin domains, and incorporate only the complementarity-determining regions of the animal-derived antibody. This is accomplished by carefully examining the sequence of the hyper-variable loops of the variable regions of the monoclonal antibody, and fitting them to the structure of the human antibody chains. Although facially complex, the process is straightforward in practice. See, e.g., U.S. Pat. No. 6,187,287, incorporated fully herein by reference. [0235]
  • Alternatively, polyclonal or monoclonal antibodies may be produced from animals which have been genetically altered to produce human immunoglobulins, such as the Abgenix XenoMouse or the Medarex HuMAb® technology. The transgenic animal may be produced by initially producing a “knock-out” animal which does not produce the animal's natural antibodies, and stably transforming the animal with a human antibody locus (e.g., by the use of a human artificial chromosome). Only human antibodies are then made by the animal. Techniques for generating such animals, and deriving antibodies therefrom, are described in U.S. Pat. Nos. 6,162,963 and 6,150,584, incorporated fully herein by reference. Such fully human xenogenic antibodies are a preferred antibody for use in the methods and compositions of the present invention. [0236]
  • Alternatively, single chain antibodies (Fv, as described below) can be produced from phage libraries containing human variable regions. See U.S. Pat. No. 6,174,708, incorporated fully herein by reference. Also see Kuan, C. T., Reist, C. J., Foulon, C. F., Lorimer, I. A., Archer, G., Pegram, C. N., Pastan, I., Zalutsky, M. R., and Bigner, D. D. (1999). 125I-labeled anti-epidermal growth factor receptor-viii single-chain Fv exhibits specific and high-level targeting of glioma xenografts. [0237] Clin Cancer Res. 5, 1539-49;Lorimer, I. A., Keppler-Hafkemeyer, A., Beers, R. A., Pegram, C. N., Bigner, D. D., and Pastan, I. (1996). Recombinant immunotoxins specific for a mutant epidermal growth factor receptor: targeting with a single chain antibody variable domain isolated by phage display. Proc. Nat. Acad. Sci. USA 93, 14815-20; Pastan, I. H., Archer, G. E., McLendon, R. E., Friedman, H. S., Fuchs, H. E., Wang, Q. C., Pai, L. H., Herndon, J., and Bigner, D. D. (1995). Intrathecal administration of single-chain immunotoxin, LMB-7 [B3(Fv)-PE38], produces cures of carcinomatous meningitis in a rat model. Proc Natl. Acad. Sci USA 92, 2765-9, all of which are incorporated by reference fully herein.
  • In addition to entire immunoglobulins (or their recombinant counterparts), immunoglobulin fragments comprising the epitope binding site (e.g., Fab′, F(ab′)[0238] 2, or other fragments) are useful as antibody moieties in the present invention. Such antibody fragments may be generated from whole immunoglobulins by ficin, pepsin, papain, or other protease cleavage. “Fragment,” or minimal immunoglobulins may be designed utilizing recombinant immunoglobulin techniques. For instance “Fv” immunoglobulins for use in the present invention may be produced by linking a variable light chain region to a variable heavy chain region via a peptide linker (e.g., poly-glycine or another sequence which does not form an alpha helix or beta sheet motif).
  • Fv fragments are heterodimers of the variable heavy chain domain (V[0239] H) and the variable light chain domain (VL). The heterodimers of heavy and light chain domains that occur in whole IgG, for example, are connected by a disulfide bond. Recombinant Fvs in which VH and VL are connected by a peptide linker are typically stable, see, for example, Huston et al., Proc. Natl. Acad, Sci. USA 85:5879-5883 (1988) and Bird et al., Science 242:423-426 (1988), both fully incorporated herein, by reference. These are single chain Fvs which have been found to retain specificity and affinity and have been shown to be useful for imaging tumors and to make recombinant immunotoxins for tumor therapy. However, researchers have bound that some of the single chain Fvs have a reduced affinity for antigen and the peptide linker can interfere with binding. Improved Fv's have been also been made which comprise stabilizing disulfide bonds between the VH and VL regions, as described in U.S. Pat. No. 6,147,203, incorporated fully herein by reference. Any of these minimal antibodies may be utilized in the present invention, and those which are humanized to avoid HAMA reactions are preferred for use in embodiments of the invention.
  • In addition, derivatized immunoglobulins with added chemical linkers, detectable moieties [fluorescent dyes, enzymes, substrates, chemiluminescent moieties], or specific binding moieties [such as streptavidin, avidin, or biotin] may be utilized in the methods and compositions of the present invention. For convenience, the term “antibody” or “antibody moiety” will be used throughout to generally refer to molecules which specifically bind to an epitope of the brain tumor protein targets, although the term will encompass all immunoglobulins, derivatives, fragments, recombinant or engineered immunoglobulins, and modified immunoglobulins, as described above. [0240]
  • Candidate anti-T[0241] BT antibodies can be tested for anti-TBT activity by any suitable standard means. As a first screen, the antibodies may be tested for binding against the brain tumor protein target antigen utilized to produce them, or against the entire brain tumor protein target extracellular domain or protein. As a second screen, anti-TBT candidates may be tested for binding to an appropriate glioblastoma cell line (i.e., one which approximates primary tumor brain tumor protein target expression), or to primary tumor tissue samples. For these screens, the anti-TBT candidate antibody may be labeled for detection (e.g., with fluorescein or another fluorescent moiety, or with an enzyme such as horseradish peroxidase). After selective binding to the brain tumor protein target is established, the candidate antibody, or an antibody conjugate produced as described below, may be tested for appropriate activity (i.e., the ability to decrease tumor cell growth and/or to aid in visualizing tumor cells) in an in vivo model, such as an appropriate glioblastoma cell line, or in a mouse or rat human brain tumor model, as described below.
  • General Functional Assay Methods for Antibodies for Use in the Invention [0242]
  • In addition to the specific binding assays and protein-specific functional assays described for individual proteins above, antibodies which are useful for altering the function of ARP-2, SPARC, c-MET, BEHAB, CD-44, TSPN3, PTN, OPN, VIPR-2, or PTPζ may be assayed in functional formats, such as glioblastoma cell culture or mouse/rat CNS tumor model studies. In glioblastoma cell models of activity, expression of the protein is first verified in the particular cell strain to be used. If necessary, the cell line may be stably transfected with a coding sequence of the protein under the control of an appropriate constituent promoter, in order to express the protein at a level comparable to that found in primary tumors. The ability of the glioblastoma cells to survive in the presence of the candidate function-altering anti-protein antibody is then determined. In addition to cell-survival assays, cell migration assays, as described below in Example 1, may be utilized to determine the effect of the candidate antibody therapeutic agent on the tumor-like behavior of the cells. Alternatively, if the brain tumor protein target is involved in angiogenesis, or endothelial cell sprouting assays such as described in Example 2 may be utilized to determine the ability of the candidate antibody therapeutic to inhibit vascular neogenesis, an important function in tumor biology. [0243]
  • Similarly, in vivo models for human brain tumors, particularly nude mice/SCID mice model or rat models, have been described [Antunes, L., Angioi-Duprez, K. S., Bracard, S. R., Klein-Monhoven, N. A., Le Faou, A. E., Duprez, A. M., and Plenat, F. M. (2000). Analysis of tissue chimerism in nude mouse brain and abdominal xenograft models of human glioblastoma multiforme: what does it tell us about the models and about glioblastoma biology and therapy? [0244] J Histochem Cytochem 48, 847-58; Price, A., Shi, Q., Morris, D., Wilcox, M. E., Brasher, P. M., Rewcastle, N. B., Shalinsky, D., Zou, H., Appelt, K., Johnston, R. N., Yong, V. W., Edwards, D., and Forsyth, P. (1999). Marked inhibition of tumor growth in a malignant glioma tumor model by a novel synthetic matrix metalloproteinase inhibitor AG3340. Clin Cancer Res 5, 845-54; and Senner, V., Sturm, A., Hoess, N., Wassmann, H., and Paulus, W. (2000). In vivo glioma model enabling regulated gene expression. Acta Neuropathol (Berl) 99, 603-8.] Once correct expression of the protein in the tumor model is verified, the effect of the candidate anti-protein antibodies on the tumor masses in these models can be evaluated, wherein the ability of the anti-protein antibody candidates to alter protein activity is indicated by a decrease in tumor growth or a reduction in the tumor mass. Thus, antibodies that exhibit the appropriate anti-tumor effect may be selected without direct knowledge of the particular biomolecular role of the protein in oncogenesis.
  • Therapeutic and Imaging Moieties and Methods for Conjugating Them Qith anti-PTPζ Antibodies to Use in the Compositions and Methods of the Invention [0245]
  • As described above, the anti-T[0246] BT antibodies for use in the present invention may have utility without conjugation when the native activity of the brain tumor protein target is altered in the tumor cell. Such antibodies, which may be selected as described above, may be utilized without further modification to include a cytotoxic or imaging moiety. These types of compositions have the advantage of reduced toxicity (in that only the toxicity of the antibody moieties themselves must be taken into account when dosing), and are simpler to manufacture. Thus, non-conjugated activity altering anti-TBT antibody therapeutics are a preferred embodiment of the invention. However, the conjugation of cytotoxic or imaging agents is yet another preferred embodiment when utilizing these antibodies because the added moieties add functionality to the therapeutic.
  • Thus, in many preferred embodiments of the invention, the anti-T[0247] BT antibodies may be coupled or conjugated to one or more therapeutic cytotoxic or imaging moieties. As used herein, “cytotoxic moiety” (C) simply means a moiety which inhibits cell growth or promotes cell death when proximate to or absorbed by the cell. Suitable cytotoxic moieties in this regard include radioactive isotopes (radionuclides), chemotoxic agents such as differentiation inducers and small chemotoxic drugs, toxin proteins, and derivatives thereof. As utilized herein, “imaging moiety” (I) means a moiety which can be utilized to increase contrast between a tumor and the surrounding healthy tissue in a visualization technique (e.g., radiography, positron-emission tomography, magnetic resonance imaging, direct or indirect visual inspection). Thus, suitable imaging moieties include radiography moieties (e.g. heavy metals and radiation emitting moieties), positron emitting moieties, magnetic resonance contrast moieties, and optically visible moieties (e.g., fluorescent or visible-spectrum dyes, visible particles, etc.). It will be appreciated by one of ordinary skill that some overlap exists between what is a therapeutic moiety and what is an imaging moiety. For instance 212Pb and 212Bi are both useful radioisotopes for therapeutic compositions, but are also electron-dense, and thus provide contrast for X-ray radiographic imaging techniques, and can also be utilized in scintillation imaging techniques.
  • In general, therapeutic or imaging agents may be conjugated to the anti-PTPζ moiety by any suitable technique, with appropriate consideration of the need for pharmokinetic stability and reduced overall toxicity to the patient. A therapeutic agent may be coupled to a suitable antibody moiety either directly or indirectly (e.g. via a linker group). A direct reaction between an agent and an antibody is possible when each possesses a functional group capable of reacting with the other. For example, a nucleophilic group, such as an amino or sulfhydryl group, may be capable of reacting with a carbonyl-containing group, such as an anhydride or an acid halide, or with an alkyl group containing a good leaving group (e.g., a halide). Alternatively, a suitable chemical linker group may be used. A linker group can function as a spacer to distance an antibody from an agent in order to avoid interference with binding capabilities. A linker group can also serve to increase the chemical reactivity of a substituent on a moiety or an antibody, and thus increase the coupling efficiency. An increase in chemical reactivity may also facilitate the use of moieties, or functional groups on moieties, which otherwise would not be possible. [0248]
  • Suitable linkage chemistries include maleimidyl linkers and alkyl halide linkers (which react with a sulfhydryl on the antibody moiety) and succinimidyl linkers (which react with a primary amine on the antibody moiety). Several primary amine and sulfhydryl groups are present on immunoglobulins, and additional groups may be designed into recombinant immunoglobulin molecules. It will be evident to those skilled in the art that a variety of bifunctional or polyfunctional reagents, both homo- and hetero-functional (such as those described in the catalog of the Pierce Chemical Co., Rockford, Ill.), may be employed as a linker group. Coupling may be effected, for example, through amino groups, carboxyl groups, sulfhydryl groups or oxidized carbohydrate residues. There are numerous references describing such methodology, e.g., U.S. Pat. No. 4,671,958. As an alternative coupling method, cytotoxic or imaging moieties may be coupled to the anti-T[0249] BT antibody moiety through a an oxidized carbohydrate group at a glycosylation site, as described in U.S. Pat. Nos. 5,057,313 and 5,156,840. Yet another alternative method of coupling the antibody moiety to the cytotoxic or imaging moiety is by the use of a non-covalent binding pair, such as streptavidin/biotin, or avidin/biotin. In these embodiments, one member of the pair is covalently coupled to the antibody moiety and the other member of the binding pair is covalently coupled to the cytotoxic or imaging moiety.
  • Where a cytotoxic moiety is more potent when free from the antibody portion of the immunoconjugates of the present invention, it may be desirable to use a linker group which is cleavable during or upon internalization into a cell, or which is gradually cleavable over time in the extracellular environment. A number of different cleavable linker groups have been described. The mechanisms for the intracellular release of a cytotoxic moiety agent from these linker groups include cleavage by reduction of a disulfide bond (e.g., U.S. Pat. No. 4,489,710), by irradiation of a photolabile bond (e.g., U.S. Pat. No. 4,625,014), by hydrolysis of derivatized amino acid side chains (e.g., U.S. Pat. No. 4,638,045), by serum complement-mediated hydrolysis (e.g., U.S. Pat. No. 4,671,958), and acid-catalyzed hydrolysis (e.g., U.S. Pat. No. 4,569,789). [0250]
  • It may be desirable to couple more than one cytotoxic and/or imaging moiety to an antibody. By poly-derivatizing the anti-T[0251] BT antibody, several cytotoxic strategies may be simultaneously implemented, an antibody may be made useful as a contrasting agent for several visualization techniques, or a therapeutic antibody may be labeled for tracking by a visualization technique. In one embodiment, multiple molecules of an imaging or cytotoxic moiety are coupled to one antibody molecule. In another embodiment, more than one type of moiety may be coupled to one antibody. Regardless of the particular embodiment, immunoconjugates with more than one moiety may be prepared in a variety of ways. For example, more than one moiety may be coupled directly to an antibody molecule, or linkers which provide multiple sites for attachment (e.g., dendrimers) can be used. Alternatively, a carrier with the capacity to hold more than one cytotoxic or imaging moiety can be used.
  • A carrier may bear the agents in a variety of ways, including covalent bonding either directly or via a linker group, and non-covalent associations. Suitable covalent-bond carriers include proteins such as albumins (e.g., U.S. Pat. No. 4,507,234), peptides, and polysaccharides such as aminodextran (e.g., U.S. Pat. No. 4,699,784), each of which have multiple sites for the attachment of moieties. A carrier may also bear an agent by non-covalent associations, such as non-covalent bonding or by encapsulation, such as within a liposome vesicle (e.g., U.S. Pat. Nos. 4,429,008 and 4,873,088). Encapsulation carriers are especially useful for imaging moiety conjugation to anti-T[0252] BT antibody moieties for use in the invention, as a sufficient amount of the imaging moiety (dye, magnetic resonance contrast reagent, etc.) for detection may be more easily associated with the antibody moiety. In addition, encapsulation carriers are also useful in chemotoxic therapeutic embodiments, as they can allow the therapeutic compositions to gradually release a chemotoxic moiety over time while concentrating it in the vicinity of the tumor cells.
  • Carriers and linkers specific for radionuclide agents (both for use as cytotoxic moieties or positron-emission imaging moieties) include radiohalogenated small molecules and chelating compounds. For example, U.S. Pat. No. 4,735,792 discloses representative radiohalogenated small molecules and their synthesis. A radionuclide chelate may be formed from chelating compounds that include those containing nitrogen and sulfur atoms as the donor atoms for binding the metal, or metal oxide, radionuclide. For example, U.S. Pat. No. 4,673,562, to Davison et al. discloses representative chelating compounds and their synthesis. Such chelation carriers are also useful for magnetic spin contrast ions for use in magnetic resonance imaging tumor visualization methods, and for the chelation of heavy metal ions for use in radiographic visualization methods. [0253]
  • Preferred radionuclides for use as cytotoxic moieties are radionuclides which are suitable for pharmacological administration. Such radionuclides include [0254] 123I, 125I, 131I, 90Y, 211At, 67Cu, 186Re, 188Re, 212Pb, and 212Bi. Iodine and astatine isotopes are more preferred radionuclides for use in the therapeutic compositions of the present invention, as a large body of literature has been accumulated regarding their use. 131I is particularly preferred, as are other β-radiation emitting nuclides, which have an effective range of several millimeters. 123I, 125I, 131I, or 211At may be conjugated to antibody moieties for use in the compositions and methods utilizing any of several known conjugation reagents, including Iodogen, N-succinimidyl 3-[211At]astatobenzoate, N-succinimidyl 3-[133I]iodobenzoate (SIB), and, N-succinimidyl 5-[131I]iodob-3-pyridinecarboxylate (SIPC). Any iodine isotope may be utilized in the recited iodo-reagents. For example, a suitable antibody for use in the present invention may be easily made by coupling an Fab fragment of the BD Transduction Labs R20720 anti-PTPζ MAb with 131I Iodogen according to the manufacturer's instructions. Other radionuclides may be conjugated to anti-TBTantibody moieties by suitable chelation agents known to those of skill in the nuclear medicine arts.
  • Preferred chemotoxic agents include small-molecule drugs such as carboplatin, cisplatin, vincristine, taxanes such as paclitaxel and doceltaxel, hydroxyurea, gemcitabine, vinorelbine, irinotecan, tirapazamine, matrilysin, methotrexate, pyrimidine and purine analogs, and other suitable small toxins known in the art. Preferred chemotoxin differentiation inducers include phorbol esters and butyric acid. Chemotoxic moieties may be directly conjugated to the anti-T[0255] BTantibody moiety via a chemical linker, or may encapsulated in a carrier, which is in turn coupled to the anti-TBT antibody moiety.
  • Preferred toxin proteins for use as cytotoxic moieties include ricins A and B, abrin, diphtheria toxin, bryodin 1 and 2, momordin, trichokirin, cholera toxin, gelonin, Pseudomonas exotoxin, Shigella toxin, pokeweed antiviral protein, and other toxin proteins known in the medicinal biochemistry arts. As these toxin agents may elicit undesirable immune responses in the patient, especially if injected intravascularly, it is preferred that they be encapsulated in a carrier for coupling to the anti-T[0256] BT antibody moiety.
  • Preferred radiographic moieties for use as imaging moieties in the present invention include compounds and chelates with relatively large atoms, such as gold, iridium, technetium, barium, thallium, iodine, and their isotopes. It is preferred that less toxic radiographic imaging moieties, such as iodine or iodine isotopes, be utilized in the compositions and methods of the invention. Examples of such compositions which may be utilized for x-ray radiography are described in U.S. Pat. No. 5,709,846, incorporated fully herein by reference. Such moieties may be conjugated to the anti-T[0257] BT antibody moiety through an acceptable chemical linker or chelation carrier. In addition, radionuclides which emit radiation capable of penetrating the scull may be useful for scintillation imaging techniques. Suitable radionuclides for conjugation include 99Tc, 111In, and 67Ga. Positron emitting moieties for use in the present invention include 18F, which can be easily conjugated by a fluorination reaction with the anti-TBT antibody moiety according to the method described in U.S. Pat. No. 6,187,284.
  • Preferred magnetic resonance contrast moieties include chelates of chromium(III), manganese(II), iron(II), nickel(II), copper(II), praseodymium(III), neodymium(III), samarium(III) and ytterbium(III) ion. Because of their very strong magnetic moment, the gadolinium(III), terbium(III), dysprosium(III), holmium(III), erbium(III), and iron(III) ions are especially preferred. Examples of such chelates, suitable for magnetic resonance spin imaging, are described in U.S. Pat. No. 5,733,522, incorporated fully herein by reference. Nuclear spin contrast chelates may be conjugated to the anti-T[0258] BT antibody moieties through a suitable chemical linker.
  • Optically visible moieties for use as imaging moieties include fluorescent dyes, or visible-spectrum dyes, visible particles, and other visible labeling moieties. Fluorescent dyes such as fluorescein, coumarin, rhodamine, bodipy Texas red, and cyanine dyes, are useful when sufficient excitation energy can be provided to the site to be inspected visually. Endoscopic visualization procedures may be more compatible with the use of such labels. For many procedures where imaging agents are useful, such as during an operation to resect a brain tumor, visible spectrum dyes are preferred. Acceptable dyes include FDA-approved food dyes and colors, which are non-toxic, although pharmaceutically acceptable dyes which have been approved for internal administration are preferred. In preferred embodiments, such dyes are encapsulated in carrier moieties, which are in turn conjugated to the anti-T[0259] BT antibody. Alternatively, visible particles, such as colloidal gold particles or latex particles, may be coupled to the anti-TBT antibody moiety via a suitable chemical linker.
  • Delivery of Therapeutic and Imaging Agents to the Patient: [0260]
  • The Blood Brain Barrier (BBB) and Administration Strategies: [0261]
  • At one time, the BBB was not considered to present a problem in the diagnosis and treatment of brain tumors, because early scans of human brain tumors suggested that the BTB (blood tumor barrier) was “leaky.” However, as the size of the molecule increases, the rate of movement across the barrier decreases. The BBB has been demonstrated to be heterogeneous in experimental human tumor xenograft animal models and in human patients. This lack of uniformity is because of the reduced integrity of tight junctions in the capillary endothelial cells of the tumor neovasculature, intratumoral variation in permeability, and altered intratumoral blood flow (Fuchs et al, 1990[0262] , Cancer research 50, 1954-59, Groothuis et al., 1984, Prog. Exp. Tumor Res.) Thus, although the BBB may not pose a delivery problem for some tumors in some patients, this cannot be said for all brain tumors. In addition, a preferred mode of administration of the therapeutics of the invention is after removal of the main tumor mass (resection of the tumor), which destroys much of the “leaky” neovasculature. Moreover, as brain carcinomas are usually pervasive throughout the organ, therapies which are directed towards eradicating all tumor-producing cells cannot rely exclusively on the localized tumor neovasculature.
  • A first strategy for drug delivery through the BBB entails disruption of the BBB, either by osmotic means such as mannitol or leukotrienes, or biochemically by the use of vasoactive substances such as bradykinin. The potential for using BBB opening to target specific agents to brain tumors is also an option. In preferred embodiments, a BBB disrupting agent is co-administered with the therapeutic or imaging compositions of the invention when the compositions are administered by intravascular injection. Other strategies to go through the BBB may entail the use of endogenous transport systems, including carrier-mediated transporters such as glucose and amino acid carriers, receptor-mediated transcytosis for insulin or transferrin, and active efflux transporters such as p-glycoprotein. Active transport moieties may also be conjugated to the therapeutic or imaging compounds for use in the invention to facilitate transport across the epithelial wall of the blood vessel. However, the best current strategy for drug delivery behind the BBB is by intrathecal delivery of therapeutics or imaging agents directly to the cranium, as through an Ommaya reservoir. [0263]
  • Delivery/Administration of Therapeutic Antibodies: [0264]
  • For administration, the antibody-therapeutic or antibody-imaging agent will generally be mixed, prior to administration, with a non-toxic, pharmaceutically acceptable carrier substance. Usually, this will be an aqueous solution, such as normal saline or phosphate-buffered saline (PBS), Ringer's solution, lactate-Ringer's solution, or any isotonic physiologically acceptable solution for administration by the chosen means. Preferably, the solution is sterile and pyrogen-free, and is manufactured and packaged under current Good Manufacturing Processes (GMPs), as approved by the FDA. The clinician of ordinary skill is familiar with appropriate ranges for pH, tonicity, and additives or preservatives when formulating pharmaceutical compositions for administration by intravascular injection, intrathecal injection, injection into the cerebro-spinal fluid, direct injection into the tumor, or by other routes. In addition to additives for adjusting pH or tonicity, the antibody-therapeutics and antibody-imaging agents may be stabilized against aggregation and polymerization with amino acids and non-ionic detergents, polysorbate, and polyethylene glycol. Optionally, additional stabilizers may include various physiologically-acceptable carbohydrates and salts. Also, polyvinylpyrrolidone may be added in addition to the amino acid. Suitable therapeutic immunoglobulin solutions which are stabilized for storage and administration to humans are described in U.S. Pat. No. 5,945,098, incorporated fully herein by reference. Other agents, such as human serum albumin (HSA), may be added to the therapeutic or imaging composition to stabilize the antibody conjugates. [0265]
  • The compositions of the invention may be administered using any medically appropriate procedure, e.g., intravascular (intravenous, intraarterial, intracapillary) administration, injection into the cerebrospinal fluid, intracavity or direct injection in the tumor. Intrathecal administration maybe carried out through the use of an Ommaya reservoir, in accordance with known techniques. (F. Balis et al., Am J. Pediatr. Hematol. Oncol. 11, 74, 76 (1989). For the imaging compositions of the invention, administration via intravascular injection is preferred for pre-operative visualization of the tumor. Post-operative visualization or visualization concurrent with an operation may be through intrathecal or intracavity administration, as through an Ommaya reservoir, or also by intravascular administration. [0266]
  • Intravascular injection may be by intravenous or intraarterial injection: carotid artery injection is thought to assist in administration to the brain, and is thus preferred. Antibody-agents injected into the blood stream have been shown to cross the blood-brain barrier and to infiltrate the cranial cavity to some extent, usually in the range of 10[0267] −4 to 10−3% [?UNITS?] injected dose per gram. This rate of uptake may be sufficient for imaging reagents, and also may be useful for tumor cell specific cytotoxic agents (e.g, those specifically directed to the inhibition of the function of tumor-cell overexpressed proteins). However, in order to achieve therapeutic concentrations of the antibody-therapeutic agents without unacceptable toxicity to the patient, it is preferred that the therapeutics compositions be administered by intrathecal injection, direct injection, or injection into the cerebro-spinal fluid.
  • Thus, a preferred method for administration of the therapeutic compositions of the invention is by depositing it into the inner cavity of a cystic tumor by any suitable technique, such as by direct injection (aided by stereotaxic positioning of an injection syringe, if necessary) or by placing the tip of an Ommaya reservoir into a cavity, or cyst, for administration. Where the tumor is a solid tumor, the antibody may be administered by first creating a resection cavity in the location of the tumor. This procedure differs from an ordinary craniotomy and tumor resection only in a few minor respects. As tumor resection is a common treatment procedure, and is often indicated to relieve pressure, administration of the therapeutic compositions of the invention following tumor resection is a preferred embodiment of the treatment methods of the invention. Following gross total resection in a standard neurosurgical fashion, the cavity is preferable rinsed with saline until all bleeding is stopped by cauterization. Next the pia-arachnoid membrane, surrounding the tumor cavity at the surface, is cauterized to enhance the formation of fibroblastic reaction and scarring in the pia-arachnoid area. The result is the formation of an enclosed, fluid-filled cavity within the brain tissue at the location from where the tumor was removed. After the cyst has been formed, either the tip of an Ommaya reservoir or a micro catheter, which is connected to a pump device and allows the continues infusion of an antibody solution into the cavity, can be placed into the cavity. See, e.g., U.S. Pat. No. 5,558,852, incorporated fully herein by reference. [0268]
  • Alternatively, a convection-enhanced delivery catheter may be implanted directly into the tumor mass, into a natural or surgically created cyst, or into the normal brain mass. Such convection-enhanced pharmaceutical composition delivery devices greatly improve the diffusion of the composition throughout the brain mass. The implanted catheters of these delivery devices utilize high-flow microinfusion (with flow rates in the range of about 0.5 to 15.0 μl/minute), rather than diffusive flow, to deliver the therapeutic or imaging composition to the brain and/or tumor mass. Such devices are described in U.S. Pat. No. 5,720,720, incorporated fully herein by reference. [0269]
  • The effective amount of the therapeutic antibody-conjugate composition or of the imaging antibody-conjugate compositions to be given to a particular patient will depend on a variety of factors, several of which will be different from patient to patient. A competent clinician will be able to determine an effective amount of a therapeutic antibody-conjugate composition to administer to a patient to retard the growth and promote the death of tumor cells, or an effective amount of an imaging composition to administer to a patient to facilitate the visualization of a tumor. Dosage of the antibody-conjugate will depend on the treatment of the tumor, route of administration, the nature of the therapeutics, sensitivity of the tumor to the therapeutics, etc. Utilizing LD[0270] 50 animal data, and other information available for the conjugated cytotoxic or imaging moiety, a clinician can determine the maximum safe dose for an individual, depending on the route of administration. For instance, an intravenously administered dose may be more than an intrathecally administered dose, given the greater body of fluid into which the therapeutic composition is being administered. Similarly, compositions which are rapidly cleared from the body may be administered at higher doses, or in repeated doses, in order to maintain a therapeutic concentration. Imaging moieties are typically less toxic than cytotoxic moieties and may be administered in higher doses in some embodiments. Utilizing ordinary skill, the competent clinician will be able to optimize the dosage of a particular therapeutic or imaging composition in the course of routine clinical trials.
  • Typically the dosage will be 0.001 to 100 milligrams of conjugate per kilogram subject body weight. Doses in the range of 0.01 to 1 mg per kilogram of patient body weight may be utilized for a radionuclide therapeutic composition which is administered intrathecally. Relatively large doses, in the range of 0.1 to 10 mg per kilogram of patient body weight, may used for imaging conjugates with a relatively non-toxic imaging moiety. The amount utilized will depend on the sensitivity of the imaging method, and the relative toxicity of the imaging moiety. In a therapeutic example, where the therapeutic composition comprises a [0271] 131I cytotoxic moiety, the dosage to the patient will typically start at a lower range of 10 mCi, and go up to 100, 300 or even 500 mCi. Stated otherwise, where the therapeutic agent is 131I, the dosage to the patient will typically be from 5,000 Rads to 100,000 Rads (preferably at least 13,000 Rads, or even at least 50,000 Rads). Doses for other radionuclides are typically selected so that the tumoricidal dose will be equivalent to the foregoing range for 131I. Similarly, chemotoxic or toxin protein doses may be scaled accordingly.
  • The antibody conjugate can be administered to the subject in a series of more than one administration. For therapeutic compositions, regular periodic administration (e.g., every 2-3 days) will sometimes be required, or may be desirable to reduce toxicity. For therapeutic compositions which will be utilized in repeated-dose regimens, antibody moieties which do not provoke HAMA or other immune responses are preferred. The imaging antibody conjugate compositions may be administered at an appropriate time before the visualization technique. For example, administration within an hour before direct visual inspection may be appropriate, or administration within twelve hours before an MRI scan may be appropriate. Care should be taken, however, to not allow too much time to pass between administration and visualization, as the imaging compound may eventually be cleared from the patient's system. [0272]
  • In addition to the use of imaging antibody conjugates for simple visualization, these compositions may be utilized as a “dry run” for more toxic cytotoxic antibody conjugates. If the same antibody moiety is utilized for the imaging conjugate as for the therapeutic conjugate, the physician may first use a visualization technique to determine precisely where in the brain the cytotoxic conjugate will concentrate. If a sufficient degree of tissue selectivity is not achieved (e.g, if the tumor cells are too disperse in the normal tissue, or if the particular brain tumor protein target chosen is not sufficiently overexpressed in the particular patient's tumor cells), then the physician may choose another brain tumor protein target. The provision of numerous brain tumor protein targets by the present invention, along with both imaging and therapeutic agents, allows a high degree of flexibility in designing an effective treatment regimen for the individual patient. [0273]
  • Combination Therapies of the Invention [0274]
  • As mentioned previously, brain tumors tend to be heterogeneous in character, and pervasive throughout the brain tissue. This combination often makes them difficult to treat, as individual portions of the tumor cells in any particular patient may have differing biological characteristic. Thus, in some cases, it may be preferred to use various combinations of therapeutic or imaging agents, as described above in the Summary of Invention, in order to more fully target all of the cells exhibiting tumorigenic characteristics. Such combination treatments may be by administering blended antibody therapeutic or imaging compositions, individually prepared as described above, and administering the blended therapeutic to the patient as described. The skilled administering physician will be able to take such factors as combined toxicity, and individual antibody agent efficacy, into account when administering such combined agents. Additionally, those of skill in the art will be able to screen the antibodies to avoid potential cross-reaction with each other, in order to assure full efficacy of each antibody therapeutic or imaging agent. [0275]
  • Alternatively, several individual brain tumor protein target compositions may be administered simultaneously or in succession for a combined therapy. This may be desirable to avoid accumulated toxicity from several antibody conjugate reagents, or to more closely monitor potential adverse reactions to the individual antibody reagents. Thus, cycles such as where a first antibody therapeutic agent is administered on day one, followed by a second on day two, then a period with out administration, followed by re-administration of the antibody therapeutics on different successive days, is comprehended within the present invention. [0276]
  • EXAMPLES Example 1 Identification of Two New Splicing Variant Isoforms of PTPζ: PTPζ SM1 and SM2
  • The mRNA nucleotide sequence for PTPζ SM1 was identified in a human fetal brain phage cDNA library by sequencing. [0277]
  • The mRNA nucleotide sequence for PTPζ SM2 was identified by PCR amplification of adult human brain cDNA, and sequencing of the resulting nucleic acids. [0278]
  • For the RT-PCR analyses performed below, total RNA was isolated from either cells (glioblastoma cultured lines) or tissue using Trizole (Gibco Life Technologies, Inc.), following the manufacture's protocol. cDNA was generated from total RNA using the 1[0279] st Strand synthesis kit from Gibco Life Technologies, Inc., and an oligo dT30 anchored primer. For each RT-PCR reaction, 1 μl of cDNA was utilized. The PCR reaction was carried out using an Advantage 2 kit (Clontech) under standard conditions. The products of the PCR reactions were confirmed via sequencing.
  • Both clones were verified by RT-PCR analysis of glioblastoma cell lines and primary tumors. For PTPζ SM1, primers CAGCAGTTGGATGGAAGAGGAC [SEQ ID NO. 28] and CACTGAGATTCTGGCACTATTC [SEQ ID NO. 29] were used, producing an identifiable 1116 bp product. RT-PCR analysis was performed, confirming expression of the SM1 splice variant in 11 of 17 different glioblastoma cell lines tested, fetal brain, and adult brain, using the unique 3 end and portion of the 3′ untranslated region as the hybridization target for the probe. In addition, RT-PCR analysis was performed on 28 primary brain tumor samples, confirming expression of the PTPζ SM1 variant in 16 of the 28 tumors. [0280]
  • For PTPζ SM2, primers AACAATTCCAGGGTCTCACTC [SEQ ID NO. 30] and TTGACTGGCTCAGGAGTATAG [SEQ ID NO. 31] were used, which produce a 130 bp product when the extra exon 23a is present, and a no product when the exon 23a is absent. RT-PCR analysis was performed, confirming expression in 6 of 17 different glioblastoma cell lines tested. In addition, RT-PCR analysis was performed on 28 primary brain tumor samples, confirming expression of the PTPζ SM1 variant in 19 of the 28 tumors. [0281]
  • For comparison, RT-PCR analysis was also done for the expression of PTPζ-α (primers CTGATAATGAGGGCTCCCAAC [SEQ ID NO. 32] and CTCTGCACTTCCTGGTAAAACTCT [SEQ ID NO. 33]) and PTPζ-β (primers CAGCAGTTGGATGGAAGAGGAC [SEQ ID NO. 34] and CTCTGCACTTCCTGGTAAAACTCT [SEQ ID NO. 35]) in the 28 brain tumor tissue samples. PTPζ-α was shown to be expressed in 16 of the 28 samples, and the short form PTPζ-β was shown to be expressed in 19 of the 28 samples. [0282]
  • The nucleotide sequence alignment of the two new splice variants with the reference sequence for PTPζ-α is shown in the following table: [0283]
    TABLE 2
    PTP 5′ PTP 3′ PAC 1 5′ PAC 1 3′ Corresponding Exon Key:
    1 48 87274 87321 5′ UTR PAC 1: RPS-1062J16
    BAC: RP11-384A20
    70 205 87343 87487  1 PAC 2: RPS-1049N15
    205 272 142076 142143  2
    BAC 5′ BAC 3′
    291 451 24001 24161 *  3 *88 nt deletion seen in 5′ PCR
    clone from PTP 363-451
    450 603 28570 28723  4
    602 701 32814 32888  5
    698 772 32814 32888  6
    766 924 39695 39853  7
    922 1075 39995 40148  8
    1074 1261 52411 52598 *  9 *not spliced at 1261 in phage
    library clones
    1260 1387 53910 54037 10
    1387 1435 60644 60692 11
    1432 2346 66362 67276  5′ 12(end of BAC)
    PAC 2 5′ PAC 2 3′
    2147 4409 1 2263 mid 12
    4437 4987 2294 2844  3′ 12
    4925 5133 8027 8224 13
    5131 5224 17505 17598 14
    5223 5310 20427 20514 15
    5309 5332 23048 23071 16
    5329 5428 23234 23333 17
    5429 5512 25555 25638 18
    5512 5646 27710 27844 19
    5572 5602 42925 42955 * Duplicate of mid 19 *duplicated regions of exons 19
    5646 5768 28408 28530 most of 20 (−12 bp 3′) and 26 vary by one aa/two nt
    5791 5945 29770 29934 21 (−10 bp 5′)
    5943 6082 31560 31699 22
    6080 6228 33375 33523 ˜ 23 ˜116 nt insert seen b/w exons
    23 & 24 in 3′ PCR clone:
    maps to PAC b/w 23 & 24
    6225 6322 40379 40476 ˜ 24 PTP location PAC 2 5′ PAC 2 3′
    6322 6397 40820 40895 25 6228 36744 36629
    6396 6526 42864 42994 26
    6457 6487 27770 27800 * Duplicate of mid 26
    6525 6673 43895 44043 27
    6671 6816 47753 47898 28
    6816 6952 48708 48844 29
  • Example 2 Cell Migration Assay For Determining Antibody Activity on Protein Targets
  • Tumor cells are known to migrate more rapidly towards chemoattractants. The cell migration assay measures the ability of a cell to migrate. The ability to migrate is taken as a measure of tumorigenicity. Chemoattractants generally used are fetal bovine serum, pleiotrophin, bFGF, and VEGF. Thus, this assay can be used to determine migration capability of a cell in which the gene has been knocked down or the gene of interest is being overexpressed. [0284]
  • The ChemoTx® disposable chemotaxis system (Neuroprobe, Inc., Gaithersburg, Md.) is used according to the manufacturer's instructions, with a few modifications. Briefly, glioblastoma cultured cells from cell line G55T2 are prepared by splitting the cells the day before the assay is performed. A ChemoTx® chamber with the following specifications is used: [0285] Pore size 8 μm, exposed filter area 8 mm, exposed filter area diameter 3.2 mm2. The plate configuration is: 30 μl per well, 96 well plate. The membrane type is: Track-etched polycarbonate.
  • In preparation for the assays, the filter membrane is coated in 100 ml PBS containing 0.1% acetic acid and 3.5 ml Vitrogen 100 (from Cohesion) at 37° C. overnight. About 30 minutes before starting the assay the coated membrane is washed and rinsed with PBS containing 0.1% BSA. Cells are harvested by using the standard technique (trypsin-EDTA). The cells are washed once with DMEM 10% FBS, and then spun at 1000 RPM, for 5 minutes at room temperature. The pellet is resuspended in DMEM without serum, containing 0.1% BSA (serum free medium). The cells are spun and resuspended again in serum free medium, and then spun and resuspended in the amount of serum free medium needed to provide a concentration of 1 mio. cells/ml, or 25,000 cells per 25 ul. Just prior to the assay, a suitable amount of the antibody to be tested for anti-target function activity is added to the cell suspension. [0286]
  • For the assay, a standard chemoattractant is used to measure the mobility of the cells. The chemoattractants are diluted in serum free medium. A suitable unspecific chemoattractant is DMEM with 5% FBS. The chemoattractant solutions and control solutions without chemoattractant are pipetted (29 μl) into the lower plate wells. After placing and securing the filter plate over the lower wells, ensuring contact with the solution in the bottom wells, serial dilutions of the cell suspension are pipetted onto each site on the filter top. The plates are them covered and incubated at 37° C., 5% CO[0287] 2, for 3-4 hours.
  • After incubation, the upper filter side is rinsed with PBS and exposed upper filter areas are cleaned with wet cotton swabs. The filter is stained using Diff-Quik™ (VWR) dye kit, according to the manufacturer's instructions. The migrated cells are counted on the lower filter side using a microscope (Magnification 200×), by counting of 5 high power field sections per well. [0288]
  • Example 2 HUVEC(Human Umbilical Vein Endothelial Cells) Endothelial Sprouting Assay For Determining Antibody Activity on Protein Targets
  • Cell-sprouting morphology can be utilized as an easily visualized assay to determine the inhibitory effect of a candidate antibody on the protein target function for protein targets which stimulate endothelial cell sprouting, such as ARP2. Such assays have been described extensively in the literature (Nehls, V., et al., [0289] Histochem. Cell Biol. 104: 459-466 (1995); Koblizek, T. I., et al., Curr. Biol. 8: 529-532 (1988); and Kwak, H. J., et al., FEBS Lett. 448: 249-253). Briefly, a endothelial cells from a suitable source, such as HUVECs or PPAECs (porcine pulmonary artery endothelial cells) are grown to confluence on microcarrier (MC) beads (diameter 175 μm, available from Sigma) and placed into a 2.5 mg/ml fibrinogen gel containing the protein target at an appropriate effective concentration (200 ng/ml is an suitable starting concentration, which the skilled practitioner may optimize) and the antibody in an appropriate range of concentrations (this will depend on antibody titer and affinity for the target), and 200 units/ml Trasylol (available from Bayer). Fibrin gels are incubated in M-199 with a daily supplement of the same amount of recombinant protein and antibody, 2.0% heat-inactivated fetal bovine serum, and 200 units/ml Trasylol. After three days, the extent of sprouting is determined using a phase-contrast microscope (such as those available from Zeiss). A decrease in cell sprouting as compared to controls without antibody indicates a reduction in protein target activity by the antibody.
  • The foregoing is intended to be illustrative of the embodiments of the present invention, and are not intended to limit the invention in any way. Although the invention has been described with respect to specific modifications, the details thereof are not to be construed as limitations, for it will be apparent that various equivalents, changes and modifications may be resorted to without departing from the spirit and scope thereof and it is understood that such equivalent embodiments are to be included herein. All publications and patent applications are herein incorporated by reference to the same extent as if each individual publication or patent application was specifically and individually indicated to be incorporated by reference. [0290]
  • 1 35 1 3091 DNA Homo sapiens gene (1)..(3091) PTP-zeta SM1 exon 9 variant 1 cacacatacg cacgcacgat ctcacttcga tctatacact ggaggattaa aacaaacaaa 60 caaaaaaaac atttccttcg ctccccctcc ctctccactc tgagaagcag aggagccgca 120 cggcgagggg ccgcagaccg tctggaa atg cga atc cta aag cgt ttc ctc gct 174 Met Arg Ile Leu Lys Arg Phe Leu Ala 1 5 tgc att cag ctc ctc tgt gtt tgc cgc ctg gat tgg gct aat gga tac 222 Cys Ile Gln Leu Leu Cys Val Cys Arg Leu Asp Trp Ala Asn Gly Tyr 10 15 20 25 tac aga caa cag aga aaa ctt gtt gaa gag att ggc tgg tcc tat aca 270 Tyr Arg Gln Gln Arg Lys Leu Val Glu Glu Ile Gly Trp Ser Tyr Thr 30 35 40 gga gca ctg aat caa aaa aat tgg gga aag aaa tat cca aca tgt aat 318 Gly Ala Leu Asn Gln Lys Asn Trp Gly Lys Lys Tyr Pro Thr Cys Asn 45 50 55 agc cca aaa caa tct cct atc aat att gat gaa gat ctt aca caa gta 366 Ser Pro Lys Gln Ser Pro Ile Asn Ile Asp Glu Asp Leu Thr Gln Val 60 65 70 aat gtg aat ctt aag aaa ctt aaa ttt cag ggt tgg gat aaa aca tca 414 Asn Val Asn Leu Lys Lys Leu Lys Phe Gln Gly Trp Asp Lys Thr Ser 75 80 85 ttg gaa aac aca ttc att cat aac act ggg aaa aca gtg gaa att aat 462 Leu Glu Asn Thr Phe Ile His Asn Thr Gly Lys Thr Val Glu Ile Asn 90 95 100 105 ctc act aat gac tac cgt gtc agc gga gga gtt tca gaa atg gtg ttt 510 Leu Thr Asn Asp Tyr Arg Val Ser Gly Gly Val Ser Glu Met Val Phe 110 115 120 aaa gca agc aag ata act ttt cac tgg gga aaa tgc aat atg tca tct 558 Lys Ala Ser Lys Ile Thr Phe His Trp Gly Lys Cys Asn Met Ser Ser 125 130 135 gat gga tca gag cat agt tta gaa gga caa aaa ttt cca ctt gag atg 606 Asp Gly Ser Glu His Ser Leu Glu Gly Gln Lys Phe Pro Leu Glu Met 140 145 150 caa atc tac tgc ttt gat gcg gac cga ttt tca agt ttt gag gaa gca 654 Gln Ile Tyr Cys Phe Asp Ala Asp Arg Phe Ser Ser Phe Glu Glu Ala 155 160 165 gtc aaa gga aaa ggg aag tta aga gct tta tcc att ttg ttt gag gtt 702 Val Lys Gly Lys Gly Lys Leu Arg Ala Leu Ser Ile Leu Phe Glu Val 170 175 180 185 ggg aca gaa gaa aat ttg gat ttc aaa gcg att att gat gga gtc gaa 750 Gly Thr Glu Glu Asn Leu Asp Phe Lys Ala Ile Ile Asp Gly Val Glu 190 195 200 agt gtt agt cgt ttt ggg aag cag gct gct tta gat cca ttc ata ctg 798 Ser Val Ser Arg Phe Gly Lys Gln Ala Ala Leu Asp Pro Phe Ile Leu 205 210 215 ttg aac ctt ctg cca aac tca act gac aag tat tac att tac aat ggc 846 Leu Asn Leu Leu Pro Asn Ser Thr Asp Lys Tyr Tyr Ile Tyr Asn Gly 220 225 230 tca ttg aca tct cct ccc tgc aca gac aca gtt gac tgg att gtt ttt 894 Ser Leu Thr Ser Pro Pro Cys Thr Asp Thr Val Asp Trp Ile Val Phe 235 240 245 aaa gat aca gtt agc atc tct gaa agc cag ttg gct gtt ttt tgt gaa 942 Lys Asp Thr Val Ser Ile Ser Glu Ser Gln Leu Ala Val Phe Cys Glu 250 255 260 265 gtt ctt aca atg caa caa tct ggt tat gtc atg ctg atg gac tac tta 990 Val Leu Thr Met Gln Gln Ser Gly Tyr Val Met Leu Met Asp Tyr Leu 270 275 280 caa aac aat ttt cga gag caa cag tac aag ttc tct aga cag gtg ttt 1038 Gln Asn Asn Phe Arg Glu Gln Gln Tyr Lys Phe Ser Arg Gln Val Phe 285 290 295 tcc tca tac act gga aag gaa gag att cat gaa gca gtt tgt agt tca 1086 Ser Ser Tyr Thr Gly Lys Glu Glu Ile His Glu Ala Val Cys Ser Ser 300 305 310 gaa cca gaa aat gtt cag gct gac cca gag aat tat acc agc ctt ctt 1134 Glu Pro Glu Asn Val Gln Ala Asp Pro Glu Asn Tyr Thr Ser Leu Leu 315 320 325 gtt aca tgg gaa aga cct cga gtc gtt tat gat acc atg att gag aag 1182 Val Thr Trp Glu Arg Pro Arg Val Val Tyr Asp Thr Met Ile Glu Lys 330 335 340 345 ttt gca gtt ttg tac cag cag ttg gat gga gag gac caa acc aag cat 1230 Phe Ala Val Leu Tyr Gln Gln Leu Asp Gly Glu Asp Gln Thr Lys His 350 355 360 gaa ttt ttg aca gat ggc tat caa gac ttg gta act ata tga 1272 Glu Phe Leu Thr Asp Gly Tyr Gln Asp Leu Val Thr Ile 365 370 tcagttgttt tacatagggt aacattataa tttaatttcc aaggtaagaa cttacaaatg 1332 gttgtatatt attttcctcc attactttta gactttatgt gaaggtgggg taggctgagt 1392 atttttaaat ttaaaaaaaa attttaaatt agaagctata ctaaattatg tttaaagtta 1452 catttaatta aatggatatc ataactttgc caacaataac actatagagt agatacatat 1512 gacttatgaa ctggagatca tttagtgtgg cctttcttaa gatttcagtt gtagaatagt 1572 gccagaatct cagtgccctg atacatttta tattgtgtct tccattacgc tatatcagca 1632 caggaaaagt agagtagggg acatacaagt cctctttgtt gcaccaaaaa attttcagat 1692 aacagctggg aagtcatgat tgggtcagaa ctttggggat gtaagaaaac atttcttaca 1752 aaaagatcca cccctgcctc cctccaccag cgcatgcgaa taaagtacag attccctttg 1812 tggcctgagc atgtcagtat taaactttgc tctggtaggg aagtgttggc catagattag 1872 ggtgtagttg acaaaccttc atctggatgt aggtccagaa agtccccact gcaggttaaa 1932 ggacactgga ctctgcactc aggcacctag agtcctgcaa gtcctgggaa cctgcattta 1992 aataaaaatg cactattaat tatgtttcat atcatgtgga caaaatggat aaaattttag 2052 taacctttta attcagttgc ctggaatatg gagacacaat gacctgggaa aatcgtgaaa 2112 taaatagtaa taaaaatgtt tatttcataa ttacgtgaag aagataattc tattactgtt 2172 cttgcatata tattgtcaag aaaaagagat aacttagttg ttcacttttt cacattgctc 2232 cttgtttgca aatgcccccc atttatttgt ctaaaatatt aatttttagt ttgtagtact 2292 aatttatgaa tttgatgagt tctggctaaa aatgaaactt cctgaaacta aatctgattt 2352 ttaaaaagca aaaaaaaaaa aaaagcctag ctttccagtt cttcataatt cacaaatacc 2412 acaagtttaa ctaagcaaca ttgcataaac ttttccttag gttaataaaa tagaagtatt 2472 ttccacggac cagggagaaa aagttttcta ggaaagatac ctagtgtgtt ggtagtccta 2532 tgagaataac atttgtataa ttactaacat ctttctttta gggtgctatt ctcaataatt 2592 tgctacccaa tatgagttat gttcttcaga tagtagccat atgcactaat ggcttatatg 2652 gaaaatacag cgaccaactg attgtcgaca tgcctactga taatcctggt aagtgccacc 2712 agatacatct atatattaac tcaataaatg aggttagttt aattactgta tgcattgatg 2772 ctttctctct atattctttt ggccaaaagg caaagtgatt ttctcttaag tctggattgc 2832 cgggtaattt tttggggcat gggacccatt tctcattcag caggtctggt gccagacaat 2892 aagtaaactt atccttaata ttggagttta ccatttgtaa aataagagtg actaaacata 2952 tttataacat tgtaataatc attaaatgaa aattgctatg taaatgttga gactgttatt 3012 ttggataatt aagagttggt ttaatttgta tttatttcct cttttcagcc cccaaagcat 3072 tatgtagtaa gtgtataca 3091 2 374 PRT Homo sapiens gene (1)..(374) PTP-zeta SM1 exon 9 variant 2 Met Arg Ile Leu Lys Arg Phe Leu Ala Cys Ile Gln Leu Leu Cys Val 1 5 10 15 Cys Arg Leu Asp Trp Ala Asn Gly Tyr Tyr Arg Gln Gln Arg Lys Leu 20 25 30 Val Glu Glu Ile Gly Trp Ser Tyr Thr Gly Ala Leu Asn Gln Lys Asn 35 40 45 Trp Gly Lys Lys Tyr Pro Thr Cys Asn Ser Pro Lys Gln Ser Pro Ile 50 55 60 Asn Ile Asp Glu Asp Leu Thr Gln Val Asn Val Asn Leu Lys Lys Leu 65 70 75 80 Lys Phe Gln Gly Trp Asp Lys Thr Ser Leu Glu Asn Thr Phe Ile His 85 90 95 Asn Thr Gly Lys Thr Val Glu Ile Asn Leu Thr Asn Asp Tyr Arg Val 100 105 110 Ser Gly Gly Val Ser Glu Met Val Phe Lys Ala Ser Lys Ile Thr Phe 115 120 125 His Trp Gly Lys Cys Asn Met Ser Ser Asp Gly Ser Glu His Ser Leu 130 135 140 Glu Gly Gln Lys Phe Pro Leu Glu Met Gln Ile Tyr Cys Phe Asp Ala 145 150 155 160 Asp Arg Phe Ser Ser Phe Glu Glu Ala Val Lys Gly Lys Gly Lys Leu 165 170 175 Arg Ala Leu Ser Ile Leu Phe Glu Val Gly Thr Glu Glu Asn Leu Asp 180 185 190 Phe Lys Ala Ile Ile Asp Gly Val Glu Ser Val Ser Arg Phe Gly Lys 195 200 205 Gln Ala Ala Leu Asp Pro Phe Ile Leu Leu Asn Leu Leu Pro Asn Ser 210 215 220 Thr Asp Lys Tyr Tyr Ile Tyr Asn Gly Ser Leu Thr Ser Pro Pro Cys 225 230 235 240 Thr Asp Thr Val Asp Trp Ile Val Phe Lys Asp Thr Val Ser Ile Ser 245 250 255 Glu Ser Gln Leu Ala Val Phe Cys Glu Val Leu Thr Met Gln Gln Ser 260 265 270 Gly Tyr Val Met Leu Met Asp Tyr Leu Gln Asn Asn Phe Arg Glu Gln 275 280 285 Gln Tyr Lys Phe Ser Arg Gln Val Phe Ser Ser Tyr Thr Gly Lys Glu 290 295 300 Glu Ile His Glu Ala Val Cys Ser Ser Glu Pro Glu Asn Val Gln Ala 305 310 315 320 Asp Pro Glu Asn Tyr Thr Ser Leu Leu Val Thr Trp Glu Arg Pro Arg 325 330 335 Val Val Tyr Asp Thr Met Ile Glu Lys Phe Ala Val Leu Tyr Gln Gln 340 345 350 Leu Asp Gly Glu Asp Gln Thr Lys His Glu Phe Leu Thr Asp Gly Tyr 355 360 365 Gln Asp Leu Val Thr Ile 370 3 8058 DNA Homo sapiens gene (1)..(8058) PTP-zeta SM2 exon 23a variant 3 cacacatacg cacgcacgat ctcacttcga tctatacact ggaggattaa aacaaacaaa 60 caaaaaaaac atttccttcg ctccccctcc ctctccactc tgagaagcag aggagccgca 120 cggcgagggg ccgcagaccg tctggaaatg cgaatcctaa agcgtttcct cgcttgcatt 180 cagctcctct gtgtttgccg cctggattgg gctaatggat actacagaca acagagaaaa 240 cttgttgaag agattggctg gtcctataca ggagcactga atcaaaaaaa ttggggaaag 300 aaatatccaa catgtaatag cccaaaacaa tctcctatca atattgatga agatcttaca 360 caagtaaatg tgaatcttaa gaaacttaaa tttcagggtt gggataaaac atcattggaa 420 aacacattca ttcataacac tgggaaaaca gtggaaatta atctcactaa tgactaccgt 480 gtcagcggag gagtttcaga aatggtgttt aaagcaagca agataacttt tcactgggga 540 aaatgcaata tgtcatctga tggatcagag catagtttag aaggacaaaa atttccactt 600 gagatgcaaa tctactgctt tgatgcggac cgattttcaa gttttgagga agcagtcaaa 660 ggaaaaggga agttaagagc tttatccatt ttgtttgagg ttgggacaga agaaaatttg 720 gatttcaaag cgattattga tggagtcgaa agtgttagtc gttttgggaa gcaggctgct 780 ttagatccat tcatactgtt gaaccttctg ccaaactcaa ctgacaagta ttacatttac 840 aatggctcat tgacatctcc tccctgcaca gacacagttg actggattgt ttttaaagat 900 acagttagca tctctgaaag ccagttggct gttttttgtg aagttcttac aatgcaacaa 960 tctggttatg tcatgctgat ggactactta caaaacaatt ttcgagagca acagtacaag 1020 ttctctagac aggtgttttc ctcatacact ggaaaggaag agattcatga agcagtttgt 1080 agttcagaac cagaaaatgt tcaggctgac ccagagaatt ataccagcct tcttgttaca 1140 tgggaaagac ctcgagtcgt ttatgatacc atgattgaga agtttgcagt tttgtaccag 1200 cagttggatg gagaggacca aaccaagcat gaatttttga cagatggcta tcaagacttg 1260 ggtgctattc tcaataattt gctacccaat atgagttatg ttcttcagat agtagccata 1320 tgcactaatg gcttatatgg aaaatacagc gaccaactga ttgtcgacat gcctactgat 1380 aatcctgaac ttgatctttt ccctgaatta attggaactg aagaaataat caaggaggag 1440 gaagagggaa aagacattga agaaggcgct attgtgaatc ctggtagaga cagtgctaca 1500 aaccaaatca ggaaaaagga accccagatt tctaccacaa cacactacaa tcgcataggg 1560 acgaaataca atgaagccaa gactaaccga tccccaacaa gaggaagtga attctctgga 1620 aagggtgatg ttcccaatac atctttaaat tccacttccc aaccagtcac taaattagcc 1680 acagaaaaag atatttcctt gacttctcag actgtgactg aactgccacc tcacactgtg 1740 gaaggtactt cagcctcttt aaatgatggc tctaaaactg ttcttagatc tccacatatg 1800 aacttgtcgg ggactgcaga atccttaaat acagtttcta taacagaata tgaggaggag 1860 agtttattga ccagtttcaa gcttgatact ggagctgaag attcttcagg ctccagtccc 1920 gcaacttctg ctatcccatt catctctgag aacatatccc aagggtatat attttcctcc 1980 gaaaacccag agacaataac atatgatgtc cttataccag aatctgctag aaatgcttcc 2040 gaagattcaa cttcatcagg ttcagaagaa tcactaaagg atccttctat ggagggaaat 2100 gtgtggtttc ctagctctac agacataaca gcacagcccg atgttggatc aggcagagag 2160 agctttctcc agactaatta cactgagata cgtgttgatg aatctgagaa gacaaccaag 2220 tccttttctg caggcccagt gatgtcacag ggtccctcag ttacagatct ggaaatgcca 2280 cattattcta cctttgccta cttcccaact gaggtaacac ctcatgcttt taccccatcc 2340 tccagacaac aggatttggt ctccacggtc aacgtggtat actcgcagac aacccaaccg 2400 gtatacaatg gtgagacacc tcttcaacct tcctacagta gtgaagtctt tcctctagtc 2460 acccctttgt tgcttgacaa tcagatcctc aacactaccc ctgctgcttc aagtagtgat 2520 tcggccttgc atgctacgcc tgtatttccc agtgtcgatg tgtcatttga atccatcctg 2580 tcttcctatg atggtgcacc tttgcttcca ttttcctctg cttccttcag tagtgaattg 2640 tttcgccatc tgcatacagt ttctcaaatc cttccacaag ttacttcagc taccgagagt 2700 gataaggtgc ccttgcatgc ttctctgcca gtggctgggg gtgatttgct attagagccc 2760 agccttgctc agtattctga tgtgctgtcc actactcatg ctgcttcaga gacgctggaa 2820 tttggtagtg aatctggtgt tctttataaa acgcttatgt tttctcaagt tgaaccaccc 2880 agcagtgatg ccatgatgca tgcacgttct tcagggcctg aaccttctta tgccttgtct 2940 gataatgagg gctcccaaca catcttcact gtttcttaca gttctgcaat acctgtgcat 3000 gattctgtgg gtgtaactta tcagggttcc ttatttagcg gccctagcca tataccaata 3060 cctaagtctt cgttaataac cccaactgca tcattactgc agcctactca tgccctctct 3120 ggtgatgggg aatggtctgg agcctcttct gatagtgaat ttcttttacc tgacacagat 3180 gggctgacag cccttaacat ttcttcacct gtttctgtag ctgaatttac atatacaaca 3240 tctgtgtttg gtgatgataa taaggcgctt tctaaaagtg aaataatata tggaaatgag 3300 actgaactgc aaattccttc tttcaatgag atggtttacc cttctgaaag cacagtcatg 3360 cccaacatgt atgataatgt aaataagttg aatgcgtctt tacaagaaac ctctgtttcc 3420 atttctagca ccaagggcat gtttccaggg tcccttgctc ataccaccac taaggttttt 3480 gatcatgaga ttagtcaagt tccagaaaat aacttttcag ttcaacctac acatactgtc 3540 tctcaagcat ctggtgacac ttcgcttaaa cctgtgctta gtgcaaactc agagccagca 3600 tcctctgacc ctgcttctag tgaaatgtta tctccttcaa ctcagctctt attttatgag 3660 acctcagctt cttttagtac tgaagtattg ctacaacctt cctttcaggc ttctgatgtt 3720 gacaccttgc ttaaaactgt tcttccagct gtgcccagtg atccaatatt ggttgaaacc 3780 cccaaagttg ataaaattag ttctacaatg ttgcatctca ttgtatcaaa ttctgcttca 3840 agtgaaaaca tgctgcactc tacatctgta ccagtttttg atgtgtcgcc tacttctcat 3900 atgcactctg cttcacttca aggtttgacc atttcctatg caagtgagaa atatgaacca 3960 gttttgttaa aaagtgaaag ttcccaccaa gtggtacctt ctttgtacag taatgatgag 4020 ttgttccaaa cggccaattt ggagattaac caggcccatc ccccaaaagg aaggcatgta 4080 tttgctacac ctgttttatc aattgatgaa ccattaaata cactaataaa taagcttata 4140 cattccgatg aaattttaac ctccaccaaa agttctgtta ctggtaaggt atttgctggt 4200 attccaacag ttgcttctga tacatttgta tctactgatc attctgttcc tataggaaat 4260 gggcatgttg ccattacagc tgtttctccc cacagagatg gttctgtaac ctcaacaaag 4320 ttgctgtttc cttctaaggc aacttctgag ctgagtcata gtgccaaatc tgatgccggt 4380 ttagtgggtg gtggtgaaga tggtgacact gatgatgatg gtgatgatga tgatgacaga 4440 gatagtgatg gcttatccat tcataagtgt atgtcatgct catcctatag agaatcacag 4500 gaaaaggtaa tgaatgattc agacacccac gaaaacagtc ttatggatca gaataatcca 4560 atctcatact cactatctga gaattctgaa gaagataata gagtcacaag tgtatcctca 4620 gacagtcaaa ctggtatgga cagaagtcct ggtaaatcac catcagcaaa tgggctatcc 4680 caaaagcaca atgatggaaa agaggaaaat gacattcaga ctggtagtgc tctgcttcct 4740 ctcagccctg aatctaaagc atgggcagtt ctgacaagtg atgaagaaag tggatcaggg 4800 caaggtacct cagatagcct taatgagaat gagacttcca cagatttcag ttttgcagac 4860 actaatgaaa aagatgctga tgggatcctg gcagcaggtg actcagaaat aactcctgga 4920 ttcccacagt ccccaacatc atctgttact agcgagaact cagaagtgtt ccacgtttca 4980 gaggcagagg ccagtaatag tagccatgag tctcgtattg gtctagctga ggggttggaa 5040 tccgagaaga aggcagttat accccttgtg atcgtgtcag ccctgacttt tatctgtcta 5100 gtggttcttg tgggtattct catctactgg aggaaatgct tccagactgc acacttttac 5160 ttagaggaca gtacatcccc tagagttata tccacacctc caacacctat ctttccaatt 5220 tcagatgatg tcggagcaat tccaataaag cactttccaa agcatgttgc agatttacat 5280 gcaagtagtg ggtttactga agaatttgag acactgaaag agttttacca ggaagtgcag 5340 agctgtactg ttgacttagg tattacagca gacagctcca accacccaga caacaagcac 5400 aagaatcgat acataaatat cgttgcctat gatcatagca gggttaagct agcacagctt 5460 gctgaaaagg atggcaaact gactgattat atcaatgcca attatgttga tggctacaac 5520 agaccaaaag cttatattgc tgcccaaggc ccactgaaat ccacagctga agatttctgg 5580 agaatgatat gggaacataa tgtggaagtt attgtcatga taacaaacct cgtggagaaa 5640 ggaaggagaa aatgtgatca gtactggcct gccgatggga gtgaggagta cgggaacttt 5700 ctggtcactc agaagagtgt gcaagtgctt gcctattata ctgtgaggaa ttttactcta 5760 agaaacacaa aaataaaaaa gggctcccag aaaggaagac ccagtggacg tgtggtcaca 5820 cagtatcact acacgcagtg gcctgacatg ggagtaccag agtactccct gccagtgctg 5880 acctttgtga gaaaggcagc ctatgccaag cgccatgcag tggggcctgt tgtcgtccac 5940 tgcagtgctg gagttggaag aacaggcaca tatattgtgc tagacagtat gttgcagcag 6000 attcaacacg aaggaactgt caacatattt ggcttcttaa aacacatccg ttcacaaaga 6060 aattatttgg tacaaactga ggagcaatat gtcttcattc atgatacact ggttgaggcc 6120 atacttagta aagaaactga ggtgctggac agtcatattc atgcctatgt taatgcactc 6180 ctcattcctg gaccagcagg caaaacaaag ctagagaaac aattccaggg tctcactctg 6240 tcacccaggc tggagtgcag aggcacaatc tcggctcact gcaaccttcc tctccctggc 6300 ttaactgatc ctcctacctc agcctcccga gtggctggga ctatactcct gagccagtca 6360 aatatacagc agagtgacta ttctgcagcc ctaaagcaat gcaacaggga aaagaatcga 6420 acttcttcta tcatccctgt ggaaagatca agggttggca tttcatccct gagtggagaa 6480 ggcacagact acatcaatgc ctcctatatc atgggctatt accagagcaa tgaattcatc 6540 attacccagc accctctcct tcataccatc aaggatttct ggaggatgat atgggaccat 6600 aatgcccaac tggtggttat gattcctgat ggccaaaaca tggcagaaga tgaatttgtt 6660 tactggccaa ataaagatga gcctataaat tgtgagagct ttaaggtcac tcttatggct 6720 gaagaacaca aatgtctatc taatgaggaa aaacttataa ttcaggactt tatcttagaa 6780 gctacacagg atgattatgt acttgaagtg aggcactttc agtgtcctaa atggccaaat 6840 ccagatagcc ccattagtaa aacttttgaa cttataagtg ttataaaaga agaagctgcc 6900 aatagggatg ggcctatgat tgttcatgat gagcatggag gagtgacggc aggaactttc 6960 tgtgctctga caacccttat gcaccaacta gaaaaagaaa attccgtgga tgtttaccag 7020 gtagccaaga tgatcaatct gatgaggcca ggagtctttg ctgacattga gcagtatcag 7080 tttctctaca aagtgatcct cagccttgtg agcacaaggc aggaagagaa tccatccacc 7140 tctctggaca gtaatggtgc agcattgcct gatggaaata tagctgagag cttagagtct 7200 ttagtttaac acagaaaggg gtggggggac tcacatctga gcattgtttt cctcttccta 7260 aaattaggca ggaaaatcag tctagttctg ttatctgttg atttcccatc acctgacagt 7320 aactttcatg acataggatt ctgccgccaa atttatatca ttaacaatgt gtgccttttt 7380 gcaagacttg taatttactt attatgtttg aactaaaatg attgaatttt acagtatttc 7440 taagaatgga attgtggtat ttttttctgt attgatttta acagaaaatt tcaatttata 7500 gaggttagga attccaaact acagaaaatg tttgttttta gtgtcaaatt tttagctgta 7560 tttgtagcaa ttatcaggtt tgctagaaat ataactttta atacagtagc ctgtaaataa 7620 aacactcttc catatgatat tcaacatttt acaactgcag tattcaccta aagtagaaat 7680 aatctgttac ttattgtaaa tactgcccta gtgtctccat ggaccaaatt tatatttata 7740 attgtagatt tttatatttt actactgagt caagttttct agttctgtgt aattgtttag 7800 tttaatgacg tagttcatta gctggtctta ctctaccagt tttctgacat tgtattgtgt 7860 tacctaagtc attaactttg tttcagcatg taattttaac ttttgtggaa aatagaaata 7920 ccttcatttt gaaagaagtt tttatgagaa taacacctta ccaaacattg ttcaaatggt 7980 ttttatccaa ggaattgcaa aaataaatat aaatattgcc attaaaaaaa aaaaaaaaaa 8040 aaaaaaaaaa aaaaaaaa 8058 4 2353 PRT Homo sapiens gene (1)..(2353) PTP-zeta SM2 23a exon variant 4 Met Arg Ile Leu Lys Arg Phe Leu Ala Cys Ile Gln Leu Leu Cys Val 1 5 10 15 Cys Arg Leu Asp Trp Ala Asn Gly Tyr Tyr Arg Gln Gln Arg Lys Leu 20 25 30 Val Glu Glu Ile Gly Trp Ser Tyr Thr Gly Ala Leu Asn Gln Lys Asn 35 40 45 Trp Gly Lys Lys Tyr Pro Thr Cys Asn Ser Pro Lys Gln Ser Pro Ile 50 55 60 Asn Ile Asp Glu Asp Leu Thr Gln Val Asn Val Asn Leu Lys Lys Leu 65 70 75 80 Lys Phe Gln Gly Trp Asp Lys Thr Ser Leu Glu Asn Thr Phe Ile His 85 90 95 Asn Thr Gly Lys Thr Val Glu Ile Asn Leu Thr Asn Asp Tyr Arg Val 100 105 110 Ser Gly Gly Val Ser Glu Met Val Phe Lys Ala Ser Lys Ile Thr Phe 115 120 125 His Trp Gly Lys Cys Asn Met Ser Ser Asp Gly Ser Glu His Ser Leu 130 135 140 Glu Gly Gln Lys Phe Pro Leu Glu Met Gln Ile Tyr Cys Phe Asp Ala 145 150 155 160 Asp Arg Phe Ser Ser Phe Glu Glu Ala Val Lys Gly Lys Gly Lys Leu 165 170 175 Arg Ala Leu Ser Ile Leu Phe Glu Val Gly Thr Glu Glu Asn Leu Asp 180 185 190 Phe Lys Ala Ile Ile Asp Gly Val Glu Ser Val Ser Arg Phe Gly Lys 195 200 205 Gln Ala Ala Leu Asp Pro Phe Ile Leu Leu Asn Leu Leu Pro Asn Ser 210 215 220 Thr Asp Lys Tyr Tyr Ile Tyr Asn Gly Ser Leu Thr Ser Pro Pro Cys 225 230 235 240 Thr Asp Thr Val Asp Trp Ile Val Phe Lys Asp Thr Val Ser Ile Ser 245 250 255 Glu Ser Gln Leu Ala Val Phe Cys Glu Val Leu Thr Met Gln Gln Ser 260 265 270 Gly Tyr Val Met Leu Met Asp Tyr Leu Gln Asn Asn Phe Arg Glu Gln 275 280 285 Gln Tyr Lys Phe Ser Arg Gln Val Phe Ser Ser Tyr Thr Gly Lys Glu 290 295 300 Glu Ile His Glu Ala Val Cys Ser Ser Glu Pro Glu Asn Val Gln Ala 305 310 315 320 Asp Pro Glu Asn Tyr Thr Ser Leu Leu Val Thr Trp Glu Arg Pro Arg 325 330 335 Val Val Tyr Asp Thr Met Ile Glu Lys Phe Ala Val Leu Tyr Gln Gln 340 345 350 Leu Asp Gly Glu Asp Gln Thr Lys His Glu Phe Leu Thr Asp Gly Tyr 355 360 365 Gln Asp Leu Gly Ala Ile Leu Asn Asn Leu Leu Pro Asn Met Ser Tyr 370 375 380 Val Leu Gln Ile Val Ala Ile Cys Thr Asn Gly Leu Tyr Gly Lys Tyr 385 390 395 400 Ser Asp Gln Leu Ile Val Asp Met Pro Thr Asp Asn Pro Glu Leu Asp 405 410 415 Leu Phe Pro Glu Leu Ile Gly Thr Glu Glu Ile Ile Lys Glu Glu Glu 420 425 430 Glu Gly Lys Asp Ile Glu Glu Gly Ala Ile Val Asn Pro Gly Arg Asp 435 440 445 Ser Ala Thr Asn Gln Ile Arg Lys Lys Glu Pro Gln Ile Ser Thr Thr 450 455 460 Thr His Tyr Asn Arg Ile Gly Thr Lys Tyr Asn Glu Ala Lys Thr Asn 465 470 475 480 Arg Ser Pro Thr Arg Gly Ser Glu Phe Ser Gly Lys Gly Asp Val Pro 485 490 495 Asn Thr Ser Leu Asn Ser Thr Ser Gln Pro Val Thr Lys Leu Ala Thr 500 505 510 Glu Lys Asp Ile Ser Leu Thr Ser Gln Thr Val Thr Glu Leu Pro Pro 515 520 525 His Thr Val Glu Gly Thr Ser Ala Ser Leu Asn Asp Gly Ser Lys Thr 530 535 540 Val Leu Arg Ser Pro His Met Asn Leu Ser Gly Thr Ala Glu Ser Leu 545 550 555 560 Asn Thr Val Ser Ile Thr Glu Tyr Glu Glu Glu Ser Leu Leu Thr Ser 565 570 575 Phe Lys Leu Asp Thr Gly Ala Glu Asp Ser Ser Gly Ser Ser Pro Ala 580 585 590 Thr Ser Ala Ile Pro Phe Ile Ser Glu Asn Ile Ser Gln Gly Tyr Ile 595 600 605 Phe Ser Ser Glu Asn Pro Glu Thr Ile Thr Tyr Asp Val Leu Ile Pro 610 615 620 Glu Ser Ala Arg Asn Ala Ser Glu Asp Ser Thr Ser Ser Gly Ser Glu 625 630 635 640 Glu Ser Leu Lys Asp Pro Ser Met Glu Gly Asn Val Trp Phe Pro Ser 645 650 655 Ser Thr Asp Ile Thr Ala Gln Pro Asp Val Gly Ser Gly Arg Glu Ser 660 665 670 Phe Leu Gln Thr Asn Tyr Thr Glu Ile Arg Val Asp Glu Ser Glu Lys 675 680 685 Thr Thr Lys Ser Phe Ser Ala Gly Pro Val Met Ser Gln Gly Pro Ser 690 695 700 Val Thr Asp Leu Glu Met Pro His Tyr Ser Thr Phe Ala Tyr Phe Pro 705 710 715 720 Thr Glu Val Thr Pro His Ala Phe Thr Pro Ser Ser Arg Gln Gln Asp 725 730 735 Leu Val Ser Thr Val Asn Val Val Tyr Ser Gln Thr Thr Gln Pro Val 740 745 750 Tyr Asn Gly Glu Thr Pro Leu Gln Pro Ser Tyr Ser Ser Glu Val Phe 755 760 765 Pro Leu Val Thr Pro Leu Leu Leu Asp Asn Gln Ile Leu Asn Thr Thr 770 775 780 Pro Ala Ala Ser Ser Ser Asp Ser Ala Leu His Ala Thr Pro Val Phe 785 790 795 800 Pro Ser Val Asp Val Ser Phe Glu Ser Ile Leu Ser Ser Tyr Asp Gly 805 810 815 Ala Pro Leu Leu Pro Phe Ser Ser Ala Ser Phe Ser Ser Glu Leu Phe 820 825 830 Arg His Leu His Thr Val Ser Gln Ile Leu Pro Gln Val Thr Ser Ala 835 840 845 Thr Glu Ser Asp Lys Val Pro Leu His Ala Ser Leu Pro Val Ala Gly 850 855 860 Gly Asp Leu Leu Leu Glu Pro Ser Leu Ala Gln Tyr Ser Asp Val Leu 865 870 875 880 Ser Thr Thr His Ala Ala Ser Glu Thr Leu Glu Phe Gly Ser Glu Ser 885 890 895 Gly Val Leu Tyr Lys Thr Leu Met Phe Ser Gln Val Glu Pro Pro Ser 900 905 910 Ser Asp Ala Met Met His Ala Arg Ser Ser Gly Pro Glu Pro Ser Tyr 915 920 925 Ala Leu Ser Asp Asn Glu Gly Ser Gln His Ile Phe Thr Val Ser Tyr 930 935 940 Ser Ser Ala Ile Pro Val His Asp Ser Val Gly Val Thr Tyr Gln Gly 945 950 955 960 Ser Leu Phe Ser Gly Pro Ser His Ile Pro Ile Pro Lys Ser Ser Leu 965 970 975 Ile Thr Pro Thr Ala Ser Leu Leu Gln Pro Thr His Ala Leu Ser Gly 980 985 990 Asp Gly Glu Trp Ser Gly Ala Ser Ser Asp Ser Glu Phe Leu Leu Pro 995 1000 1005 Asp Thr Asp Gly Leu Thr Ala Leu Asn Ile Ser Ser Pro Val Ser 1010 1015 1020 Val Ala Glu Phe Thr Tyr Thr Thr Ser Val Phe Gly Asp Asp Asn 1025 1030 1035 Lys Ala Leu Ser Lys Ser Glu Ile Ile Tyr Gly Asn Glu Thr Glu 1040 1045 1050 Leu Gln Ile Pro Ser Phe Asn Glu Met Val Tyr Pro Ser Glu Ser 1055 1060 1065 Thr Val Met Pro Asn Met Tyr Asp Asn Val Asn Lys Leu Asn Ala 1070 1075 1080 Ser Leu Gln Glu Thr Ser Val Ser Ile Ser Ser Thr Lys Gly Met 1085 1090 1095 Phe Pro Gly Ser Leu Ala His Thr Thr Thr Lys Val Phe Asp His 1100 1105 1110 Glu Ile Ser Gln Val Pro Glu Asn Asn Phe Ser Val Gln Pro Thr 1115 1120 1125 His Thr Val Ser Gln Ala Ser Gly Asp Thr Ser Leu Lys Pro Val 1130 1135 1140 Leu Ser Ala Asn Ser Glu Pro Ala Ser Ser Asp Pro Ala Ser Ser 1145 1150 1155 Glu Met Leu Ser Pro Ser Thr Gln Leu Leu Phe Tyr Glu Thr Ser 1160 1165 1170 Ala Ser Phe Ser Thr Glu Val Leu Leu Gln Pro Ser Phe Gln Ala 1175 1180 1185 Ser Asp Val Asp Thr Leu Leu Lys Thr Val Leu Pro Ala Val Pro 1190 1195 1200 Ser Asp Pro Ile Leu Val Glu Thr Pro Lys Val Asp Lys Ile Ser 1205 1210 1215 Ser Thr Met Leu His Leu Ile Val Ser Asn Ser Ala Ser Ser Glu 1220 1225 1230 Asn Met Leu His Ser Thr Ser Val Pro Val Phe Asp Val Ser Pro 1235 1240 1245 Thr Ser His Met His Ser Ala Ser Leu Gln Gly Leu Thr Ile Ser 1250 1255 1260 Tyr Ala Ser Glu Lys Tyr Glu Pro Val Leu Leu Lys Ser Glu Ser 1265 1270 1275 Ser His Gln Val Val Pro Ser Leu Tyr Ser Asn Asp Glu Leu Phe 1280 1285 1290 Gln Thr Ala Asn Leu Glu Ile Asn Gln Ala His Pro Pro Lys Gly 1295 1300 1305 Arg His Val Phe Ala Thr Pro Val Leu Ser Ile Asp Glu Pro Leu 1310 1315 1320 Asn Thr Leu Ile Asn Lys Leu Ile His Ser Asp Glu Ile Leu Thr 1325 1330 1335 Ser Thr Lys Ser Ser Val Thr Gly Lys Val Phe Ala Gly Ile Pro 1340 1345 1350 Thr Val Ala Ser Asp Thr Phe Val Ser Thr Asp His Ser Val Pro 1355 1360 1365 Ile Gly Asn Gly His Val Ala Ile Thr Ala Val Ser Pro His Arg 1370 1375 1380 Asp Gly Ser Val Thr Ser Thr Lys Leu Leu Phe Pro Ser Lys Ala 1385 1390 1395 Thr Ser Glu Leu Ser His Ser Ala Lys Ser Asp Ala Gly Leu Val 1400 1405 1410 Gly Gly Gly Glu Asp Gly Asp Thr Asp Asp Asp Gly Asp Asp Asp 1415 1420 1425 Asp Asp Arg Asp Ser Asp Gly Leu Ser Ile His Lys Cys Met Ser 1430 1435 1440 Cys Ser Ser Tyr Arg Glu Ser Gln Glu Lys Val Met Asn Asp Ser 1445 1450 1455 Asp Thr His Glu Asn Ser Leu Met Asp Gln Asn Asn Pro Ile Ser 1460 1465 1470 Tyr Ser Leu Ser Glu Asn Ser Glu Glu Asp Asn Arg Val Thr Ser 1475 1480 1485 Val Ser Ser Asp Ser Gln Thr Gly Met Asp Arg Ser Pro Gly Lys 1490 1495 1500 Ser Pro Ser Ala Asn Gly Leu Ser Gln Lys His Asn Asp Gly Lys 1505 1510 1515 Glu Glu Asn Asp Ile Gln Thr Gly Ser Ala Leu Leu Pro Leu Ser 1520 1525 1530 Pro Glu Ser Lys Ala Trp Ala Val Leu Thr Ser Asp Glu Glu Ser 1535 1540 1545 Gly Ser Gly Gln Gly Thr Ser Asp Ser Leu Asn Glu Asn Glu Thr 1550 1555 1560 Ser Thr Asp Phe Ser Phe Ala Asp Thr Asn Glu Lys Asp Ala Asp 1565 1570 1575 Gly Ile Leu Ala Ala Gly Asp Ser Glu Ile Thr Pro Gly Phe Pro 1580 1585 1590 Gln Ser Pro Thr Ser Ser Val Thr Ser Glu Asn Ser Glu Val Phe 1595 1600 1605 His Val Ser Glu Ala Glu Ala Ser Asn Ser Ser His Glu Ser Arg 1610 1615 1620 Ile Gly Leu Ala Glu Gly Leu Glu Ser Glu Lys Lys Ala Val Ile 1625 1630 1635 Pro Leu Val Ile Val Ser Ala Leu Thr Phe Ile Cys Leu Val Val 1640 1645 1650 Leu Val Gly Ile Leu Ile Tyr Trp Arg Lys Cys Phe Gln Thr Ala 1655 1660 1665 His Phe Tyr Leu Glu Asp Ser Thr Ser Pro Arg Val Ile Ser Thr 1670 1675 1680 Pro Pro Thr Pro Ile Phe Pro Ile Ser Asp Asp Val Gly Ala Ile 1685 1690 1695 Pro Ile Lys His Phe Pro Lys His Val Ala Asp Leu His Ala Ser 1700 1705 1710 Ser Gly Phe Thr Glu Glu Phe Glu Thr Leu Lys Glu Phe Tyr Gln 1715 1720 1725 Glu Val Gln Ser Cys Thr Val Asp Leu Gly Ile Thr Ala Asp Ser 1730 1735 1740 Ser Asn His Pro Asp Asn Lys His Lys Asn Arg Tyr Ile Asn Ile 1745 1750 1755 Val Ala Tyr Asp His Ser Arg Val Lys Leu Ala Gln Leu Ala Glu 1760 1765 1770 Lys Asp Gly Lys Leu Thr Asp Tyr Ile Asn Ala Asn Tyr Val Asp 1775 1780 1785 Gly Tyr Asn Arg Pro Lys Ala Tyr Ile Ala Ala Gln Gly Pro Leu 1790 1795 1800 Lys Ser Thr Ala Glu Asp Phe Trp Arg Met Ile Trp Glu His Asn 1805 1810 1815 Val Glu Val Ile Val Met Ile Thr Asn Leu Val Glu Lys Gly Arg 1820 1825 1830 Arg Lys Cys Asp Gln Tyr Trp Pro Ala Asp Gly Ser Glu Glu Tyr 1835 1840 1845 Gly Asn Phe Leu Val Thr Gln Lys Ser Val Gln Val Leu Ala Tyr 1850 1855 1860 Tyr Thr Val Arg Asn Phe Thr Leu Arg Asn Thr Lys Ile Lys Lys 1865 1870 1875 Gly Ser Gln Lys Gly Arg Pro Ser Gly Arg Val Val Thr Gln Tyr 1880 1885 1890 His Tyr Thr Gln Trp Pro Asp Met Gly Val Pro Glu Tyr Ser Leu 1895 1900 1905 Pro Val Leu Thr Phe Val Arg Lys Ala Ala Tyr Ala Lys Arg His 1910 1915 1920 Ala Val Gly Pro Val Val Val His Cys Ser Ala Gly Val Gly Arg 1925 1930 1935 Thr Gly Thr Tyr Ile Val Leu Asp Ser Met Leu Gln Gln Ile Gln 1940 1945 1950 His Glu Gly Thr Val Asn Ile Phe Gly Phe Leu Lys His Ile Arg 1955 1960 1965 Ser Gln Arg Asn Tyr Leu Val Gln Thr Glu Glu Gln Tyr Val Phe 1970 1975 1980 Ile His Asp Thr Leu Val Glu Ala Ile Leu Ser Lys Glu Thr Glu 1985 1990 1995 Val Leu Asp Ser His Ile His Ala Tyr Val Asn Ala Leu Leu Ile 2000 2005 2010 Pro Gly Pro Ala Gly Lys Thr Lys Leu Glu Lys Gln Phe Gln Gly 2015 2020 2025 Leu Thr Leu Ser Pro Arg Leu Glu Cys Arg Gly Thr Ile Ser Ala 2030 2035 2040 His Cys Asn Leu Pro Leu Pro Gly Leu Thr Asp Pro Pro Thr Ser 2045 2050 2055 Ala Ser Arg Val Ala Gly Thr Ile Leu Leu Ser Gln Ser Asn Ile 2060 2065 2070 Gln Gln Ser Asp Tyr Ser Ala Ala Leu Lys Gln Cys Asn Arg Glu 2075 2080 2085 Lys Asn Arg Thr Ser Ser Ile Ile Pro Val Glu Arg Ser Arg Val 2090 2095 2100 Gly Ile Ser Ser Leu Ser Gly Glu Gly Thr Asp Tyr Ile Asn Ala 2105 2110 2115 Ser Tyr Ile Met Gly Tyr Tyr Gln Ser Asn Glu Phe Ile Ile Thr 2120 2125 2130 Gln His Pro Leu Leu His Thr Ile Lys Asp Phe Trp Arg Met Ile 2135 2140 2145 Trp Asp His Asn Ala Gln Leu Val Val Met Ile Pro Asp Gly Gln 2150 2155 2160 Asn Met Ala Glu Asp Glu Phe Val Tyr Trp Pro Asn Lys Asp Glu 2165 2170 2175 Pro Ile Asn Cys Glu Ser Phe Lys Val Thr Leu Met Ala Glu Glu 2180 2185 2190 His Lys Cys Leu Ser Asn Glu Glu Lys Leu Ile Ile Gln Asp Phe 2195 2200 2205 Ile Leu Glu Ala Thr Gln Asp Asp Tyr Val Leu Glu Val Arg His 2210 2215 2220 Phe Gln Cys Pro Lys Trp Pro Asn Pro Asp Ser Pro Ile Ser Lys 2225 2230 2235 Thr Phe Glu Leu Ile Ser Val Ile Lys Glu Glu Ala Ala Asn Arg 2240 2245 2250 Asp Gly Pro Met Ile Val His Asp Glu His Gly Gly Val Thr Ala 2255 2260 2265 Gly Thr Phe Cys Ala Leu Thr Thr Leu Met His Gln Leu Glu Lys 2270 2275 2280 Glu Asn Ser Val Asp Val Tyr Gln Val Ala Lys Met Ile Asn Leu 2285 2290 2295 Met Arg Pro Gly Val Phe Ala Asp Ile Glu Gln Tyr Gln Phe Leu 2300 2305 2310 Tyr Lys Val Ile Leu Ser Leu Val Ser Thr Arg Gln Glu Glu Asn 2315 2320 2325 Pro Ser Thr Ser Leu Asp Ser Asn Gly Ala Ala Leu Pro Asp Gly 2330 2335 2340 Asn Ile Ala Glu Ser Leu Glu Ser Leu Val 2345 2350 5 7941 DNA Homo sapiens CDS (148)..(7092) 5 cacacatacg cacgcacgat ctcacttcga tctatacact ggaggattaa aacaaacaaa 60 caaaaaaaac atttccttcg ctccccctcc ctctccactc tgagaagcag aggagccgca 120 cggcgagggg ccgcagaccg tctggaa atg cga atc cta aag cgt ttc ctc gct 174 Met Arg Ile Leu Lys Arg Phe Leu Ala 1 5 tgc att cag ctc ctc tgt gtt tgc cgc ctg gat tgg gct aat gga tac 222 Cys Ile Gln Leu Leu Cys Val Cys Arg Leu Asp Trp Ala Asn Gly Tyr 10 15 20 25 tac aga caa cag aga aaa ctt gtt gaa gag att ggc tgg tcc tat aca 270 Tyr Arg Gln Gln Arg Lys Leu Val Glu Glu Ile Gly Trp Ser Tyr Thr 30 35 40 gga gca ctg aat caa aaa aat tgg gga aag aaa tat cca aca tgt aat 318 Gly Ala Leu Asn Gln Lys Asn Trp Gly Lys Lys Tyr Pro Thr Cys Asn 45 50 55 agc cca aaa caa tct cct atc aat att gat gaa gat ctt aca caa gta 366 Ser Pro Lys Gln Ser Pro Ile Asn Ile Asp Glu Asp Leu Thr Gln Val 60 65 70 aat gtg aat ctt aag aaa ctt aaa ttt cag ggt tgg gat aaa aca tca 414 Asn Val Asn Leu Lys Lys Leu Lys Phe Gln Gly Trp Asp Lys Thr Ser 75 80 85 ttg gaa aac aca ttc att cat aac act ggg aaa aca gtg gaa att aat 462 Leu Glu Asn Thr Phe Ile His Asn Thr Gly Lys Thr Val Glu Ile Asn 90 95 100 105 ctc act aat gac tac cgt gtc agc gga gga gtt tca gaa atg gtg ttt 510 Leu Thr Asn Asp Tyr Arg Val Ser Gly Gly Val Ser Glu Met Val Phe 110 115 120 aaa gca agc aag ata act ttt cac tgg gga aaa tgc aat atg tca tct 558 Lys Ala Ser Lys Ile Thr Phe His Trp Gly Lys Cys Asn Met Ser Ser 125 130 135 gat gga tca gag cat agt tta gaa gga caa aaa ttt cca ctt gag atg 606 Asp Gly Ser Glu His Ser Leu Glu Gly Gln Lys Phe Pro Leu Glu Met 140 145 150 caa atc tac tgc ttt gat gcg gac cga ttt tca agt ttt gag gaa gca 654 Gln Ile Tyr Cys Phe Asp Ala Asp Arg Phe Ser Ser Phe Glu Glu Ala 155 160 165 gtc aaa gga aaa ggg aag tta aga gct tta tcc att ttg ttt gag gtt 702 Val Lys Gly Lys Gly Lys Leu Arg Ala Leu Ser Ile Leu Phe Glu Val 170 175 180 185 ggg aca gaa gaa aat ttg gat ttc aaa gcg att att gat gga gtc gaa 750 Gly Thr Glu Glu Asn Leu Asp Phe Lys Ala Ile Ile Asp Gly Val Glu 190 195 200 agt gtt agt cgt ttt ggg aag cag gct gct tta gat cca ttc ata ctg 798 Ser Val Ser Arg Phe Gly Lys Gln Ala Ala Leu Asp Pro Phe Ile Leu 205 210 215 ttg aac ctt ctg cca aac tca act gac aag tat tac att tac aat ggc 846 Leu Asn Leu Leu Pro Asn Ser Thr Asp Lys Tyr Tyr Ile Tyr Asn Gly 220 225 230 tca ttg aca tct cct ccc tgc aca gac aca gtt gac tgg att gtt ttt 894 Ser Leu Thr Ser Pro Pro Cys Thr Asp Thr Val Asp Trp Ile Val Phe 235 240 245 aaa gat aca gtt agc atc tct gaa agc cag ttg gct gtt ttt tgt gaa 942 Lys Asp Thr Val Ser Ile Ser Glu Ser Gln Leu Ala Val Phe Cys Glu 250 255 260 265 gtt ctt aca atg caa caa tct ggt tat gtc atg ctg atg gac tac tta 990 Val Leu Thr Met Gln Gln Ser Gly Tyr Val Met Leu Met Asp Tyr Leu 270 275 280 caa aac aat ttt cga gag caa cag tac aag ttc tct aga cag gtg ttt 1038 Gln Asn Asn Phe Arg Glu Gln Gln Tyr Lys Phe Ser Arg Gln Val Phe 285 290 295 tcc tca tac act gga aag gaa gag att cat gaa gca gtt tgt agt tca 1086 Ser Ser Tyr Thr Gly Lys Glu Glu Ile His Glu Ala Val Cys Ser Ser 300 305 310 gaa cca gaa aat gtt cag gct gac cca gag aat tat acc agc ctt ctt 1134 Glu Pro Glu Asn Val Gln Ala Asp Pro Glu Asn Tyr Thr Ser Leu Leu 315 320 325 gtt aca tgg gaa aga cct cga gtc gtt tat gat acc atg att gag aag 1182 Val Thr Trp Glu Arg Pro Arg Val Val Tyr Asp Thr Met Ile Glu Lys 330 335 340 345 ttt gca gtt ttg tac cag cag ttg gat gga gag gac caa acc aag cat 1230 Phe Ala Val Leu Tyr Gln Gln Leu Asp Gly Glu Asp Gln Thr Lys His 350 355 360 gaa ttt ttg aca gat ggc tat caa gac ttg ggt gct att ctc aat aat 1278 Glu Phe Leu Thr Asp Gly Tyr Gln Asp Leu Gly Ala Ile Leu Asn Asn 365 370 375 ttg cta ccc aat atg agt tat gtt ctt cag ata gta gcc ata tgc act 1326 Leu Leu Pro Asn Met Ser Tyr Val Leu Gln Ile Val Ala Ile Cys Thr 380 385 390 aat ggc tta tat gga aaa tac agc gac caa ctg att gtc gac atg cct 1374 Asn Gly Leu Tyr Gly Lys Tyr Ser Asp Gln Leu Ile Val Asp Met Pro 395 400 405 act gat aat cct gaa ctt gat ctt ttc cct gaa tta att gga act gaa 1422 Thr Asp Asn Pro Glu Leu Asp Leu Phe Pro Glu Leu Ile Gly Thr Glu 410 415 420 425 gaa ata atc aag gag gag gaa gag gga aaa gac att gaa gaa ggc gct 1470 Glu Ile Ile Lys Glu Glu Glu Glu Gly Lys Asp Ile Glu Glu Gly Ala 430 435 440 att gtg aat cct ggt aga gac agt gct aca aac caa atc agg aaa aag 1518 Ile Val Asn Pro Gly Arg Asp Ser Ala Thr Asn Gln Ile Arg Lys Lys 445 450 455 gaa ccc cag att tct acc aca aca cac tac aat cgc ata ggg acg aaa 1566 Glu Pro Gln Ile Ser Thr Thr Thr His Tyr Asn Arg Ile Gly Thr Lys 460 465 470 tac aat gaa gcc aag act aac cga tcc cca aca aga gga agt gaa ttc 1614 Tyr Asn Glu Ala Lys Thr Asn Arg Ser Pro Thr Arg Gly Ser Glu Phe 475 480 485 tct gga aag ggt gat gtt ccc aat aca tct tta aat tcc act tcc caa 1662 Ser Gly Lys Gly Asp Val Pro Asn Thr Ser Leu Asn Ser Thr Ser Gln 490 495 500 505 cca gtc act aaa tta gcc aca gaa aaa gat att tcc ttg act tct cag 1710 Pro Val Thr Lys Leu Ala Thr Glu Lys Asp Ile Ser Leu Thr Ser Gln 510 515 520 act gtg act gaa ctg cca cct cac act gtg gaa ggt act tca gcc tct 1758 Thr Val Thr Glu Leu Pro Pro His Thr Val Glu Gly Thr Ser Ala Ser 525 530 535 tta aat gat ggc tct aaa act gtt ctt aga tct cca cat atg aac ttg 1806 Leu Asn Asp Gly Ser Lys Thr Val Leu Arg Ser Pro His Met Asn Leu 540 545 550 tcg ggg act gca gaa tcc tta aat aca gtt tct ata aca gaa tat gag 1854 Ser Gly Thr Ala Glu Ser Leu Asn Thr Val Ser Ile Thr Glu Tyr Glu 555 560 565 gag gag agt tta ttg acc agt ttc aag ctt gat act gga gct gaa gat 1902 Glu Glu Ser Leu Leu Thr Ser Phe Lys Leu Asp Thr Gly Ala Glu Asp 570 575 580 585 tct tca ggc tcc agt ccc gca act tct gct atc cca ttc atc tct gag 1950 Ser Ser Gly Ser Ser Pro Ala Thr Ser Ala Ile Pro Phe Ile Ser Glu 590 595 600 aac ata tcc caa ggg tat ata ttt tcc tcc gaa aac cca gag aca ata 1998 Asn Ile Ser Gln Gly Tyr Ile Phe Ser Ser Glu Asn Pro Glu Thr Ile 605 610 615 aca tat gat gtc ctt ata cca gaa tct gct aga aat gct tcc gaa gat 2046 Thr Tyr Asp Val Leu Ile Pro Glu Ser Ala Arg Asn Ala Ser Glu Asp 620 625 630 tca act tca tca ggt tca gaa gaa tca cta aag gat cct tct atg gag 2094 Ser Thr Ser Ser Gly Ser Glu Glu Ser Leu Lys Asp Pro Ser Met Glu 635 640 645 gga aat gtg tgg ttt cct agc tct aca gac ata aca gca cag ccc gat 2142 Gly Asn Val Trp Phe Pro Ser Ser Thr Asp Ile Thr Ala Gln Pro Asp 650 655 660 665 gtt gga tca ggc aga gag agc ttt ctc cag act aat tac act gag ata 2190 Val Gly Ser Gly Arg Glu Ser Phe Leu Gln Thr Asn Tyr Thr Glu Ile 670 675 680 cgt gtt gat gaa tct gag aag aca acc aag tcc ttt tct gca ggc cca 2238 Arg Val Asp Glu Ser Glu Lys Thr Thr Lys Ser Phe Ser Ala Gly Pro 685 690 695 gtg atg tca cag ggt ccc tca gtt aca gat ctg gaa atg cca cat tat 2286 Val Met Ser Gln Gly Pro Ser Val Thr Asp Leu Glu Met Pro His Tyr 700 705 710 tct acc ttt gcc tac ttc cca act gag gta aca cct cat gct ttt acc 2334 Ser Thr Phe Ala Tyr Phe Pro Thr Glu Val Thr Pro His Ala Phe Thr 715 720 725 cca tcc tcc aga caa cag gat ttg gtc tcc acg gtc aac gtg gta tac 2382 Pro Ser Ser Arg Gln Gln Asp Leu Val Ser Thr Val Asn Val Val Tyr 730 735 740 745 tcg cag aca acc caa ccg gta tac aat ggt gag aca cct ctt caa cct 2430 Ser Gln Thr Thr Gln Pro Val Tyr Asn Gly Glu Thr Pro Leu Gln Pro 750 755 760 tcc tac agt agt gaa gtc ttt cct cta gtc acc cct ttg ttg ctt gac 2478 Ser Tyr Ser Ser Glu Val Phe Pro Leu Val Thr Pro Leu Leu Leu Asp 765 770 775 aat cag atc ctc aac act acc cct gct gct tca agt agt gat tcg gcc 2526 Asn Gln Ile Leu Asn Thr Thr Pro Ala Ala Ser Ser Ser Asp Ser Ala 780 785 790 ttg cat gct acg cct gta ttt ccc agt gtc gat gtg tca ttt gaa tcc 2574 Leu His Ala Thr Pro Val Phe Pro Ser Val Asp Val Ser Phe Glu Ser 795 800 805 atc ctg tct tcc tat gat ggt gca cct ttg ctt cca ttt tcc tct gct 2622 Ile Leu Ser Ser Tyr Asp Gly Ala Pro Leu Leu Pro Phe Ser Ser Ala 810 815 820 825 tcc ttc agt agt gaa ttg ttt cgc cat ctg cat aca gtt tct caa atc 2670 Ser Phe Ser Ser Glu Leu Phe Arg His Leu His Thr Val Ser Gln Ile 830 835 840 ctt cca caa gtt act tca gct acc gag agt gat aag gtg ccc ttg cat 2718 Leu Pro Gln Val Thr Ser Ala Thr Glu Ser Asp Lys Val Pro Leu His 845 850 855 gct tct ctg cca gtg gct ggg ggt gat ttg cta tta gag ccc agc ctt 2766 Ala Ser Leu Pro Val Ala Gly Gly Asp Leu Leu Leu Glu Pro Ser Leu 860 865 870 gct cag tat tct gat gtg ctg tcc act act cat gct gct tca gag acg 2814 Ala Gln Tyr Ser Asp Val Leu Ser Thr Thr His Ala Ala Ser Glu Thr 875 880 885 ctg gaa ttt ggt agt gaa tct ggt gtt ctt tat aaa acg ctt atg ttt 2862 Leu Glu Phe Gly Ser Glu Ser Gly Val Leu Tyr Lys Thr Leu Met Phe 890 895 900 905 tct caa gtt gaa cca ccc agc agt gat gcc atg atg cat gca cgt tct 2910 Ser Gln Val Glu Pro Pro Ser Ser Asp Ala Met Met His Ala Arg Ser 910 915 920 tca ggg cct gaa cct tct tat gcc ttg tct gat aat gag ggc tcc caa 2958 Ser Gly Pro Glu Pro Ser Tyr Ala Leu Ser Asp Asn Glu Gly Ser Gln 925 930 935 cac atc ttc act gtt tct tac agt tct gca ata cct gtg cat gat tct 3006 His Ile Phe Thr Val Ser Tyr Ser Ser Ala Ile Pro Val His Asp Ser 940 945 950 gtg ggt gta act tat cag ggt tcc tta ttt agc ggc cct agc cat ata 3054 Val Gly Val Thr Tyr Gln Gly Ser Leu Phe Ser Gly Pro Ser His Ile 955 960 965 cca ata cct aag tct tcg tta ata acc cca act gca tca tta ctg cag 3102 Pro Ile Pro Lys Ser Ser Leu Ile Thr Pro Thr Ala Ser Leu Leu Gln 970 975 980 985 cct act cat gcc ctc tct ggt gat ggg gaa tgg tct gga gcc tct tct 3150 Pro Thr His Ala Leu Ser Gly Asp Gly Glu Trp Ser Gly Ala Ser Ser 990 995 1000 gat agt gaa ttt ctt tta cct gac aca gat ggg ctg aca gcc ctt 3195 Asp Ser Glu Phe Leu Leu Pro Asp Thr Asp Gly Leu Thr Ala Leu 1005 1010 1015 aac att tct tca cct gtt tct gta gct gaa ttt aca tat aca aca 3240 Asn Ile Ser Ser Pro Val Ser Val Ala Glu Phe Thr Tyr Thr Thr 1020 1025 1030 tct gtg ttt ggt gat gat aat aag gcg ctt tct aaa agt gaa ata 3285 Ser Val Phe Gly Asp Asp Asn Lys Ala Leu Ser Lys Ser Glu Ile 1035 1040 1045 ata tat gga aat gag act gaa ctg caa att cct tct ttc aat gag 3330 Ile Tyr Gly Asn Glu Thr Glu Leu Gln Ile Pro Ser Phe Asn Glu 1050 1055 1060 atg gtt tac cct tct gaa agc aca gtc atg ccc aac atg tat gat 3375 Met Val Tyr Pro Ser Glu Ser Thr Val Met Pro Asn Met Tyr Asp 1065 1070 1075 aat gta aat aag ttg aat gcg tct tta caa gaa acc tct gtt tcc 3420 Asn Val Asn Lys Leu Asn Ala Ser Leu Gln Glu Thr Ser Val Ser 1080 1085 1090 att tct agc acc aag ggc atg ttt cca ggg tcc ctt gct cat acc 3465 Ile Ser Ser Thr Lys Gly Met Phe Pro Gly Ser Leu Ala His Thr 1095 1100 1105 acc act aag gtt ttt gat cat gag att agt caa gtt cca gaa aat 3510 Thr Thr Lys Val Phe Asp His Glu Ile Ser Gln Val Pro Glu Asn 1110 1115 1120 aac ttt tca gtt caa cct aca cat act gtc tct caa gca tct ggt 3555 Asn Phe Ser Val Gln Pro Thr His Thr Val Ser Gln Ala Ser Gly 1125 1130 1135 gac act tcg ctt aaa cct gtg ctt agt gca aac tca gag cca gca 3600 Asp Thr Ser Leu Lys Pro Val Leu Ser Ala Asn Ser Glu Pro Ala 1140 1145 1150 tcc tct gac cct gct tct agt gaa atg tta tct cct tca act cag 3645 Ser Ser Asp Pro Ala Ser Ser Glu Met Leu Ser Pro Ser Thr Gln 1155 1160 1165 ctc tta ttt tat gag acc tca gct tct ttt agt act gaa gta ttg 3690 Leu Leu Phe Tyr Glu Thr Ser Ala Ser Phe Ser Thr Glu Val Leu 1170 1175 1180 cta caa cct tcc ttt cag gct tct gat gtt gac acc ttg ctt aaa 3735 Leu Gln Pro Ser Phe Gln Ala Ser Asp Val Asp Thr Leu Leu Lys 1185 1190 1195 act gtt ctt cca gct gtg ccc agt gat cca ata ttg gtt gaa acc 3780 Thr Val Leu Pro Ala Val Pro Ser Asp Pro Ile Leu Val Glu Thr 1200 1205 1210 ccc aaa gtt gat aaa att agt tct aca atg ttg cat ctc att gta 3825 Pro Lys Val Asp Lys Ile Ser Ser Thr Met Leu His Leu Ile Val 1215 1220 1225 tca aat tct gct tca agt gaa aac atg ctg cac tct aca tct gta 3870 Ser Asn Ser Ala Ser Ser Glu Asn Met Leu His Ser Thr Ser Val 1230 1235 1240 cca gtt ttt gat gtg tcg cct act tct cat atg cac tct gct tca 3915 Pro Val Phe Asp Val Ser Pro Thr Ser His Met His Ser Ala Ser 1245 1250 1255 ctt caa ggt ttg acc att tcc tat gca agt gag aaa tat gaa cca 3960 Leu Gln Gly Leu Thr Ile Ser Tyr Ala Ser Glu Lys Tyr Glu Pro 1260 1265 1270 gtt ttg tta aaa agt gaa agt tcc cac caa gtg gta cct tct ttg 4005 Val Leu Leu Lys Ser Glu Ser Ser His Gln Val Val Pro Ser Leu 1275 1280 1285 tac agt aat gat gag ttg ttc caa acg gcc aat ttg gag att aac 4050 Tyr Ser Asn Asp Glu Leu Phe Gln Thr Ala Asn Leu Glu Ile Asn 1290 1295 1300 cag gcc cat ccc cca aaa gga agg cat gta ttt gct aca cct gtt 4095 Gln Ala His Pro Pro Lys Gly Arg His Val Phe Ala Thr Pro Val 1305 1310 1315 tta tca att gat gaa cca tta aat aca cta ata aat aag ctt ata 4140 Leu Ser Ile Asp Glu Pro Leu Asn Thr Leu Ile Asn Lys Leu Ile 1320 1325 1330 cat tcc gat gaa att tta acc tcc acc aaa agt tct gtt act ggt 4185 His Ser Asp Glu Ile Leu Thr Ser Thr Lys Ser Ser Val Thr Gly 1335 1340 1345 aag gta ttt gct ggt att cca aca gtt gct tct gat aca ttt gta 4230 Lys Val Phe Ala Gly Ile Pro Thr Val Ala Ser Asp Thr Phe Val 1350 1355 1360 tct act gat cat tct gtt cct ata gga aat ggg cat gtt gcc att 4275 Ser Thr Asp His Ser Val Pro Ile Gly Asn Gly His Val Ala Ile 1365 1370 1375 aca gct gtt tct ccc cac aga gat ggt tct gta acc tca aca aag 4320 Thr Ala Val Ser Pro His Arg Asp Gly Ser Val Thr Ser Thr Lys 1380 1385 1390 ttg ctg ttt cct tct aag gca act tct gag ctg agt cat agt gcc 4365 Leu Leu Phe Pro Ser Lys Ala Thr Ser Glu Leu Ser His Ser Ala 1395 1400 1405 aaa tct gat gcc ggt tta gtg ggt ggt ggt gaa gat ggt gac act 4410 Lys Ser Asp Ala Gly Leu Val Gly Gly Gly Glu Asp Gly Asp Thr 1410 1415 1420 gat gat gat ggt gat gat gat gat gac aga gat agt gat ggc tta 4455 Asp Asp Asp Gly Asp Asp Asp Asp Asp Arg Asp Ser Asp Gly Leu 1425 1430 1435 tcc att cat aag tgt atg tca tgc tca tcc tat aga gaa tca cag 4500 Ser Ile His Lys Cys Met Ser Cys Ser Ser Tyr Arg Glu Ser Gln 1440 1445 1450 gaa aag gta atg aat gat tca gac acc cac gaa aac agt ctt atg 4545 Glu Lys Val Met Asn Asp Ser Asp Thr His Glu Asn Ser Leu Met 1455 1460 1465 gat cag aat aat cca atc tca tac tca cta tct gag aat tct gaa 4590 Asp Gln Asn Asn Pro Ile Ser Tyr Ser Leu Ser Glu Asn Ser Glu 1470 1475 1480 gaa gat aat aga gtc aca agt gta tcc tca gac agt caa act ggt 4635 Glu Asp Asn Arg Val Thr Ser Val Ser Ser Asp Ser Gln Thr Gly 1485 1490 1495 atg gac aga agt cct ggt aaa tca cca tca gca aat ggg cta tcc 4680 Met Asp Arg Ser Pro Gly Lys Ser Pro Ser Ala Asn Gly Leu Ser 1500 1505 1510 caa aag cac aat gat gga aaa gag gaa aat gac att cag act ggt 4725 Gln Lys His Asn Asp Gly Lys Glu Glu Asn Asp Ile Gln Thr Gly 1515 1520 1525 agt gct ctg ctt cct ctc agc cct gaa tct aaa gca tgg gca gtt 4770 Ser Ala Leu Leu Pro Leu Ser Pro Glu Ser Lys Ala Trp Ala Val 1530 1535 1540 ctg aca agt gat gaa gaa agt gga tca ggg caa ggt acc tca gat 4815 Leu Thr Ser Asp Glu Glu Ser Gly Ser Gly Gln Gly Thr Ser Asp 1545 1550 1555 agc ctt aat gag aat gag act tcc aca gat ttc agt ttt gca gac 4860 Ser Leu Asn Glu Asn Glu Thr Ser Thr Asp Phe Ser Phe Ala Asp 1560 1565 1570 act aat gaa aaa gat gct gat ggg atc ctg gca gca ggt gac tca 4905 Thr Asn Glu Lys Asp Ala Asp Gly Ile Leu Ala Ala Gly Asp Ser 1575 1580 1585 gaa ata act cct gga ttc cca cag tcc cca aca tca tct gtt act 4950 Glu Ile Thr Pro Gly Phe Pro Gln Ser Pro Thr Ser Ser Val Thr 1590 1595 1600 agc gag aac tca gaa gtg ttc cac gtt tca gag gca gag gcc agt 4995 Ser Glu Asn Ser Glu Val Phe His Val Ser Glu Ala Glu Ala Ser 1605 1610 1615 aat agt agc cat gag tct cgt att ggt cta gct gag ggg ttg gaa 5040 Asn Ser Ser His Glu Ser Arg Ile Gly Leu Ala Glu Gly Leu Glu 1620 1625 1630 tcc gag aag aag gca gtt ata ccc ctt gtg atc gtg tca gcc ctg 5085 Ser Glu Lys Lys Ala Val Ile Pro Leu Val Ile Val Ser Ala Leu 1635 1640 1645 act ttt atc tgt cta gtg gtt ctt gtg ggt att ctc atc tac tgg 5130 Thr Phe Ile Cys Leu Val Val Leu Val Gly Ile Leu Ile Tyr Trp 1650 1655 1660 agg aaa tgc ttc cag act gca cac ttt tac tta gag gac agt aca 5175 Arg Lys Cys Phe Gln Thr Ala His Phe Tyr Leu Glu Asp Ser Thr 1665 1670 1675 tcc cct aga gtt ata tcc aca cct cca aca cct atc ttt cca att 5220 Ser Pro Arg Val Ile Ser Thr Pro Pro Thr Pro Ile Phe Pro Ile 1680 1685 1690 tca gat gat gtc gga gca att cca ata aag cac ttt cca aag cat 5265 Ser Asp Asp Val Gly Ala Ile Pro Ile Lys His Phe Pro Lys His 1695 1700 1705 gtt gca gat tta cat gca agt agt ggg ttt act gaa gaa ttt gag 5310 Val Ala Asp Leu His Ala Ser Ser Gly Phe Thr Glu Glu Phe Glu 1710 1715 1720 aca ctg aaa gag ttt tac cag gaa gtg cag agc tgt act gtt gac 5355 Thr Leu Lys Glu Phe Tyr Gln Glu Val Gln Ser Cys Thr Val Asp 1725 1730 1735 tta ggt att aca gca gac agc tcc aac cac cca gac aac aag cac 5400 Leu Gly Ile Thr Ala Asp Ser Ser Asn His Pro Asp Asn Lys His 1740 1745 1750 aag aat cga tac ata aat atc gtt gcc tat gat cat agc agg gtt 5445 Lys Asn Arg Tyr Ile Asn Ile Val Ala Tyr Asp His Ser Arg Val 1755 1760 1765 aag cta gca cag ctt gct gaa aag gat ggc aaa ctg act gat tat 5490 Lys Leu Ala Gln Leu Ala Glu Lys Asp Gly Lys Leu Thr Asp Tyr 1770 1775 1780 atc aat gcc aat tat gtt gat ggc tac aac aga cca aaa gct tat 5535 Ile Asn Ala Asn Tyr Val Asp Gly Tyr Asn Arg Pro Lys Ala Tyr 1785 1790 1795 att gct gcc caa ggc cca ctg aaa tcc aca gct gaa gat ttc tgg 5580 Ile Ala Ala Gln Gly Pro Leu Lys Ser Thr Ala Glu Asp Phe Trp 1800 1805 1810 aga atg ata tgg gaa cat aat gtg gaa gtt att gtc atg ata aca 5625 Arg Met Ile Trp Glu His Asn Val Glu Val Ile Val Met Ile Thr 1815 1820 1825 aac ctc gtg gag aaa gga agg aga aaa tgt gat cag tac tgg cct 5670 Asn Leu Val Glu Lys Gly Arg Arg Lys Cys Asp Gln Tyr Trp Pro 1830 1835 1840 gcc gat ggg agt gag gag tac ggg aac ttt ctg gtc act cag aag 5715 Ala Asp Gly Ser Glu Glu Tyr Gly Asn Phe Leu Val Thr Gln Lys 1845 1850 1855 agt gtg caa gtg ctt gcc tat tat act gtg agg aat ttt act cta 5760 Ser Val Gln Val Leu Ala Tyr Tyr Thr Val Arg Asn Phe Thr Leu 1860 1865 1870 aga aac aca aaa ata aaa aag ggc tcc cag aaa gga aga ccc agt 5805 Arg Asn Thr Lys Ile Lys Lys Gly Ser Gln Lys Gly Arg Pro Ser 1875 1880 1885 gga cgt gtg gtc aca cag tat cac tac acg cag tgg cct gac atg 5850 Gly Arg Val Val Thr Gln Tyr His Tyr Thr Gln Trp Pro Asp Met 1890 1895 1900 gga gta cca gag tac tcc ctg cca gtg ctg acc ttt gtg aga aag 5895 Gly Val Pro Glu Tyr Ser Leu Pro Val Leu Thr Phe Val Arg Lys 1905 1910 1915 gca gcc tat gcc aag cgc cat gca gtg ggg cct gtt gtc gtc cac 5940 Ala Ala Tyr Ala Lys Arg His Ala Val Gly Pro Val Val Val His 1920 1925 1930 tgc agt gct gga gtt gga aga aca ggc aca tat att gtg cta gac 5985 Cys Ser Ala Gly Val Gly Arg Thr Gly Thr Tyr Ile Val Leu Asp 1935 1940 1945 agt atg ttg cag cag att caa cac gaa gga act gtc aac ata ttt 6030 Ser Met Leu Gln Gln Ile Gln His Glu Gly Thr Val Asn Ile Phe 1950 1955 1960 ggc ttc tta aaa cac atc cgt tca caa aga aat tat ttg gta caa 6075 Gly Phe Leu Lys His Ile Arg Ser Gln Arg Asn Tyr Leu Val Gln 1965 1970 1975 act gag gag caa tat gtc ttc att cat gat aca ctg gtt gag gcc 6120 Thr Glu Glu Gln Tyr Val Phe Ile His Asp Thr Leu Val Glu Ala 1980 1985 1990 ata ctt agt aaa gaa act gag gtg ctg gac agt cat att cat gcc 6165 Ile Leu Ser Lys Glu Thr Glu Val Leu Asp Ser His Ile His Ala 1995 2000 2005 tat gtt aat gca ctc ctc att cct gga cca gca ggc aaa aca aag 6210 Tyr Val Asn Ala Leu Leu Ile Pro Gly Pro Ala Gly Lys Thr Lys 2010 2015 2020 cta gag aaa caa ttc cag ctc ctg agc cag tca aat ata cag cag 6255 Leu Glu Lys Gln Phe Gln Leu Leu Ser Gln Ser Asn Ile Gln Gln 2025 2030 2035 agt gac tat tct gca gcc cta aag caa tgc aac agg gaa aag aat 6300 Ser Asp Tyr Ser Ala Ala Leu Lys Gln Cys Asn Arg Glu Lys Asn 2040 2045 2050 cga act tct tct atc atc cct gtg gaa aga tca agg gtt ggc att 6345 Arg Thr Ser Ser Ile Ile Pro Val Glu Arg Ser Arg Val Gly Ile 2055 2060 2065 tca tcc ctg agt gga gaa ggc aca gac tac atc aat gcc tcc tat 6390 Ser Ser Leu Ser Gly Glu Gly Thr Asp Tyr Ile Asn Ala Ser Tyr 2070 2075 2080 atc atg ggc tat tac cag agc aat gaa ttc atc att acc cag cac 6435 Ile Met Gly Tyr Tyr Gln Ser Asn Glu Phe Ile Ile Thr Gln His 2085 2090 2095 cct ctc ctt cat acc atc aag gat ttc tgg agg atg ata tgg gac 6480 Pro Leu Leu His Thr Ile Lys Asp Phe Trp Arg Met Ile Trp Asp 2100 2105 2110 cat aat gcc caa ctg gtg gtt atg att cct gat ggc caa aac atg 6525 His Asn Ala Gln Leu Val Val Met Ile Pro Asp Gly Gln Asn Met 2115 2120 2125 gca gaa gat gaa ttt gtt tac tgg cca aat aaa gat gag cct ata 6570 Ala Glu Asp Glu Phe Val Tyr Trp Pro Asn Lys Asp Glu Pro Ile 2130 2135 2140 aat tgt gag agc ttt aag gtc act ctt atg gct gaa gaa cac aaa 6615 Asn Cys Glu Ser Phe Lys Val Thr Leu Met Ala Glu Glu His Lys 2145 2150 2155 tgt cta tct aat gag gaa aaa ctt ata att cag gac ttt atc tta 6660 Cys Leu Ser Asn Glu Glu Lys Leu Ile Ile Gln Asp Phe Ile Leu 2160 2165 2170 gaa gct aca cag gat gat tat gta ctt gaa gtg agg cac ttt cag 6705 Glu Ala Thr Gln Asp Asp Tyr Val Leu Glu Val Arg His Phe Gln 2175 2180 2185 tgt cct aaa tgg cca aat cca gat agc ccc att agt aaa act ttt 6750 Cys Pro Lys Trp Pro Asn Pro Asp Ser Pro Ile Ser Lys Thr Phe 2190 2195 2200 gaa ctt ata agt gtt ata aaa gaa gaa gct gcc aat agg gat ggg 6795 Glu Leu Ile Ser Val Ile Lys Glu Glu Ala Ala Asn Arg Asp Gly 2205 2210 2215 cct atg att gtt cat gat gag cat gga gga gtg acg gca gga act 6840 Pro Met Ile Val His Asp Glu His Gly Gly Val Thr Ala Gly Thr 2220 2225 2230 ttc tgt gct ctg aca acc ctt atg cac caa cta gaa aaa gaa aat 6885 Phe Cys Ala Leu Thr Thr Leu Met His Gln Leu Glu Lys Glu Asn 2235 2240 2245 tcc gtg gat gtt tac cag gta gcc aag atg atc aat ctg atg agg 6930 Ser Val Asp Val Tyr Gln Val Ala Lys Met Ile Asn Leu Met Arg 2250 2255 2260 cca gga gtc ttt gct gac att gag cag tat cag ttt ctc tac aaa 6975 Pro Gly Val Phe Ala Asp Ile Glu Gln Tyr Gln Phe Leu Tyr Lys 2265 2270 2275 gtg atc ctc agc ctt gtg agc aca agg cag gaa gag aat cca tcc 7020 Val Ile Leu Ser Leu Val Ser Thr Arg Gln Glu Glu Asn Pro Ser 2280 2285 2290 acc tct ctg gac agt aat ggt gca gca ttg cct gat gga aat ata 7065 Thr Ser Leu Asp Ser Asn Gly Ala Ala Leu Pro Asp Gly Asn Ile 2295 2300 2305 gct gag agc tta gag tct tta gtt taa cacagaaagg ggtgggggga 7112 Ala Glu Ser Leu Glu Ser Leu Val 2310 ctcacatctg agcattgttt tcctcttcct aaaattaggc aggaaaatca gtctagttct 7172 gttatctgtt gatttcccat cacctgacag taactttcat gacataggat tctgccgcca 7232 aatttatatc attaacaatg tgtgcctttt tgcaagactt gtaatttact tattatgttt 7292 gaactaaaat gattgaattt tacagtattt ctaagaatgg aattgtggta tttttttctg 7352 tattgatttt aacagaaaat ttcaatttat agaggttagg aattccaaac tacagaaaat 7412 gtttgttttt agtgtcaaat ttttagctgt atttgtagca attatcaggt ttgctagaaa 7472 tataactttt aatacagtag cctgtaaata aaacactctt ccatatgata ttcaacattt 7532 tacaactgca gtattcacct aaagtagaaa taatctgtta cttattgtaa atactgccct 7592 agtgtctcca tggaccaaat ttatatttat aattgtagat ttttatattt tactactgag 7652 tcaagttttc tagttctgtg taattgttta gtttaatgac gtagttcatt agctggtctt 7712 actctaccag ttttctgaca ttgtattgtg ttacctaagt cattaacttt gtttcagcat 7772 gtaattttaa cttttgtgga aaatagaaat accttcattt tgaaagaagt ttttatgaga 7832 ataacacctt accaaacatt gttcaaatgg tttttatcca aggaattgca aaaataaata 7892 taaatattgc cattaaaaaa aaaaaaaaaa aaaaaaaaaa aaaaaaaaa 7941 6 2314 PRT Homo sapiens gene (1)..(2314) PTP-zeta 6 Met Arg Ile Leu Lys Arg Phe Leu Ala Cys Ile Gln Leu Leu Cys Val 1 5 10 15 Cys Arg Leu Asp Trp Ala Asn Gly Tyr Tyr Arg Gln Gln Arg Lys Leu 20 25 30 Val Glu Glu Ile Gly Trp Ser Tyr Thr Gly Ala Leu Asn Gln Lys Asn 35 40 45 Trp Gly Lys Lys Tyr Pro Thr Cys Asn Ser Pro Lys Gln Ser Pro Ile 50 55 60 Asn Ile Asp Glu Asp Leu Thr Gln Val Asn Val Asn Leu Lys Lys Leu 65 70 75 80 Lys Phe Gln Gly Trp Asp Lys Thr Ser Leu Glu Asn Thr Phe Ile His 85 90 95 Asn Thr Gly Lys Thr Val Glu Ile Asn Leu Thr Asn Asp Tyr Arg Val 100 105 110 Ser Gly Gly Val Ser Glu Met Val Phe Lys Ala Ser Lys Ile Thr Phe 115 120 125 His Trp Gly Lys Cys Asn Met Ser Ser Asp Gly Ser Glu His Ser Leu 130 135 140 Glu Gly Gln Lys Phe Pro Leu Glu Met Gln Ile Tyr Cys Phe Asp Ala 145 150 155 160 Asp Arg Phe Ser Ser Phe Glu Glu Ala Val Lys Gly Lys Gly Lys Leu 165 170 175 Arg Ala Leu Ser Ile Leu Phe Glu Val Gly Thr Glu Glu Asn Leu Asp 180 185 190 Phe Lys Ala Ile Ile Asp Gly Val Glu Ser Val Ser Arg Phe Gly Lys 195 200 205 Gln Ala Ala Leu Asp Pro Phe Ile Leu Leu Asn Leu Leu Pro Asn Ser 210 215 220 Thr Asp Lys Tyr Tyr Ile Tyr Asn Gly Ser Leu Thr Ser Pro Pro Cys 225 230 235 240 Thr Asp Thr Val Asp Trp Ile Val Phe Lys Asp Thr Val Ser Ile Ser 245 250 255 Glu Ser Gln Leu Ala Val Phe Cys Glu Val Leu Thr Met Gln Gln Ser 260 265 270 Gly Tyr Val Met Leu Met Asp Tyr Leu Gln Asn Asn Phe Arg Glu Gln 275 280 285 Gln Tyr Lys Phe Ser Arg Gln Val Phe Ser Ser Tyr Thr Gly Lys Glu 290 295 300 Glu Ile His Glu Ala Val Cys Ser Ser Glu Pro Glu Asn Val Gln Ala 305 310 315 320 Asp Pro Glu Asn Tyr Thr Ser Leu Leu Val Thr Trp Glu Arg Pro Arg 325 330 335 Val Val Tyr Asp Thr Met Ile Glu Lys Phe Ala Val Leu Tyr Gln Gln 340 345 350 Leu Asp Gly Glu Asp Gln Thr Lys His Glu Phe Leu Thr Asp Gly Tyr 355 360 365 Gln Asp Leu Gly Ala Ile Leu Asn Asn Leu Leu Pro Asn Met Ser Tyr 370 375 380 Val Leu Gln Ile Val Ala Ile Cys Thr Asn Gly Leu Tyr Gly Lys Tyr 385 390 395 400 Ser Asp Gln Leu Ile Val Asp Met Pro Thr Asp Asn Pro Glu Leu Asp 405 410 415 Leu Phe Pro Glu Leu Ile Gly Thr Glu Glu Ile Ile Lys Glu Glu Glu 420 425 430 Glu Gly Lys Asp Ile Glu Glu Gly Ala Ile Val Asn Pro Gly Arg Asp 435 440 445 Ser Ala Thr Asn Gln Ile Arg Lys Lys Glu Pro Gln Ile Ser Thr Thr 450 455 460 Thr His Tyr Asn Arg Ile Gly Thr Lys Tyr Asn Glu Ala Lys Thr Asn 465 470 475 480 Arg Ser Pro Thr Arg Gly Ser Glu Phe Ser Gly Lys Gly Asp Val Pro 485 490 495 Asn Thr Ser Leu Asn Ser Thr Ser Gln Pro Val Thr Lys Leu Ala Thr 500 505 510 Glu Lys Asp Ile Ser Leu Thr Ser Gln Thr Val Thr Glu Leu Pro Pro 515 520 525 His Thr Val Glu Gly Thr Ser Ala Ser Leu Asn Asp Gly Ser Lys Thr 530 535 540 Val Leu Arg Ser Pro His Met Asn Leu Ser Gly Thr Ala Glu Ser Leu 545 550 555 560 Asn Thr Val Ser Ile Thr Glu Tyr Glu Glu Glu Ser Leu Leu Thr Ser 565 570 575 Phe Lys Leu Asp Thr Gly Ala Glu Asp Ser Ser Gly Ser Ser Pro Ala 580 585 590 Thr Ser Ala Ile Pro Phe Ile Ser Glu Asn Ile Ser Gln Gly Tyr Ile 595 600 605 Phe Ser Ser Glu Asn Pro Glu Thr Ile Thr Tyr Asp Val Leu Ile Pro 610 615 620 Glu Ser Ala Arg Asn Ala Ser Glu Asp Ser Thr Ser Ser Gly Ser Glu 625 630 635 640 Glu Ser Leu Lys Asp Pro Ser Met Glu Gly Asn Val Trp Phe Pro Ser 645 650 655 Ser Thr Asp Ile Thr Ala Gln Pro Asp Val Gly Ser Gly Arg Glu Ser 660 665 670 Phe Leu Gln Thr Asn Tyr Thr Glu Ile Arg Val Asp Glu Ser Glu Lys 675 680 685 Thr Thr Lys Ser Phe Ser Ala Gly Pro Val Met Ser Gln Gly Pro Ser 690 695 700 Val Thr Asp Leu Glu Met Pro His Tyr Ser Thr Phe Ala Tyr Phe Pro 705 710 715 720 Thr Glu Val Thr Pro His Ala Phe Thr Pro Ser Ser Arg Gln Gln Asp 725 730 735 Leu Val Ser Thr Val Asn Val Val Tyr Ser Gln Thr Thr Gln Pro Val 740 745 750 Tyr Asn Gly Glu Thr Pro Leu Gln Pro Ser Tyr Ser Ser Glu Val Phe 755 760 765 Pro Leu Val Thr Pro Leu Leu Leu Asp Asn Gln Ile Leu Asn Thr Thr 770 775 780 Pro Ala Ala Ser Ser Ser Asp Ser Ala Leu His Ala Thr Pro Val Phe 785 790 795 800 Pro Ser Val Asp Val Ser Phe Glu Ser Ile Leu Ser Ser Tyr Asp Gly 805 810 815 Ala Pro Leu Leu Pro Phe Ser Ser Ala Ser Phe Ser Ser Glu Leu Phe 820 825 830 Arg His Leu His Thr Val Ser Gln Ile Leu Pro Gln Val Thr Ser Ala 835 840 845 Thr Glu Ser Asp Lys Val Pro Leu His Ala Ser Leu Pro Val Ala Gly 850 855 860 Gly Asp Leu Leu Leu Glu Pro Ser Leu Ala Gln Tyr Ser Asp Val Leu 865 870 875 880 Ser Thr Thr His Ala Ala Ser Glu Thr Leu Glu Phe Gly Ser Glu Ser 885 890 895 Gly Val Leu Tyr Lys Thr Leu Met Phe Ser Gln Val Glu Pro Pro Ser 900 905 910 Ser Asp Ala Met Met His Ala Arg Ser Ser Gly Pro Glu Pro Ser Tyr 915 920 925 Ala Leu Ser Asp Asn Glu Gly Ser Gln His Ile Phe Thr Val Ser Tyr 930 935 940 Ser Ser Ala Ile Pro Val His Asp Ser Val Gly Val Thr Tyr Gln Gly 945 950 955 960 Ser Leu Phe Ser Gly Pro Ser His Ile Pro Ile Pro Lys Ser Ser Leu 965 970 975 Ile Thr Pro Thr Ala Ser Leu Leu Gln Pro Thr His Ala Leu Ser Gly 980 985 990 Asp Gly Glu Trp Ser Gly Ala Ser Ser Asp Ser Glu Phe Leu Leu Pro 995 1000 1005 Asp Thr Asp Gly Leu Thr Ala Leu Asn Ile Ser Ser Pro Val Ser 1010 1015 1020 Val Ala Glu Phe Thr Tyr Thr Thr Ser Val Phe Gly Asp Asp Asn 1025 1030 1035 Lys Ala Leu Ser Lys Ser Glu Ile Ile Tyr Gly Asn Glu Thr Glu 1040 1045 1050 Leu Gln Ile Pro Ser Phe Asn Glu Met Val Tyr Pro Ser Glu Ser 1055 1060 1065 Thr Val Met Pro Asn Met Tyr Asp Asn Val Asn Lys Leu Asn Ala 1070 1075 1080 Ser Leu Gln Glu Thr Ser Val Ser Ile Ser Ser Thr Lys Gly Met 1085 1090 1095 Phe Pro Gly Ser Leu Ala His Thr Thr Thr Lys Val Phe Asp His 1100 1105 1110 Glu Ile Ser Gln Val Pro Glu Asn Asn Phe Ser Val Gln Pro Thr 1115 1120 1125 His Thr Val Ser Gln Ala Ser Gly Asp Thr Ser Leu Lys Pro Val 1130 1135 1140 Leu Ser Ala Asn Ser Glu Pro Ala Ser Ser Asp Pro Ala Ser Ser 1145 1150 1155 Glu Met Leu Ser Pro Ser Thr Gln Leu Leu Phe Tyr Glu Thr Ser 1160 1165 1170 Ala Ser Phe Ser Thr Glu Val Leu Leu Gln Pro Ser Phe Gln Ala 1175 1180 1185 Ser Asp Val Asp Thr Leu Leu Lys Thr Val Leu Pro Ala Val Pro 1190 1195 1200 Ser Asp Pro Ile Leu Val Glu Thr Pro Lys Val Asp Lys Ile Ser 1205 1210 1215 Ser Thr Met Leu His Leu Ile Val Ser Asn Ser Ala Ser Ser Glu 1220 1225 1230 Asn Met Leu His Ser Thr Ser Val Pro Val Phe Asp Val Ser Pro 1235 1240 1245 Thr Ser His Met His Ser Ala Ser Leu Gln Gly Leu Thr Ile Ser 1250 1255 1260 Tyr Ala Ser Glu Lys Tyr Glu Pro Val Leu Leu Lys Ser Glu Ser 1265 1270 1275 Ser His Gln Val Val Pro Ser Leu Tyr Ser Asn Asp Glu Leu Phe 1280 1285 1290 Gln Thr Ala Asn Leu Glu Ile Asn Gln Ala His Pro Pro Lys Gly 1295 1300 1305 Arg His Val Phe Ala Thr Pro Val Leu Ser Ile Asp Glu Pro Leu 1310 1315 1320 Asn Thr Leu Ile Asn Lys Leu Ile His Ser Asp Glu Ile Leu Thr 1325 1330 1335 Ser Thr Lys Ser Ser Val Thr Gly Lys Val Phe Ala Gly Ile Pro 1340 1345 1350 Thr Val Ala Ser Asp Thr Phe Val Ser Thr Asp His Ser Val Pro 1355 1360 1365 Ile Gly Asn Gly His Val Ala Ile Thr Ala Val Ser Pro His Arg 1370 1375 1380 Asp Gly Ser Val Thr Ser Thr Lys Leu Leu Phe Pro Ser Lys Ala 1385 1390 1395 Thr Ser Glu Leu Ser His Ser Ala Lys Ser Asp Ala Gly Leu Val 1400 1405 1410 Gly Gly Gly Glu Asp Gly Asp Thr Asp Asp Asp Gly Asp Asp Asp 1415 1420 1425 Asp Asp Arg Asp Ser Asp Gly Leu Ser Ile His Lys Cys Met Ser 1430 1435 1440 Cys Ser Ser Tyr Arg Glu Ser Gln Glu Lys Val Met Asn Asp Ser 1445 1450 1455 Asp Thr His Glu Asn Ser Leu Met Asp Gln Asn Asn Pro Ile Ser 1460 1465 1470 Tyr Ser Leu Ser Glu Asn Ser Glu Glu Asp Asn Arg Val Thr Ser 1475 1480 1485 Val Ser Ser Asp Ser Gln Thr Gly Met Asp Arg Ser Pro Gly Lys 1490 1495 1500 Ser Pro Ser Ala Asn Gly Leu Ser Gln Lys His Asn Asp Gly Lys 1505 1510 1515 Glu Glu Asn Asp Ile Gln Thr Gly Ser Ala Leu Leu Pro Leu Ser 1520 1525 1530 Pro Glu Ser Lys Ala Trp Ala Val Leu Thr Ser Asp Glu Glu Ser 1535 1540 1545 Gly Ser Gly Gln Gly Thr Ser Asp Ser Leu Asn Glu Asn Glu Thr 1550 1555 1560 Ser Thr Asp Phe Ser Phe Ala Asp Thr Asn Glu Lys Asp Ala Asp 1565 1570 1575 Gly Ile Leu Ala Ala Gly Asp Ser Glu Ile Thr Pro Gly Phe Pro 1580 1585 1590 Gln Ser Pro Thr Ser Ser Val Thr Ser Glu Asn Ser Glu Val Phe 1595 1600 1605 His Val Ser Glu Ala Glu Ala Ser Asn Ser Ser His Glu Ser Arg 1610 1615 1620 Ile Gly Leu Ala Glu Gly Leu Glu Ser Glu Lys Lys Ala Val Ile 1625 1630 1635 Pro Leu Val Ile Val Ser Ala Leu Thr Phe Ile Cys Leu Val Val 1640 1645 1650 Leu Val Gly Ile Leu Ile Tyr Trp Arg Lys Cys Phe Gln Thr Ala 1655 1660 1665 His Phe Tyr Leu Glu Asp Ser Thr Ser Pro Arg Val Ile Ser Thr 1670 1675 1680 Pro Pro Thr Pro Ile Phe Pro Ile Ser Asp Asp Val Gly Ala Ile 1685 1690 1695 Pro Ile Lys His Phe Pro Lys His Val Ala Asp Leu His Ala Ser 1700 1705 1710 Ser Gly Phe Thr Glu Glu Phe Glu Thr Leu Lys Glu Phe Tyr Gln 1715 1720 1725 Glu Val Gln Ser Cys Thr Val Asp Leu Gly Ile Thr Ala Asp Ser 1730 1735 1740 Ser Asn His Pro Asp Asn Lys His Lys Asn Arg Tyr Ile Asn Ile 1745 1750 1755 Val Ala Tyr Asp His Ser Arg Val Lys Leu Ala Gln Leu Ala Glu 1760 1765 1770 Lys Asp Gly Lys Leu Thr Asp Tyr Ile Asn Ala Asn Tyr Val Asp 1775 1780 1785 Gly Tyr Asn Arg Pro Lys Ala Tyr Ile Ala Ala Gln Gly Pro Leu 1790 1795 1800 Lys Ser Thr Ala Glu Asp Phe Trp Arg Met Ile Trp Glu His Asn 1805 1810 1815 Val Glu Val Ile Val Met Ile Thr Asn Leu Val Glu Lys Gly Arg 1820 1825 1830 Arg Lys Cys Asp Gln Tyr Trp Pro Ala Asp Gly Ser Glu Glu Tyr 1835 1840 1845 Gly Asn Phe Leu Val Thr Gln Lys Ser Val Gln Val Leu Ala Tyr 1850 1855 1860 Tyr Thr Val Arg Asn Phe Thr Leu Arg Asn Thr Lys Ile Lys Lys 1865 1870 1875 Gly Ser Gln Lys Gly Arg Pro Ser Gly Arg Val Val Thr Gln Tyr 1880 1885 1890 His Tyr Thr Gln Trp Pro Asp Met Gly Val Pro Glu Tyr Ser Leu 1895 1900 1905 Pro Val Leu Thr Phe Val Arg Lys Ala Ala Tyr Ala Lys Arg His 1910 1915 1920 Ala Val Gly Pro Val Val Val His Cys Ser Ala Gly Val Gly Arg 1925 1930 1935 Thr Gly Thr Tyr Ile Val Leu Asp Ser Met Leu Gln Gln Ile Gln 1940 1945 1950 His Glu Gly Thr Val Asn Ile Phe Gly Phe Leu Lys His Ile Arg 1955 1960 1965 Ser Gln Arg Asn Tyr Leu Val Gln Thr Glu Glu Gln Tyr Val Phe 1970 1975 1980 Ile His Asp Thr Leu Val Glu Ala Ile Leu Ser Lys Glu Thr Glu 1985 1990 1995 Val Leu Asp Ser His Ile His Ala Tyr Val Asn Ala Leu Leu Ile 2000 2005 2010 Pro Gly Pro Ala Gly Lys Thr Lys Leu Glu Lys Gln Phe Gln Leu 2015 2020 2025 Leu Ser Gln Ser Asn Ile Gln Gln Ser Asp Tyr Ser Ala Ala Leu 2030 2035 2040 Lys Gln Cys Asn Arg Glu Lys Asn Arg Thr Ser Ser Ile Ile Pro 2045 2050 2055 Val Glu Arg Ser Arg Val Gly Ile Ser Ser Leu Ser Gly Glu Gly 2060 2065 2070 Thr Asp Tyr Ile Asn Ala Ser Tyr Ile Met Gly Tyr Tyr Gln Ser 2075 2080 2085 Asn Glu Phe Ile Ile Thr Gln His Pro Leu Leu His Thr Ile Lys 2090 2095 2100 Asp Phe Trp Arg Met Ile Trp Asp His Asn Ala Gln Leu Val Val 2105 2110 2115 Met Ile Pro Asp Gly Gln Asn Met Ala Glu Asp Glu Phe Val Tyr 2120 2125 2130 Trp Pro Asn Lys Asp Glu Pro Ile Asn Cys Glu Ser Phe Lys Val 2135 2140 2145 Thr Leu Met Ala Glu Glu His Lys Cys Leu Ser Asn Glu Glu Lys 2150 2155 2160 Leu Ile Ile Gln Asp Phe Ile Leu Glu Ala Thr Gln Asp Asp Tyr 2165 2170 2175 Val Leu Glu Val Arg His Phe Gln Cys Pro Lys Trp Pro Asn Pro 2180 2185 2190 Asp Ser Pro Ile Ser Lys Thr Phe Glu Leu Ile Ser Val Ile Lys 2195 2200 2205 Glu Glu Ala Ala Asn Arg Asp Gly Pro Met Ile Val His Asp Glu 2210 2215 2220 His Gly Gly Val Thr Ala Gly Thr Phe Cys Ala Leu Thr Thr Leu 2225 2230 2235 Met His Gln Leu Glu Lys Glu Asn Ser Val Asp Val Tyr Gln Val 2240 2245 2250 Ala Lys Met Ile Asn Leu Met Arg Pro Gly Val Phe Ala Asp Ile 2255 2260 2265 Glu Gln Tyr Gln Phe Leu Tyr Lys Val Ile Leu Ser Leu Val Ser 2270 2275 2280 Thr Arg Gln Glu Glu Asn Pro Ser Thr Ser Leu Asp Ser Asn Gly 2285 2290 2295 Ala Ala Leu Pro Asp Gly Asn Ile Ala Glu Ser Leu Glu Ser Leu 2300 2305 2310 Val 7 1518 DNA Homo sapiens gene (1)..(1518) Angiopoietin-like 2 (ANGPTL2), mRNA 7 aaccaccatt ttgcaaggac c atg agg cca ctg tgc gtg aca tgc tgg tgg 51 Met Arg Pro Leu Cys Val Thr Cys Trp Trp 1 5 10 ctc gga ctg ctg gct gcc atg gga gct gtt gca ggc cag gag gac ggt 99 Leu Gly Leu Leu Ala Ala Met Gly Ala Val Ala Gly Gln Glu Asp Gly 15 20 25 ttt gag ggc act gag gag ggc tcg cca aga gag ttc att tac cta aac 147 Phe Glu Gly Thr Glu Glu Gly Ser Pro Arg Glu Phe Ile Tyr Leu Asn 30 35 40 agg tac aag cgg gcg ggc gag tcc cag gac aag tgc acc tac acc ttc 195 Arg Tyr Lys Arg Ala Gly Glu Ser Gln Asp Lys Cys Thr Tyr Thr Phe 45 50 55 att gtg ccc cag cag cgg gtc acg ggt gcc atc tgc gtc aac tcc aag 243 Ile Val Pro Gln Gln Arg Val Thr Gly Ala Ile Cys Val Asn Ser Lys 60 65 70 gag cct gag gtg ctt ctg gag aac cga gtg cat aag cag gag cta gag 291 Glu Pro Glu Val Leu Leu Glu Asn Arg Val His Lys Gln Glu Leu Glu 75 80 85 90 ctg ctc aac aat gag ctg ctc aag cag aag cgg cag atc gag aca ctg 339 Leu Leu Asn Asn Glu Leu Leu Lys Gln Lys Arg Gln Ile Glu Thr Leu 95 100 105 cag cag ctg gtg gag gtg gac ggc ggc att gtg agc gag gtg aag ctg 387 Gln Gln Leu Val Glu Val Asp Gly Gly Ile Val Ser Glu Val Lys Leu 110 115 120 ctg cgc aag gag agc cgc aac atg aac tcg cgg gtc acg cag ctc tac 435 Leu Arg Lys Glu Ser Arg Asn Met Asn Ser Arg Val Thr Gln Leu Tyr 125 130 135 atg cag ctc ctg cac gag atc atc cgc aag cgg gac aac gcg ttg gag 483 Met Gln Leu Leu His Glu Ile Ile Arg Lys Arg Asp Asn Ala Leu Glu 140 145 150 ctc tcc cag ctg gag aac agg atc ctg aac cag aca gcc gac atg ctg 531 Leu Ser Gln Leu Glu Asn Arg Ile Leu Asn Gln Thr Ala Asp Met Leu 155 160 165 170 cag ctg gcc agc aag tac aag gac ctg gag cac aag tac cag cac ctg 579 Gln Leu Ala Ser Lys Tyr Lys Asp Leu Glu His Lys Tyr Gln His Leu 175 180 185 gcc aca ctg gcc cac aac caa tca gag atc atc gcg cag ctt gag gag 627 Ala Thr Leu Ala His Asn Gln Ser Glu Ile Ile Ala Gln Leu Glu Glu 190 195 200 cac tgc cag agg gtg ccc tcg gcc agg ccc gtc ccc cag cca ccc ccc 675 His Cys Gln Arg Val Pro Ser Ala Arg Pro Val Pro Gln Pro Pro Pro 205 210 215 gct gcc ccg ccc cgg gtc tac caa cca ccc acc tac aac cgc atc atc 723 Ala Ala Pro Pro Arg Val Tyr Gln Pro Pro Thr Tyr Asn Arg Ile Ile 220 225 230 aac cag atc tct acc aac gag atc cag agt gac cag aac ctg aag gtg 771 Asn Gln Ile Ser Thr Asn Glu Ile Gln Ser Asp Gln Asn Leu Lys Val 235 240 245 250 ctg cca ccc cct ctg ccc act atg ccc act ctc acc agc ctc cca tct 819 Leu Pro Pro Pro Leu Pro Thr Met Pro Thr Leu Thr Ser Leu Pro Ser 255 260 265 tcc acc gac aag ccg tcg ggc cca tgg aga gac tgc ctg cag gcc ctg 867 Ser Thr Asp Lys Pro Ser Gly Pro Trp Arg Asp Cys Leu Gln Ala Leu 270 275 280 gag gat ggc cac gac acc agc tcc atc tac ctg gtg aag ccg gag aac 915 Glu Asp Gly His Asp Thr Ser Ser Ile Tyr Leu Val Lys Pro Glu Asn 285 290 295 acc aac cgc ctc atg cag gtg tgg tgc gac cag aga cac gac ccc ggg 963 Thr Asn Arg Leu Met Gln Val Trp Cys Asp Gln Arg His Asp Pro Gly 300 305 310 ggc tgg acc gtc atc cag aga cgc ctg gat ggc tct gtt aac ttc ttc 1011 Gly Trp Thr Val Ile Gln Arg Arg Leu Asp Gly Ser Val Asn Phe Phe 315 320 325 330 agg aac tgg gag acg tac aag caa ggg ttt ggg aac att gat ggc gaa 1059 Arg Asn Trp Glu Thr Tyr Lys Gln Gly Phe Gly Asn Ile Asp Gly Glu 335 340 345 tac tgg ctg ggc ctg gag aac att tac tgg ctg acg aac caa ggc aac 1107 Tyr Trp Leu Gly Leu Glu Asn Ile Tyr Trp Leu Thr Asn Gln Gly Asn 350 355 360 tac aaa ctc ctg gtg acc atg gag gac tgg tcc ggc cgc aaa gtc ttt 1155 Tyr Lys Leu Leu Val Thr Met Glu Asp Trp Ser Gly Arg Lys Val Phe 365 370 375 gca gaa tac gcc agt ttc cgc ctg gaa cct gag agc gag tat tat aag 1203 Ala Glu Tyr Ala Ser Phe Arg Leu Glu Pro Glu Ser Glu Tyr Tyr Lys 380 385 390 ctg cgg ctg ggg cgc tac cat ggc aat gcg ggt gac tcc ttt aca tgg 1251 Leu Arg Leu Gly Arg Tyr His Gly Asn Ala Gly Asp Ser Phe Thr Trp 395 400 405 410 cac aac ggc aag cag ttc acc acc ctg gac aga gat cat gat gtc tac 1299 His Asn Gly Lys Gln Phe Thr Thr Leu Asp Arg Asp His Asp Val Tyr 415 420 425 aca gga aac tgt gcc cac tac cag aag gga ggc tgg tgg tat aac gcc 1347 Thr Gly Asn Cys Ala His Tyr Gln Lys Gly Gly Trp Trp Tyr Asn Ala 430 435 440 tgt gcc cac tcc aac ctc aac ggg gtc tgg tac cgc ggg ggc cat tac 1395 Cys Ala His Ser Asn Leu Asn Gly Val Trp Tyr Arg Gly Gly His Tyr 445 450 455 cgg agc cgc tac cag gac gga gtc tac tgg gct gag ttc cga gga ggc 1443 Arg Ser Arg Tyr Gln Asp Gly Val Tyr Trp Ala Glu Phe Arg Gly Gly 460 465 470 tct tac tca ctc aag aaa gtg gtg atg atg atc cga ccg aac ccc aac 1491 Ser Tyr Ser Leu Lys Lys Val Val Met Met Ile Arg Pro Asn Pro Asn 475 480 485 490 acc ttc cac taa gccagctccc cctcc 1518 Thr Phe His 8 493 PRT Homo sapiens gene (1)..(493) Angiopoietin-like 2 (ANGPTL2), protein 8 Met Arg Pro Leu Cys Val Thr Cys Trp Trp Leu Gly Leu Leu Ala Ala 1 5 10 15 Met Gly Ala Val Ala Gly Gln Glu Asp Gly Phe Glu Gly Thr Glu Glu 20 25 30 Gly Ser Pro Arg Glu Phe Ile Tyr Leu Asn Arg Tyr Lys Arg Ala Gly 35 40 45 Glu Ser Gln Asp Lys Cys Thr Tyr Thr Phe Ile Val Pro Gln Gln Arg 50 55 60 Val Thr Gly Ala Ile Cys Val Asn Ser Lys Glu Pro Glu Val Leu Leu 65 70 75 80 Glu Asn Arg Val His Lys Gln Glu Leu Glu Leu Leu Asn Asn Glu Leu 85 90 95 Leu Lys Gln Lys Arg Gln Ile Glu Thr Leu Gln Gln Leu Val Glu Val 100 105 110 Asp Gly Gly Ile Val Ser Glu Val Lys Leu Leu Arg Lys Glu Ser Arg 115 120 125 Asn Met Asn Ser Arg Val Thr Gln Leu Tyr Met Gln Leu Leu His Glu 130 135 140 Ile Ile Arg Lys Arg Asp Asn Ala Leu Glu Leu Ser Gln Leu Glu Asn 145 150 155 160 Arg Ile Leu Asn Gln Thr Ala Asp Met Leu Gln Leu Ala Ser Lys Tyr 165 170 175 Lys Asp Leu Glu His Lys Tyr Gln His Leu Ala Thr Leu Ala His Asn 180 185 190 Gln Ser Glu Ile Ile Ala Gln Leu Glu Glu His Cys Gln Arg Val Pro 195 200 205 Ser Ala Arg Pro Val Pro Gln Pro Pro Pro Ala Ala Pro Pro Arg Val 210 215 220 Tyr Gln Pro Pro Thr Tyr Asn Arg Ile Ile Asn Gln Ile Ser Thr Asn 225 230 235 240 Glu Ile Gln Ser Asp Gln Asn Leu Lys Val Leu Pro Pro Pro Leu Pro 245 250 255 Thr Met Pro Thr Leu Thr Ser Leu Pro Ser Ser Thr Asp Lys Pro Ser 260 265 270 Gly Pro Trp Arg Asp Cys Leu Gln Ala Leu Glu Asp Gly His Asp Thr 275 280 285 Ser Ser Ile Tyr Leu Val Lys Pro Glu Asn Thr Asn Arg Leu Met Gln 290 295 300 Val Trp Cys Asp Gln Arg His Asp Pro Gly Gly Trp Thr Val Ile Gln 305 310 315 320 Arg Arg Leu Asp Gly Ser Val Asn Phe Phe Arg Asn Trp Glu Thr Tyr 325 330 335 Lys Gln Gly Phe Gly Asn Ile Asp Gly Glu Tyr Trp Leu Gly Leu Glu 340 345 350 Asn Ile Tyr Trp Leu Thr Asn Gln Gly Asn Tyr Lys Leu Leu Val Thr 355 360 365 Met Glu Asp Trp Ser Gly Arg Lys Val Phe Ala Glu Tyr Ala Ser Phe 370 375 380 Arg Leu Glu Pro Glu Ser Glu Tyr Tyr Lys Leu Arg Leu Gly Arg Tyr 385 390 395 400 His Gly Asn Ala Gly Asp Ser Phe Thr Trp His Asn Gly Lys Gln Phe 405 410 415 Thr Thr Leu Asp Arg Asp His Asp Val Tyr Thr Gly Asn Cys Ala His 420 425 430 Tyr Gln Lys Gly Gly Trp Trp Tyr Asn Ala Cys Ala His Ser Asn Leu 435 440 445 Asn Gly Val Trp Tyr Arg Gly Gly His Tyr Arg Ser Arg Tyr Gln Asp 450 455 460 Gly Val Tyr Trp Ala Glu Phe Arg Gly Gly Ser Tyr Ser Leu Lys Lys 465 470 475 480 Val Val Met Met Ile Arg Pro Asn Pro Asn Thr Phe His 485 490 9 2133 DNA Homo sapiens gene (1)..(2133) Human SPARC/osteonectin mRNA 9 cgggagagcg cgctctgcct gccgcctgcc tgcctgccac tgagggttcc cagcacc 57 atg agg gcc tgg atc ttc ttt ctc ctt tgc ctg gcc ggg agg gcc ttg 105 Met Arg Ala Trp Ile Phe Phe Leu Leu Cys Leu Ala Gly Arg Ala Leu 1 5 10 15 gca gcc cct cag caa gaa gcc ctg cct gat gag aca gag gtg gtg gaa 153 Ala Ala Pro Gln Gln Glu Ala Leu Pro Asp Glu Thr Glu Val Val Glu 20 25 30 gaa act gtg gca gag gtg act gag gta tct gtg gga gct aat cct gtc 201 Glu Thr Val Ala Glu Val Thr Glu Val Ser Val Gly Ala Asn Pro Val 35 40 45 cag gtg gaa gta gga gaa ttt gat gat ggt gca gag gaa acc gaa gag 249 Gln Val Glu Val Gly Glu Phe Asp Asp Gly Ala Glu Glu Thr Glu Glu 50 55 60 gag gtg gtg gcg gaa aat ccc tgc cag aac cac cac tgc aaa cac ggc 297 Glu Val Val Ala Glu Asn Pro Cys Gln Asn His His Cys Lys His Gly 65 70 75 80 aag gtg tgc gag ctg gat gag aac aac acc ccc atg tgc gtg tgc cag 345 Lys Val Cys Glu Leu Asp Glu Asn Asn Thr Pro Met Cys Val Cys Gln 85 90 95 gac ccc acc agc tgc cca gcc ccc att ggc gag ttt gag aag gtg tgc 393 Asp Pro Thr Ser Cys Pro Ala Pro Ile Gly Glu Phe Glu Lys Val Cys 100 105 110 agc aat gac aac aag acc ttc gac tct tcc tgc cac ttc ttt gcc aca 441 Ser Asn Asp Asn Lys Thr Phe Asp Ser Ser Cys His Phe Phe Ala Thr 115 120 125 aag tgc acc ctg gag ggc acc aag aag ggc cac aag ctc cac ctg gac 489 Lys Cys Thr Leu Glu Gly Thr Lys Lys Gly His Lys Leu His Leu Asp 130 135 140 tac atc ggg cct tgc aaa tac atc ccc cct tgc ctg gac tct gag ctg 537 Tyr Ile Gly Pro Cys Lys Tyr Ile Pro Pro Cys Leu Asp Ser Glu Leu 145 150 155 160 acc gaa ttc ccc ctg cgc atg cgg gac tgg ctc aag aac gtc ctg gtc 585 Thr Glu Phe Pro Leu Arg Met Arg Asp Trp Leu Lys Asn Val Leu Val 165 170 175 acc ctg tat gag agg gat gag gac aac aac ctt ctg act gag aag cag 633 Thr Leu Tyr Glu Arg Asp Glu Asp Asn Asn Leu Leu Thr Glu Lys Gln 180 185 190 aag ctg cgg gtg aag aag atc cat gag aat gag aag cgc ctg gag gca 681 Lys Leu Arg Val Lys Lys Ile His Glu Asn Glu Lys Arg Leu Glu Ala 195 200 205 gga gac cac ccc gtg gag ctg ctg gcc cgg gac ttc gag aag aac tat 729 Gly Asp His Pro Val Glu Leu Leu Ala Arg Asp Phe Glu Lys Asn Tyr 210 215 220 aac atg tac atc ttc cct gta cac tgg cag ttc ggc cag ctg gac cag 777 Asn Met Tyr Ile Phe Pro Val His Trp Gln Phe Gly Gln Leu Asp Gln 225 230 235 240 cac ccc att gac ggg tac ctc tcc cac acc gag ctg gct cca ctg cgt 825 His Pro Ile Asp Gly Tyr Leu Ser His Thr Glu Leu Ala Pro Leu Arg 245 250 255 gct ccc ctc atc ccc atg gag cat tgc acc acc cgc ttt ttc gag acc 873 Ala Pro Leu Ile Pro Met Glu His Cys Thr Thr Arg Phe Phe Glu Thr 260 265 270 tgt gac ctg gac aat gac aag tac atc gcc ctg gat gag tgg gcc ggc 921 Cys Asp Leu Asp Asn Asp Lys Tyr Ile Ala Leu Asp Glu Trp Ala Gly 275 280 285 tgc ttc ggc atc aag cag aag gat atc gac aag gat ctt gtg atc taa 969 Cys Phe Gly Ile Lys Gln Lys Asp Ile Asp Lys Asp Leu Val Ile 290 295 300 atccactcct tccacagtac cggattctct ctttaaccct ccccttcgtg tttcccccaa 1029 tgtttaaaat gtttggatgg tttgttgttc tgcctggaga caaggtgcta acatagattt 1089 aagtgaatac attaacggtg ctaaaaatga aaattctaac ccaagacatg acattcttag 1149 ctgtaactta actattaagg ccttttccac acgcattaat agtcccattt ttctcttgcc 1209 atttgtagct ttgcccattg tcttattggc acatgggtgg acacggatct gctgggctct 1269 gccttaaaca cacattgcag cttcaacttt tctctttagt gttctgtttg aaactaatac 1329 ttaccgagtc agactttgtg ttcatttcat ttcagggtct tggctgcctg tgggcttccc 1389 caggtggcct ggaggtgggc aaagggaagt aacagacaca cgatgttgtc aaggatggtt 1449 ttgggactag aggctcagtg gtgggagaga tccctgcaga atccaccaac cagaacgtgg 1509 tttgcctgag gctgtaactg agagaaagat tctggggctg tcttatgaaa atatagacat 1569 tctcacataa gcccagttca tcaccatttc ctcctttacc tttcagtgca gtttcttttc 1629 acattaggct gttggttcaa acttttggga gcacggactg tcagttctct gggaagtggt 1689 cagcgcatcc tgcagggctt ctcctcctct gtcttttgga gaaccagggc tcttctcagg 1749 ggctctaggg actgccaggc tgtttcagcc aggaaggcca aaatcaagag tgagatgtag 1809 aaagttgtaa aatagaaaaa gtggagttgg tgaatcggtt gttctttcct cacatttgga 1869 tgattgtcat aaggttttta gcatgttcct ccttttcttc accctcccct ttgttcttct 1929 attaatcaag agaaacttca aagttaatgg gatggtcgga tctcacaggc tgagaactcg 1989 ttcacctcca agcatttcat gaaaaagctg cttcttatta atcatacaaa ctctcaccat 2049 gatgtgaaga gtttcacaaa tctttcaaaa taaaaagtaa tgacttagaa actgaaaaaa 2109 aaaaaaaaaa aaaaaaaaaa aaaa 2133 10 303 PRT Homo sapiens SIGNAL (1)..(17) 10 Met Arg Ala Trp Ile Phe Phe Leu Leu Cys Leu Ala Gly Arg Ala Leu 1 5 10 15 Ala Ala Pro Gln Gln Glu Ala Leu Pro Asp Glu Thr Glu Val Val Glu 20 25 30 Glu Thr Val Ala Glu Val Thr Glu Val Ser Val Gly Ala Asn Pro Val 35 40 45 Gln Val Glu Val Gly Glu Phe Asp Asp Gly Ala Glu Glu Thr Glu Glu 50 55 60 Glu Val Val Ala Glu Asn Pro Cys Gln Asn His His Cys Lys His Gly 65 70 75 80 Lys Val Cys Glu Leu Asp Glu Asn Asn Thr Pro Met Cys Val Cys Gln 85 90 95 Asp Pro Thr Ser Cys Pro Ala Pro Ile Gly Glu Phe Glu Lys Val Cys 100 105 110 Ser Asn Asp Asn Lys Thr Phe Asp Ser Ser Cys His Phe Phe Ala Thr 115 120 125 Lys Cys Thr Leu Glu Gly Thr Lys Lys Gly His Lys Leu His Leu Asp 130 135 140 Tyr Ile Gly Pro Cys Lys Tyr Ile Pro Pro Cys Leu Asp Ser Glu Leu 145 150 155 160 Thr Glu Phe Pro Leu Arg Met Arg Asp Trp Leu Lys Asn Val Leu Val 165 170 175 Thr Leu Tyr Glu Arg Asp Glu Asp Asn Asn Leu Leu Thr Glu Lys Gln 180 185 190 Lys Leu Arg Val Lys Lys Ile His Glu Asn Glu Lys Arg Leu Glu Ala 195 200 205 Gly Asp His Pro Val Glu Leu Leu Ala Arg Asp Phe Glu Lys Asn Tyr 210 215 220 Asn Met Tyr Ile Phe Pro Val His Trp Gln Phe Gly Gln Leu Asp Gln 225 230 235 240 His Pro Ile Asp Gly Tyr Leu Ser His Thr Glu Leu Ala Pro Leu Arg 245 250 255 Ala Pro Leu Ile Pro Met Glu His Cys Thr Thr Arg Phe Phe Glu Thr 260 265 270 Cys Asp Leu Asp Asn Asp Lys Tyr Ile Ala Leu Asp Glu Trp Ala Gly 275 280 285 Cys Phe Gly Ile Lys Gln Lys Asp Ile Asp Lys Asp Leu Val Ile 290 295 300 11 4586 DNA Homo sapiens gene (1)..(4586) mRNA for met proto-oncogene 11 gaattccgcc ctcgccgccc gcggcgcccc gagcgctttg tgagcagatg cggagccgag 60 tggagggcgc gagccagatg cggggcgaca gctgacttgc tgagaggagg cggggaggcg 120 cggagcgcgc gtgtggtcct tgcgccgctg acttctccac tggttcctgg gcaccgaaag 180 ataaacctct cata atg aag gcc ccc gct gtg ctt gca cct ggc atc ctc 230 Met Lys Ala Pro Ala Val Leu Ala Pro Gly Ile Leu 1 5 10 gtg ctc ctg ttt acc ttg gtg cag agg agc aat ggg gag tgt aaa gag 278 Val Leu Leu Phe Thr Leu Val Gln Arg Ser Asn Gly Glu Cys Lys Glu 15 20 25 gca cta gca aag tcc gag atg aat gtg aat atg aag tat cag ctt ccc 326 Ala Leu Ala Lys Ser Glu Met Asn Val Asn Met Lys Tyr Gln Leu Pro 30 35 40 aac ttc acc gcg gaa aca ccc atc cag aat gtc att cta cat gag cat 374 Asn Phe Thr Ala Glu Thr Pro Ile Gln Asn Val Ile Leu His Glu His 45 50 55 60 cac att ttc ctt ggt gcc act aac tac att tat gtt tta aat gag gaa 422 His Ile Phe Leu Gly Ala Thr Asn Tyr Ile Tyr Val Leu Asn Glu Glu 65 70 75 gac ctt cag aag gtt gct gag tac aag act ggg cct gtg ctg gaa cac 470 Asp Leu Gln Lys Val Ala Glu Tyr Lys Thr Gly Pro Val Leu Glu His 80 85 90 cca gat tgt ttc cca tgt cag gac tgc agc agc aaa gcc aat tta tca 518 Pro Asp Cys Phe Pro Cys Gln Asp Cys Ser Ser Lys Ala Asn Leu Ser 95 100 105 gga ggt gtt tgg aaa gat aac atc aac atg gct cta gtt gtc gac acc 566 Gly Gly Val Trp Lys Asp Asn Ile Asn Met Ala Leu Val Val Asp Thr 110 115 120 tac tat gat gat caa ctc att agc tgt ggc agc gtc aac aga ggg acc 614 Tyr Tyr Asp Asp Gln Leu Ile Ser Cys Gly Ser Val Asn Arg Gly Thr 125 130 135 140 tgc cag cga cat gtc ttt ccc cac aat cat act gct gac ata cag tcg 662 Cys Gln Arg His Val Phe Pro His Asn His Thr Ala Asp Ile Gln Ser 145 150 155 gag gtt cac tgc ata ttc tcc cca cag ata gaa gag ccc agc cag tgt 710 Glu Val His Cys Ile Phe Ser Pro Gln Ile Glu Glu Pro Ser Gln Cys 160 165 170 cct gac tgt gtg gtg agc gcc ctg gga gcc aaa gtc ctt tca tct gta 758 Pro Asp Cys Val Val Ser Ala Leu Gly Ala Lys Val Leu Ser Ser Val 175 180 185 aag gac cgg ttc atc aac ttc ttt gta ggc aat acc ata aat tct tct 806 Lys Asp Arg Phe Ile Asn Phe Phe Val Gly Asn Thr Ile Asn Ser Ser 190 195 200 tat ttc cca gat cat cca ttg cat tcg ata tca gtg aga agg cta aag 854 Tyr Phe Pro Asp His Pro Leu His Ser Ile Ser Val Arg Arg Leu Lys 205 210 215 220 gaa acg aaa gat ggt ttt atg ttt ttg acg gac cag tcc tac att gat 902 Glu Thr Lys Asp Gly Phe Met Phe Leu Thr Asp Gln Ser Tyr Ile Asp 225 230 235 gtt tta cct gag ttc aga gat tct tac ccc att aag tat gtc cat gcc 950 Val Leu Pro Glu Phe Arg Asp Ser Tyr Pro Ile Lys Tyr Val His Ala 240 245 250 ttt gaa agc aac aat ttt att tac ttc ttg acg gtc caa agg gaa act 998 Phe Glu Ser Asn Asn Phe Ile Tyr Phe Leu Thr Val Gln Arg Glu Thr 255 260 265 cta gat gct cag act ttt cac aca aga ata atc agg ttc tgt tcc ata 1046 Leu Asp Ala Gln Thr Phe His Thr Arg Ile Ile Arg Phe Cys Ser Ile 270 275 280 aac tct gga ttg cat tcc tac atg gaa atg cct ctg gag tgt att ctc 1094 Asn Ser Gly Leu His Ser Tyr Met Glu Met Pro Leu Glu Cys Ile Leu 285 290 295 300 aca gaa aag aga aaa aag aga tcc aca aag aag gaa gtg ttt aat ata 1142 Thr Glu Lys Arg Lys Lys Arg Ser Thr Lys Lys Glu Val Phe Asn Ile 305 310 315 ctt cag gct gcg tat gtc agc aag cct ggg gcc cag ctt gct aga caa 1190 Leu Gln Ala Ala Tyr Val Ser Lys Pro Gly Ala Gln Leu Ala Arg Gln 320 325 330 ata gga gcc agc ctg aat gat gac att ctt ttc ggg gtg ttc gca caa 1238 Ile Gly Ala Ser Leu Asn Asp Asp Ile Leu Phe Gly Val Phe Ala Gln 335 340 345 agc aag cca gat tct gcc gaa cca atg gat cga tct gcc atg tgt gca 1286 Ser Lys Pro Asp Ser Ala Glu Pro Met Asp Arg Ser Ala Met Cys Ala 350 355 360 ttc cct atc aaa tat gtc aac gac ttc ttc aac aag atc gtc aac aaa 1334 Phe Pro Ile Lys Tyr Val Asn Asp Phe Phe Asn Lys Ile Val Asn Lys 365 370 375 380 aac aat gtg aga tgt ctc cag cat ttt tac gga ccc aat cat gag cac 1382 Asn Asn Val Arg Cys Leu Gln His Phe Tyr Gly Pro Asn His Glu His 385 390 395 tgc ttt aat agg aca ctt ctg aga aat tca tca ggc tgt gaa gcg cgc 1430 Cys Phe Asn Arg Thr Leu Leu Arg Asn Ser Ser Gly Cys Glu Ala Arg 400 405 410 cgt gat gaa tat cga aca gag ttt acc aca gct ttg cag cgc gtt gac 1478 Arg Asp Glu Tyr Arg Thr Glu Phe Thr Thr Ala Leu Gln Arg Val Asp 415 420 425 tta ttc atg ggt caa ttc agc gaa gtc ctc tta aca tct ata tcc acc 1526 Leu Phe Met Gly Gln Phe Ser Glu Val Leu Leu Thr Ser Ile Ser Thr 430 435 440 ttc att aaa gga gac ctc acc ata gct aat ctt ggg aca tca gag ggt 1574 Phe Ile Lys Gly Asp Leu Thr Ile Ala Asn Leu Gly Thr Ser Glu Gly 445 450 455 460 cgc ttc atg cag gtt gtg gtt tct cga tca gga cca tca acc cct cat 1622 Arg Phe Met Gln Val Val Val Ser Arg Ser Gly Pro Ser Thr Pro His 465 470 475 gtg aat ttt ctc ctg gac tcc cat cca gtg tct cca gaa gtg att gtg 1670 Val Asn Phe Leu Leu Asp Ser His Pro Val Ser Pro Glu Val Ile Val 480 485 490 gag cat aca tta aac caa aat ggc tac aca ctg gtt atc act ggg aag 1718 Glu His Thr Leu Asn Gln Asn Gly Tyr Thr Leu Val Ile Thr Gly Lys 495 500 505 aag atc acg aag atc cca ttg aat ggc ttg ggc tgc aga cat ttc cag 1766 Lys Ile Thr Lys Ile Pro Leu Asn Gly Leu Gly Cys Arg His Phe Gln 510 515 520 tcc tgc agt caa tgc ctc tct gcc cca ccc ttt gtt cag tgt ggc tgg 1814 Ser Cys Ser Gln Cys Leu Ser Ala Pro Pro Phe Val Gln Cys Gly Trp 525 530 535 540 tgc cac gac aaa tgt gtg cga tcg gag gaa tgc ctg agc ggg aca tgg 1862 Cys His Asp Lys Cys Val Arg Ser Glu Glu Cys Leu Ser Gly Thr Trp 545 550 555 act caa cag atc tgt ctg cct gca atc tac aag gtt ttc cca aat agt 1910 Thr Gln Gln Ile Cys Leu Pro Ala Ile Tyr Lys Val Phe Pro Asn Ser 560 565 570 gca ccc ctt gaa gga ggg aca agg ctg acc ata tgt ggc tgg gac ttt 1958 Ala Pro Leu Glu Gly Gly Thr Arg Leu Thr Ile Cys Gly Trp Asp Phe 575 580 585 gga ttt cgg agg aat aat aaa ttt gat tta aag aaa act aga gtt ctc 2006 Gly Phe Arg Arg Asn Asn Lys Phe Asp Leu Lys Lys Thr Arg Val Leu 590 595 600 ctt gga aat gag agc tgc acc ttg act tta agt gag agc acg atg aat 2054 Leu Gly Asn Glu Ser Cys Thr Leu Thr Leu Ser Glu Ser Thr Met Asn 605 610 615 620 aca ttg aaa tgc aca gtt ggt cct gcc atg aat aag cat ttc aat atg 2102 Thr Leu Lys Cys Thr Val Gly Pro Ala Met Asn Lys His Phe Asn Met 625 630 635 tcc ata att att tca aat ggc cac ggg aca aca caa tac agt aca ttc 2150 Ser Ile Ile Ile Ser Asn Gly His Gly Thr Thr Gln Tyr Ser Thr Phe 640 645 650 tcc tat gtg gat cct gta ata aca agt att tcg ccg aaa tac ggt cct 2198 Ser Tyr Val Asp Pro Val Ile Thr Ser Ile Ser Pro Lys Tyr Gly Pro 655 660 665 atg gct ggt ggc act tta ctt act tta act gga aat tac cta aac agt 2246 Met Ala Gly Gly Thr Leu Leu Thr Leu Thr Gly Asn Tyr Leu Asn Ser 670 675 680 ggg aat tct aga cac att tca att ggt gga aaa aca tgt act tta aaa 2294 Gly Asn Ser Arg His Ile Ser Ile Gly Gly Lys Thr Cys Thr Leu Lys 685 690 695 700 agt gtg tca aac agt att ctt gaa tgt tat acc cca gcc caa acc att 2342 Ser Val Ser Asn Ser Ile Leu Glu Cys Tyr Thr Pro Ala Gln Thr Ile 705 710 715 tca act gag ttt gct gtt aaa ttg aaa att gac tta gcc aac cga gag 2390 Ser Thr Glu Phe Ala Val Lys Leu Lys Ile Asp Leu Ala Asn Arg Glu 720 725 730 aca agc atc ttc agt tac cgt gaa gat ccc att gtc tat gaa att cat 2438 Thr Ser Ile Phe Ser Tyr Arg Glu Asp Pro Ile Val Tyr Glu Ile His 735 740 745 cca acc aaa tct ttt att agt ggt ggg agc aca ata aca ggt gtt ggg 2486 Pro Thr Lys Ser Phe Ile Ser Gly Gly Ser Thr Ile Thr Gly Val Gly 750 755 760 aaa aac ctg aat tca gtt agt gtc ccg aga atg gtc ata aat gtg cat 2534 Lys Asn Leu Asn Ser Val Ser Val Pro Arg Met Val Ile Asn Val His 765 770 775 780 gaa gca gga agg aac ttt aca gtg gca tgt caa cat cgc tct aat tca 2582 Glu Ala Gly Arg Asn Phe Thr Val Ala Cys Gln His Arg Ser Asn Ser 785 790 795 gag ata atc tgt tgt acc act cct tcc ctg caa cag ctg aat ctg caa 2630 Glu Ile Ile Cys Cys Thr Thr Pro Ser Leu Gln Gln Leu Asn Leu Gln 800 805 810 ctc ccc ctg aaa acc aaa gcc ttt ttc atg tta gat ggg atc ctt tcc 2678 Leu Pro Leu Lys Thr Lys Ala Phe Phe Met Leu Asp Gly Ile Leu Ser 815 820 825 aaa tac ttt gat ctc att tat gta cat aat cct gtg ttt aag cct ttt 2726 Lys Tyr Phe Asp Leu Ile Tyr Val His Asn Pro Val Phe Lys Pro Phe 830 835 840 gaa aag cca gtg atg atc tca atg ggc aat gaa aat gta ctg gaa att 2774 Glu Lys Pro Val Met Ile Ser Met Gly Asn Glu Asn Val Leu Glu Ile 845 850 855 860 aag gga aat gat att gac cct gaa gca gtt aaa ggt gaa gtg tta aaa 2822 Lys Gly Asn Asp Ile Asp Pro Glu Ala Val Lys Gly Glu Val Leu Lys 865 870 875 gtt gga aat aag agc tgt gag aat ata cac tta cat tct gaa gcc gtt 2870 Val Gly Asn Lys Ser Cys Glu Asn Ile His Leu His Ser Glu Ala Val 880 885 890 tta tgc acg gtc ccc aat gac ctg ctg aaa ttg aac agc gag cta aat 2918 Leu Cys Thr Val Pro Asn Asp Leu Leu Lys Leu Asn Ser Glu Leu Asn 895 900 905 ata gag tgg aag caa gca att tct tca acc gtc ctt gga aaa gta ata 2966 Ile Glu Trp Lys Gln Ala Ile Ser Ser Thr Val Leu Gly Lys Val Ile 910 915 920 gtt caa cca gat cag aat ttc aca gga ttg att gct ggt gtt gtc tca 3014 Val Gln Pro Asp Gln Asn Phe Thr Gly Leu Ile Ala Gly Val Val Ser 925 930 935 940 ata tca aca gca ctg tta tta cta ctt ggg ttt ttc ctg tgg ctg aaa 3062 Ile Ser Thr Ala Leu Leu Leu Leu Leu Gly Phe Phe Leu Trp Leu Lys 945 950 955 aag aga aag caa att aaa gat ctg ggc agt gaa tta gtt cgc tac gat 3110 Lys Arg Lys Gln Ile Lys Asp Leu Gly Ser Glu Leu Val Arg Tyr Asp 960 965 970 gca aga gta cac act cct cat ttg gat agg ctt gta agt gcc cga agt 3158 Ala Arg Val His Thr Pro His Leu Asp Arg Leu Val Ser Ala Arg Ser 975 980 985 gta agc cca act aca gaa atg gtt tca aat gaa tct gta gac tac cga 3206 Val Ser Pro Thr Thr Glu Met Val Ser Asn Glu Ser Val Asp Tyr Arg 990 995 1000 gct act ttt cca gaa gat cag ttt cct aat tca tct cag aac ggt 3251 Ala Thr Phe Pro Glu Asp Gln Phe Pro Asn Ser Ser Gln Asn Gly 1005 1010 1015 tca tgc cga caa gtg cag tat cct ctg aca gac atg tcc ccc atc 3296 Ser Cys Arg Gln Val Gln Tyr Pro Leu Thr Asp Met Ser Pro Ile 1020 1025 1030 cta act agt ggg gac tct gat ata tcc agt cca tta ctg caa aat 3341 Leu Thr Ser Gly Asp Ser Asp Ile Ser Ser Pro Leu Leu Gln Asn 1035 1040 1045 act gtc cac att gac ctc agt gct cta aat cca gag ctg gtc cag 3386 Thr Val His Ile Asp Leu Ser Ala Leu Asn Pro Glu Leu Val Gln 1050 1055 1060 gca gtg cag cat gta gtg att ggg ccc agt agc ctg att gtg cat 3431 Ala Val Gln His Val Val Ile Gly Pro Ser Ser Leu Ile Val His 1065 1070 1075 ttc aat gaa gtc ata gga aga ggg cat ttt ggt tgt gta tat cat 3476 Phe Asn Glu Val Ile Gly Arg Gly His Phe Gly Cys Val Tyr His 1080 1085 1090 ggg act ttg ttg gac aat gat ggc aag aaa att cac tgt gct gtg 3521 Gly Thr Leu Leu Asp Asn Asp Gly Lys Lys Ile His Cys Ala Val 1095 1100 1105 aaa tcc ttg aac aga atc act gac ata gga gaa gtt tcc caa ttt 3566 Lys Ser Leu Asn Arg Ile Thr Asp Ile Gly Glu Val Ser Gln Phe 1110 1115 1120 ctg acc gag gga atc atc atg aaa gat ttt agt cat ccc aat gtc 3611 Leu Thr Glu Gly Ile Ile Met Lys Asp Phe Ser His Pro Asn Val 1125 1130 1135 ctc tcg ctc ctg gga atc tgc ctg cga agt gaa ggg tct ccg ctg 3656 Leu Ser Leu Leu Gly Ile Cys Leu Arg Ser Glu Gly Ser Pro Leu 1140 1145 1150 gtg gtc cta cca tac atg aaa cat gga gat ctt cga aat ttc att 3701 Val Val Leu Pro Tyr Met Lys His Gly Asp Leu Arg Asn Phe Ile 1155 1160 1165 cga aat gag act cat aat cca act gta aaa gat ctt att ggc ttt 3746 Arg Asn Glu Thr His Asn Pro Thr Val Lys Asp Leu Ile Gly Phe 1170 1175 1180 ggt ctt caa gta gcc aaa ggc atg aaa tat ctt gca agc aaa aag 3791 Gly Leu Gln Val Ala Lys Gly Met Lys Tyr Leu Ala Ser Lys Lys 1185 1190 1195 ttt gtc cac aga gac ttg gct gca aga aac tgt atg ctg gat gaa 3836 Phe Val His Arg Asp Leu Ala Ala Arg Asn Cys Met Leu Asp Glu 1200 1205 1210 aaa ttc aca gtc aag gtt gct gat ttt ggt ctt gcc aga gac atg 3881 Lys Phe Thr Val Lys Val Ala Asp Phe Gly Leu Ala Arg Asp Met 1215 1220 1225 tat gat aaa gaa tac tat agt gta cac aac aaa aca ggt gca aag 3926 Tyr Asp Lys Glu Tyr Tyr Ser Val His Asn Lys Thr Gly Ala Lys 1230 1235 1240 ctg cca gtg aag tgg atg gct ttg gaa agt ctg caa act caa aag 3971 Leu Pro Val Lys Trp Met Ala Leu Glu Ser Leu Gln Thr Gln Lys 1245 1250 1255 ttt acc acc aag tca gat gtg tgg tcc ttt ggc gtc gtc ctc tgg 4016 Phe Thr Thr Lys Ser Asp Val Trp Ser Phe Gly Val Val Leu Trp 1260 1265 1270 gag ctg atg aca aga gga gcc cca cct tat cct gac gta aac acc 4061 Glu Leu Met Thr Arg Gly Ala Pro Pro Tyr Pro Asp Val Asn Thr 1275 1280 1285 ttt gat ata act gtt tac ttg ttg caa ggg aga aga ctc cta caa 4106 Phe Asp Ile Thr Val Tyr Leu Leu Gln Gly Arg Arg Leu Leu Gln 1290 1295 1300 ccc gaa tac tgc cca gac ccc tta tat gaa gta atg cta aaa tgc 4151 Pro Glu Tyr Cys Pro Asp Pro Leu Tyr Glu Val Met Leu Lys Cys 1305 1310 1315 tgg cac cct aaa gcc gaa atg cgc cca tcc ttt tct gaa ctg gtg 4196 Trp His Pro Lys Ala Glu Met Arg Pro Ser Phe Ser Glu Leu Val 1320 1325 1330 tcc cgg ata tca gcg atc ttc tct act ttc att ggg gag cac tat 4241 Ser Arg Ile Ser Ala Ile Phe Ser Thr Phe Ile Gly Glu His Tyr 1335 1340 1345 gtc cat gtg aac gct act tat gtg aac gta aaa tgt gtc gct ccg 4286 Val His Val Asn Ala Thr Tyr Val Asn Val Lys Cys Val Ala Pro 1350 1355 1360 tat cct tct ctg ttg tca tca gaa gat aac gct gat gat gag gtg 4331 Tyr Pro Ser Leu Leu Ser Ser Glu Asp Asn Ala Asp Asp Glu Val 1365 1370 1375 gac aca cga cca gcc tcc ttc tgg gag aca tca tag tgctagtact 4377 Asp Thr Arg Pro Ala Ser Phe Trp Glu Thr Ser 1380 1385 1390 atgtcaaagc aacagtccac actttgtcca atggtttttt cactgcctga cctttaaaag 4437 gccatcgata ttctttgctc cttgccaaat tgcactatta ataggacttg tattgttatt 4497 taaattactg gattctaagg aatttcttat ctgacagagc atcagaacca gaggcttggt 4557 cccacaggcc agggaccaat gcgctgcag 4586 12 1390 PRT Homo sapiens SIGNAL (1)..(24) 12 Met Lys Ala Pro Ala Val Leu Ala Pro Gly Ile Leu Val Leu Leu Phe 1 5 10 15 Thr Leu Val Gln Arg Ser Asn Gly Glu Cys Lys Glu Ala Leu Ala Lys 20 25 30 Ser Glu Met Asn Val Asn Met Lys Tyr Gln Leu Pro Asn Phe Thr Ala 35 40 45 Glu Thr Pro Ile Gln Asn Val Ile Leu His Glu His His Ile Phe Leu 50 55 60 Gly Ala Thr Asn Tyr Ile Tyr Val Leu Asn Glu Glu Asp Leu Gln Lys 65 70 75 80 Val Ala Glu Tyr Lys Thr Gly Pro Val Leu Glu His Pro Asp Cys Phe 85 90 95 Pro Cys Gln Asp Cys Ser Ser Lys Ala Asn Leu Ser Gly Gly Val Trp 100 105 110 Lys Asp Asn Ile Asn Met Ala Leu Val Val Asp Thr Tyr Tyr Asp Asp 115 120 125 Gln Leu Ile Ser Cys Gly Ser Val Asn Arg Gly Thr Cys Gln Arg His 130 135 140 Val Phe Pro His Asn His Thr Ala Asp Ile Gln Ser Glu Val His Cys 145 150 155 160 Ile Phe Ser Pro Gln Ile Glu Glu Pro Ser Gln Cys Pro Asp Cys Val 165 170 175 Val Ser Ala Leu Gly Ala Lys Val Leu Ser Ser Val Lys Asp Arg Phe 180 185 190 Ile Asn Phe Phe Val Gly Asn Thr Ile Asn Ser Ser Tyr Phe Pro Asp 195 200 205 His Pro Leu His Ser Ile Ser Val Arg Arg Leu Lys Glu Thr Lys Asp 210 215 220 Gly Phe Met Phe Leu Thr Asp Gln Ser Tyr Ile Asp Val Leu Pro Glu 225 230 235 240 Phe Arg Asp Ser Tyr Pro Ile Lys Tyr Val His Ala Phe Glu Ser Asn 245 250 255 Asn Phe Ile Tyr Phe Leu Thr Val Gln Arg Glu Thr Leu Asp Ala Gln 260 265 270 Thr Phe His Thr Arg Ile Ile Arg Phe Cys Ser Ile Asn Ser Gly Leu 275 280 285 His Ser Tyr Met Glu Met Pro Leu Glu Cys Ile Leu Thr Glu Lys Arg 290 295 300 Lys Lys Arg Ser Thr Lys Lys Glu Val Phe Asn Ile Leu Gln Ala Ala 305 310 315 320 Tyr Val Ser Lys Pro Gly Ala Gln Leu Ala Arg Gln Ile Gly Ala Ser 325 330 335 Leu Asn Asp Asp Ile Leu Phe Gly Val Phe Ala Gln Ser Lys Pro Asp 340 345 350 Ser Ala Glu Pro Met Asp Arg Ser Ala Met Cys Ala Phe Pro Ile Lys 355 360 365 Tyr Val Asn Asp Phe Phe Asn Lys Ile Val Asn Lys Asn Asn Val Arg 370 375 380 Cys Leu Gln His Phe Tyr Gly Pro Asn His Glu His Cys Phe Asn Arg 385 390 395 400 Thr Leu Leu Arg Asn Ser Ser Gly Cys Glu Ala Arg Arg Asp Glu Tyr 405 410 415 Arg Thr Glu Phe Thr Thr Ala Leu Gln Arg Val Asp Leu Phe Met Gly 420 425 430 Gln Phe Ser Glu Val Leu Leu Thr Ser Ile Ser Thr Phe Ile Lys Gly 435 440 445 Asp Leu Thr Ile Ala Asn Leu Gly Thr Ser Glu Gly Arg Phe Met Gln 450 455 460 Val Val Val Ser Arg Ser Gly Pro Ser Thr Pro His Val Asn Phe Leu 465 470 475 480 Leu Asp Ser His Pro Val Ser Pro Glu Val Ile Val Glu His Thr Leu 485 490 495 Asn Gln Asn Gly Tyr Thr Leu Val Ile Thr Gly Lys Lys Ile Thr Lys 500 505 510 Ile Pro Leu Asn Gly Leu Gly Cys Arg His Phe Gln Ser Cys Ser Gln 515 520 525 Cys Leu Ser Ala Pro Pro Phe Val Gln Cys Gly Trp Cys His Asp Lys 530 535 540 Cys Val Arg Ser Glu Glu Cys Leu Ser Gly Thr Trp Thr Gln Gln Ile 545 550 555 560 Cys Leu Pro Ala Ile Tyr Lys Val Phe Pro Asn Ser Ala Pro Leu Glu 565 570 575 Gly Gly Thr Arg Leu Thr Ile Cys Gly Trp Asp Phe Gly Phe Arg Arg 580 585 590 Asn Asn Lys Phe Asp Leu Lys Lys Thr Arg Val Leu Leu Gly Asn Glu 595 600 605 Ser Cys Thr Leu Thr Leu Ser Glu Ser Thr Met Asn Thr Leu Lys Cys 610 615 620 Thr Val Gly Pro Ala Met Asn Lys His Phe Asn Met Ser Ile Ile Ile 625 630 635 640 Ser Asn Gly His Gly Thr Thr Gln Tyr Ser Thr Phe Ser Tyr Val Asp 645 650 655 Pro Val Ile Thr Ser Ile Ser Pro Lys Tyr Gly Pro Met Ala Gly Gly 660 665 670 Thr Leu Leu Thr Leu Thr Gly Asn Tyr Leu Asn Ser Gly Asn Ser Arg 675 680 685 His Ile Ser Ile Gly Gly Lys Thr Cys Thr Leu Lys Ser Val Ser Asn 690 695 700 Ser Ile Leu Glu Cys Tyr Thr Pro Ala Gln Thr Ile Ser Thr Glu Phe 705 710 715 720 Ala Val Lys Leu Lys Ile Asp Leu Ala Asn Arg Glu Thr Ser Ile Phe 725 730 735 Ser Tyr Arg Glu Asp Pro Ile Val Tyr Glu Ile His Pro Thr Lys Ser 740 745 750 Phe Ile Ser Gly Gly Ser Thr Ile Thr Gly Val Gly Lys Asn Leu Asn 755 760 765 Ser Val Ser Val Pro Arg Met Val Ile Asn Val His Glu Ala Gly Arg 770 775 780 Asn Phe Thr Val Ala Cys Gln His Arg Ser Asn Ser Glu Ile Ile Cys 785 790 795 800 Cys Thr Thr Pro Ser Leu Gln Gln Leu Asn Leu Gln Leu Pro Leu Lys 805 810 815 Thr Lys Ala Phe Phe Met Leu Asp Gly Ile Leu Ser Lys Tyr Phe Asp 820 825 830 Leu Ile Tyr Val His Asn Pro Val Phe Lys Pro Phe Glu Lys Pro Val 835 840 845 Met Ile Ser Met Gly Asn Glu Asn Val Leu Glu Ile Lys Gly Asn Asp 850 855 860 Ile Asp Pro Glu Ala Val Lys Gly Glu Val Leu Lys Val Gly Asn Lys 865 870 875 880 Ser Cys Glu Asn Ile His Leu His Ser Glu Ala Val Leu Cys Thr Val 885 890 895 Pro Asn Asp Leu Leu Lys Leu Asn Ser Glu Leu Asn Ile Glu Trp Lys 900 905 910 Gln Ala Ile Ser Ser Thr Val Leu Gly Lys Val Ile Val Gln Pro Asp 915 920 925 Gln Asn Phe Thr Gly Leu Ile Ala Gly Val Val Ser Ile Ser Thr Ala 930 935 940 Leu Leu Leu Leu Leu Gly Phe Phe Leu Trp Leu Lys Lys Arg Lys Gln 945 950 955 960 Ile Lys Asp Leu Gly Ser Glu Leu Val Arg Tyr Asp Ala Arg Val His 965 970 975 Thr Pro His Leu Asp Arg Leu Val Ser Ala Arg Ser Val Ser Pro Thr 980 985 990 Thr Glu Met Val Ser Asn Glu Ser Val Asp Tyr Arg Ala Thr Phe Pro 995 1000 1005 Glu Asp Gln Phe Pro Asn Ser Ser Gln Asn Gly Ser Cys Arg Gln 1010 1015 1020 Val Gln Tyr Pro Leu Thr Asp Met Ser Pro Ile Leu Thr Ser Gly 1025 1030 1035 Asp Ser Asp Ile Ser Ser Pro Leu Leu Gln Asn Thr Val His Ile 1040 1045 1050 Asp Leu Ser Ala Leu Asn Pro Glu Leu Val Gln Ala Val Gln His 1055 1060 1065 Val Val Ile Gly Pro Ser Ser Leu Ile Val His Phe Asn Glu Val 1070 1075 1080 Ile Gly Arg Gly His Phe Gly Cys Val Tyr His Gly Thr Leu Leu 1085 1090 1095 Asp Asn Asp Gly Lys Lys Ile His Cys Ala Val Lys Ser Leu Asn 1100 1105 1110 Arg Ile Thr Asp Ile Gly Glu Val Ser Gln Phe Leu Thr Glu Gly 1115 1120 1125 Ile Ile Met Lys Asp Phe Ser His Pro Asn Val Leu Ser Leu Leu 1130 1135 1140 Gly Ile Cys Leu Arg Ser Glu Gly Ser Pro Leu Val Val Leu Pro 1145 1150 1155 Tyr Met Lys His Gly Asp Leu Arg Asn Phe Ile Arg Asn Glu Thr 1160 1165 1170 His Asn Pro Thr Val Lys Asp Leu Ile Gly Phe Gly Leu Gln Val 1175 1180 1185 Ala Lys Gly Met Lys Tyr Leu Ala Ser Lys Lys Phe Val His Arg 1190 1195 1200 Asp Leu Ala Ala Arg Asn Cys Met Leu Asp Glu Lys Phe Thr Val 1205 1210 1215 Lys Val Ala Asp Phe Gly Leu Ala Arg Asp Met Tyr Asp Lys Glu 1220 1225 1230 Tyr Tyr Ser Val His Asn Lys Thr Gly Ala Lys Leu Pro Val Lys 1235 1240 1245 Trp Met Ala Leu Glu Ser Leu Gln Thr Gln Lys Phe Thr Thr Lys 1250 1255 1260 Ser Asp Val Trp Ser Phe Gly Val Val Leu Trp Glu Leu Met Thr 1265 1270 1275 Arg Gly Ala Pro Pro Tyr Pro Asp Val Asn Thr Phe Asp Ile Thr 1280 1285 1290 Val Tyr Leu Leu Gln Gly Arg Arg Leu Leu Gln Pro Glu Tyr Cys 1295 1300 1305 Pro Asp Pro Leu Tyr Glu Val Met Leu Lys Cys Trp His Pro Lys 1310 1315 1320 Ala Glu Met Arg Pro Ser Phe Ser Glu Leu Val Ser Arg Ile Ser 1325 1330 1335 Ala Ile Phe Ser Thr Phe Ile Gly Glu His Tyr Val His Val Asn 1340 1345 1350 Ala Thr Tyr Val Asn Val Lys Cys Val Ala Pro Tyr Pro Ser Leu 1355 1360 1365 Leu Ser Ser Glu Asp Asn Ala Asp Asp Glu Val Asp Thr Arg Pro 1370 1375 1380 Ala Ser Phe Trp Glu Thr Ser 1385 1390 13 2558 DNA Homo sapiens gene (1)..(2558) Chondroitin sulfate proteoglycan BEHAB/brevican mRNA, GPI isoform, complete cds 13 tgtggcactg cctgcgtacc caaccccagc cctgggtagc ctgcagc atg gcc cag 56 Met Ala Gln 1 ctg ttc ctg ccc ctg ctg gca gcc ctg gtc ctg gcc cag gct cct gca 104 Leu Phe Leu Pro Leu Leu Ala Ala Leu Val Leu Ala Gln Ala Pro Ala 5 10 15 gct tta gca gat gtt ctg gaa gga gac agc tca gag gac cgc gct ttt 152 Ala Leu Ala Asp Val Leu Glu Gly Asp Ser Ser Glu Asp Arg Ala Phe 20 25 30 35 cgc gtg cgc atc gcg ggc gac gcg cca ctg cag ggc gtg ctc ggc ggc 200 Arg Val Arg Ile Ala Gly Asp Ala Pro Leu Gln Gly Val Leu Gly Gly 40 45 50 gcc ctc acc atc cct tgc cac gtc cac tac ctg cgg cca ccg ccg agc 248 Ala Leu Thr Ile Pro Cys His Val His Tyr Leu Arg Pro Pro Pro Ser 55 60 65 cgc cgg gct gtg ctg ggc tct ccg cgg gtc aag tgg act ttc ctg tcc 296 Arg Arg Ala Val Leu Gly Ser Pro Arg Val Lys Trp Thr Phe Leu Ser 70 75 80 cgg ggc cgg gag gca gag gtg ctg gtg gcg cgg gga gtg cgc gtc aag 344 Arg Gly Arg Glu Ala Glu Val Leu Val Ala Arg Gly Val Arg Val Lys 85 90 95 gtg aac gag gcc tac cgg ttc cgc gtg gca ctg cct gcg tac cca gcg 392 Val Asn Glu Ala Tyr Arg Phe Arg Val Ala Leu Pro Ala Tyr Pro Ala 100 105 110 115 tcg ctc acc gac gtc tcc ctg gcg ctg agc gag ctg cgc ccc aac gac 440 Ser Leu Thr Asp Val Ser Leu Ala Leu Ser Glu Leu Arg Pro Asn Asp 120 125 130 tca ggt atc tat cgc tgt gag gtc cag cac ggc atc gat gac agc agc 488 Ser Gly Ile Tyr Arg Cys Glu Val Gln His Gly Ile Asp Asp Ser Ser 135 140 145 gac gct gtg gag gtc aag gtc aaa ggg gtc gtc ttt ctc tac cga gag 536 Asp Ala Val Glu Val Lys Val Lys Gly Val Val Phe Leu Tyr Arg Glu 150 155 160 ggc tct gcc cgc tat gct ttc tcc ttt tct ggg gcc cag gag gcc tgt 584 Gly Ser Ala Arg Tyr Ala Phe Ser Phe Ser Gly Ala Gln Glu Ala Cys 165 170 175 gcc cgc att gga gcc cac atc gcc acc ccg gag cag ctc tat gcc gcc 632 Ala Arg Ile Gly Ala His Ile Ala Thr Pro Glu Gln Leu Tyr Ala Ala 180 185 190 195 tac ctt ggg ggc tat gag caa tgt gat gct ggc tgg ctg tcg gat cag 680 Tyr Leu Gly Gly Tyr Glu Gln Cys Asp Ala Gly Trp Leu Ser Asp Gln 200 205 210 acc gtg agg tat ccc atc cag acc cca cga gag gcc tgt tac gga gac 728 Thr Val Arg Tyr Pro Ile Gln Thr Pro Arg Glu Ala Cys Tyr Gly Asp 215 220 225 atg gat ggc ttc ccc ggg gtc cgg aac tat ggt gtg gtg gac ccg gat 776 Met Asp Gly Phe Pro Gly Val Arg Asn Tyr Gly Val Val Asp Pro Asp 230 235 240 gac ctc tat gat gtg tac tgt tat gct gaa gac cta aat gga gaa ttg 824 Asp Leu Tyr Asp Val Tyr Cys Tyr Ala Glu Asp Leu Asn Gly Glu Leu 245 250 255 ttc ctg ggt gac cct cca gag aag ctg aca ttg gag gaa gca cgg gcg 872 Phe Leu Gly Asp Pro Pro Glu Lys Leu Thr Leu Glu Glu Ala Arg Ala 260 265 270 275 tac tgc cag gag cgg ggt gca gag att gcc acc acg ggc caa ctg tat 920 Tyr Cys Gln Glu Arg Gly Ala Glu Ile Ala Thr Thr Gly Gln Leu Tyr 280 285 290 gca gcc tgg gat ggt ggc ctg gac cac tgc agc cca ggg tgg cta gct 968 Ala Ala Trp Asp Gly Gly Leu Asp His Cys Ser Pro Gly Trp Leu Ala 295 300 305 gat ggc agt gtg cgc tac ccc atc gtc aca ccc agc cag cgc tgt ggt 1016 Asp Gly Ser Val Arg Tyr Pro Ile Val Thr Pro Ser Gln Arg Cys Gly 310 315 320 ggg ggc ttg cct ggt gtc aag act ctc ttc ctc ttc ccc aac cag act 1064 Gly Gly Leu Pro Gly Val Lys Thr Leu Phe Leu Phe Pro Asn Gln Thr 325 330 335 ggc ttc ccc aat aag cac agc cgc ttc aac gtc tac tgc ttc cga gac 1112 Gly Phe Pro Asn Lys His Ser Arg Phe Asn Val Tyr Cys Phe Arg Asp 340 345 350 355 tcg gcc cag cct tct gcc atc cct gag gcc tcc aac cca gcc tcc aac 1160 Ser Ala Gln Pro Ser Ala Ile Pro Glu Ala Ser Asn Pro Ala Ser Asn 360 365 370 cca gcc tct gat gga cta gag gct atc gtc aca gtg aca gag acc ctg 1208 Pro Ala Ser Asp Gly Leu Glu Ala Ile Val Thr Val Thr Glu Thr Leu 375 380 385 gag gaa ctg cag ctg cct cag gaa gcc aca gag agt gaa tcc cgt ggg 1256 Glu Glu Leu Gln Leu Pro Gln Glu Ala Thr Glu Ser Glu Ser Arg Gly 390 395 400 gcc atc tac tcc atc ccc atc atg gag gac gga gga ggt gga agc tcc 1304 Ala Ile Tyr Ser Ile Pro Ile Met Glu Asp Gly Gly Gly Gly Ser Ser 405 410 415 act cca gaa gac cca gca gag gcc cct agg acg ctc cta gaa ttt gaa 1352 Thr Pro Glu Asp Pro Ala Glu Ala Pro Arg Thr Leu Leu Glu Phe Glu 420 425 430 435 aca caa tcc atg gta ccg ccc acg ggg ttc tca gaa gag gaa ggt aag 1400 Thr Gln Ser Met Val Pro Pro Thr Gly Phe Ser Glu Glu Glu Gly Lys 440 445 450 gca ttg gag gaa gaa gag aaa tat gaa gat gaa gaa gag aaa gag gag 1448 Ala Leu Glu Glu Glu Glu Lys Tyr Glu Asp Glu Glu Glu Lys Glu Glu 455 460 465 gaa gaa gaa gag gag gag gtg gag gat gag gct ctg tgg gca tgg ccc 1496 Glu Glu Glu Glu Glu Glu Val Glu Asp Glu Ala Leu Trp Ala Trp Pro 470 475 480 agc gag ctc agc agc ccg ggc cct gag gcc tct ctc ccc act gag cca 1544 Ser Glu Leu Ser Ser Pro Gly Pro Glu Ala Ser Leu Pro Thr Glu Pro 485 490 495 gca gcc cag gag gag tca ctc tcc cag gcg cca gca agg gca gtc ctg 1592 Ala Ala Gln Glu Glu Ser Leu Ser Gln Ala Pro Ala Arg Ala Val Leu 500 505 510 515 cag cct ggt gca tca cca ctt cct gat gga gag tca gaa gct tcc agg 1640 Gln Pro Gly Ala Ser Pro Leu Pro Asp Gly Glu Ser Glu Ala Ser Arg 520 525 530 cct cca agg gtc cat gga cca cct act gag act ctg ccc act ccc agg 1688 Pro Pro Arg Val His Gly Pro Pro Thr Glu Thr Leu Pro Thr Pro Arg 535 540 545 gag agg aac cta gca tcc cca tca cct tcc act ctg gtt gag gca aga 1736 Glu Arg Asn Leu Ala Ser Pro Ser Pro Ser Thr Leu Val Glu Ala Arg 550 555 560 gag gtg ggg gag gca act ggt ggt cct gag cta tct ggg gtc cct cga 1784 Glu Val Gly Glu Ala Thr Gly Gly Pro Glu Leu Ser Gly Val Pro Arg 565 570 575 gga gag agc gag gag aca gga agc tcc gag ggt gcc cct tcc ctg ctt 1832 Gly Glu Ser Glu Glu Thr Gly Ser Ser Glu Gly Ala Pro Ser Leu Leu 580 585 590 595 cca gcc aca cgg gcc cct gag ggt acc agg gag ctg gag gcc ccc tct 1880 Pro Ala Thr Arg Ala Pro Glu Gly Thr Arg Glu Leu Glu Ala Pro Ser 600 605 610 gaa gat aat tct gga aga act gcc cca gca ggg acc tca gtg cag gcc 1928 Glu Asp Asn Ser Gly Arg Thr Ala Pro Ala Gly Thr Ser Val Gln Ala 615 620 625 cag cca gtg ctg ccc act gac agc gcc agc cga ggt gga gtg gcc gtg 1976 Gln Pro Val Leu Pro Thr Asp Ser Ala Ser Arg Gly Gly Val Ala Val 630 635 640 gtc ccc gca tca ggt aat tct gcc caa ggc tca act gcc ctc tct atc 2024 Val Pro Ala Ser Gly Asn Ser Ala Gln Gly Ser Thr Ala Leu Ser Ile 645 650 655 cta ctc ctt ttc ttc ccc ctg cag ctc tgg gtc acc tga cctgtagtcc 2073 Leu Leu Leu Phe Phe Pro Leu Gln Leu Trp Val Thr 660 665 670 tttaacccac catcatccca aactctcctg tcctttgcct tcattctctt acccacctct 2133 acctatgggt ctccaatctc ggatatccac cttgtgggta tctcagctct ccgcgtcttt 2193 accctgtgat cccagccccg ccactgacca tctgtgaccc ttccctgcca ttgggccctc 2253 cacctgtggc tcacatctcg ccagccccac agagcatcct caggcctctc caagggtcct 2313 catcacctat tgcagccttc agggctcggc ctattttcca ctactccctt catccgcctg 2373 tgtgccgtcc cctttagctg cctcctattg atctcaggga agcctgggag tcccttctca 2433 cccctcaacc tccggagtcc aggagaaccc gtacccccac agagccttaa gcaactactt 2493 ctgtgaagta ttttttgact gtttcatgga aaacaagcct tggaaataaa tctctattaa 2553 accgc 2558 14 671 PRT Homo sapiens gene (1)..(671) Chondroitin sulfate proteoglycan BEHAB/brevican 14 Met Ala Gln Leu Phe Leu Pro Leu Leu Ala Ala Leu Val Leu Ala Gln 1 5 10 15 Ala Pro Ala Ala Leu Ala Asp Val Leu Glu Gly Asp Ser Ser Glu Asp 20 25 30 Arg Ala Phe Arg Val Arg Ile Ala Gly Asp Ala Pro Leu Gln Gly Val 35 40 45 Leu Gly Gly Ala Leu Thr Ile Pro Cys His Val His Tyr Leu Arg Pro 50 55 60 Pro Pro Ser Arg Arg Ala Val Leu Gly Ser Pro Arg Val Lys Trp Thr 65 70 75 80 Phe Leu Ser Arg Gly Arg Glu Ala Glu Val Leu Val Ala Arg Gly Val 85 90 95 Arg Val Lys Val Asn Glu Ala Tyr Arg Phe Arg Val Ala Leu Pro Ala 100 105 110 Tyr Pro Ala Ser Leu Thr Asp Val Ser Leu Ala Leu Ser Glu Leu Arg 115 120 125 Pro Asn Asp Ser Gly Ile Tyr Arg Cys Glu Val Gln His Gly Ile Asp 130 135 140 Asp Ser Ser Asp Ala Val Glu Val Lys Val Lys Gly Val Val Phe Leu 145 150 155 160 Tyr Arg Glu Gly Ser Ala Arg Tyr Ala Phe Ser Phe Ser Gly Ala Gln 165 170 175 Glu Ala Cys Ala Arg Ile Gly Ala His Ile Ala Thr Pro Glu Gln Leu 180 185 190 Tyr Ala Ala Tyr Leu Gly Gly Tyr Glu Gln Cys Asp Ala Gly Trp Leu 195 200 205 Ser Asp Gln Thr Val Arg Tyr Pro Ile Gln Thr Pro Arg Glu Ala Cys 210 215 220 Tyr Gly Asp Met Asp Gly Phe Pro Gly Val Arg Asn Tyr Gly Val Val 225 230 235 240 Asp Pro Asp Asp Leu Tyr Asp Val Tyr Cys Tyr Ala Glu Asp Leu Asn 245 250 255 Gly Glu Leu Phe Leu Gly Asp Pro Pro Glu Lys Leu Thr Leu Glu Glu 260 265 270 Ala Arg Ala Tyr Cys Gln Glu Arg Gly Ala Glu Ile Ala Thr Thr Gly 275 280 285 Gln Leu Tyr Ala Ala Trp Asp Gly Gly Leu Asp His Cys Ser Pro Gly 290 295 300 Trp Leu Ala Asp Gly Ser Val Arg Tyr Pro Ile Val Thr Pro Ser Gln 305 310 315 320 Arg Cys Gly Gly Gly Leu Pro Gly Val Lys Thr Leu Phe Leu Phe Pro 325 330 335 Asn Gln Thr Gly Phe Pro Asn Lys His Ser Arg Phe Asn Val Tyr Cys 340 345 350 Phe Arg Asp Ser Ala Gln Pro Ser Ala Ile Pro Glu Ala Ser Asn Pro 355 360 365 Ala Ser Asn Pro Ala Ser Asp Gly Leu Glu Ala Ile Val Thr Val Thr 370 375 380 Glu Thr Leu Glu Glu Leu Gln Leu Pro Gln Glu Ala Thr Glu Ser Glu 385 390 395 400 Ser Arg Gly Ala Ile Tyr Ser Ile Pro Ile Met Glu Asp Gly Gly Gly 405 410 415 Gly Ser Ser Thr Pro Glu Asp Pro Ala Glu Ala Pro Arg Thr Leu Leu 420 425 430 Glu Phe Glu Thr Gln Ser Met Val Pro Pro Thr Gly Phe Ser Glu Glu 435 440 445 Glu Gly Lys Ala Leu Glu Glu Glu Glu Lys Tyr Glu Asp Glu Glu Glu 450 455 460 Lys Glu Glu Glu Glu Glu Glu Glu Glu Val Glu Asp Glu Ala Leu Trp 465 470 475 480 Ala Trp Pro Ser Glu Leu Ser Ser Pro Gly Pro Glu Ala Ser Leu Pro 485 490 495 Thr Glu Pro Ala Ala Gln Glu Glu Ser Leu Ser Gln Ala Pro Ala Arg 500 505 510 Ala Val Leu Gln Pro Gly Ala Ser Pro Leu Pro Asp Gly Glu Ser Glu 515 520 525 Ala Ser Arg Pro Pro Arg Val His Gly Pro Pro Thr Glu Thr Leu Pro 530 535 540 Thr Pro Arg Glu Arg Asn Leu Ala Ser Pro Ser Pro Ser Thr Leu Val 545 550 555 560 Glu Ala Arg Glu Val Gly Glu Ala Thr Gly Gly Pro Glu Leu Ser Gly 565 570 575 Val Pro Arg Gly Glu Ser Glu Glu Thr Gly Ser Ser Glu Gly Ala Pro 580 585 590 Ser Leu Leu Pro Ala Thr Arg Ala Pro Glu Gly Thr Arg Glu Leu Glu 595 600 605 Ala Pro Ser Glu Asp Asn Ser Gly Arg Thr Ala Pro Ala Gly Thr Ser 610 615 620 Val Gln Ala Gln Pro Val Leu Pro Thr Asp Ser Ala Ser Arg Gly Gly 625 630 635 640 Val Ala Val Val Pro Ala Ser Gly Asn Ser Ala Gln Gly Ser Thr Ala 645 650 655 Leu Ser Ile Leu Leu Leu Phe Phe Pro Leu Gln Leu Trp Val Thr 660 665 670 15 2316 DNA Homo sapiens gene (1)..(2316) Human mRNA for CD44E (epithelial form) 15 agcggacccc agcctctgcc aggttcggtc cgccatcctc gtcccgtcct ccgccggccc 60 ctgccccgcg cccagggatc ctccagctcc tttcgcccgc gccctccgtt cgctccggac 120 acc atg gac aag ttt tgg tgg cac gca gcc tgg gga ctc tgc ctc gtg 168 Met Asp Lys Phe Trp Trp His Ala Ala Trp Gly Leu Cys Leu Val 1 5 10 15 ccg ctg agc ctg gcg cag atc gat ttg aat ata acc tgc cgc ttt gca 216 Pro Leu Ser Leu Ala Gln Ile Asp Leu Asn Ile Thr Cys Arg Phe Ala 20 25 30 ggt gta ttc cac gtg gag aaa aat ggt cgc tac agc atc tct cgg acg 264 Gly Val Phe His Val Glu Lys Asn Gly Arg Tyr Ser Ile Ser Arg Thr 35 40 45 gag gcc gct gac ctc tgc aag gct ttc aat agc acc ttg ccc aca atg 312 Glu Ala Ala Asp Leu Cys Lys Ala Phe Asn Ser Thr Leu Pro Thr Met 50 55 60 gcc cag atg gag aaa gct ctg agc atc gga ttt gag acc tgc agg tat 360 Ala Gln Met Glu Lys Ala Leu Ser Ile Gly Phe Glu Thr Cys Arg Tyr 65 70 75 ggg ttc ata gaa ggg cat gtg gtg att ccc cgg atc cac ccc aac tcc 408 Gly Phe Ile Glu Gly His Val Val Ile Pro Arg Ile His Pro Asn Ser 80 85 90 95 atc tgt gca gca aac aac aca ggg gtg tac atc ctc aca tac aac acc 456 Ile Cys Ala Ala Asn Asn Thr Gly Val Tyr Ile Leu Thr Tyr Asn Thr 100 105 110 tcc cag tat gac aca tat tgc ttc aat gct tca gct cca cct gaa gaa 504 Ser Gln Tyr Asp Thr Tyr Cys Phe Asn Ala Ser Ala Pro Pro Glu Glu 115 120 125 gat tgt aca tca gtc aca gac ctg ccc aat gcc ttt gat gga cca att 552 Asp Cys Thr Ser Val Thr Asp Leu Pro Asn Ala Phe Asp Gly Pro Ile 130 135 140 acc ata act att gtt aac cgt gat ggc acc cgc tat gtc cag aaa gga 600 Thr Ile Thr Ile Val Asn Arg Asp Gly Thr Arg Tyr Val Gln Lys Gly 145 150 155 gaa tac aga acg aat cct gaa gac atc tac ccc agc aac cct act gat 648 Glu Tyr Arg Thr Asn Pro Glu Asp Ile Tyr Pro Ser Asn Pro Thr Asp 160 165 170 175 gat gac gtg agc agc ggc tcc tcc agt gaa agg agc agc act tca gga 696 Asp Asp Val Ser Ser Gly Ser Ser Ser Glu Arg Ser Ser Thr Ser Gly 180 185 190 ggt tac atc ttt tac acc ttt tct act gta cac ccc atc cca gac gaa 744 Gly Tyr Ile Phe Tyr Thr Phe Ser Thr Val His Pro Ile Pro Asp Glu 195 200 205 gac agt ccc tgg atc acc gac agc aca gac aga atc cct cgt acc aat 792 Asp Ser Pro Trp Ile Thr Asp Ser Thr Asp Arg Ile Pro Arg Thr Asn 210 215 220 atg gac tcc agt cat agt aca acg ctt cag cct act gca aat cca aac 840 Met Asp Ser Ser His Ser Thr Thr Leu Gln Pro Thr Ala Asn Pro Asn 225 230 235 aca ggt ttg gtg gaa gat ttg gac agg aca gga cct ctt tca atg aca 888 Thr Gly Leu Val Glu Asp Leu Asp Arg Thr Gly Pro Leu Ser Met Thr 240 245 250 255 acg cag cag agt aat tct cag agc ttc tct aca tca cat gaa ggc ttg 936 Thr Gln Gln Ser Asn Ser Gln Ser Phe Ser Thr Ser His Glu Gly Leu 260 265 270 gaa gaa gat aaa gac cat cca aca act tct act ctg aca tca agc aat 984 Glu Glu Asp Lys Asp His Pro Thr Thr Ser Thr Leu Thr Ser Ser Asn 275 280 285 agg aat gat gtc aca ggt gga aga aga gac cca aat cat tct gaa ggc 1032 Arg Asn Asp Val Thr Gly Gly Arg Arg Asp Pro Asn His Ser Glu Gly 290 295 300 tca act cat tta ctg gaa ggt tat acc tct cat tac cca cac acg aag 1080 Ser Thr His Leu Leu Glu Gly Tyr Thr Ser His Tyr Pro His Thr Lys 305 310 315 gaa agc agg acc ttc atc cca gtg acc tca gct aag act ggg tcc ttt 1128 Glu Ser Arg Thr Phe Ile Pro Val Thr Ser Ala Lys Thr Gly Ser Phe 320 325 330 335 gga gtt act gca gtt act gtt gga gat tcc aac tct aat gtc aat cgt 1176 Gly Val Thr Ala Val Thr Val Gly Asp Ser Asn Ser Asn Val Asn Arg 340 345 350 tcc tta tca gga gac caa gac aca ttc cac ccc agt ggg ggg tcc cat 1224 Ser Leu Ser Gly Asp Gln Asp Thr Phe His Pro Ser Gly Gly Ser His 355 360 365 acc act cat gga tct gaa tca gat gga cac tca cat ggg agt caa gaa 1272 Thr Thr His Gly Ser Glu Ser Asp Gly His Ser His Gly Ser Gln Glu 370 375 380 ggt gga gca aac aca acc tct ggt cct ata agg aca ccc caa att cca 1320 Gly Gly Ala Asn Thr Thr Ser Gly Pro Ile Arg Thr Pro Gln Ile Pro 385 390 395 gaa tgg ctg atc atc ttg gca tcc ctc ttg gcc ttg gct ttg att ctt 1368 Glu Trp Leu Ile Ile Leu Ala Ser Leu Leu Ala Leu Ala Leu Ile Leu 400 405 410 415 gca gtt tgc att gca gtc aac agt cga aga agg tgt ggg cag aag aaa 1416 Ala Val Cys Ile Ala Val Asn Ser Arg Arg Arg Cys Gly Gln Lys Lys 420 425 430 aag cta gtg atc aac agt ggc aat gga gct gtg gag gac aga aag cca 1464 Lys Leu Val Ile Asn Ser Gly Asn Gly Ala Val Glu Asp Arg Lys Pro 435 440 445 agt gga ctc aac gga gag gcc agc aag tct cag gaa atg gtg cat ttg 1512 Ser Gly Leu Asn Gly Glu Ala Ser Lys Ser Gln Glu Met Val His Leu 450 455 460 gtg aac aag gag tcg tca gaa act cca gac cag ttt atg aca gct gat 1560 Val Asn Lys Glu Ser Ser Glu Thr Pro Asp Gln Phe Met Thr Ala Asp 465 470 475 gag aca agg aac ctg cag aat gtg gac atg aag att ggg gtg taa 1605 Glu Thr Arg Asn Leu Gln Asn Val Asp Met Lys Ile Gly Val 480 485 490 cacctacacc attatcttgg aaagaaacaa cgttggaaac ataaccatta caggggagct 1665 gggacactta acagatgcaa tgtgctactg attgtttcat ttcgaatcta taatagcata 1725 aaattttcta ctctttttgt tttttgtgtt ttgttcttta aagtcaggtc caatttgtaa 1785 aaacagcatt gctttctgaa attagggccc aattaataat cagcaagaat tttgatcgtt 1845 tcagttcccc acttggaggc ctttcatccc tcgggtgtgc tatggatggc ttctaacaaa 1905 aacctaccac atagttattc ctgatcgcca accttgcccc ccaccagcta aggacatttc 1965 cagggttaat agggcctggt cctgggagga aatttgaatg ggtcattttg cccttccatt 2025 agcctaatcc ctgggcattg ctttccactg aggttggggg ttggggtgta ctagttacac 2085 atcttcaaca gaccccctct agaaattttt cagatgcttc tgggagacac ccaaagggta 2145 agtctattta tctgtagtaa actatttatc tgtgtttttg aaatattaaa ccctggatca 2205 gtccttttat tcagtataat tttttaaagt tactttgtca gaggcacaaa aagggtttaa 2265 actgattcat aataaatatc tgtaccttct tcgaaaaaaa aaaaaaaaaa a 2316 16 742 PRT Homo sapiens SIGNAL (1)..(20) BY SIMILARITY 16 Met Asp Lys Phe Trp Trp His Ala Ala Trp Gly Leu Cys Leu Val Pro 1 5 10 15 Leu Ser Leu Ala Gln Ile Asp Leu Asn Ile Thr Cys Arg Phe Ala Gly 20 25 30 Val Phe His Val Glu Lys Asn Gly Arg Tyr Ser Ile Ser Arg Thr Glu 35 40 45 Ala Ala Asp Leu Cys Lys Ala Phe Asn Ser Thr Leu Pro Thr Met Ala 50 55 60 Gln Met Glu Lys Ala Leu Ser Ile Gly Phe Glu Thr Cys Arg Tyr Gly 65 70 75 80 Phe Ile Glu Gly His Val Val Ile Pro Arg Ile His Pro Asn Ser Ile 85 90 95 Cys Ala Ala Asn Asn Thr Gly Val Tyr Ile Leu Thr Ser Asn Thr Ser 100 105 110 Gln Tyr Asp Thr Tyr Cys Phe Asn Ala Ser Ala Pro Pro Glu Glu Asp 115 120 125 Cys Thr Ser Val Thr Asp Leu Pro Asn Ala Phe Asp Gly Pro Ile Thr 130 135 140 Ile Thr Ile Val Asn Arg Asp Gly Thr Arg Tyr Val Gln Lys Gly Glu 145 150 155 160 Tyr Arg Thr Asn Pro Glu Asp Ile Tyr Pro Ser Asn Pro Thr Asp Asp 165 170 175 Asp Val Ser Ser Gly Ser Ser Ser Glu Arg Ser Ser Thr Ser Gly Gly 180 185 190 Tyr Ile Phe Tyr Thr Phe Ser Thr Val His Pro Ile Pro Asp Glu Asp 195 200 205 Ser Pro Trp Ile Thr Asp Ser Thr Asp Arg Ile Pro Ala Thr Thr Leu 210 215 220 Met Ser Thr Ser Ala Thr Ala Thr Glu Thr Ala Thr Lys Arg Gln Glu 225 230 235 240 Thr Trp Asp Trp Phe Ser Trp Leu Phe Leu Pro Ser Glu Ser Lys Asn 245 250 255 His Leu His Thr Thr Thr Gln Met Ala Gly Thr Ser Ser Asn Thr Ile 260 265 270 Ser Ala Gly Trp Glu Pro Asn Glu Glu Asn Glu Asp Glu Arg Asp Arg 275 280 285 His Leu Ser Phe Ser Gly Ser Gly Ile Asp Asp Asp Glu Asp Phe Ile 290 295 300 Ser Ser Thr Ile Ser Thr Thr Pro Arg Ala Phe Asp His Thr Lys Gln 305 310 315 320 Asn Gln Asp Trp Thr Gln Trp Asn Pro Ser His Ser Asn Pro Glu Val 325 330 335 Leu Leu Gln Thr Thr Thr Arg Met Thr Asp Val Asp Arg Asn Gly Thr 340 345 350 Thr Ala Tyr Glu Gly Asn Trp Asn Pro Glu Ala His Pro Pro Leu Ile 355 360 365 His His Glu His His Glu Glu Glu Glu Thr Pro His Ser Thr Ser Thr 370 375 380 Ile Gln Ala Thr Pro Ser Ser Thr Thr Glu Glu Thr Ala Thr Gln Lys 385 390 395 400 Glu Gln Trp Phe Gly Asn Arg Trp His Glu Gly Tyr Arg Gln Thr Pro 405 410 415 Arg Glu Asp Ser His Ser Thr Thr Gly Thr Ala Ala Ala Ser Ala His 420 425 430 Thr Ser His Pro Met Gln Gly Arg Thr Thr Pro Ser Pro Glu Asp Ser 435 440 445 Ser Trp Thr Asp Phe Phe Asn Pro Ile Ser His Pro Met Gly Arg Gly 450 455 460 His Gln Ala Gly Arg Arg Met Asp Met Asp Ser Ser His Ser Thr Thr 465 470 475 480 Leu Gln Pro Thr Ala Asn Pro Asn Thr Gly Leu Val Glu Asp Leu Asp 485 490 495 Arg Thr Gly Pro Leu Ser Met Thr Thr Gln Gln Ser Asn Ser Gln Ser 500 505 510 Phe Ser Thr Ser His Glu Gly Leu Glu Glu Asp Lys Asp His Pro Thr 515 520 525 Thr Ser Thr Leu Thr Ser Ser Asn Arg Asn Asp Val Thr Gly Gly Arg 530 535 540 Arg Asp Pro Asn His Ser Glu Gly Ser Thr Thr Leu Leu Glu Gly Tyr 545 550 555 560 Thr Ser His Tyr Pro His Thr Lys Glu Ser Arg Thr Phe Ile Pro Val 565 570 575 Thr Ser Ala Lys Thr Gly Ser Phe Gly Val Thr Ala Val Thr Val Gly 580 585 590 Asp Ser Asn Ser Asn Val Asn Arg Ser Leu Ser Gly Asp Gln Asp Thr 595 600 605 Phe His Pro Ser Gly Gly Ser His Thr Thr His Gly Ser Glu Ser Asp 610 615 620 Gly His Ser His Gly Ser Gln Glu Gly Gly Ala Asn Thr Thr Ser Gly 625 630 635 640 Pro Ile Arg Thr Pro Gln Ile Pro Glu Trp Leu Ile Ile Leu Ala Ser 645 650 655 Leu Leu Ala Leu Ala Leu Ile Leu Ala Val Cys Ile Ala Val Asn Ser 660 665 670 Arg Arg Arg Cys Gly Gln Lys Lys Lys Leu Val Ile Asn Ser Gly Asn 675 680 685 Gly Ala Val Glu Asp Arg Lys Pro Ser Gly Leu Asn Gly Glu Ala Ser 690 695 700 Lys Ser Gln Glu Met Val His Leu Val Asn Lys Glu Ser Ser Glu Thr 705 710 715 720 Pro Asp Gln Phe Met Thr Ala Asp Glu Thr Arg Asn Leu Gln Asn Val 725 730 735 Asp Met Lys Ile Gly Val 740 17 930 DNA Homo sapiens Gene (1)..(930) Tetraspan TM4SF (TSPAN-3), mRNA 17 atg ggc cag tgc ggc atc acc tcc tcc aag acc gtg ctg gtc ttt ctc 48 Met Gly Gln Cys Gly Ile Thr Ser Ser Lys Thr Val Leu Val Phe Leu 1 5 10 15 aac ctc atc ttc tgg ggg gca gct ggc att tta tgc tat gtg gga gcc 96 Asn Leu Ile Phe Trp Gly Ala Ala Gly Ile Leu Cys Tyr Val Gly Ala 20 25 30 tat gtc ttc atc act tat gat gac tat gac cac ttc ttt gaa gat gtg 144 Tyr Val Phe Ile Thr Tyr Asp Asp Tyr Asp His Phe Phe Glu Asp Val 35 40 45 tac acg ctc atc cct gct gta gtg atc ata gct gta gga gcc ctg ctt 192 Tyr Thr Leu Ile Pro Ala Val Val Ile Ile Ala Val Gly Ala Leu Leu 50 55 60 ttc atc att ggg cta att ggc tgc tgt gcc aca atc cgg gaa agt cgc 240 Phe Ile Ile Gly Leu Ile Gly Cys Cys Ala Thr Ile Arg Glu Ser Arg 65 70 75 80 tgt gga ctt gcc acg ttt gtc atc atc ctg ctc ttg gtt ttt gtc aca 288 Cys Gly Leu Ala Thr Phe Val Ile Ile Leu Leu Leu Val Phe Val Thr 85 90 95 gaa gtt gtt gta gtg gtt ttg gga tat gtt tac aga gca aag gtg gaa 336 Glu Val Val Val Val Val Leu Gly Tyr Val Tyr Arg Ala Lys Val Glu 100 105 110 aat gag gtt gat cgc agc att cag aaa gtg tat aag acc tac aat gga 384 Asn Glu Val Asp Arg Ser Ile Gln Lys Val Tyr Lys Thr Tyr Asn Gly 115 120 125 acc aac cct gat gct gct agc cgg gct att gat tat gta cag aga cag 432 Thr Asn Pro Asp Ala Ala Ser Arg Ala Ile Asp Tyr Val Gln Arg Gln 130 135 140 ctg cat tgt tgt gga att cac aac tac tca gac tgg gaa aat aca gat 480 Leu His Cys Cys Gly Ile His Asn Tyr Ser Asp Trp Glu Asn Thr Asp 145 150 155 160 tgg ttc aaa gaa acc aaa aac cag agt gtc cct ctt agc tgc tgc aga 528 Trp Phe Lys Glu Thr Lys Asn Gln Ser Val Pro Leu Ser Cys Cys Arg 165 170 175 gag act gcc agc aat tgt aat ggc agc ctg gcc cac cct tcc gac ctc 576 Glu Thr Ala Ser Asn Cys Asn Gly Ser Leu Ala His Pro Ser Asp Leu 180 185 190 tat gct gag ggg tgt gag gct cta gtt gtg aag aag cta caa gaa atc 624 Tyr Ala Glu Gly Cys Glu Ala Leu Val Val Lys Lys Leu Gln Glu Ile 195 200 205 atg atg cat gtg atc tgg gcc gca ctg gca ttt gca gct att cag ctg 672 Met Met His Val Ile Trp Ala Ala Leu Ala Phe Ala Ala Ile Gln Leu 210 215 220 ctg ggc atg ctg tgt gct tgc atc gtg ttg tgc aga agg agt aga gat 720 Leu Gly Met Leu Cys Ala Cys Ile Val Leu Cys Arg Arg Ser Arg Asp 225 230 235 240 cct gct tac gag ctc ctc atc act ggc gga acc tat gca tag 762 Pro Ala Tyr Glu Leu Leu Ile Thr Gly Gly Thr Tyr Ala 245 250 ttgacaactc ttgcctgagc tttttggtct tgttctgatt tggaaggtga attgagcagg 822 tctgctgctg ttggcctctg gagttcattt agttaaagca catgtacact ggtgttggac 882 agagcagctt ggcttttcat gtgcccaact acttactact actgcgat 930 18 253 PRT Homo sapiens DOMAIN (1)..(11) Cytoplasmic (Potential) 18 Met Gly Gln Cys Gly Ile Thr Ser Ser Lys Thr Val Leu Val Phe Leu 1 5 10 15 Asn Leu Ile Phe Trp Gly Ala Ala Gly Ile Leu Cys Tyr Val Gly Ala 20 25 30 Tyr Val Phe Ile Thr Tyr Asp Asp Tyr Asp His Phe Phe Glu Asp Val 35 40 45 Tyr Thr Leu Ile Pro Ala Val Val Ile Ile Ala Val Gly Ala Leu Leu 50 55 60 Phe Ile Ile Gly Leu Ile Gly Cys Cys Ala Thr Ile Arg Glu Ser Arg 65 70 75 80 Cys Gly Leu Ala Thr Phe Val Ile Ile Leu Leu Leu Val Phe Val Thr 85 90 95 Glu Val Val Val Val Val Leu Gly Tyr Val Tyr Arg Ala Lys Val Glu 100 105 110 Asn Glu Val Asp Arg Ser Ile Gln Lys Val Tyr Lys Thr Tyr Asn Gly 115 120 125 Thr Asn Pro Asp Ala Ala Ser Arg Ala Ile Asp Tyr Val Gln Arg Gln 130 135 140 Leu His Cys Cys Gly Ile His Asn Tyr Ser Asp Trp Glu Asn Thr Asp 145 150 155 160 Trp Phe Lys Glu Thr Lys Asn Gln Ser Val Pro Leu Ser Cys Cys Arg 165 170 175 Glu Thr Ala Ser Asn Cys Asn Gly Ser Leu Ala His Pro Ser Asp Leu 180 185 190 Tyr Ala Glu Gly Cys Glu Ala Leu Val Val Lys Lys Leu Gln Glu Ile 195 200 205 Met Met His Val Ile Trp Ala Ala Leu Ala Phe Ala Ala Ile Gln Leu 210 215 220 Leu Gly Met Leu Cys Ala Cys Ile Val Leu Cys Arg Arg Ser Arg Asp 225 230 235 240 Pro Ala Tyr Glu Leu Leu Ile Thr Gly Gly Thr Tyr Ala 245 250 19 1317 DNA Homo sapiens Gene (1)..(1317) Vasoactive Intestinal Peptide Receptor-2 19 atg cgg acg ctg ctg cct ccc gcg ctg ctg acc tgc tgg ctg ctc gcc 48 Met Arg Thr Leu Leu Pro Pro Ala Leu Leu Thr Cys Trp Leu Leu Ala 1 5 10 15 ccc gtg aac agc att cac cca gaa tgc cga ttt cat ctg gaa ata cag 96 Pro Val Asn Ser Ile His Pro Glu Cys Arg Phe His Leu Glu Ile Gln 20 25 30 gag gaa gaa aca aaa tgt aca gag ctt ctg agg tct caa aca gaa aaa 144 Glu Glu Glu Thr Lys Cys Thr Glu Leu Leu Arg Ser Gln Thr Glu Lys 35 40 45 cac aaa gcc tgc agt ggc gtc tgg gac aac atc acg tgc tgg cgg cct 192 His Lys Ala Cys Ser Gly Val Trp Asp Asn Ile Thr Cys Trp Arg Pro 50 55 60 gcc aat gtg gga gag acc gtc acg gtg ccc tgc cca aaa gtc ttc agc 240 Ala Asn Val Gly Glu Thr Val Thr Val Pro Cys Pro Lys Val Phe Ser 65 70 75 80 aat ttt tac agc aaa gca gga aac ata agc aaa aac tgt acg agt gac 288 Asn Phe Tyr Ser Lys Ala Gly Asn Ile Ser Lys Asn Cys Thr Ser Asp 85 90 95 gga tgg tca gag acg ttc cca gat ttc gtc gat gcc tgt ggc tac agc 336 Gly Trp Ser Glu Thr Phe Pro Asp Phe Val Asp Ala Cys Gly Tyr Ser 100 105 110 gac ccg gag gat gag agc aag atc acg ttt tat att ctg gtg aag gcc 384 Asp Pro Glu Asp Glu Ser Lys Ile Thr Phe Tyr Ile Leu Val Lys Ala 115 120 125 att tat acc ctg ggc tac agt gtc tct ctg atg tct ctt gca aca gga 432 Ile Tyr Thr Leu Gly Tyr Ser Val Ser Leu Met Ser Leu Ala Thr Gly 130 135 140 agc ata att ctg tgc ctc ttc agg aag ctg cac tgc acc agg aat tac 480 Ser Ile Ile Leu Cys Leu Phe Arg Lys Leu His Cys Thr Arg Asn Tyr 145 150 155 160 atc cac ctg aac ctg ttc ctg tcc ttc atc ctg aga gcc atc tca gtg 528 Ile His Leu Asn Leu Phe Leu Ser Phe Ile Leu Arg Ala Ile Ser Val 165 170 175 ctg gtc aag gac gac gtt ctc tac tcc agc tct ggc acg ttg cac tgc 576 Leu Val Lys Asp Asp Val Leu Tyr Ser Ser Ser Gly Thr Leu His Cys 180 185 190 cct gac cag cca tcc tcc tgg gtg ggc tgc aag ctg agc ctg gtc ttc 624 Pro Asp Gln Pro Ser Ser Trp Val Gly Cys Lys Leu Ser Leu Val Phe 195 200 205 ctg cag tac tgc atc atg gcc aac ttc ttc tgg ctg ctg gtg gag ggg 672 Leu Gln Tyr Cys Ile Met Ala Asn Phe Phe Trp Leu Leu Val Glu Gly 210 215 220 ctc tac ctc cac acc ctc ctg gtg gcc atg ctc ccc cct aga agg tgc 720 Leu Tyr Leu His Thr Leu Leu Val Ala Met Leu Pro Pro Arg Arg Cys 225 230 235 240 ttc ctg gcc tac ctc ctg atc gga tgg ggc ctc ccc acc gtc tgc atc 768 Phe Leu Ala Tyr Leu Leu Ile Gly Trp Gly Leu Pro Thr Val Cys Ile 245 250 255 ggt gca tgg act gcg gcc agg ctc tac tta gaa gac acc ggt tgc tgg 816 Gly Ala Trp Thr Ala Ala Arg Leu Tyr Leu Glu Asp Thr Gly Cys Trp 260 265 270 gat aca aac gac cac agt gtg ccc tgg tgg gtc ata cga ata ccg att 864 Asp Thr Asn Asp His Ser Val Pro Trp Trp Val Ile Arg Ile Pro Ile 275 280 285 tta att tcc atc atc gtc aat ttt gtc ctt ttc att agt att ata cga 912 Leu Ile Ser Ile Ile Val Asn Phe Val Leu Phe Ile Ser Ile Ile Arg 290 295 300 att ttg ctg cag aag tta aca tcc cca gat gtc ggc ggc aac gac cag 960 Ile Leu Leu Gln Lys Leu Thr Ser Pro Asp Val Gly Gly Asn Asp Gln 305 310 315 320 tct cag tac aag agg ctg gcc aag tcc acg ctc ctg ctt atc ccg ctg 1008 Ser Gln Tyr Lys Arg Leu Ala Lys Ser Thr Leu Leu Leu Ile Pro Leu 325 330 335 ttc ggc gtc cac tac atg gtg ttt gcc gtg ttt ccc atc agc atc tcc 1056 Phe Gly Val His Tyr Met Val Phe Ala Val Phe Pro Ile Ser Ile Ser 340 345 350 tcc aaa tac cag ata ctg ttt gag ctg tgc ctc ggg tcg ttc cag ggc 1104 Ser Lys Tyr Gln Ile Leu Phe Glu Leu Cys Leu Gly Ser Phe Gln Gly 355 360 365 ctg gtg gtg gcc gtc ctc tac tgt ttc ctg aac agt gag gtg cag tgc 1152 Leu Val Val Ala Val Leu Tyr Cys Phe Leu Asn Ser Glu Val Gln Cys 370 375 380 gag ctg aag cga aaa tgg cga agc cgg tgc ccg acc ccg tcc gcg agc 1200 Glu Leu Lys Arg Lys Trp Arg Ser Arg Cys Pro Thr Pro Ser Ala Ser 385 390 395 400 cgg gat tac agg gtc tgc ggt tcc tcc ttc tcc cac aac ggc tcg gag 1248 Arg Asp Tyr Arg Val Cys Gly Ser Ser Phe Ser His Asn Gly Ser Glu 405 410 415 ggc gcc ctg cag ttc cac cgc gcg tcc cga gcc cag tcc ttc ctg caa 1296 Gly Ala Leu Gln Phe His Arg Ala Ser Arg Ala Gln Ser Phe Leu Gln 420 425 430 acg gag acc tcg gtc atc tag 1317 Thr Glu Thr Ser Val Ile 435 20 438 PRT Homo sapiens SIGNAL (1)..(23) Potential 20 Met Arg Thr Leu Leu Pro Pro Ala Leu Leu Thr Cys Trp Leu Leu Ala 1 5 10 15 Pro Val Asn Ser Ile His Pro Glu Cys Arg Phe His Leu Glu Ile Gln 20 25 30 Glu Glu Glu Thr Lys Cys Ala Glu Leu Leu Arg Ser Gln Thr Glu Lys 35 40 45 His Lys Ala Cys Ser Gly Val Trp Asp Asn Ile Thr Cys Trp Arg Pro 50 55 60 Ala Asn Val Gly Glu Thr Val Thr Val Pro Cys Pro Lys Val Phe Ser 65 70 75 80 Asn Phe Tyr Ser Lys Ala Gly Asn Ile Ser Lys Asn Cys Thr Ser Asp 85 90 95 Gly Trp Ser Glu Thr Phe Pro Asp Phe Val Asp Ala Cys Gly Tyr Ser 100 105 110 Asp Pro Glu Asp Glu Ser Lys Ile Thr Phe Tyr Ile Leu Val Lys Ala 115 120 125 Ile Tyr Thr Leu Gly Tyr Ser Val Ser Leu Met Ser Leu Ala Thr Gly 130 135 140 Ser Ile Ile Leu Cys Leu Phe Arg Lys Leu His Cys Thr Arg Asn Tyr 145 150 155 160 Ile His Leu Asn Leu Phe Leu Ser Phe Ile Leu Arg Ala Ile Ser Val 165 170 175 Leu Val Lys Asp Asp Val Leu Tyr Ser Ser Ser Gly Thr Leu His Cys 180 185 190 Pro Asp Gln Pro Ser Ser Trp Val Gly Cys Lys Leu Ser Leu Val Phe 195 200 205 Leu Gln Tyr Cys Ile Met Ala Asn Phe Phe Trp Leu Leu Val Glu Gly 210 215 220 Leu Tyr Leu His Thr Leu Leu Val Ala Met Leu Pro Pro Arg Arg Cys 225 230 235 240 Phe Leu Ala Tyr Leu Leu Ile Gly Trp Gly Leu Pro Thr Val Cys Ile 245 250 255 Gly Ala Trp Thr Ala Ala Arg Leu Tyr Leu Glu Asp Thr Gly Cys Trp 260 265 270 Asp Thr Asn Asp His Ser Val Pro Trp Trp Val Ile Arg Ile Pro Ile 275 280 285 Leu Ile Ser Ile Ile Val Asn Phe Val Leu Phe Ile Ser Ile Ile Arg 290 295 300 Ile Leu Leu Gln Lys Leu Thr Ser Pro Asp Val Gly Gly Asn Asp Gln 305 310 315 320 Ser Gln Tyr Lys Arg Leu Ala Lys Ser Thr Leu Leu Leu Ile Pro Leu 325 330 335 Phe Gly Val His Tyr Met Val Phe Ala Val Phe Pro Ile Ser Ile Ser 340 345 350 Ser Lys Tyr Gln Ile Leu Phe Glu Leu Cys Leu Gly Ser Phe Gln Gly 355 360 365 Leu Val Val Ala Val Leu Tyr Cys Phe Leu Asn Ser Glu Val Gln Cys 370 375 380 Glu Leu Lys Arg Lys Trp Arg Ser Arg Cys Pro Thr Pro Ser Ala Ser 385 390 395 400 Arg Asp Tyr Arg Val Cys Gly Ser Ser Phe Ser Arg Asn Gly Ser Glu 405 410 415 Gly Ala Leu Gln Phe His Arg Gly Ser Arg Ala Gln Ser Phe Leu Gln 420 425 430 Thr Glu Thr Ser Val Ile 435 21 889 DNA Homo sapiens Gene (1)..(889) Pleiotrophin 21 gtcaaaggca ggatcaggtt ccccgccttc cagtccaaaa atcccgccaa gagagcccca 60 gagcagagga aaatccaaag tggagagagg ggaagaaaga gaccagtgag tcatccgtcc 120 agaaggcggg gagagcagca gcggcccaag caggagctgc agcgagccgg gtacctggac 180 tcagcggtag caacctcgcc ccttgcaaca aaggcagact gagcgccaga gaggacgttt 240 ccaactcaaa a atg cag gct caa cag tac cag cag cag cgt cga aaa ttt 290 Met Gln Ala Gln Gln Tyr Gln Gln Gln Arg Arg Lys Phe 1 5 10 gca gct gcc ttc ttg gca ttc att ttc ata ctg gca gct gtg gat act 338 Ala Ala Ala Phe Leu Ala Phe Ile Phe Ile Leu Ala Ala Val Asp Thr 15 20 25 gct gaa gca ggg aag aaa gag aaa cca gaa aaa aaa gtg aag aag tct 386 Ala Glu Ala Gly Lys Lys Glu Lys Pro Glu Lys Lys Val Lys Lys Ser 30 35 40 45 gac tgt gga gaa tgg cag tgg agt gtg tgt gtg ccc acc agt gga gac 434 Asp Cys Gly Glu Trp Gln Trp Ser Val Cys Val Pro Thr Ser Gly Asp 50 55 60 tgt ggg ctg ggc aca cgg gag ggc act cgg act gga gct gag tgc aag 482 Cys Gly Leu Gly Thr Arg Glu Gly Thr Arg Thr Gly Ala Glu Cys Lys 65 70 75 caa acc atg aag acc cag aga tgt aag atc ccc tgc aac tgg aag aag 530 Gln Thr Met Lys Thr Gln Arg Cys Lys Ile Pro Cys Asn Trp Lys Lys 80 85 90 caa ttt ggc gcg gag tgc aaa tac cag ttc cag gcc tgg gga gaa tgt 578 Gln Phe Gly Ala Glu Cys Lys Tyr Gln Phe Gln Ala Trp Gly Glu Cys 95 100 105 gac ctg aac aca gcc ctg aag acc aga act gga agt ctg aag cga gcc 626 Asp Leu Asn Thr Ala Leu Lys Thr Arg Thr Gly Ser Leu Lys Arg Ala 110 115 120 125 ctg cac aat gcc gaa tgc cag aag act gtc acc atc tcc aag ccc tgt 674 Leu His Asn Ala Glu Cys Gln Lys Thr Val Thr Ile Ser Lys Pro Cys 130 135 140 ggc aaa ctg acc aag ccc aaa cct caa gca gaa tct aag aag aag aaa 722 Gly Lys Leu Thr Lys Pro Lys Pro Gln Ala Glu Ser Lys Lys Lys Lys 145 150 155 aag gaa ggc aag aaa cag gag aag atg ctg gat taa aagatgtcac 768 Lys Glu Gly Lys Lys Gln Glu Lys Met Leu Asp 160 165 ctgtggaaca taaaaaggac atcagcaaac aggatcagtt aactattgca tttatatgta 828 ccgtaggctt tgtattcaaa aattatctat agctaagtac acaataagca aaaacaaaaa 888 g 889 22 168 PRT Homo sapiens SIGNAL (1)..(32) 22 Met Gln Ala Gln Gln Tyr Gln Gln Gln Arg Arg Lys Phe Ala Ala Ala 1 5 10 15 Phe Leu Ala Phe Ile Phe Ile Leu Ala Ala Val Asp Thr Ala Glu Ala 20 25 30 Gly Lys Lys Glu Lys Pro Glu Lys Lys Val Lys Lys Ser Asp Cys Gly 35 40 45 Glu Trp Gln Trp Ser Val Cys Val Pro Thr Ser Gly Asp Cys Gly Leu 50 55 60 Gly Thr Arg Glu Gly Thr Arg Thr Gly Ala Glu Cys Lys Gln Thr Met 65 70 75 80 Lys Thr Gln Arg Cys Lys Ile Pro Cys Asn Trp Lys Lys Gln Phe Gly 85 90 95 Ala Glu Cys Lys Tyr Gln Phe Gln Ala Trp Gly Glu Cys Asp Leu Asn 100 105 110 Thr Ala Leu Lys Thr Arg Thr Gly Ser Leu Lys Arg Ala Leu His Asn 115 120 125 Ala Glu Cys Gln Lys Thr Val Thr Ile Ser Lys Pro Cys Gly Lys Leu 130 135 140 Thr Lys Pro Lys Pro Gln Ala Glu Ser Lys Lys Lys Lys Lys Glu Gly 145 150 155 160 Lys Lys Gln Glu Lys Met Leu Asp 165 23 3143 DNA Homo sapiens Gene (1)..(3143) Osteopontin 23 ggggaagtgt gggagcaggt gggctgggca gtggcagaaa cctgatgaca caatctcgcc 60 gcctccctgt gttggtggag gatgtctgca gcagcattta aattctggga gggcttggtt 120 gtcagcagca gcaggaggag gcagagacag catcgtcggg accagactcg tctcaggcca 180 gttgcagcct tctcagccaa acgccgacca aggtacagct tcagtttgct actgggttgt 240 gcattcagct gaatttcatg gggaagtcca aattctaagg aaaaaaatgt ggtagtataa 300 aaaggtatca ctgttgtaac ctatgaagat gtcagctatt cctttgaaat attttgcagg 360 aaaactcact acc atg aga att gca gtg att tgc ttt tgc ctc cta ggc 409 Met Arg Ile Ala Val Ile Cys Phe Cys Leu Leu Gly 1 5 10 atc acc tgt gcc ata cca gtgagtacag ttgcatctta aagaaaattc 457 Ile Thr Cys Ala Ile Pro 15 ctgaaaataa ctgaattgtg tgcttccatg tgctaggagg acattcttgt aatctttctt 517 catcttttct gtttctaag gtt aaa cag gct gat tct gga agt tct gag gaa 569 Val Lys Gln Ala Asp Ser Gly Ser Ser Glu Glu 20 25 aag cag gtaagcatct tttatgtttt tatatagtta aatcatttac tcaattatgg 625 Lys Gln 30 cgagaggtgc aagaaacgta tttgctgcga tcaaatgagt tcatatttgt aaagcaattt 685 gaaagagtgc ctagcccaca gtaagtgcta cataagagtt tgttaaatga atctgcaaaa 745 aaaaaaaaaa ttacaaaaag gtacctaagg gtccgggtga ctatatgctt ccatcaagac 805 tagtgaagaa tggttgtttt ttccattcat ccctacattt ctttttttaa taatgataaa 865 catgcaactt ttttgtag ctt tac aac aaa tac cca gat gct gtg gcc aca 916 Leu Tyr Asn Lys Tyr Pro Asp Ala Val Ala Thr 35 40 tgg cta aac cct gac cca tct cag aag cag aat ctc cta gcc cca cag 964 Trp Leu Asn Pro Asp Pro Ser Gln Lys Gln Asn Leu Leu Ala Pro Gln 45 50 55 gtatttttaa acttctcata attaaactac agtgatgaaa gatagccaca ctcaggccat 1024 ttgggctgct cagatgaatc ctgccctgcc tgctggcaaa catgtgctta ggacattgac 1084 tgatctgcca tgttggcttc tctctgtgtt aagccatcca cagatgaggc tgaaaaataa 1144 aaactgcttt ggattaaaaa ggttaacttt tgaataaaaa agctaggcat gtgtgatgcg 1204 cactaacacg tgccattcct tcttcag aat gct gtg tcc tct gaa gaa acc aat 1258 Asn Ala Val Ser Ser Glu Glu Thr Asn 60 65 gac ttt aaa caa gag gtaagttctc attttcaatc agaggcccat catgccttga 1313 Asp Phe Lys Gln Glu 70 agagatgaaa gaaggcattg cctggattct cttctgatga aatttcatta gcaagttttc 1373 cagctaattg gcagtctaaa acttgctcat aaataaaaca tgtatttact aaatatcaga 1433 aatactaggt ttcctcggat aacctaaaag ccatggtatg tactgtgaat gcaaagattc 1493 tgaaactaaa taaaaagaaa gatagtaaaa gactaatgtg ctataaaggc taagggaaaa 1553 taaaaaccca tatattaatt ttcccggcca tcttaatttt cag acc ctt cca agt 1608 Thr Leu Pro Ser 75 aag tcc aac gaa agc cat gac cac atg gat gat atg gat gat gaa gat 1656 Lys Ser Asn Glu Ser His Asp His Met Asp Asp Met Asp Asp Glu Asp 80 85 90 gat gat gac cat gtg gac agc cag gac tcc att gac tcg aac gac tct 1704 Asp Asp Asp His Val Asp Ser Gln Asp Ser Ile Asp Ser Asn Asp Ser 95 100 105 gat gat gta gat gac act gat gat tct cac cag tct gat gag tct cac 1752 Asp Asp Val Asp Asp Thr Asp Asp Ser His Gln Ser Asp Glu Ser His 110 115 120 cat tct gat gaa tct gat gaa ctg gtc act gat ttt ccc acg gac ctg 1800 His Ser Asp Glu Ser Asp Glu Leu Val Thr Asp Phe Pro Thr Asp Leu 125 130 135 140 cca gca acc gaa gtt ttc act cca gtt gtc ccc aca gta gac aca tat 1848 Pro Ala Thr Glu Val Phe Thr Pro Val Val Pro Thr Val Asp Thr Tyr 145 150 155 gat ggc cga ggt gat agt gtg gtt tat gga ctg agg tca aaa tct aag 1896 Asp Gly Arg Gly Asp Ser Val Val Tyr Gly Leu Arg Ser Lys Ser Lys 160 165 170 aag ttt cgc aga cct gac atc cag gtaaatcctt taacagacac acctgatggt 1950 Lys Phe Arg Arg Pro Asp Ile Gln 175 180 tctgactagc gctcaagtct aggaaaccac agtttgcata ttcattcatt cattcatcca 2010 ttcattcatc cattcagcaa gaattcattc atattctact ttatgaccat tgaatacaaa 2070 tctttttctg cttggcggtt tttgtaagtc tacataattt ctctctagat ttgattctca 2130 aacacaattc tactttttga aatcctggat caaagtaaca tgctagtatt atttcagcca 2190 gatttagaca atttttagta taagatgacc taaaagctag agagtggaaa aggattacca 2250 tattcccatc cctagccgtt catataatta ttcttcattt gtgccgtgat tcag tac 2307 Tyr cct gat gct aca gac gag gac atc acc tca cac atg gaa agc gag gag 2355 Pro Asp Ala Thr Asp Glu Asp Ile Thr Ser His Met Glu Ser Glu Glu 185 190 195 ttg aat ggt gca tac aag gcc atc ccc gtt gcc cag gac ctg aac gcg 2403 Leu Asn Gly Ala Tyr Lys Ala Ile Pro Val Ala Gln Asp Leu Asn Ala 200 205 210 cct tct gat tgg gac agc cgt ggg aag gac agt tat gaa acg agt cag 2451 Pro Ser Asp Trp Asp Ser Arg Gly Lys Asp Ser Tyr Glu Thr Ser Gln 215 220 225 ctg gat gac cag agt gct gaa acc cac agc cac aag cag tcc aga tta 2499 Leu Asp Asp Gln Ser Ala Glu Thr His Ser His Lys Gln Ser Arg Leu 230 235 240 245 tat aag cgg aaa gcc aat gat gag agc aat gag cat tcc gat gtg att 2547 Tyr Lys Arg Lys Ala Asn Asp Glu Ser Asn Glu His Ser Asp Val Ile 250 255 260 gat agt cag gaa ctt tcc aaa gtc agc cgt gaa ttc cac agc cat gaa 2595 Asp Ser Gln Glu Leu Ser Lys Val Ser Arg Glu Phe His Ser His Glu 265 270 275 ttt cac agc cat gaa gat atg ctg gtt gta gac ccc aaa agt aag gaa 2643 Phe His Ser His Glu Asp Met Leu Val Val Asp Pro Lys Ser Lys Glu 280 285 290 gaa gat aaa cac ctg aaa ttt cgt att tct cat gaa tta gat agt gca 2691 Glu Asp Lys His Leu Lys Phe Arg Ile Ser His Glu Leu Asp Ser Ala 295 300 305 tct tct gag gtc aat taa aaggagaaaa aatacaattt ctcactttgc 2739 Ser Ser Glu Val Asn 310 atttagtcaa aagaaaaaat gctttatagc aaaatgaaag agaacatgaa atgcttcttt 2799 ctcagtttat tggttgaatg tgtatctatt tgagtctgga aataactaat gtgtttgata 2859 attagtttag tttgtggctt catggaaact ccctgtaaac aaaagcttca gggttatgtc 2919 tatgttcatt ctatagaaga aatgcaaact atcactgtat tttaatattt gttattctct 2979 catgaataga aatttatgta gaagcaaaca aaatactttt acccacttaa aaagagaata 3039 taacatttta tgtcactata atcttttgtt ttttaagtta gtgtatattt tgttgtgatt 3099 atcttttgtg gtgtgaataa atcttttatc ttgaatgtaa taag 3143 24 314 PRT Homo sapiens SIGNAL (1)..(16) Potential 24 Met Arg Ile Ala Val Ile Cys Phe Cys Leu Leu Gly Ile Thr Cys Ala 1 5 10 15 Ile Pro Val Lys Gln Ala Asp Ser Gly Ser Ser Glu Glu Lys Gln Leu 20 25 30 Tyr Asn Lys Tyr Pro Asp Ala Val Ala Thr Trp Leu Asn Pro Asp Pro 35 40 45 Ser Gln Lys Gln Asn Leu Leu Ala Pro Gln Asn Ala Val Ser Ser Glu 50 55 60 Glu Thr Asn Asp Phe Lys Gln Glu Thr Leu Pro Ser Lys Ser Asn Glu 65 70 75 80 Ser His Asp His Met Asp Asp Met Asp Asp Glu Asp Asp Asp Asp His 85 90 95 Val Asp Ser Gln Asp Ser Ile Asp Ser Asn Asp Ser Asp Asp Val Asp 100 105 110 Asp Thr Asp Asp Ser His Gln Ser Asp Glu Ser His His Ser Asp Glu 115 120 125 Ser Asp Glu Leu Val Thr Asp Phe Pro Thr Asp Leu Pro Ala Thr Glu 130 135 140 Val Phe Thr Pro Val Val Pro Thr Val Asp Thr Tyr Asp Gly Arg Gly 145 150 155 160 Asp Ser Val Val Tyr Gly Leu Arg Ser Lys Ser Lys Lys Phe Arg Arg 165 170 175 Pro Asp Ile Gln Tyr Pro Asp Ala Thr Asp Glu Asp Ile Thr Ser His 180 185 190 Met Glu Ser Glu Glu Leu Asn Gly Ala Tyr Lys Ala Ile Pro Val Ala 195 200 205 Gln Asp Leu Asn Ala Pro Ser Asp Trp Asp Ser Arg Gly Lys Asp Ser 210 215 220 Tyr Glu Thr Ser Gln Leu Asp Asp Gln Ser Ala Glu Thr His Ser His 225 230 235 240 Lys Gln Ser Arg Leu Tyr Lys Arg Lys Ala Asn Asp Glu Ser Asn Glu 245 250 255 His Ser Asp Val Ile Asp Ser Gln Glu Leu Ser Lys Val Ser Arg Glu 260 265 270 Phe His Ser His Glu Phe His Ser His Glu Asp Met Leu Val Val Asp 275 280 285 Pro Lys Ser Lys Glu Glu Asp Lys His Leu Lys Phe Arg Ile Ser His 290 295 300 Glu Leu Asp Ser Ala Ser Ser Glu Val Asn 305 310 25 259 PRT Homo sapiens Gene (1)..(259) Carbonic Anhydrase domain of human carbonic anhydrase III 25 Ala Lys Glu Trp Gly Tyr Ala Ser His Asn Gly Pro Asp His Trp His 1 5 10 15 Glu Leu Phe Pro Asn Ala Lys Gly Glu Asn Gln Ser Pro Ile Glu Leu 20 25 30 His Thr Lys Asp Ile Arg His Asp Pro Ser Leu Gln Pro Trp Ser Val 35 40 45 Ser Tyr Asp Gly Gly Ser Ala Lys Thr Ile Leu Asn Asn Gly Lys Thr 50 55 60 Cys Arg Val Val Phe Asp Asp Thr Tyr Asp Arg Ser Met Leu Arg Gly 65 70 75 80 Gly Pro Leu Pro Gly Pro Tyr Arg Leu Arg Gln Phe His Leu His Trp 85 90 95 Gly Ser Ser Asp Asp His Gly Ser Glu His Thr Val Asp Gly Val Lys 100 105 110 Tyr Ala Ala Glu Leu His Leu Val His Trp Asn Pro Lys Tyr Asn Thr 115 120 125 Phe Lys Glu Ala Leu Lys Gln Arg Asp Gly Ile Ala Val Ile Gly Ile 130 135 140 Phe Leu Lys Ile Gly His Glu Asn Gly Glu Phe Gln Ile Phe Leu Asp 145 150 155 160 Ala Leu Asp Lys Ile Lys Thr Lys Gly Lys Glu Ala Pro Phe Thr Lys 165 170 175 Phe Asp Pro Ser Cys Leu Phe Pro Ala Cys Arg Asp Tyr Trp Thr Tyr 180 185 190 Gln Gly Ser Phe Thr Thr Pro Pro Cys Glu Glu Cys Ile Val Trp Leu 195 200 205 Leu Leu Lys Glu Pro Met Thr Val Ser Ser Asp Gln Met Ala Lys Leu 210 215 220 Arg Ser Leu Leu Ser Ser Ala Glu Asn Glu Pro Pro Val Pro Leu Val 225 230 235 240 Ser Asn Trp Arg Pro Pro Gln Pro Ile Asn Asn Arg Val Val Arg Ala 245 250 255 Ser Phe Lys 26 260 PRT Homo sapiens Gene (1)..(260) Carbonic anhydrase domain of human carbonic anhydrase I 26 Ala Ser Pro Asp Trp Gly Tyr Asp Asp Lys Asn Gly Pro Glu Gln Trp 1 5 10 15 Ser Lys Leu Tyr Pro Ile Ala Asn Gly Asn Asn Gln Ser Pro Val Asp 20 25 30 Ile Lys Thr Ser Glu Thr Lys His Asp Thr Ser Leu Lys Pro Ile Ser 35 40 45 Val Ser Tyr Asn Pro Ala Thr Ala Lys Glu Ile Ile Asn Val Gly His 50 55 60 Ser Phe His Val Asn Phe Glu Asp Asn Asp Asn Arg Ser Val Leu Lys 65 70 75 80 Gly Gly Pro Phe Ser Asp Ser Tyr Arg Leu Phe Gln Phe His Phe His 85 90 95 Trp Gly Ser Thr Asn Glu His Gly Ser Glu His Thr Val Asp Gly Val 100 105 110 Lys Tyr Ser Ala Glu Leu His Val Ala His Trp Asn Ser Ala Lys Tyr 115 120 125 Ser Ser Leu Ala Glu Ala Ala Ser Lys Ala Asp Gly Leu Ala Val Ile 130 135 140 Gly Val Leu Met Lys Val Gly Glu Ala Asn Pro Lys Leu Gln Lys Val 145 150 155 160 Leu Asp Ala Leu Gln Ala Ile Lys Thr Lys Gly Lys Arg Ala Pro Phe 165 170 175 Thr Asn Phe Asp Pro Ser Thr Leu Leu Pro Ser Ser Leu Asp Phe Trp 180 185 190 Thr Tyr Pro Gly Ser Leu Thr His Pro Pro Leu Tyr Glu Ser Val Thr 195 200 205 Trp Ile Ile Cys Lys Glu Ser Ile Ser Val Ser Ser Glu Gln Leu Ala 210 215 220 Gln Phe Arg Ser Leu Leu Ser Asn Val Glu Gly Asp Asn Ala Val Pro 225 230 235 240 Met Gln His Asn Asn Arg Pro Thr Gln Pro Leu Lys Gly Arg Thr Val 245 250 255 Arg Ala Ser Phe 260 27 337 PRT Homo sapiens Gene (1)..(337) Carbonic anhydrase domain of human carbonic anhydrase VIX 27 Met Leu Phe Ser Ala Leu Leu Leu Glu Val Ile Trp Ile Leu Ala Ala 1 5 10 15 Asp Gly Gly Gln His Trp Thr Tyr Glu Gly Pro His Gly Gln Asp His 20 25 30 Trp Pro Ala Ser Tyr Pro Glu Cys Gly Asn Asn Ala Gln Ser Pro Ile 35 40 45 Asp Ile Gln Thr Asp Ser Val Thr Phe Asp Pro Asp Leu Pro Ala Leu 50 55 60 Gln Pro His Gly Tyr Asp Gln Pro Gly Thr Glu Pro Leu Asp Leu His 65 70 75 80 Asn Asn Gly His Thr Val Gln Leu Ser Leu Pro Ser Thr Leu Tyr Leu 85 90 95 Gly Gly Leu Pro Arg Lys Tyr Val Ala Ala Gln Leu His Leu His Trp 100 105 110 Gly Gln Lys Gly Ser Pro Gly Gly Ser Glu His Gln Ile Asn Ser Glu 115 120 125 Ala Thr Phe Ala Glu Leu His Ile Val His Tyr Asp Ser Asp Ser Tyr 130 135 140 Asp Ser Leu Ser Glu Ala Ala Glu Arg Pro Gln Gly Leu Ala Val Leu 145 150 155 160 Gly Ile Leu Ile Glu Val Gly Glu Thr Lys Asn Ile Ala Tyr Glu His 165 170 175 Ile Leu Ser His Leu His Glu Val Arg His Lys Asp Gln Lys Thr Ser 180 185 190 Val Pro Pro Phe Asn Leu Arg Glu Leu Leu Pro Lys Gln Leu Gly Gln 195 200 205 Tyr Phe Arg Tyr Asn Gly Ser Leu Thr Thr Pro Pro Cys Tyr Gln Ser 210 215 220 Val Leu Trp Thr Val Phe Tyr Arg Arg Ser Gln Ile Ser Met Glu Gln 225 230 235 240 Leu Glu Lys Leu Gln Gly Thr Leu Phe Ser Thr Glu Glu Glu Pro Ser 245 250 255 Lys Leu Leu Val Gln Asn Tyr Arg Ala Leu Gln Pro Leu Asn Gln Arg 260 265 270 Met Val Phe Ala Ser Phe Ile Gln Ala Gly Ser Ser Tyr Thr Thr Gly 275 280 285 Glu Met Leu Ser Leu Gly Val Gly Ile Leu Val Gly Cys Leu Cys Leu 290 295 300 Leu Leu Ala Val Tyr Phe Ile Ala Arg Lys Ile Arg Lys Lys Arg Leu 305 310 315 320 Glu Asn Arg Lys Ser Val Val Phe Thr Ser Ala Gln Ala Thr Thr Glu 325 330 335 Ala 28 22 DNA Artificial sequence Primer 28 cagcagttgg atggaagagg ac 22 29 22 DNA Artificial sequence Primer 29 cactgagatt ctggcactat tc 22 30 21 DNA Artificial sequence Primer 30 aacaattcca gggtctcact c 21 31 21 DNA Artificial sequence Primer 31 ttgactggct caggagtata g 21 32 21 DNA Artificial sequence Primer 32 ctgataatga gggctcccaa c 21 33 24 DNA Artificial sequence Primer 33 ctctgcactt cctggtaaaa ctct 24 34 22 DNA Artificial sequence Primer 34 cagcagttgg atggaagagg ac 22 35 24 DNA Artificial sequence Primer 35 ctctgcactt cctggtaaaa ctct 24

Claims (17)

We claim:
1. A method to treat a brain tumor comprising administering a therapeutic amount of a composition comprising:
a compound of the general formula α(VIPR2), wherein α(VIPR2) is one or more moieties which specifically binds to a human vasoactive intestinal peptide receptor-2, wherein the binding of α(VIPR2) alters the function of the vasoactive intestinal peptide receptor-2,
and a pharmaceutically acceptable carrier.
2. The method of claim 1 wherein the therapeutic composition is administered by intrathecal administration.
3. The method of claim 1 wherein the therapeutic composition is administered by intravascular administration.
4. The method of claim 1 wherein the brain tumor is a glioblastoma.
5. The method of claim 1 wherein α(VIPR2) is selected from the group consisting of an antibody and an antibody fragment.
6. The method of claim 1 wherein the composition is administered as a single dose.
7. The method of claim 1 wherein the composition is administered as a series of doses over a period of time.
8. The method of claim 1 wherein α(VIPR2) further comprises attached thereto one or more cytotoxic moieties C.
9. The method of claim 8 wherein the therapeutic composition is administered by intrathecal administration.
10. The method of claim 8 wherein the therapeutic composition is administered by intravascular administration.
11. The method of claim 8 wherein the brain tumor is a glioblastoma.
12. The method of claim 8 wherein α(VIPR2) is selected from the group consisting of an antibody and an antibody fragment.
13. The method of claim 8 wherein C is a radioactive moiety.
14. The method of claim 8 wherein C is a chemotoxic moiety.
15. The method of claim 8 wherein C is a toxin protein moiety.
16. The method of claim 8 wherein the composition is administered as a single dose.
17. The method of claim 8 wherein the composition is administered as a series of doses over a period of time.
US09/983,000 2001-03-23 2001-10-17 Use of protein biomolecular targets in the treatment and visualization of brain tumors Abandoned US20030118585A1 (en)

Priority Applications (7)

Application Number Priority Date Filing Date Title
US09/983,000 US20030118585A1 (en) 2001-10-17 2001-10-17 Use of protein biomolecular targets in the treatment and visualization of brain tumors
JP2002575022A JP2004537282A (en) 2001-03-23 2002-03-22 Use of biomolecular targets in the treatment and visualization of brain tumors
PCT/US2002/008992 WO2002076510A1 (en) 2001-03-23 2002-03-22 Use of biomolecular targets in the treatment and visualization of brain tumors
CA002439630A CA2439630A1 (en) 2001-03-23 2002-03-22 Use of biomolecular targets in the treatment and visualization of brain tumors
EP02726668A EP1383543A4 (en) 2001-03-23 2002-03-22 Use of biomolecular targets in the treatment and visualization of brain tumors
US10/652,981 US7060275B2 (en) 2001-10-17 2003-08-28 Use of protein biomolecular targets in the treatment and visualization of brain tumors
US11/097,728 US20050260132A1 (en) 2001-10-17 2005-03-31 Monoclonal antibodies directed to receptor protein tyrosine phosphatase zeta

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
US09/983,000 US20030118585A1 (en) 2001-10-17 2001-10-17 Use of protein biomolecular targets in the treatment and visualization of brain tumors

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US10/652,981 Continuation US7060275B2 (en) 2001-10-17 2003-08-28 Use of protein biomolecular targets in the treatment and visualization of brain tumors

Publications (1)

Publication Number Publication Date
US20030118585A1 true US20030118585A1 (en) 2003-06-26

Family

ID=25529729

Family Applications (2)

Application Number Title Priority Date Filing Date
US09/983,000 Abandoned US20030118585A1 (en) 2001-03-23 2001-10-17 Use of protein biomolecular targets in the treatment and visualization of brain tumors
US10/652,981 Expired - Fee Related US7060275B2 (en) 2001-10-17 2003-08-28 Use of protein biomolecular targets in the treatment and visualization of brain tumors

Family Applications After (1)

Application Number Title Priority Date Filing Date
US10/652,981 Expired - Fee Related US7060275B2 (en) 2001-10-17 2003-08-28 Use of protein biomolecular targets in the treatment and visualization of brain tumors

Country Status (1)

Country Link
US (2) US20030118585A1 (en)

Cited By (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040248157A1 (en) * 2001-09-14 2004-12-09 Michal Ayalon-Soffer Novel polynucleotides encoding soluble polypeptides and methods using same
US20040265799A1 (en) * 2003-06-24 2004-12-30 Compugen Ltd. Human-virus homologous sequences and uses thereof
US20050123538A1 (en) * 2003-10-03 2005-06-09 Ronen Shemesh Polynucleotides encoding novel ErbB-2 polypeptides and kits and methods using same
US20050186600A1 (en) * 2004-01-13 2005-08-25 Osnat Sella-Tavor Polynucleotides encoding novel UbcH10 polypeptides and kits and methods using same
US20060068405A1 (en) * 2004-01-27 2006-03-30 Alex Diber Methods and systems for annotating biomolecular sequences
US20060165679A1 (en) * 2002-05-08 2006-07-27 Bayer Healthcare Ag Diagnostics and therapeutics for diseases associated with vasoactive intestinal peptide receptor 2 (vpac2)
US20060199180A1 (en) * 2002-08-06 2006-09-07 Macina Roberto A Compositions and methods relating to ovarian specific genes and proteins
US20070083334A1 (en) * 2001-09-14 2007-04-12 Compugen Ltd. Methods and systems for annotating biomolecular sequences
US20070082337A1 (en) * 2004-01-27 2007-04-12 Compugen Ltd. Methods of identifying putative gene products by interspecies sequence comparison and biomolecular sequences uncovered thereby
WO2009015843A1 (en) * 2007-07-27 2009-02-05 Immatics Biotechnologies Gmbh Novel immunotherapy against neuronal and brain tumors
EP2105501A1 (en) * 2008-03-27 2009-09-30 Immatics Biotechnologies GmbH Novel immunotherapy against neuronal and brain tumors
US20110092452A1 (en) * 2008-03-05 2011-04-21 The Regents Of The University Of Michigan Compositions and methods for diagnosing and treating pancreatic cancer
US10420800B2 (en) 2007-07-27 2019-09-24 Immatics Biotechnologies Gmbh Immunotherapy against neuronal and brain tumors
CN111748574A (en) * 2020-06-16 2020-10-09 闽江学院 Method for improving cadmium tolerance of plants by over-expressing Kandelia candel KoCSD3
WO2022147334A1 (en) * 2020-12-31 2022-07-07 Encodia, Inc. Metalloenzymes for biomolecular recognition of n-terminal modified peptides

Families Citing this family (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2061904A2 (en) * 2006-09-06 2009-05-27 Ortho-McNeil Pharmaceutical, Inc. Biomarkers for assessing response to c-met treatment
EP2373331A4 (en) * 2008-12-05 2015-11-18 Abraxis Bioscience Llc Albumin binding peptide-mediated disease targeting
DE102009030321A1 (en) * 2009-06-24 2011-01-05 Siemens Aktiengesellschaft Method for imaging tumor tissue
WO2011020090A2 (en) * 2009-08-14 2011-02-17 Case Western Reserve University Compositions and methods for treating cancer
MY196837A (en) 2015-12-11 2023-05-03 Immatics Biotechnologies Gmbh Novel peptides and combination of peptides for use in immunotherapy against various cancers
GB201521894D0 (en) 2015-12-11 2016-01-27 Immatics Biotechnologies Gmbh Novel peptides and combination of peptides for use in immunotherapy against various cancers
US20230391865A1 (en) * 2020-10-15 2023-12-07 The Regents Of The University Of California Anti-cancer inhibitory antibodies

Citations (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4565789A (en) * 1983-04-04 1986-01-21 The United States Of America As Represented By The Department Of Health And Human Services Cell matrix receptor system and use in cancer diagnosis and management
US4861710A (en) * 1986-09-26 1989-08-29 The United States Of America As Represented By The Department Of Health And Human Services Recombinant DNA clone encoding laminin receptor
US5648273A (en) * 1989-12-27 1997-07-15 The United States Of America, As Represented By The Department Of Health And Human Services Hepatic growth factor receptor is the MET proto-oncogene
US5686292A (en) * 1995-06-02 1997-11-11 Genentech, Inc. Hepatocyte growth factor receptor antagonist antibodies and uses thereof
US5720720A (en) * 1993-08-27 1998-02-24 The United States Of America As Represented By The Department Of Health And Human Services Convection-enhanced drug delivery
US5855866A (en) * 1992-03-05 1999-01-05 Board Of Regenis, The University Of Texas System Methods for treating the vasculature of solid tumors
US5871959A (en) * 1989-12-27 1999-02-16 The United States Of America As Represented By The Department Of Health And Human Services Method of producing hepatocycte growth factor/scatter factor and related cell lines
US5945098A (en) * 1990-02-01 1999-08-31 Baxter International Inc. Stable intravenously-administrable immune globulin preparation
US6034218A (en) * 1996-03-15 2000-03-07 Corixa Corporation Compounds and methods for immunotherapy and immunodiagnosis of prostate cancer
US6051230A (en) * 1992-03-05 2000-04-18 Board Of Regents, The University Of Texas System Compositions for targeting the vasculature of solid tumors
US6063905A (en) * 1997-01-07 2000-05-16 Board Of Regents, The University Of Texas System Recombinant human IGA-J. chain dimer
US6147203A (en) * 1993-06-14 2000-11-14 The United States Of America As Represented By The Department Of Health And Human Services Recombinant disulfide-stabilized polypeptide fragments having binding specificity
US6174708B1 (en) * 1995-12-04 2001-01-16 Pasteur Merieux Serums & Vaccins Preparation of a multicombinatorial library of antibody gene expression vectors
US6187287B1 (en) * 1994-08-12 2001-02-13 Immunomedics, Inc. Immunoconjugates and humanized antibodies specific for B-cell lymphoma and leukemia cells
US6750194B1 (en) * 2000-10-23 2004-06-15 The Procter & Gamble Company Methods of identifying compounds for regulating muscle mass or function using vasoactive intestinal peptide receptors

Family Cites Families (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5362716A (en) 1989-12-27 1994-11-08 The United States Of America As Represented By The Department Of Health And Human Services Methods for stimulating hematopoietic progenitors using hepatocyte growth factor and lymphokines
US6344321B1 (en) 1990-06-11 2002-02-05 Gilead Sciences, Inc. Nucleic acid ligands which bind to hepatocyte growth factor/scatter factor (HGF/SF) or its receptor c-met
DK0584125T3 (en) 1991-05-10 1997-09-08 Pharmacia & Upjohn Spa Truncated forms of hepatocyte growth factor (HGF) receptor
GB9323884D0 (en) * 1993-11-19 1994-01-05 Eisai London Res Lab Ltd Physiological modulation
US20010053371A1 (en) 1999-01-07 2001-12-20 Waldemar Debinski Method for diagnosing, imaging, and treating tumors using restrictive receptor for interleukin 13
JP3832674B2 (en) 1995-10-24 2006-10-11 敏一 中村 Anticancer drug
US6020179A (en) * 1996-10-03 2000-02-01 Incyte Pharmaceuticals, Inc. Nucleic acids encoding human tyrosine phosphatases
WO1998019696A1 (en) 1996-11-05 1998-05-14 Smithkline Beecham Corporation Hepatocyte growth factor antagonists
EP0950097A2 (en) 1996-12-27 1999-10-20 Instituto de Investigaciones Bioquimicas Fundacion Campomar Compositions and methods for tumour therapy
ATE359298T1 (en) 1997-05-30 2007-05-15 Stefan Weiss SOLUBLE LAMININ RECEPTOR PRECURSOR AND METHOD FOR PREVENTING LAMININ INTERACTIONS
JP4094814B2 (en) 1998-04-28 2008-06-04 敏一 中村 Angiogenesis inhibitor
AU4320499A (en) 1998-06-02 1999-12-20 Eli Lilly And Company Angiopoietin related gene sequence scarface 1
AU5571799A (en) 1998-08-20 2000-03-14 University Of Vermont And State Agricultural College, The Angiogenesis inhibitors and uses thereof
EP1121598A4 (en) 1998-10-06 2002-01-30 Univ Georgetown Detection of pleiotrophin
EP1171459A1 (en) 1999-04-01 2002-01-16 Genetics Institute, Inc. Angiopoietin-like proteins, crisp proteins and polynucleotides encoding same
AU5022200A (en) 1999-05-19 2000-12-05 Human Genome Sciences, Inc. Tm4sf receptors
IL146794A0 (en) 1999-05-28 2002-07-25 Univ Virginia Osteonectin based toxic gene therapy for the treatment of calcified tumors and tissues
WO2000078361A2 (en) 1999-06-17 2000-12-28 Regeneron Pharmaceuticals, Inc. Methods of imaging and targeting tumor vasculature
WO2001013105A1 (en) 1999-07-30 2001-02-22 Agy Therapeutics, Inc. Techniques for facilitating identification of candidate genes
US6455026B1 (en) * 2001-03-23 2002-09-24 Agy Therapeutics, Inc. Use of protein tyrosine phosphatase zeta as a biomolecular target in the treatment and visualization of brain tumors

Patent Citations (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4565789A (en) * 1983-04-04 1986-01-21 The United States Of America As Represented By The Department Of Health And Human Services Cell matrix receptor system and use in cancer diagnosis and management
US4861710A (en) * 1986-09-26 1989-08-29 The United States Of America As Represented By The Department Of Health And Human Services Recombinant DNA clone encoding laminin receptor
US5871959A (en) * 1989-12-27 1999-02-16 The United States Of America As Represented By The Department Of Health And Human Services Method of producing hepatocycte growth factor/scatter factor and related cell lines
US5648273A (en) * 1989-12-27 1997-07-15 The United States Of America, As Represented By The Department Of Health And Human Services Hepatic growth factor receptor is the MET proto-oncogene
US5945098A (en) * 1990-02-01 1999-08-31 Baxter International Inc. Stable intravenously-administrable immune globulin preparation
US6051230A (en) * 1992-03-05 2000-04-18 Board Of Regents, The University Of Texas System Compositions for targeting the vasculature of solid tumors
US5855866A (en) * 1992-03-05 1999-01-05 Board Of Regenis, The University Of Texas System Methods for treating the vasculature of solid tumors
US6147203A (en) * 1993-06-14 2000-11-14 The United States Of America As Represented By The Department Of Health And Human Services Recombinant disulfide-stabilized polypeptide fragments having binding specificity
US5720720A (en) * 1993-08-27 1998-02-24 The United States Of America As Represented By The Department Of Health And Human Services Convection-enhanced drug delivery
US6187287B1 (en) * 1994-08-12 2001-02-13 Immunomedics, Inc. Immunoconjugates and humanized antibodies specific for B-cell lymphoma and leukemia cells
US5686292A (en) * 1995-06-02 1997-11-11 Genentech, Inc. Hepatocyte growth factor receptor antagonist antibodies and uses thereof
US6174708B1 (en) * 1995-12-04 2001-01-16 Pasteur Merieux Serums & Vaccins Preparation of a multicombinatorial library of antibody gene expression vectors
US6034218A (en) * 1996-03-15 2000-03-07 Corixa Corporation Compounds and methods for immunotherapy and immunodiagnosis of prostate cancer
US6063905A (en) * 1997-01-07 2000-05-16 Board Of Regents, The University Of Texas System Recombinant human IGA-J. chain dimer
US6750194B1 (en) * 2000-10-23 2004-06-15 The Procter & Gamble Company Methods of identifying compounds for regulating muscle mass or function using vasoactive intestinal peptide receptors

Cited By (26)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7745391B2 (en) 2001-09-14 2010-06-29 Compugen Ltd. Human thrombospondin polypeptide
US20040248157A1 (en) * 2001-09-14 2004-12-09 Michal Ayalon-Soffer Novel polynucleotides encoding soluble polypeptides and methods using same
US20070083334A1 (en) * 2001-09-14 2007-04-12 Compugen Ltd. Methods and systems for annotating biomolecular sequences
US20060165679A1 (en) * 2002-05-08 2006-07-27 Bayer Healthcare Ag Diagnostics and therapeutics for diseases associated with vasoactive intestinal peptide receptor 2 (vpac2)
US7678889B2 (en) * 2002-08-06 2010-03-16 Diadexus, Inc. Compositions and methods relating to ovarian specific genes and proteins
US20060199180A1 (en) * 2002-08-06 2006-09-07 Macina Roberto A Compositions and methods relating to ovarian specific genes and proteins
US20040265799A1 (en) * 2003-06-24 2004-12-30 Compugen Ltd. Human-virus homologous sequences and uses thereof
US20050123538A1 (en) * 2003-10-03 2005-06-09 Ronen Shemesh Polynucleotides encoding novel ErbB-2 polypeptides and kits and methods using same
US20050186600A1 (en) * 2004-01-13 2005-08-25 Osnat Sella-Tavor Polynucleotides encoding novel UbcH10 polypeptides and kits and methods using same
US20070082337A1 (en) * 2004-01-27 2007-04-12 Compugen Ltd. Methods of identifying putative gene products by interspecies sequence comparison and biomolecular sequences uncovered thereby
US20060068405A1 (en) * 2004-01-27 2006-03-30 Alex Diber Methods and systems for annotating biomolecular sequences
KR101290892B1 (en) 2007-07-27 2013-07-31 이매틱스 바이오테크놀로지스 게엠베하 Novel immunotherapy against neuronal and brain tumors
US9175040B2 (en) 2007-07-27 2015-11-03 Immatics Biotechnologies Gmbh Immunotherapeutic peptides against neuronal and brain tumors and related methods and compositions
US20090123489A1 (en) * 2007-07-27 2009-05-14 Immatics Biotechnologies Gmhh Novel immunotherapy against neuronal and brain tumors
US11648276B2 (en) 2007-07-27 2023-05-16 Immatics Biotechnologies Gmbh Immunotherapy against neuronal and brain tumors
WO2009015843A1 (en) * 2007-07-27 2009-02-05 Immatics Biotechnologies Gmbh Novel immunotherapy against neuronal and brain tumors
KR101317989B1 (en) 2007-07-27 2013-10-14 이매틱스 바이오테크놀로지스 게엠베하 Novel immunotherapy against neuronal and brain tumors
EP2660248A1 (en) * 2007-07-27 2013-11-06 Immatics Biotechnologies GmbH Novel immunotherapy against neuronal and brain tumors
US10709735B2 (en) 2007-07-27 2020-07-14 Immatics Biotechnologies Gmbh Immunotherapy against neuronal and brain tumors
EA029831B1 (en) * 2007-07-27 2018-05-31 Имматикс Байотекнолоджиз Гмбх Peptide inducing cytotoxic t lymphocytes and stimulating anti-tumor immune responses
US10420800B2 (en) 2007-07-27 2019-09-24 Immatics Biotechnologies Gmbh Immunotherapy against neuronal and brain tumors
US10434120B2 (en) 2007-07-27 2019-10-08 Immatics Biotechnologies Gmbh Immunotherapy against neuronal and brain tumors
US20110092452A1 (en) * 2008-03-05 2011-04-21 The Regents Of The University Of Michigan Compositions and methods for diagnosing and treating pancreatic cancer
EP2105501A1 (en) * 2008-03-27 2009-09-30 Immatics Biotechnologies GmbH Novel immunotherapy against neuronal and brain tumors
CN111748574A (en) * 2020-06-16 2020-10-09 闽江学院 Method for improving cadmium tolerance of plants by over-expressing Kandelia candel KoCSD3
WO2022147334A1 (en) * 2020-12-31 2022-07-07 Encodia, Inc. Metalloenzymes for biomolecular recognition of n-terminal modified peptides

Also Published As

Publication number Publication date
US7060275B2 (en) 2006-06-13
US20050074400A1 (en) 2005-04-07

Similar Documents

Publication Publication Date Title
US7060275B2 (en) Use of protein biomolecular targets in the treatment and visualization of brain tumors
US5624659A (en) Method of treating brain tumors expressing tenascin
ES2286831T3 (en) ANTIBODIES FOR THE FIBRONECTINE ED-B DOMAIN, THE CONSTRUCTION OF THESE AND ITS USES.
US6528481B1 (en) NG2/HM proteoglycan-binding peptides that home to angiogenic vasculature and related methods
Neri et al. Tumour vascular targeting
US5489525A (en) Monoclonal antibodies to prostate cells
ES2557769T3 (en) Antibodies against the T cell immunoglobulin domain antigen and mucin domain 1 (TIM-1) and uses thereof
US7332568B2 (en) Q3 SPARC deletion mutant and uses thereof
AU2010325969B2 (en) J591 minibodies and cys-diabodies for targeting human prostate specific membrane antigen
ES2534726T3 (en) ED-A fibrinogen antigen that is associated with lymphomas
US20030082188A1 (en) Treatment of prostate cancer by inhibitors of NCAM2
JP2005527488A (en) Monoclonal antibody imaging and treatment of tumors expressing Met and binding to hepatocyte growth factor
US6455026B1 (en) Use of protein tyrosine phosphatase zeta as a biomolecular target in the treatment and visualization of brain tumors
US8093010B2 (en) Angiogenesis inhibiting molecules, their selection, production and their use in the treatment of cancer
US6958140B2 (en) Methods of imaging and targeting vasculature
EA010653B1 (en) Targeting of tumor vasculature using radiolabelled antibody l19 against fibronectin ed-b
US20070178104A1 (en) Methods and means for treating solid tumors
US20050260132A1 (en) Monoclonal antibodies directed to receptor protein tyrosine phosphatase zeta
JP2004537282A (en) Use of biomolecular targets in the treatment and visualization of brain tumors
RU2753677C2 (en) Antibody polypeptides and their application
CA2341586A1 (en) Targeting of molecules to large vessel endothelium using epcr
WO2022246511A1 (en) Improved immunoglobulin ii
Ayriss et al. Molecular targets for antibody-mediated immunotherapy of malignant glioma
AU2002257085A1 (en) Use of biomolecular targets in the treatment and visualization of brain tumours
US20070179105A1 (en) Method and means for treating solid tumors

Legal Events

Date Code Title Description
AS Assignment

Owner name: AGY THERAPEUTICS, CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:MULLER, SABINE;MELCHER, THORSTEN;CHIN, DANIEL;REEL/FRAME:012533/0899;SIGNING DATES FROM 20011129 TO 20020104

STCB Information on status: application discontinuation

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

AS Assignment

Owner name: GENERAL ELECTRIC CAPITAL CORPORATION, CONNECTICUT

Free format text: SECURITY AGREEMENT;ASSIGNOR:AGY THERAPEUTICS, INC.;REEL/FRAME:017015/0108

Effective date: 20050920

AS Assignment

Owner name: AGY THERAPEUTICS, INC., CALIFORNIA

Free format text: RELEASE BY SECURED PARTY;ASSIGNOR:GENERAL ELECTRIC CAPITAL CORPORATION;REEL/FRAME:017366/0336

Effective date: 20060327