CA2397207C - Novel stra6 polypeptides - Google Patents

Novel stra6 polypeptides Download PDF

Info

Publication number
CA2397207C
CA2397207C CA2397207A CA2397207A CA2397207C CA 2397207 C CA2397207 C CA 2397207C CA 2397207 A CA2397207 A CA 2397207A CA 2397207 A CA2397207 A CA 2397207A CA 2397207 C CA2397207 C CA 2397207C
Authority
CA
Canada
Prior art keywords
polypeptide
sequence identity
amino acid
antibody
sequence
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.)
Expired - Fee Related
Application number
CA2397207A
Other languages
French (fr)
Other versions
CA2397207A1 (en
Inventor
Diane Pennica
Victoria Smith
William I. Wood
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.)
Genentech Inc
Original Assignee
Genentech 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
Application filed by Genentech Inc filed Critical Genentech Inc
Publication of CA2397207A1 publication Critical patent/CA2397207A1/en
Application granted granted Critical
Publication of CA2397207C publication Critical patent/CA2397207C/en
Anticipated expiration legal-status Critical
Expired - Fee Related legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/10Processes for the isolation, preparation or purification of DNA or RNA
    • C12N15/1034Isolating an individual clone by screening libraries
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/1703Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • A61K38/1709Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/569Single domain, e.g. dAb, sdAb, VHH, VNAR or nanobody®
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide

Abstract

The present invention is directed to novel polypeptides having sequence similarity to Stra6, a murine retinoic acid responsive protein, and to nucleic acid molecules encoding those polypeptides. Also provided herein are vectors and host cells comprising those nucleic acid sequences, chimeric polypeptide molecules comprising the polypeptides of the present invention fused to heterologous polypeptide sequences, antibodies which bind to the polypeptides of the present invention and to methods for producing the polypeptides of the present invention.

Description

FIELD OF THE INVENTION
The present invention relates generally to the identification and isolation of novel DNA and to the recombinant production of novel polypeptides having sequence similarity to murine Stra6, a retinoic acid responsive protein. Some of these molecules were earlier designated as "PRO10282", but will hereinafter also be referred to as "Stra6" polypeptides.
BACKGROUND OF THE INVENTION
Membrane-bound Proteins Membrane-bound proteins and receptors can play important roles in, among other things, the formation, differentiation and maintenance of multicellular organisms. The fate of many individual cells, e.g., proliferation, migration, differentiation, or interaction with other cells, is typically governed by information received from other cells and/or the immediate environment. This information is often transmitted by secreted polypeptides (for instance, mitogenic factors, survival factors, cytotoxic factors, differentiation factors, neuropeptides, and hormones) which are, in turn, received and interpreted by diverse cell receptors or membrane-bound proteins. Such membrane-bound proteins and cell receptors include, but are not limited to, cytokine receptors, receptor kinases, receptor phosphatases, receptors involved in cell-cell interactions, and cellular adhesin molecules like selectins and integrins. For instance, transduction of signals that regulate cell growth and differentiation is regulated in part by phospharylation of various cellular proteins. Protein tyrosine kinases, 2 0 enzymes that catalyze that process, can also act as growth factor receptors. Examples include fibroblast growth factor receptor and nerve growth factor receptor.
Membrane-bound proteins and receptor molecules have various industrial applications, including as pharmaceutical and diagnostic agents. Receptor immunoadhesins, for instance, can be employed as therapeutic agents to block receptor-ligand interactions. The membrane-bound proteins can also be employed for screening of potential peptide 2 5 or small molecule inhibitors of the relevant receptor/ligand interaction.
Efforts are being undertaken by both industry and academia to identify new, native receptor or membrane-bound proteins. Many efforts are focused on the screening of mammalian recombinant DNA libraries to identify the coding sequences for novel receptor or membrane-bound proteins.
The Stra6 polypeptides of the present invention share sequence homology (73%
identity and 81% similarity) 3 0 with the murine protein Stra6, whose expression is induced by retinoic acid (Bouillet et al., Dev. Biol. 170:420-433 [1995];
Bouillet et al., Mech. Day. 63: 173-186 [1997]; Chazaud et al., Day. Genet.
19: 66-73 [1996]). Since retinoic acid is an important signaling molecule during vertebrate development, the genes induced in response to retinoic acid are thought to play a crucial role in growth and differentiation during embryonic development. The murine Stra6 cDNA was isolated from P19 mine embyonal carcinoma cells using a subtractive hybridization approach designed to identify and isolate retinoic 3 5 acid inducible genes, and does not show similarity with previously characterized proteins. It contains highly hydrophobic stretches of amino acid residues that correspond to multiple trans-membrane domains, a characteristic of a membrane integral protein. Based on its expression pattern, Stra6 is thought to play an important role in early dorsoventral limb patterning during embyonic development and later in the control of endochondral ossification (Chazaud et al., Dev. Genet.
19: 66-73 [19961 The Stra6 polypeptides disclosed herein contain multiple highly hydrophobic regions that likely constitute trans-membrane domains indicating that the Stra6 polypeptides are membrane integral proteins. They may function as receptors for an unknown ligand and may be a part of signal transduction pathway with impact on cell growth, development or differentiation.
Gene Amplification in Tumor Cells Malignant tumors (cancers) are the second leading cause of death in the United States, after heart disease (Boring et al., CA Cancel J. Clin., 43:7 [1993]).
Cancer is characterized by an increase in the number of abnormal, or neoplastic cells derived from a normal tissue which proliferate to form a tumor mass, the invasion of adjacent tissues by these neoplastic tumor cells, and the generation of malignant cells which eventually spread via the blood or lymphatic system to regional lymph nodes and to distant sites (metastasis). In a cancerous state, a cell proliferates under conditions in which normal cells would not grow. Cancer manifests itself in a wide variety of forms, characterized by different degrees of invasiveness and aggressiveness.
Alteration of gene expression is intimately related to the uncontrolled cell growth and de-differentiation, which are a common feature of all cancers. The genomes of certain well studied tumors have been found to show decreased expression of recessive genes, usually referred to as tumor suppression genes, which would normally function to prevent malignant cell growth, and/or overexpression of certain dominant genes, such as oncogenes, that act to promote malignant growth. Each of these genetic changes appears to be responsible for importing some of the traits that, in aggregate, represent the full neoplastic phenotype (Hunter, Cell, 64:1129 [1991] and Bishop, Cell, 64:235-248 [19911).
A well-known mechanism of gene (e.g., oncogene) overexpression in cancer cells is gene amplification. This is a process where in the chromosome of the ancestral cell multiple copies of a particular gene are produced. The process involves unscheduled replication of the region of chromosome comprising the gene, followed by recombination of the replicated segments back into the chromosome (Alitalo et al., Adv. Cancer Res., 47:235-281 [1986]). It is believed that the overexpression of the gene parallels gene amplification, i.e., is proportionate to the number of copies made.
Proto-oncogenes that encode growth factors and growth factor receptors have been identified to play important roles in the pathogenesis of various human malignancies, including breast cancer. For example, it has been found that the human ErbB2 gene (erbB2, also known as her2, or c-erbB-2), which encodes a 185-kd transmembrane glycoprotein receptor (p18514'; HER2) related to the epidermal growth factor receptor EGFR), is overexpressed in about 25% to 30% of human breast cancer (Slamon et al., Science, 235:177-182 [1987]; Slamon et al., Science, 244:707-712 [19891).
It has been reported that gene amplification of a proto-oncogene is an event typically involved in the more malignant forms of cancer, and could act as a predictor of clinical outcome (Schwab et al., Genes Chromosomes Cancer, 1:181-193 [1990]; Alitalo et al., supra). Thus, erbB2 overexpression is commonly regarded as a predictor of a poor =
=

prognosis, especially in patients with primary disease that involves axillary lymph nodes (Stamen at al., (19871 and 09891, supra; Ravdin and Chamness, DArg, 159:19-27 (19951; and Hynes and Stern, Biochim. Bioehys. Acta, 1198:165-184 119941), and has been linked to sensitivity andlor resistance to hormone therapy and chemotherapeutic regimens, including CMF (cyclophosphamide, methotrexate, and fluoruracil) and anthracyclines (BaseIga at al., Oncology, 11(3 Suppl 9:43-48 119978. However, despite the association of erbB2 overexpression with poor prognosis, the odds of HER2-positive patients responding clinically to treatment with taxanes were greater than three times those of HER2-negative patients (Mid). A
recombinant humanized anti-ErbB2 (anti-HER2) monoclonal antibody (a humanized version of the murine anti-ErbB2 antibody 405, referred to as rhuMAh HER2 or HerceptinTM) has been clinically active in patients with ErbB2-overexpressing metastatic breast cancers that had received extensive prior anticancer therapy. (Basefga at of., J. Clin. Once'., 14:737-744 [1996]).
In light of the above, there is obvious interest in identifying novel molecules, methods and compositions which are useful for diagnosing and treating tumors which are associated with gene amplification.
SUMMARY OF THE INVENTION
A human cDNA clone (designated herein as DNA148380-2827) has been identified that has homology to nucleic acid encoding a mine retinoic acid responsive protein Stra6 and that encodes a 667 amino acid novel polypeptide, designated in the present application as full-length native-sequence human "PRO10282". An additional human cDNA clone (designated herein as 1JNA148389-2827-1) has been identified that encodes a 658 amino acid novel polypeptide (PRO19578), showing significant sequence homology to both murine Stra6 and native sequence human PRO10282, which is believed to be an alternatively spliced variant of native sequence human PRO10282. As discussed hereinafter, in view of their homology with murine Stra8, the PR010282 polypeptides of the present invention (including native sequence and variant molecules) will be also referred to as "Stra6 polypeptides.
In one embodiment, the invention provides an isolated nucleic acid molecule comprising a nucleotide sequence that encodes, a PRO10282 polypeptide.
In one aspect, the isolated nucleic acid molecule comprises a nucleotide sequence having at least about 80%
sequence identity, preferably at least about 81% sequence identity, more preferably at least about 82% sequence identity, yet more preferably at least about 83% sequence identity, yet more preferably at least about 84% sequence identity, yet more preferably at least about 85% sequence identity, yet more preferably at least about 86% sequence identity, yet more preferably at least about 87% sequence identity, yet more preferably at least about 88% sequence identity, yet more preferably at least about 89% sequence identity, yet more preferably at least about 90% sequence identity, yet more preferably at least about 91% sequence identity, yet more preferably at least about 92% sequence identity, yet more preferably at least about 93% sequence identity, yet more preferably at least about 94% sequence identity, yet more preferably at least about 95% sequence identity, yet more preferably at least about 96% sequence identity, yet more preferably at least about 97% sequence identity, yet more preferably at least about 98% sequence identity and yet more preferably at least about 99% sequence' identity to (a) a DNA molecule encoding a P11010282 polypeptide having the sequence of amino acid residues from about 1 to about 667, inclusive, of Figure 2 (SEG ID NO:2), or (b) a DNA molecule encoding a PRO19578 polypeptide having the sequence of amino acid residues from about 1 to about 658, inclusive, of Figure 7 (SEG ID NO: 5), or (c) the complement of the DNA molecule of (a) or (b).
In another aspect, the isolated nucleic acid molecule comprises (a) a nucleotide sequence encoding a PRO10282 polypeptide having the sequence of amino acid residues from about 1 to about 667, inclusive, of Figure 2 (SEG ID NO:2), or (b) a nucleotide sequence encoding a PRO19578 poll/peptide having the sequence of amino acid residues from about 1 to about 658, inclusive of Figure 7 (SEG ID NO: 5), or (c) the complement of the nucleotide sequence of (a) or (b).
In other aspects, the isolated nucleic acid molecule comprises a nucleotide sequence having at least about 80%
sequence identity, preferably at least about 81% sequence identity, more preferably at least about 82% sequence identity, 1 0 yet more preferably at least about 83% sequence identity, yet more preferably at least about 84% sequence identity, yet more preferably at least about 85% sequence identity, yet more preferably at least about 86% sequence identity, yet more preferably at least about 87% sequence identity, yet more preferably at least about 88% sequence identity, yet more preferably at least about 89% sequence identity, yet more preferably at least about 90% sequence identity, yet more preferably at least about 91% sequence identity, yet more preferably at least about 92% sequence identity, yet more preferably at least about 93% sequence identity, yet more preferably at least about 94% sequence identity, yet more preferably at least about 95% sequence identity, yet more preferably at least about 96% sequence identity, yet more preferably at least about 97% sequence identity, yet more preferably at least about 98% sequence identity and yet more preferably at least about 99% sequence identity to (a) a DNA molecule having the sequence of nucleotides from about 49 to about 2049, inclusive, of Figure 1 (SEG ID NO:1), or (b) a DNA molecule having the sequence of nucleotides from about 2 0 186 to about 2159, inclusive, of Figure 6 (SEG ID NO: 4), or (c) the complement of the DNA molecule of (a) or (b).
In another aspect, the isolated nucleic acid molecule comprises (a) the nucleotide sequence of from about 49 to about 2049, inclusive, of Figure 1 (SE0.1D NO:1), or (b) the nucleotide sequence of from about 186 to about 2159 of Figure 6 (SEG ID NO: 4), or (c) the complement of the nucleotide sequence of (a) or (b).
In a further aspect, the invention concerns an isolated nucleic acid molecule comprising a nucleotide sequence having at least about 80% sequence identity, preferably at least about 81%
sequence identity, more preferably at least about 82% sequence identity, yet more preferably at least about 83% sequence identity, yet more preferably at least about 84% sequence identity, yet more preferably at least about 85% sequence identity, yet more preferably at least about 86%
sequence identity, yet more preferably at least about 87% sequence identity, yet more preferably at least about 88%
sequence identity, yet more preferably at least about 89% sequence identity, yet more preferably at least about 90%
sequence identity, yet more preferably at least about 91% sequence identity, yet more preferably at least about 92%
sequence identity, yet more preferably at least about 93% sequence identity, yet more preferably at least about 94%
sequence identity, yet more preferably at least about 95% sequence identity, yet more preferably at least about 96%
sequence identity, yet more preferably at least about 97% sequence identity, yet more preferably at least about 98%
sequence identity and yet more preferably at least about 99% sequence identity to (a) a DNA molecule that encodes the same mature potypeptide encoded by the human protein cDNA deposited with the ATCC on January 11, 2000 under ATCC
Deposit No. PTA-1181 (0NA148380-2827), (b) a DNA molecule that encodes the same mature polypeptide encoded by the human protein cDNA deposited with the ATCC on February 23,2000 under ATCC
Deposit No. PTA-1405 (DNA148389-2827-1 ), or (c) the complement of the DNA molecule of (a) or (b). In a preferred embodiment, the isolated nucleic acid molecule comprises (a) a nucleotide sequence encircling the same mature polypeptide encoded by the human protein cONA
deposited with the ATCC on January 11, 2000 under ATCC Deposit No. PTA-1181 (0NA148380-2827), (hi a nucleotide sequence encoding the same mature polypeptide encoded by the lemon protein cDNA deposited with the ATCC on February 23,2000 under ATCC Deposit No. PTA-1405 (0NA148389-2827-1), or (c) the complement of the nucleotide sequence of (a) or (hi.
Inanother aspect the invention concerns an isolated nucleic acid molecule comprising a nucleotide sequence having at least about 80% sequence identity, preferably at least about 81%
sequence identity, more preferably at least about 82% sequence identity, yet more preferably at least about 83% sequence identity, yet more preferably at least about 14% sequence identity, yet more preferably at least about 85% sequence identity, yet more preferably at least about 86% -sequence identity, yet more preferably at least about 87% sequence identity, yet more preferably at least about 88%
sequence identity, yet more preferably at least about 89% sequence identity, yet more preferably at least about 90%
sequence identity, yet more preferably at least about 91% sequence identity, yet more preferably at least about 92%
sequence identity, yet more .preferably at least about 93% sequence identity, yet more preferably at least about 94%
sequence identity, yet more preferably at least about 95% sequence identity, yet more preferably at least about 96%
sequence identity, yet more preferably at least about 97% sequence identity, yet more preferably at least about 98%
sequence identity and yet more prefeiably at least about 99% sequence identity to (a) the full-length polypeptide coding sequence of the human protein cDNA deposited with the ATCC on January 11, 2000 under ATCC Deposit No. PTA-1181 (DNA148380-2827), 04 the full-length polypeptide coding sequence of the human protein cDNA deposited with the ATCC
on February 23,2000 under ATCC No. PTA-1405 (0NA148389-2827-1), or (c) the complement of the nucleotide sequence of (a) or (b).
In a preferred emboironent, the isolated nucleic acid molecule comprises (a) the full-length polypeptide coding sequence of the DNA deposited with the ATCC on January 11,2000 under ATCC
Deposit No. PT-1181 (0NA148380-2827), or (b) the fun-length polypeptide coding sequence of the DNA deposited with the ATCC on February 23,2000 under ATCC Deposit No. PTA-1405 (DNA148389-2827-1), or (c) the complement of the nucleotide sequence of (e) or OIL
In another aspect the invention concerns an isolated nucleic acid molecule which encodes an active PR010282 polypeptide as defined below comprising a nucleotide sequence that hybridizes to the complement of a nucleic acid sequence that encodes amino acids 1 to about 667, inclusive, of Figure 2 (sEa ID NO:2), or to the cornplement of a nudeic acid sequence that encodes amino adds 1 to about 658, inclusive, of Figure?
(SEQ ID NO: 5), wherein the isolated nucleic acid molecule is other than DNA encoding inurine Stra6. Preferably, hybridization occurs under high stringency (stringent) hybridization and wash conditions.
In yet another aspect the invention concerns an isolated nucleic acid molecule which encodes an active PRO10282 polypeptide as defined below comprising a nucleotide sequence that hybricraes to the complement of the nucleic acid sequence between about nucleotides 49 and about 2049, inclusive, of Figure 1 (SEG ID NO: 1), or to the complement of the nucleic acid sequenc ebetween about nucleotides 186 to about 2159, inclusive, or Figure 6 (SEG ID NO: 4), wherein the isolated nucleic acid is other than DNA encoding murine Stra 6.
Preferably, hybridization occurs under high stringency (stringent) hybridization and wash conditions.
In a further aspect, the invention concerns an isolated nucleic acid molecule having at least about 765 nucleotides and which is produced by hybridizing a test DNA molecule under stringent conditions with (a) a DNA molecule encoding a PR010282 polypeptide having the sequence of amino acid residues from about 1 to about 667, inclusive, of Figure 2 (SEG ID N0:2), or (b) a DNA molecule encoding a PR019578 polypeptide having the sequence of amino acid residues from about 1 to about 658, inclusive, of Figure 7 (SEG ID NO: 5), or (c) the complement of the DNA molecule of (a) or (b), and, if the test DNA molecule has at least about an 80% sequence identity, preferably at least about an 81%
sequence identity, more preferably at least about an 82% sequence identity, yet more preferably at least about an 83%
sequence identity, yet more preferably at least about an 84% sequence identity, yet more preferably at least about an 85%
sequence identity, yet more preferably at least about an 86% sequence identity, yet more preferably at least about an 87%
sequence identity, yet more preferably at least about an 88% sequence identity, yet more preferably at least about an 89%
sequence identity, yet more preferably at least about a 90% sequence identity, yet more preferably at least about a 91%
sequence identity, yet more preferably at least about a 92% sequence identity, yet more preferably at least about a 93%
sequence identity, yet more preferably at least about a 94% sequence identity, yet more preferably at least about a 95%
sequence identity, yet more preferably at least about a 96% sequence identity, yet more preferably at least about a 97%
sequence identity, yet more preferably at least about a 98% sequence identity and yet more preferably at least about a 2 0 99% sequence identity to (a), (b) or (c), isolating the test DNA
molecule.
In another aspect, the invention concerns an isolated nucleic acid molecule comprising a nucleotide sequence encoding a polypeptide scoring at least about 80% positives, preferably at least about 81% positives, more preferably at least about 82% positives, yet more preferably at least about 83% positives, yet more preferably at least about 84%
positives, yet more preferably at least about 85% positives, yet more preferably at least about 86% positives, yet more 2 5 preferably at least about 87% positives, yet more preferably at least about 88% positives, yet more preferably at least about 89% positives, yet more preferably at least about 90% positives, yet more preferably at least about 91% positives, yet more preferably at least about 92% positives, yet more preferably at least about 93% positives, yet more preferably at least about 94% positives, yet more preferably at least about 95%
positives, yet more preferably at least about 96%
positives, yet more preferably at least about 97% positives, yet more preferably at least about 98% positives and yet more 3 0 preferably at least about 99% positives when compared with (a) the amino acid sequence of residues about 1 to 667, inclusive, of Figure 2 (SEG ID N0:2), or (b) the amino acid sequence of residues about 1 to about 658 of Figure 7 (SEG
ID NO: 5), or (c) the complement of the nucleotide sequence of (a) or (b).
Another aspect the invention provides an isolated nucleic acid molecule comprising a nucleotide sequence encoding a PRO10282 polypeptide which is either transmembrane domain-deleted or transmembrane domain-inactivated, 3 5 or is complementary to such encoding nucleotide sequence, wherein the transmembrane domains have been tentatively identified as extending from about amino acid position 54 to about amino acid position 69, about amino acid position 102 to about amino acid position 119, about amino acid position 148 to about amino acid position 166, about amino acid position 207 to about amino acid position 222, about amino acid position 301 to about amino acid position 320, about amino acid position 364 to about amino acid position 380, about amino acid position 431 to about amino acid position 451, about amino acid position 474 to about amino acid position 489 and about amino acid position 512 to about amino acid position 531 in the sequence of Figure 2 (SEG ID N0:2); and about amino acid position 54 to about amino acid position 71, about amino acid position 93 to about amino acid position 11, about amino acid position 140 to about amino acid position 157, about amino acid position 197 to about amino acid position 214, about amino acid position 291 to about amino acid position 312, about amino acid position 356 to about amino acid position 371, about amino acid position 425 to about amino acid position 481, about amino acid position 505 to about amino acid position 522 of Figure 7 (SERG ID NO: 5).
Therefore, soluble extracellular domains of the herein described PRO10282 polypeptides are contemplated.
In this regard, another aspect of the present invention is directed to an isolated nucleic acid molecule which comprises a nucleotide sequence having at least about 80% sequence identity, preferably at least about 81% sequence identity, more preferably at least about 82% sequence identity, yet more preferably at least about 83% sequence identity, yet more preferably at least about 84% sequence identity, yet more preferably at least about 85% sequence identity, yet more preferably at least about 86% sequence identity, yet more preferably at least about 87% sequence identity, yet more preferably at least about 88% sequence identity, yet more preferably at least about 89% sequence identity, yet more preferably at least about 90% sequence identity, yet more preferably at least about 91% sequence identity, yet more preferably at least about 92% sequence identity, yet more preferably at least about 93% sequence identity, yet more preferably at least about 94% sequence identity, yet more preferably at least about 95% sequence identity, yet more preferably at least about 96% sequence identity, yet more preferably at least about 97% sequence identity, yet more preferably at least about 98% sequence identity and yet more preferably at least about 99% sequence identity to (a) a DNA
molecule encoding amino acids 1 to X of Figure 2 (SEG ID N0:2), where X is any amino acid from 49 to 59 of Figure 2 (SEG
ID NO:2), or (b) the complement of the DNA molecule of (a). In a specific aspect, the isolated nucleic acid molecule comprises a nucleotide sequence which (a) encodes amino acids 1 to X of Figure 2 (SEG ID N0:2), or of Figure 7 (SEG ID
NO: 5), where X is any amino acid from 49 to 59 of Figure 2 (SEG ID NO:2), or (b) is the complement of the DNA molecule of (a). In a specific aspect, the isolated nucleic acid molecule comprises a nucleotide sequence which (a) encodes amino acids 1 to X of Figure 2 (SEG ID NO: 2), or of Figure 7 (SEG ID NO: 5), where X is any amino acid from 49 to 59, inclusive, of Figure 2 (SEG ID NO: 2), or Figure 7 (SEG ID NO: 5), or (b) is the complement of the DNA molecule of (a).
3 0 In yet another aspect of the present invention, the isolated nucleic acid molecule (a) encodes a polypeptide scoring at least about 80% positives, preferably at least about 81% positives, more preferably at least about 82%
positives, yet more preferably at least about 83% positives, yet more preferably at least about 84% positives, yet more preferably at least about 85% positives, yet more preferably at least about 86% positives, yet more preferably at least about 87% positives, yet more preferably at least about 88% positives, yet more preferably at least about 89% positives, yet more preferably at least about 90% positives, yet more preferably at least about 91% positives, yet more preferably at least about 92% positives, yet more preferably at least about 93%
positives, yet more preferably at least about 94%
positives, yet more preferably at least about 95% positives, yet more preferably at least about 96% positives, yet more preferably at least about 97% positives, yet more preferably at least about 98% positives and yet more preferably at least about 99% positives when compared with the amino acid sequence of residues about 1 to X of Figure 2 (SEG ID NO:2), or of Figure 7 (SEG ID NO: 5), where X is any amino acid from 49 to 59 of Figure 2 (SEG ID N0:2), or (b) is the complement of the DNA molecule of (a).
Another embodiment is directed to fragments of a PR010282 polypeptide coding sequence that may find use as, for example, hybridization probes or for encoding fragments of a PRO10282 polypeptide that may optionally encode a polypeptide comprising a binding site for an anti-PRO10282 antibody. Such nucleic acid fragments are usually at least 1 0 about 20 nucleotides in length, preferably at least about 30 nucleotides in length, more preferably at least about 40 nucleotides in length, yet more preferably at least about 50 nucleotides in length, yet more preferably at least about 60 nucleotides in length, yet more preferably at least about 70 nucleotides in length, yet more preferably at least about 80 nucleotides in length, yet more preferably at least about 90 nucleotides in length, yet more preferably at least about 100 nucleotides in length, yet more preferably at least about 110 nucleotides in length, yet more preferably at least about 120 nucleotides in length, yet more preferably at least about 130 nucleotides in length, yet more preferably at least about 140 nucleotides in length, yet more preferably at least about 150 nucleotides in length, yet more preferably at least about 160 nucleotides in length, yet more preferably at least about 170 nucleotides in length, yet more preferably at least about 180 nucleotides in length, yet more preferably at least about 190 nucleotides in length, yet more preferably at least about 200 nucleotides in length, yet more preferably at least about 250 nucleotides in length, yet more preferably at least about 300 2 0 nucleotides in length, yet more preferably at least about 350 nucleotides in length, yet more preferably at least about 400 nucleotides in length, yet more preferably at least about 450 nucleotides in length, yet more preferably at least about 500 nucleotides in length, yet more preferably at least about 600 nucleotides in length, yet more preferably at least about 700 nucleotides in length, yet more preferably at least about 800 nucleotides in length, yet more preferably at least about 900 nucleotides in length and yet more preferably at least about 1000 nucleotides in length, wherein in this context the term 2 5 "about" means the referenced nucleotide sequence length plus or minus 10% of that referenced length. The nucleic acid fragments of the present invention are different from fragments of the native coding sequence of mousee Stra6. In a preferred embodiment, the nucleotide sequence fragment is derived from any coding region of the nucleotide sequence shown in Figure 1 (SEG ID N0:1), or of the nucleotide sequence shown in Figure 6 (SEG ID NO: 4). It is noted that novel fragments of a PRO10282 polypeptide-encoding nucleotide sequence may be determined in a routine manner by aligning 30 the PRO10282 polypeptide-encoding nucleotide sequence with other known nucleotide sequences using any of a number of well known sequence alignment programs and determining which PR010282 polypeptide-encoding nucleotide sequence fragment(s) are novel. All of such PRO10282 polypeptide-encoding nucleotide sequences are contemplated herein and can be determined without undue experimentation. Also contemplated are the PRO10282 polypeptide fragments encoded by these nucleotide molecule fragments, preferably those PRO10282 polypeptide fragments that comprise a binding site for 35 an anti-PR010282 antibody.
In another embodiment, the invention provides a vector comprising a nucleotide sequence encoding PRO10282 or its variants. The vector may comprise any of the isolated nucleic acid molecules hereinabove identified.
A host cell comprising such a vector is also provided. By way of example, the host cells may be CHO cells, E.
coli, or yeast. A process for producing PRO10282 polypeptides is further provided and comprises culturing host cells under conditions suitable for expression of PR010282 and recovering PR010282 from the cell culture.
In another embodiment, the invention provides isolated PRO10282 polypeptide encoded by any of the isolated nucleic acid sequences hereinabove identified.
In a specific aspect, the invention provides isolated native sequence PR010282 polypeptide, which in certain embodiments, includes an amino acid sequence comprising residues from about 1 to about 667 of Figure 2 (SEO ID NO:2), or residues from about 1 to about 658 of Figure 7 (SEG ID NO: 5).
In another aspect, the invention concerns an isolated PRO10282 polypeptide, comprising an amino acid sequence having at least about 80% sequence identity, preferably at least about 81%
sequence identity, more preferably at least about 82% sequence identity, yet more preferably at least about 83% sequence identity, yet more preferably at least about 84% sequence identity, yet more preferably at least about 85% sequence identity, yet more preferably at least about 86%
sequence identity, yet more preferably at least about 87% sequence identity, yet more preferably at least about 88%
sequence identity, yet more preferably at least about 89% sequence identity, yet more preferably at least about 90%
sequence identity, yet more preferably at least about 91% sequence identity, yet more preferably at least about 92%
sequence identity, yet more preferably at least about 93% sequence identity, yet more preferably at least about 94%
sequence identity, yet more preferably at least about 95% sequence identity, yet more preferably at least about 96%
2 0 sequence identity, yet more preferably at least about 97% sequence identity, yet more preferably at least about 98%
sequence identity and yet more preferably at least about 99% sequence identity to the sequence of amino acid residues from about 1 to about 667, inclusive, of Figure 2 (SEG ID NO:2), or to the sequence of amino acid residues from about 1 to about 658, inclusive, of Figure 7 (SEG ID NO: 5).
In a further aspect, the invention concerns an isolated PRO10282 polypeptide comprising an amino acid sequence having at least about 80% sequence identity, preferably at least about 81%
sequence identity, more preferably at least about 82% sequence identity, yet more preferably at least about 83% sequence identity, yet more preferably at least about 84% sequence identity, yet more preferably at least about 85% sequence identity, yet more preferably at least about 86%
sequence identity, yet more preferably at least about 87% sequence identity, yet more preferably at least about 88%
sequence identity, yet more preferably at least about 89% sequence identity, yet more preferably at least about 90%
sequence identity, yet more preferably at least about 91% sequence identity, yet more preferably at least about 92%
sequence identity, yet more preferably at least about 93% sequence identity, yet more preferably at least about 94%
sequence identity, yet more preferably at least about 95% sequence identity, yet more preferably at least about 96%
sequence identity, yet more preferably at least about 97% sequence identity, yet more preferably at least about 98%
sequence identity and yet more preferably at least about 99% sequence identity to an amino acid sequence encoded by __________________________________________________________________________ the human protein cDNA deposited with the ATCC on January 11, 2000 under ATCC
Deposit No. PTA-1181 (0NA148380-2827). or to an wino acid sequence encoded by the huinan protein cDNA deposited with the ATCC on February 23, 2000 under ATCC Deposit No. PTA-1405 (0NA14389-2827-1). In a preferred embodiment, the isolated PRO10282 polypeptide comprises an amino acid sequence encoded by the human protein cONA deposited with the ATCC
on January 11,2000 under ATCC Deposit No. PTA-1181 (DNA148380-2827), or by the human protein cDNA deposited with the ATCC on February 23,2000 under ATCC Deposit No. PTA-1405 (DNA 148389-2827-1).
In a further aspect, the invention concerns an isolated P11010282 polypeptide comprising an amino acid sequence scoring at least about 80% positives, preferably at least about 81%
positives, more preferably at least about 82% positives, yet more preferably at least about 83% positives, yet more preferably at least about 84% positives, yet more preferably at least about 85% positives, yet more preferably at least about 86% positives, yet more preferably at least about 87% positives, yet more preferably at least about 88% positives, yet more preferably at least about 89%
positives, yet more preferably at least about 90% positives, yet more preferably at least about 91% positives, yet more preferably at least about 92% positives, yet more preferably at least about 83% positives, yet more preferably at least about 94% positives, yet more preferably at least about 95% positives, yet more preferably at least about 96% positives, yet more preferably at least about 97% positives, yet more preferably at least about 98% positives and yet more preferably at least about 99% positives when compared with the wino acid sequence of residues from about 1 to about 667, inclusive, of Figure 2 (SED ID NO:2), or with the wino acid sequence of residues from about Ito about 659, includive, of Figure 7 (SEQ ID NO: 51.
Another aspect the invention provides an isolated PRO10282 polypeptide which is either transmembrane domain-deleted or transmernbrane domain-inactivated. Processes for producing the same are also herein described, wherein those processes coinprise culturing a host cell comprising a vector which comprises the appropriate encoding nucleic acid molecule under conditions suitable for expression of the P8010282 polypeptide and recovering the P11010282 polypeptide from the cell culture.
As such, one aspect of the present invention is directed to an isolated soluble P8010282 polypeptide which comprises an amino acid sequence having at least about 80% sequence identity, preferably at least about 81% sequence identity, more preferably at least about 82% sequence identity, yet more preferably at least about 83% sequence identity, yet more preferably at least about 84% sequence identity, yet more preferably at least about 85% sequence identity, yet more preferably at least about 86% sequence identity, yet more preferably at least about 87% sequence identity, yet more preferably at least about 88% sequence identity, yet more preferably at least about 89% sequence identity, yet more preferably at least about 90% sequence identity, yet more preferably at least about 91% sequence identity, yet more preferably at least about 92% sequence identity, yet more preferably at least about 93% sequence identity, yet more preferably at least about 94% sequence identity, yet more preferably at least about 95% sequence identity, yet more preferably at least about 96% sequence identity, yet more preferably at least about 97% sequence identity, yet mom preferably at least about 98% sequence identity and yet more preferably at least about 99% sequence identity to amino acids 1 to X of Figure 2 (SEQ ID NO:2), re of Figure 7 (SE11 ID NO: 5), where X is any anino acid from 49 to 59 of Figure 2 (Sat ID NO:2), or. of Figure 7 (sal ID NO: 6). In a preferred aspect, the isolated soluble PR010282 polypeptide comprises amino acids 1 to X of Figure 2 (SEG ID NO:2), or of Figure 7 (SEG ID
NO: 5), where X is any amino acid from 49 to 59 of Figure 2 (SEG ID NO:2) or of Figure 7 (SEG ID NO: 5).
In yet another aspect of the present invention, the isolated soluble PRO10282 polypeptide comprises an amino acid sequence which scores at least about 80% positives, preferably at least about 81% positives, more preferably at least about 82% positives, yet more preferably at least about 83% positives, yet more preferably at least about 84% positives, yet more preferably at least about 85% positives, yet more preferably at least about 86% positives, yet more preferably at least about 87% positives, yet more preferably at least about 88%
positives, yet more preferably at least about 89%
positives, yet more preferably at least about 90% positives, yet more preferably at least about 91% positives, yet more preferably at least about 92% positives, yet more preferably at least about 93% positives, yet more preferably at least about 94% positives, yet more preferably at least about 95% positives, yet more preferably at least about 96% positives, yet more preferably at least about 97% positives, yet more preferably at least about 98% positives and yet more preferably at least about 99% positives when compared with the amino acid sequence of residues about 1 to X of Figure 2 (SEG ID
NO:2), or of Figure 7 (SEG ID NO: 5), where X is any amino acid from 49 to 59 of Figure 2 (SEG ID NO:2) or of Figure 7 (SEG ID NO: 5).
In yet another aspect, the invention concerns an isolated PRO10282 polypeptide, comprising the sequence of amino acid residues from about 1 to about 667, inclusive, of Figure 2 (SEG ID
NO:2), or of amino acid residues from about 1 to about 658, inclusive, of Figure 7 (SEG ID NO: 5), or a fragment thereof which is biologically active or sufficient to provide a binding site for an anti-PRO10282 antibody, wherein the identification of PRO10282 polypeptide fragments that possess biological activity or provide a binding site for an anti-PR010282 antibody may be accomplished in a routine manner using techniques which are well known in the art. The fragment herein is other than a fragment of a mouse Stra6 polypeptide. Preferably, the PR010282 fragment retains a qualitative biological activity of a native PR010282 polypeptide.
In a still further aspect, the invention provides a polypeptide produced by (i) hybridizing a test DNA molecule under stringent conditions with (a) a DNA molecule encoding a PRO10282 polypeptide having the sequence of amino acid residues from about 1 to about 667, inclusive, of Figure 2 (SEG ID NO:2), or (b) a DNA molecule encoding a PR019578 polypeptide having the sequence of amino acid residues from about 1 to about 658, inclusive, of Figure 7 (SEU ID NO: 5),or (c) the complement of the DNA molecule of (a) or (b), and if the test DNA
molecule has at least about an 80% sequence identity, preferably at least about an 81% sequence identity, more preferably at least about an 82% sequence identity, yet more preferably at least about an 83% sequence identity, yet more preferably at least about an 84% sequence identity, 3 0 yet more preferably at least about an 85% sequence identity, yet more preferably at least about an 86% sequence identity, yet more preferably at least about an 87% sequence identity, yet more preferably at least about an 88% sequence identity, yet more preferably at least about an 89% sequence identity, yet more preferably at least about a 90% sequence identity, yet more preferably at least about a 91% sequence identity, yet more preferably at least about a 92% sequence identity, yet more preferably at least about a 93% sequence identity, yet more preferably at least about a 94% sequence identity, yet more preferably at least about a 95% sequence identity, yet more preferably at least about a 96% sequence identity, yet more preferably at least about a 97% sequence identity, yet more preferably at least about a 98% sequence identity and yet more preferably at least about a 99% sequence identity to (a), (b) or (c), (ii) culturing a host cell comprising the test DNA molecule under conditions suitable for expression of the polypeptide, and (iii) recovering the polypeptide from the cell culture.
In another embodiment, the invention provides chimeric molecules comprising a PR010282 polypeptide fused to a heterologous polypeptide or amino acid sequence, wherein the PRO10282 polypeptide may comprise any PRO10282 polypeptide, variant or fragment thereof as hereinbef ore described. An example of such a chimeric molecule comprises a PR010282 polypeptide fused to an epitope tag sequence or a Fc region of an innmunoglobulin.
In another embodiment, the invention provides an antibody as defined below which specifically binds to a 1 0 PRO10282 polypeptide as hereinbef ore described. Optionally, the antibody is a monoclonal antibody, an antibody fragment or a single chain antibody.
In yet another embodiment, the invention concerns agonists and antagonists of a native PRO10282 polypeptide as defined below. In a particular embodiment, the agonist or antagonist is an anti-PR010282 antibody or a small molecule.
In a further embodiment, the invention concerns a method of identifying agonists or antagonists to a PRO10282 polypeptide which comprise contacting the PRO10282 polypeptide with a candidate molecule and monitoring a biological activity mediated by said PRO10282 polypeptide. Preferably, the PI1010282 polypeptide is a native P11010282 polypeptide.
In a still further embodiment, the invention concerns a composition of matter comprising a PR010282 polypeptide, or an agonist or antagonist of a P11010282 polypeptide as herein described, or an anti-PR010282 antibody, in combination with a carrier. Optionally, the carrier is a pharmaceutically acceptable carrier.
Another embodiment of the present invention is directed to the use of a PRO10282 polypeptide, or an agonist or antagonist thereof as herein described, or an anti-PRO10282 antibody, for the preparation of a medicament useful in the treatment of a condition which is responsive to the PRO10282 polypeptide, an agonist or antagonist thereof or an anti-PRO10282 antibody.
In a further aspect, the invention concerns an antibody which specifically binds to a PR010282 polypeptide. The antibody prefereably induces the death of a cell that expresses the PRO10282 polypeptide. In a particularly preferred embodiment, the cell is a cancer cell that overexpresses a PRO10282 polypeptide as compared to a normal cell of the same tissue type. The antibody preferably is humanized or human, and includes antibody fragments and single-chain antibodies.
In another aspect, the invention concerns a composition of matter comprising an antibody specifically binding to a PR010282 polypeptide.
In yet another aspect, the invention concerns an isolated nucleic acid molecule encoding an antibody specifically binding to a PRO10282 polypeptide, a vector comprising such nucleic acid, and a host cell comprising such vector. The invention also concerns a method for producing anti-PRO10282 antibodies.
In a still further embodiment, the invention concerns a method for determining the presence of a PRO10282 polypeptide in a sample suspected of containing such polypeptide by exposing the sample to an anti-PRO10282 antibody (or another antagonist of PR010282), and determining the binding of such antibody (or antagonist) to the PR010282 polypeptide in the sample. In a particularly important embodiment, the sample is from a cancer cell.
In a different embodiment, the invention concerns a method of diagnosing tumor in a mammal, comprising detecting the level of expression of a gene encoding a PRO10282 polypeptide (a) in a test sample of tissue obtained from the mammal, and (b) in a control sample of known normal tissue cells of the same cell type. Higher expression level in the test sample, as compared to the control sample, indicates the presence of tumor int he mammal from whcih the test tissue cells were taken.
According to another method of the invention, tumor is diagnosed in a mammal by (a) contacting an anti-PRO10282 antibody with a test sample of tissue cells obtained from the mammal, and (b) detecting the formation of a complex between the antibody and a PRO10282 polypeptide in the test sample.
Formation of a complex is indicative of the presence of a tumor in the mammal.
In another aspect, te invention concerns a cancer diagnostic kit comprising an anti-PR010282 antibody (or a different antagonist of PRO10282) and a carrier in suitable packaging.
Optionally, the kit also contains instructions for using the antibody or other antagonist to detect the presence of a PR010282 polypeptide in a sample.
In yet another aspect, the invention concerns a method of inhibiting the growth of tumor cells by exposing tumor cells that express a PRO10282 polypeptide to an effective amount of an agent that inhibits a biological activity of the PRO10282 polypeptide, thereby inhibiting the growth of tumor cells. The agent may, for example, be an anti-PRO10282 antibody or another antagonist of PR010282. The tumor cells may be further exposed to conventional tumor treatments, such as radiation treatment, treatment with a cytotoxic agent and/or chemotherapeutic agent, etc.
In a different aspect, the invention concerns a method of inhibitingt he growth of tumor cells by exposing tumor cells that express a PRO10282 polypeptide to an effective amount of an agent that inhibits the expression of the polypeptide, thereby inhibiting tumor growth.
The invention further concerns analogous methods to prevent or slow down (lessen) the development, growth, or spread of tumor characterized by the overexpression of a PR010282 polypeptide. Such treatment may directly decrease the pathology of tumor cells, or render the tumor cells more susceptible to treatment by other therapeutic agents, e.g., radiation and/or chemotherapy. The invention specifically includes methods that control abnormal or otherwise metastasis, interference with the normal functioning of neighboring cells, release of cytokines or other secretory products at abnormal levels, suppression or aggravation of inflammatory or immunological response, etc.
The invention further concerns articles of manufacture comprising a container, a label on a container, and a composition comprising an active agent, where the composition is effective for inhibiting tumor growth and the label on the container indicates that the composition is effective for treating a condition characterized by overexpression of a PRO10282 polypeptide in tumor cells.
In a still further aspect, the invention concerns a method of identifying a compound that inhibits the biological or immunological activity of a PRO10282 polypeptide, by contacting a candidate compound with a PRO10282 polypeptide under conditions and for a time sufficient to allow the two components to interact, and determining whether a biological or immunological activity of the PR010282 polypeptide is inhibited.
In yet another aspect, the invention concerns a method of identifying a compound that inhibits an activity of a PRO10282 polypeptide, comprising the following steps: (a) contacting cells and a candidate compound to be screened in the presence of a PR010282 polypeptide under conditions suitable for the induction of a cellular response normally induced by a PR010282 polypeptide and (b) determining the induction of said cellular response to determine if the test compound is an effective antagonist. The lack of induction of a cellular response indicates that the test compound is an effective antagonist.
The invention further concerns a method for identifying a compound that inhibits the expression of a PR010282 polypeptide in cells expressing it, by contacting the cells with a candidate compound and determining whether the expression is inhibited.
The invention specifically concerns PR010282 agonists and antagonists identified by the screening methods herein. Such agonists and antagonists include, without limitation, anti-PR010282 antibodies, polypeptides, peptides, and small organic molecules with agonist or antagonist properties.
Accordingly, specific aspects of the invention include:
- an isolated nucleic acid molecule which comprises (a) a DNA
molecule encoding Stra6 polypeptide having the sequence of amino acid residues from about 1 to about 658 of SEQ ID NO:5, or (b) the complement of the DNA
molecule of (a);
- an isolated nucleic acid molecule encoding a Stra6 polypeptide comprising DNA that hybridizes under stringent hybridization conditions to the complement of the nucleic acid sequence that encodes amino acids 1 to about 658 of SEQ ID NO:5, wherein the hybridization is conducted under the following conditions:
(a) 50% (v/v) formamide with 0.1% bovine serum albumin/0.1% Fico11/0.1%

polyvinylpyrrolidone/50 mM sodium phosphate buffer at pH 6.5 with 750 mM
sodium chloride, 75 nriM sodium citrate at 42 C; (b) 50% formamide, 5 x SSC (0.75 M
NaCI, 0.075 M sodium citrate), 0.1% sodium dodecyl phosphate (SDS), 0.1% sodium pyrophosphate, 50 mM sodium phosphate, pH 6.8, 2 x Denhardt's solution, and 10%
dextran sulfate at 42 C; or (c) 50% formamide, 5 x SSC, 50 mM sodium phosphate (pH 6.8), 0.1% sodium pyrophosphate, 5 x Denhardt's solution, sonicated salmon sperm DNA (50 g/m1), 0.1% SDS, and 10% dextran sulfate at 42 C; and washing is conducted under the following conditions: (d) 0.1 x SSC and 0.1% SDS at 42 C;
(e) 0.015 M sodium chloride/0.0015 M sodium citrate/0.1% SDS at 50 C;
(f) 0.2 x SSC at 42 C; (g) 50% formamide at 55 C; or (h) 0.1 x SSC containing EDTA
at 55 C; and - an isolated Stra6 polypeptide comprising amino acid residues1 to 658 of SEQ ID NO:5.
14a BRIEF DESCRIPTION OF Dig DRAWINGS
Figure 1 shows the nucleotide sequence (SEQ ID NO:1) of a cDNA containing a nucleotide sequence (nucleotides 1-2732) encoding native sequence PR010282, wherein the nucleotide sequence (SDI ID NO:1) is a clone designated herein as "DNA148380-2827." Also presented in bold font and underlined are the positions of the respective start and stop cudons.
Figure 2 shows the amino acid sequence (sEa ID NO:2) of a native sequence P11010282 polypeptide as derived from the coding sequence of SEG ID N0:1. Also shown are the approximate locations of various other important polypeptide domains.
Figures 3A-0 show hypothetical exemplifications for using the below described method to determine % amino acid sequence identity (Figures 3A-D) and % nucleic acid sequence identity (Figures 3C-D) using the ALIGN-2 sequence comparison computer program, wherein `PRO" represents the amino acid sequence of a hypothetical PRO10282 polypeptide of interest, "Comparison Protein" represents the amino acid sequence of a polypeptide against which the 'PRO' poiypeptide of interest is being compared, 'PRO-DNA" represents a hypothetical PRO10282-encoding nucleic acid sequence of interest, "Comparison DNA' represents the nucleotide sequence of If nucleic acid molecule against which the "PRO-DNA" nucleic acid molecule of interest is being compared, 'X, "Y" and each represent different hypothetical amino acid residues and "N", "L" and "V' each represent different hypothetical nucleotides.
Figures 4A-Q provide the complete source code for the ALIGN-2 sequence comparison computer program. This source code may be routinely compiled for use one UNIX operating system to provide the ALIGN-2 sequence comparison computer program.
Figure 6 shows a nucleotide sequence designated herein as DNA100038 (8E1310 NO:3).
Figure 8 shows the nucleotide sequence (3E010 NO: 4) of a cDNA containing a nucleotide sequence (nucleotides 14b 1-2778) encoding a native sequence human Stra6 polypeptide variant, wherein the nucleotide sequence (SEG ID N0:4) is a clone designated herein as "DNA148389-2827-1." Also presented in bold font and underlined are the positions of the respective start and stop codons.
Figure 7 shows the amino acid sequence (SEG ID NO:5) of a native sequence human Stra6 polypeptide variant as derived from the coding sequence of SEG ID NO:4. Also shown are the approximate locations of various other important polypeptide domains.
Figure 8 is a schematic representation of mouse Stra6, and the human Stra6 protein encoded by DNA148380-2827 (native human PRO10282).
Figure 9 shows the hydrophobicity plot of the native sequence human Stra6 protein encoded by DNA 148380-2827 (native human PRO10282).
Figure 10 shows the relative RNA expression profile for the native sequence human Stra 6 protein encoded by DNA148380-2827 in various normal human tissues.
Figure 11 shows the RNA fold expression for the native sequence human Stra6 protein encoded by DNA 148380-2827 in human colon tumor tissue relative to RNA expression in normal mucose from the same patient assayed by quantitative PCR. The data are from one experiment done in triplicate. The experiment was repeated at least twice with a different set of PCR primers.
Figure 12A shows the RNA expression for the native sequence human Stra6 protein encoded by DNA148380-2827 in human colon tissue relative to RNA expression in normal mucose from the same patient, using the housekeeping gene, GAPDH as a control.
Figure 12B shows the localization of Stra6 to the epithelial tumor cells in a colon adenocarcinoma by in situ hybridization.
Figure 13 shows the RNA expression for the native human Stra6 protein encoded by DNA148380-2827 in human breast, kidney, colon and lung tumor cell lines, relative to corresponding normal cell lines.
Figure 14 illustrates the expression of peptide fragments derived from the native sequence human Stra 6 protein encoded by 0NA148380-2827 in E.coli.
Figure 15 illustrates Stra6 RNA expression in human colon carcinoma cells in the presence and absence of all-trans-retinoic acid (ATRA) and 9-cis-retinoic acid (9cRA), respectively.
Figure 16 In situ hybridization for Stra6 in tumor sections. Darkfield images demonstrating silver grains (A, C, E, G) are shown with corresponding hematoxylinieosin-stained brightfield images (B, D, F, H). Moderate densities of silver grains overlie tumor cells but not a blood vessel in a malignant melanoma (A, B). Neoplastic epithelium in an endometrial adenocarcinoma is moderateljr labeled whereas tumor stronna is negative (C, D). Blastemal regions in a Wilm's tumor display high expression levels whereas tumor stroma is negative (E, F). A
pheochromocytama shows very high Stra6 mRNA
expression while adjacent normal adrenal cortex is negative (G, H). Scale bars = 100 microns.
Figure 17 (A) Induction of Stra6 mRNA expression in response to 9-cis-RA or all-trans-RA in C57M13/Parent and C57MG/Wnt-1 cells. (B) Induction of Stra6 mRNA expression in C57MG(Parent cells in response to Wnt-3A conditioned media and 9-cis-RA. (C) Induction of Stra6 mRNA expression after retinoic acid treatment in HCT116 and WiDr colon adenocarcinoma cells. (0) Darkfield images demonstrating Stra6 expression by in situ hybridization in HCT116 cells before (top panel) and after (lower panel) treatment with retinoic acid. (E) Stra6 protein expression in WiDr cells before (-RA) and after (+RA) treatment with retinoic acid as visualized by Western blot with a monoclonal antibody directed against human Stra6 peptide B. (F) Stra6 membrane localization in WiDr cells untreated (left panel) or treated (right panel) with retinoic acid. Immunohistochemistry was performed with an anti-human Stra6 peptide B
monoclonal hybridonna culture supernatant (clone 12F4.2H9.105). For the experiments shown in A-F, cells were treated with retinoic acid for 48 hours. Stra6 products obtained after completion of the quantitative PCR reactions (40 cycles each) are shown below each graph A-C.
Figure 18 (A) Wnt-1 induces RARy-1 expression. Protein-equivalent amounts of whole cell lysate from tetracycline repressible C57MG1Wnt-1 cells in the absence of tetracycline for 0, 24, 48, or 72 hours, were subjected to SOS-PAGE and immunoblotted for RARy-1 and ERK2. (B) RARy-1 mRNA expression in hyperplastic mammary glands and mammary gland tumors from Wnt-1 transgenic mice. mRNA expresson was determined by quantitative RT-PCR and the data are expressed as fold expression relative to mRNA expression in wild-type mammary glands.
DETAILED DESCRIPTION OF THE PREFERRED EMBODIMENTS
Definitions The terms "PRO10282 polypeptide", "PR010282 protein", "PR010282", "Stra6 polypeptide", "Stra6 protein"
and "Stra6" are used interchangeably, and encompass native sequence PR010282 (Stra6) and PR010282 (Stra6) polypeptide variants (which are further defined herein). The PRO10282 (Stra6) polypeptide may be isolated from a variety 2 0 of sources, such as from human tissue types or from another source, or prepared by recombinant andlor synthetic methods.
A "native sequence PRO10282" or "native sequence Stra6" comprises a polypeptide having the same amino acid sequence as a P11010282 derived from nature. Such native sequence P11010282 (Stra6) can be isolated from nature or can be produced by recombinant and/or synthetic means. The term "native sequence PRO10282" or "native sequence Stra6" specifically encompasses naturally-occurring truncated or secreted forms (e.g., an extracellular domain sequence), naturally-occurring variant forms (e.g., alternatively spliced forms) and naturally-occurring allelic variants of the PR010282.
In one embodiment of the invention, the native sequence PR010282 is a mature or full-length native sequence PRO10282 comprising amino acids 1 to 667 of Figure 2 (SEG ID NO:2). In another embodiment of the invention, thenative sequence PR010282 polypeptide is a mature or full-length P11019578 polypeptide comprising amino acids 1 to 658 of SEG ID NO:
5, which is believed to be an alternatively spliced form of the native sequence PRO10282 polypeptide of SEG ID NO: 2.
Also, while the P11010282 polypeptides disclosed in Figure 2 (SEG ID NO:2) and in Figure 7, SEQ ID NO: 5 are shown to begin with the methionine residue designated herein as amino acid position 1, it is conceivable and possible that another methionine residue located either upstream or downstream from amino acid position 1 in Figure 2 (SEG ID NO:2) or in Figure 7 (SEG ID NO: 5) may be employed as the starting amino acid residue for the PRO10282 polypeptide.
The PRO10282 (Stra6) polypeptide "extracellular domain" or "ECD" refers to a form of the P11010282 (Stra6) 3 5 polypeptide which is essentially free of the transmembrane and cytoplasmic domains. Ordinarily, a PRO10282 polypeptide ECD will have less than about 1% of such transmembrane and/or cytoplasmic domains and preferably, will have less than about 0.5% of such domains. It will be understood that any transmembrane domain(s) identified for the PR010282 polypeptides of the present invention are identified pursuant to criteria routinely employed in the art for identifying that type of hydrophobic domain. The exact boundaries of a transmembrane domain may vary but most likely by no more than about 5 amino acids at either end of the domain as initially identified. As such, in one embodiment of the present invention, the extracellular domain of a PRO10282 polypeptide comprises amino acids 1 to X, wherein X is any amino acid from amino acid 49 to 59 of Figure 2 (SEG ID NO:2) or of Figure 7 (SEG ID NO: 5).
"PRO10282 variant polypeptide" or "Stra6 variant polypeptide", which terms are used interchangeably, means an active PR010282 (Stra6) polypeptide as defined below having at least about 80% amino acid sequence identity with the amino acid sequence of (a) residues 1 to 667 of the PR010282 polypeptide shown in Figure 2 (SEO ID N0:2), or residues 1 to 658 of Figure 7 (SRI ID NO: 5), (b) 1 to X of Figure 2 (SEG ID
N0:2) or Figure 7 (SEG ID NO: 5), wherein X
is any amino acid from amino acid 49 to amino acid 59 of Figure 2 (SEG ID
NO:2) or of Figure 7 (SEG ID NO: 5), or (c) another specifically derived fragment of the amino acid sequence shown in Figure 2 (SEG ID NO:2), or Figure 7 (SEG ID NO:
5). Such PR010282 (Stra6) variant polypeptides include, for instance, PR010282 (Stra6) polypeptides wherein one or more amino acid residues are added, or deleted, at the N- andlor C-terminus, as well as within one or more internal domains, of the sequence of Figure 2 (SEG ID NO:2) or Figure 7 (SEG ID NO: 5).
Ordinarily, a PR010282 variant polypeptide will have at least about 80% amino acid sequence identity, more preferably at least about 81% amino acid sequence identity, more preferably at least about 82% amino acid sequence identity, more preferably at least about 83% amino acid sequence identity, more preferably at least about 84% amino acid sequence identity, more preferably at least about 85% amino acid 2 0 sequence identity, more preferably at least about 86% amino acid sequence identity, more preferably at least about 87%
amino acid sequence identity, more preferably at least about 88% amino acid sequence identity, more preferably at least about 89% amino acid sequence identity, more preferably at least about 90%
amino acid sequence identity, more preferably at least about 91% amino acid sequence identity, more preferably at least about 92% amino acid sequence identity, more preferably at least about 93% amino acid sequence identity, more preferably at least about 94% amino acid 2 5 sequence identity, more preferably at least about 95% amino acid sequence identity, more preferably at least about 96%
amino acid sequence identity, more preferably at least about 97% amino acid sequence identity, more preferably at least about 98% amino acid sequence identity and yet more preferably at least about 99% amino acid sequence identity with (a) residues 1 to 667 of the PR010282 polypeptide shown in Figure 2 (SEG ID
NO:2) or residues 1 to 658 of the PRO19578 polypeptide of Figure 7 (SEG ID NO: 5), (b) 1 to X of Figure 2 (SEO
ID NO:2) or Figure 7 (SEG ID NO: 5), wherein 3 0 X is any amino acid from amino acid 49 to amino acid 59 of Figure 2 (SEO. ID NO:2) or Figure 7 (SEG tEl NO: 5), or (c) another specifically derived fragment of the amino acid sequence shown in Figure 2 (SEG ID NO:2) or Figure 7 (SEG ID NO:
5). PR010282 variant polypeptides do not encompass the native PRO10282 polypeptide sequence. Ordinarily, PFI010282 variant polypeptides are at least about 10 amino acids in length, often at least about 20 amino acids in length, more often at least about 30 amino acids in length, more often at least about 40 amino acids in length, more often at least about 50 35 amino acids in length, more often at least about 60 amino acids in length, more often at least about 70 amino acids in length, more often at least about 80 amino acids in length, more often at least about 90 amino acids in length, more often at least about 100 amino acids in length, more often at least about 150 amino acids in length, more often at least about 200 amino acids in length, more often at least about 250 amino acids in length, more often at least about 300 amino acids in length, or more, and are different from fragments of the murine Stra6 sequence, as disclosed in Bouillet et al., Mechanisms of Development 63, 173-186 (1997).
"Percent (%) amino acid sequence identity" with respect to the PRO10282 (Stra6) polypeptide sequences identified herein is defined as the percentage of amino acid residues in a candidate sequence that are identical with the amino acid residues in a PRO10282 sequence, after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent sequence identity, and not considering any conservative substitutions as part of the sequence identity. Alignment for purposes of determining percent amino acid sequence identity can be achieved in various ways that are within the skill in the art, for instance, using publicly available computer software such as BLAST, BLAST-2, ALIGN, ALIGN-2 or Megalign (DNASTAR) software. Those skilled in the art can determine appropriate parameters for measuring alignment, including any algorithms needed to achieve maximal alignment over the full-length of the sequences being compared. For purposes herein, however, % amino acid sequence identity values are obtained as described below by using the sequence comparison computer program ALIGN-2, wherein the complete source code for the ALIGN-2 program is provided in Figures 4A-Q. The ALIGN-2 sequence comparison computer program was authored by Genentech, Inc. and the source code shown in Figures 4A-0 has been filed with user documentation in the U.S. Copyright Office, Washington D.C., 20559, where it is registered under U.S. Copyright Registration No. TXU510087.
The ALIGN-2 program is publicly available through Genentech, Inc., South San Francisco, California or may be compiled from the source code provided in 2 0 Figures 4A-Q. The ALIGN-2 program should be compiled for use on a UNIX
operating system, preferably digital UNIX
V4.0D. All sequence comparison parameters are set by the ALIGN-2 program and do not vary.
For purposes herein, the % amino acid sequence identity of a given amino acid sequence A to, with, or against a given amino acid sequence B (which can alternatively be phrased as a given amino acid sequence A that has or comprises a certain % amino acid sequence identity to, with, or against a given amino acid sequence B) is calculated as follows:
100 times the fraction X11' where X is the number of amino acid residues scored as identical matches by the sequence alignment program ALIGN-2 in that program's alignment of A and B, and where 'I is the total number of amino acid residues in B. It will be appreciated 3 0 that where the length of amino acid sequence A is not equal to the length of amino acid sequence B, the % amino acid sequence identity of A to B will not equal the % amino acid sequence identity of B to A. As examples of % amino acid sequence identity calculations, Figures 3A-B demonstrate how to calculate the % amino acid sequence identity of the amino acid sequence designated "Comparison Protein" to the amino acid sequence designated "PRO".
Unless specifically stated otherwise, all % amino acid sequence identity values used herein are obtained as 3 5 described above using the ALIGN-2 sequence comparison computer program.
However, % amino acid sequence identity may also be determined using the sequence comparison program NCBI-BLAST2 (Altschul et al., Nucleic Acids Res.
25:3389-3402 (1997)). The NCBI-BLAST2 sequence comparison program may be downloaded from http:11www.nchi.nlm.nih.gov. NCBI-BLAST2 uses several search parameters, wherein all of those search parameters are set to default values including, for example, unmask = yes, strand = all, expected occurrences = 10, minimum low complexity length = 1515, multi-pass e-value = 0.01, constant for multi-pass =
25, dropoff for final gapped alignment = 25 and scoring matrix = BLOSUM62.
In situations where NCBI-BLAST2 is employed for amino acid sequence comparisons, the % amino acid sequence identity of a given amino acid sequence A to, with, or against a given amino acid sequence B (which can alternatively be phrased as a given amino acid sequence A that has or comprises a certain %
amino acid sequence identity to, with, or against a given amino acid sequence B) is calculated as follows:
100 times the fraction XIV
where X is the number of amino acid residues scored as identical matches by the sequence alignment program NCBI-1 5 BLAST2 in that program's alignment of A and B, and where Y is the total number of amino acid residues in B. It will be appreciated that where the length of amino acid sequence A is not equal to the length of amino acid sequence B, the %
amino acid sequence identity of A to B will not equal the % amino acid sequence identity of B to A.
"PR010282 (Stra6) variant polynucleotide" or "PR010282 (Stra6) variant nucleic acid sequence" means a nucleic acid molecule which encodes an active PRO10282 polypeptide as defined below and which has at least about 80%
nucleic acid sequence identity with either (a) a nucleic acid sequence which encodes residues 1 to 667 of the PRO10282 polypeptide shown in Figure 2 (SEG ID N0:2), or residues 1 to 658 of Figure 7 (SEG ID NO: 5), (b) a nucleic acid sequence which encodes amino acids 1 to X of Figure 2 (SEG ID NO:2) or Figure 7 (SEG ID
NO: 5), wherein X is any amino acid from amino acid 49 to amino acid 59 of Figure 2 (SEO ID NO:2) or of Figure 7 (SEG
ID NO: 5), or (c) a nucleic acid sequence which encodes another specifically derived fragment of the amino acid sequence shown in Figure 2 (SEG ID NO:2) or in Figure 7 (SEG ID NO: 5). Ordinarily, a PRO10282 variant polynucleotide will have at least about 80% nucleic acid sequence identity, more preferably at least about 81% nucleic acid sequence identity, more preferably at least about 82% nucleic acid sequence identity, more preferably at least about 83% nucleic acid sequence identity, more preferably at least about 84% nucleic acid sequence identity, more preferably at least about 85% nucleic acid sequence identity, more preferably at least about 86% nucleic acid sequence identity, more preferably at least about 87% nucleic acid sequence identity, more 3 0 preferably at least about 88% nucleic acid sequence identity, more preferably at least about 89% nucleic acid sequence identity, more preferably at least about 90% nucleic acid sequence identity, more preferably at least about 91% nucleic acid sequence identity, more preferably at least about 92% nucleic acid sequence identity, more preferably at least about 93% nucleic acid sequence identity, more preferably at least about 94% nucleic acid sequence identity, more preferably at least about 95% nucleic acid sequence identity, more preferably at least about 96% nucleic acid sequence identity, more 3 5 preferably at least about 97% nucleic acid sequence identity, more preferably at least about 98% nucleic acid sequence identity and yet more preferably at least about 99% nucleic acid sequence identity with either (a) a nucleic acid sequence which encodes residues 1 to 667 of the PRO10282 polypeptide shown in Figure 2 (SEG ID N0:2), or residues 1 to 658 of the PR019678 polypeptide shown in Figure 7 (SEG ID NO: 5), (b) a nucleic acid sequence which encodes amino acids 1 to X of Figure 2 (SEG ID NO:2) or of Figure 7 (SEG ID NO: 5), wherein X is any amino acid from amino acid 49 to amino acid 59 of Figure 2 (SEG ID NO:2) or Figure 7 (SEG ID NO: 5), or (c) a nucleic acid sequence which encodes another specifically derived fragment of the amino acid sequence shown in Figure 2 (SEG ID NO:2) or in Figure 7 (SEG ID NO: 5).
PR010282 polynucleotide variants do not encompass the native PRO10282 nucleotide sequence, or the native PR019578 nucleotide sequence.
Ordinarily, PR010282 (Stra6) variant polynucleotides are at least about 30 nucleotides in length, often at least about 60 nucleotides in length, more often at least about 90 nucleotides in length, more often at least about 120 nucleotides in length, more often at least about 150 nucleotides in length, more often at least about 180 nucleotides in length, more often at least about 210 nucleotides in length, more often at least about 240 nucleotides in length, more often at least about 270 nucleotides in length, more often at least about 300 nucleotides in length, more often at least about 450 nucleotides in length, more often at least about 600 nucleotides in length, more often at least about 900 nucleotides in length, or more, and specifically exclude polynucleotides that are included in the nucleotide sequence of murine Stra6,as disclosed in Bouillet et al., Mechanisms of Development 63, 173-186 (1997).
"Percent (%) nucleic acid sequence identity" with respect to the PRO10282 (Stra6) polypeptide-encoding nucleic acid sequences identified herein is defined as the percentage of nucleotides in a candidate sequence that are identical with the nucleotides in a PRO10282 polypeptide-encoding nucleic acid sequence, after aligning the sequences and introducing 2 0 gaps, if necessary, to achieve the maximum percent sequence identity.
Alignment for purposes of determining percent nucleic acid sequence identity can be achieved in various ways that are within the skill in the art, for instance, using publicly available computer software such as BLAST, BLAST-2, ALIGN, ALIGN-2 or Megalign (DNASTAR) software. Those skilled in the art can determine appropriate parameters for measuring alignment, including any algorithms needed to achieve maximal alignment over the full-length of the sequences being compared. For purposes herein, however, % nucleic acid 2 5 sequence identity values are obtained as described below by using the sequence comparison computer program ALIGN-2, wherein the complete source code for the ALIGN-2 program is provided in Figures 4A-0. The ALIGN-2 sequence comparison computer program was authored by Genentech, Inc. and the source code shown in Figures 4A-Q has been filed with user documentation in the U.S. Copyright Office, Washington D.C., 20559, where it is registered under U.S. Copyright Registration No. TXU510087. The ALIGN-2 program is publicly available through Genentech, Inc., South San Francisco, 3 0 California or may be compiled from the source code provided in Figures 4A-Q. The ALIGN-2 program should be compiled for use on a UNIX operating system, preferably digital UNIX V4.0D. All sequence comparison parameters are set by the ALIGN-2 program and do not vary.
For purposes herein, the % nucleic acid sequence identity of a given nucleic acid sequence C to, with, or against a given nucleic acid sequence D (which can alternatively be phrased as a given nucleic acid sequence C that has or 35 comprises a certain % nucleic acid sequence identity to, with, or against a given nucleic acid sequence D) is calculated as follows:
100 times the fraction IN/Z
where W is the number of nucleotides scored as identical matches by the sequence alignment program ALIGN-2 in that program's alignment of C and D, and where Z is the total number of nucleotides in D. It will be appreciated that where the length of nucleic acid sequence C is not equal to the length of nucleic acid sequence 0, the % nucleic acid sequence identity of C to D will not equal the % nucleic acid sequence identity of D to C. As examples of % nucleic acid sequence identity calculations, Figures 3C-D demonstrate how to calculate the % nucleic acid sequence identity of the nucleic acid sequence designated "Comparison DNA" to the nucleic acid sequence designated "PRO-DNA".
Unless specifically stated otherwise, all % nucleic acid sequence identity values used herein are obtained as described above using the ALIGN-2 sequence comparison computer program.
However, % nucleic acid sequence identity may also be determined using the sequence comparison program NCBI-BLAST2 (Altschul et al., Nucleic Acids Res.
25:3389-3402 (1997)).
The NCBI-BLAST2 sequence comparison program may be downloaded from http://www.ncbi.nlm.nih.gov. NCBI-BLAST2 uses several search parameters, wherein all of those search parameters are set to default values including, for example, unmask = yes, strand = all, expected occurrences = 10, minimum low complexity length = 15/5, multi-pass e-value = 0.01, constant for multi-pass =
25, dropoff for final gapped alignment = 25 and'scoring matrix = BLOSUM62.
In situations where NCBI-BLAST2 is employed for sequence comparisons, the %
nucleic acid sequence identity 2 0 of a given nucleic acid sequence C to, with, or against a given nucleic acid sequence D (which can alternatively be phrased as a given nucleic acid sequence C that has or comprises a certain % nucleic acid sequence identity to, with, or against a given nucleic acid sequence I)) is calculated as follows:
100 times the fraction W/Z
where W is the number of nucleotides scored as identical matches by the sequence alignment program NCBI-BLAST2 in that program's alignment of C and D, and where Z is the total number of nucleotides in D. It will be appreciated that where the length of nucleic acid sequence C is not equal to the length of nucleic acid sequence D, the % nucleic acid sequence identity of C to 0 will not equal the % nucleic acid sequence identity of 0 to C.
In other embodiments, PRO10282 (Stra6) variant polynucleotides are nucleic acid molecules that encode an active PRO10282 polypeptide and which are capable of hybridizing, preferably under stringent hybridization and wash conditions, to nucleotide sequences encoding the full-length PRO10282 polypeptides shown in Figure 2 (SEG ID NO:2) or Figure 7 (SEG If) NO: 5). PR010282 variant polypeptides may be those that are encoded by a PR010282 variant polynucleotide.
The term "positives", in the context of the amino acid sequence identity comparisons performed as described 21' above, includes amino acid residues in the sequences compared that are not only identical, but also those that have similar properties. Amino acid residues that score a positive value to an amino acid residue of interest are those that are either identical to the amino acid residue of interest or are a preferred substitution (as defined in Table 1 below) of the amino acid residue of interest.
For purposes herein, the % value of positives of a given amino acid sequence A
to, with, or against a given amino acid sequence B (which can alternatively be phrased as a given amino acid sequence A that has or comprises a certain %
positives to, with, or against a given amino acid sequence B) is calculated as follows:
100 times the fraction XfY
where X is the number of amino acid residues scoring a positive value as defined above by the sequence alignment program ALIGN-2 in that program's alignment of A and B, and where Y is the total number of amino acid residues in B. It will be appreciated that where the length of amino acid sequence A is not equal to the length of amino acid sequence B, the %
positives of A to B will not equal the % positives of B to A.
"Isolated," when used to describe the various polypeptides disclosed herein, means polypeptide that has been identified and separated andIor recovered from a component of its natural environment. Preferably, the isolated polypeptide is free of association with all components with which it is naturally associated. Contaminant components of its natural environment are materials that would typically interfere with diagnostic or therapeutic uses for the polypeptide, and may include enzymes, hormones, and other proteinaceous or non-proteinaceous solutes. In preferred embodiments, the polypeptide will be purified (1) to a degree sufficient to obtain at least 15 residues of N-terminal or internal amino acid sequence by use of a spinning cup sequenator, or (2) to homogeneity by SOS-PAGE under non-reducing or reducing conditions using Coomassie blue or, preferably, silver stain. Isolated polypeptide includes polypeptide in situ within recombinant cells, since at least one component of the PR010282 (Stra6) natural environment will not be present.
Ordinarily, however, isolated polypeptide will be prepared by at least one purification step.
An "isolated" nucleic acid molecule encoding a PRO10282 (Stra6) polypeptide is a nucleic acid molecule that is identified and separated from at least one contaminant nucleic acid molecule with which it is ordinarily associated in the natural source of the PRO10282-encoding nucleic acid. Preferably, the isolated nucleic is free of association with all components with which it is naturally associated. An isolated PRO10282-encoding nucleic acid molecule is other than in the form or setting in which it is found in nature. Isolated nucleic acid molecules therefore are distinguished from the PRO10282-encoding nucleic acid molecule as it exists in natural cells.
However, an isolated nucleic acid molecule encoding a PR010282 (Stra6) polypeptide includes PRO10282-encoding nucleic acid molecules contained in cells that ordinarily express PR010282 where, for example, the nucleic acid molecule is in a chromosomal location different from that of natural cells.
The term "control sequences" refers to DNA sequences necessary for the expression of an operably linked coding sequence in a particular host organism. The control sequences that are suitable for prokaryotes, for example, include a promoter, optionally an operator sequence, and a ribosome binding site.
Eukaryotic cells are known to utilize promoters, polyadenylation signals, and enhancers.
Nucleic acid is "operably linked" when it is placed into a functional relationship with another nucleic acid sequence. For example, DNA for a presequence or secretory leader is operably linked to DNA for a polypeptide if it is expressed as a preprotein that participates in the secretion of the polypeptide; a promoter or enhancer is operably linked to a coding sequence if it affects the transcription of the sequence; or a ribosome binding site is operably linked to a coding sequence if it is positioned so as to facilitate translation. Generally, "operably linked" means that the DNA sequences being linked are contiguous, and, in the case of a secretory leader, contiguous and in reading phase. However, enhancers do not have to be contiguous. Linking is accomplished by ligation at convenient restriction sites. If such sites do not exist, the synthetic oligonucleotide adaptors or linkers are used in accordance with conventional practice.
The term "antibody" is used in the broadest sense and specifically covers, for example, single anti-PR010282 monoclonal antibodies (including agonist, antagonist, and neutralizing antibodies), anti-PRO10282 antibody compositions with polyepitopic specificity, single chain anti-PRO10282 antibodies, and fragments of anti-PR010282 antibodies (see below). The term "monoclonal antibody" as used herein refers to an antibody obtained from a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical except for possible naturally-occurring mutations that may be present in minor amounts.
"Stringency" of hybridization reactions is readily determinable by one of ordinary skill in the art, and generally is an empirical calculation dependent upon probe length, washing temperature, and salt concentration. In general, longer probes require higher temperatures for proper annealing, while shorter probes need lower temperatures. Hybridization 2 0 generally depends on the ability of denatured DNA to reanneal when complementary strands are present in an environment below their melting temperature. The higher the degree of desired homology between the probe and hybridizable sequence, the higher the relative temperature which can be used. As a result, it follows that higher relative temperatures would tend to make the reaction conditions more stringent, while lower temperatures less so. For additional details and explanation of stringency of hybridization reactions, see Ausubel et al., Current Protocols in Molecular Biology, Wiley Interscience Publishers, (1995).
"Stringent conditions" or "high stringency conditions", as defined herein, may be identified by those that: (1) employ low ionic strength and high temperature for washing, for example 0.015 M sodium chloride10.0015 M sodium citrate10.1% sodium dodecyl sulfate at 50 C; (2) employ during hybridization a denaturing agent, such as formamide, for example, 50% (v1v) formamide with 0.1% bovine serum albumin10.1% Fico1110.1%
polyvinylpyrrolidone150mM sodium 3 0 phosphate buffer at pH 6.5 with 750 mM sodium chloride, 75 mM sodium citrate at 42 C; or (3) employ 50% formamide, 5 x SSC (0.75 M NaCI, 0.075 M sodium citrate), 50 mM sodium phosphate (pH
6.8), 0.1% sodium pyrophosphate, 5 x Denhardt's solution, sonicated salmon sperm DNA (50 gIm1), 0.1% SOS, and 10%
dextran sulfate at 42 C, with washes at 42 C in 0.2 x SSC (sodium chloridelsodium citrate) and 50% formamide at 55 C, followed by a high-stringency wash consisting of 0.1 x SSC containing EDTA at 55 C.
3 5 "Moderately stringent conditions" may be identified as described by Sambrook et al., Molecular Cloning: A

Laboratory Manual, New York: Cold Spring Harbor Press, 1989, and include the use of washing solution and hybridization conditions (e.g., temperature, ionic strength and %SDS) less stringent that those described above. An example of moderately stringent conditions is overnight incubation at 37 C in a solution comprising: 20% formamide, 5 x SSC (150 mM Na Cl, 15 mM trisodium citrate), 50 mM sodium phosphate (pH 7.6), 5 x Denhardt's solution, 10% dextran sulfate, and 20 mgiml denatured sheared salmon sperm DNA, followed by washing the filters in 1 x SSC at about 37-50 C. The skilled artisan will recognize how to adjust the temperature, ionic strength, etc. as necessary to accommodate factors such as probe length and the like.
The term "epitope tagged" when used herein refers to a chimeric polypeptide comprising a PRO10282 polypeptide fused to a "tag polypeptide". The tag polypeptide has enough residues to provide an epitope against which 1 0 an antibody can be made, yet is short enough such that it does not interfere with activity of the polypeptide to which it is fused. The tag polypeptide preferably also is fairly unique so that the antibody does not substantially cross-react with other epitopes. Suitable tag polypeptides generally have at least six amino acid residues and usually between about 8 and 50 amino acid residues (preferably, between about 10 and 20 amino acid residues).
The term "antibody" is used in the broadest sense and specifically covers, for example, single anti-PR010282 (anti-Stra6) monoclonal antibodies (including agonist, antagonist, and neutralizing antibodies), anti-PRO10282 antibody compositions with polyepitopic specificity, single chain anti-PR010282 antibodies, and fragments of anti-PR010282 antibodies (see below).
The term "monoclonal antibody" as used herein refers to an antibody obtained from a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical except for possible naturally-2 0 occurring mutations that may be present in minor amounts.
"Antibody fragments" comprise a portion of an intact antibody, preferably the antigen binding or variable region of the intact antibody. Examples of antibody fragments include Fab, Fab', F(a1312, and Fv fragments; diabodies; linear antibodies (Zapata et al., Protein Eno., 8(1O):1057-1062 [19951); single-chain antibody molecules; and multispecific antibodies formed from antibody fragments.

Papain digestion of antibodies produces two identical antigen-binding fragments, called "Fab" fragments, each with a single antigen-binding site, and a residual "Fc" fragment, whose name reflects its ability to crystallize readily.
Pepsin treatment yields an F(a13)2 fragment that has two antigen-combining sites and is still capable of cross-linking antigen.
"Fv" is the minimum antibody fragment, which contains a complete antigen-recognition and -binding site. This region consists of a dimer of one heavy- and one light-chain variable domain in tight, non-covalent association. It is in this configuration that the three CDRs of each variable domain interact to define an antigen-binding site on the surface of the VH-1/1. dimer. Collectively, the six CDRs confer antigen-binding specificity to the antibody. However, even a single variable domain (or half of an Fv comprising only three CDRs specific for an antigen) has the ability to recognize and bind antigen, although at a lower affinity than the entire binding site.
35 The Fab fragment also contains the constant domain of the light chain and the first constant domain (CH1) of the heavy chain. Fab fragments differ from Fab' fragments by the addition of a few residues at the carboxy terminus of the heavy chain CH1 domain including one or more cysteines from the antibody hinge region. Fab'-SH is the designation herein for Fab' in which the cysteine residue(s) of the constant domains bear a free thiol group. F(ab')2 antibody fragments originally were produced as pairs of Fab' fragments which have hinge cysteines between them. Other chemical couplings of antibody fragments are also known.
The "light chains" of antibodies (immunoglobulins) from any vertebrate species can be assigned to one of two clearly distinct types, called kappa ( ) and lambda R based on the amino acid sequences of their constant domains.
Depending on the amino acid sequence of the constant domain of their heavy chains, immunoglobulins can be assigned to different classes. There are five major classes of immunoglobulins: IgA, IgD, IgE, IgG, and IgM, and several of these may be further divided into subclasses (isotypes), e.g., IgG1, IgG2, IgG3, 104, IgA, and IgA2. The heavy-chain constant domains that correspond to the different classes of immunoglobulins are called õ õ and , respectively. The subunit structures and three-dimensional configurations of different classes of immunoglobulins are well known.
The term "diabodies" refers to small antibody fragments with two antigen-binding sites, which fragments comprise a heavy-chain variable domain (VH) connected to a light-chain variable domain (lid in the same polypeptide chain (VH - VL). By using a linker that is too short to allow pairing between the two domains on the same chain, the domains are forced to pair with the complementary domains of another chain and create two antigen-binding sites. Diabodies are described more fully in, for example, EP 404,097; WO 93/11161; and Hollinger et al., Proc. Natl. Acad. Sci. USA, 90:6444-6448 (1993).
An "isolated" antibody is one which has been identified and separated and/or recovered from a component of its 2 0 natural environment. Contaminant components of its natural environment are materials which would interfere with diagnostic or therapeutic uses for the antibody, and may include enzymes, hormones, and other proteinaceous or nonproteinaceous solutes. In preferred embodiments, the antibody will be purified (1) to greater than 95% by weight of antibody as determined by the Lowry method, and most preferably more than 99%
by weight, (2) to a degree sufficient to obtain at least 15 residues of N-terminal or internal amino acid sequence by use of a spinning cup sequenator, or (3) to 2 5 homogeneity by SOS-PAGE under reducing or nonreducing conditions using Coomassie blue or, preferably, silver stain.
Isolated antibody includes the antibody in situ within recombinant cells since at least one component of the antibody's natural environment will not be present. Ordinarily, however, isolated antibody will be prepared by at least one purification step.
The term "variable" refers to the fact that certain portions of the variable domains differ extensively in sequence 3 0 among antibodies and are used in the binding and specificity of each particular antibody for its particular antigen. However, the variability is not evenly distributed throughout the variable domains of antibodies. It is concentrated in three segments called complennentarity-determining regions (CDRs) or hypervariable regions both in the light-chain and the heavy-chain variable domains. The more highly conserved portions of variable domains are called the framework (FR) regions. The variable domains of native heavy and light chains each comprise four FR
regions, largely adopting a -sheet configuration, 3 5 connected by three CDRs, which form loops connecting, and in some cases forming part of, the -sheet structure. The CDRs in each chain are held together in close proximity by the FR regions and, with the CDRs from the other chain, contribute to the formation of the antigen-binding site of antibodies (see Kabat et al., N1H Publ. No.91-3242, Vol. I, pages 647-669 (1991)). The constant domains are not involved directly in binding an antibody to an antigen, but exhibit various effector functions, such as participation of the antibody in antibody-dependent cellular toxicity. .
The term "hypervariable region" when used herein refers to the amino acid residues of an antibody which are responsible for antigen-binding. The hypervariable region comprises amino acid residues from a "complementarity determining region" or "CDR" (i.e., residues 24-34 (L1), 50-56 (L2) and 89-97 (L3) in the light chain variable domain and 31-35 (H1), 50-65 (H2) and 95-102 (H3) in the heavy chain variable domain;
Kabat et al., Sequences of Proteins of Immunoloqical Interest, 5th Ed. Public Health Service, National Institute of Health, Bethesda, MD. [1991]) and/or those residues from a "hypervariable loop" (i.e., residues 26-32 (L1), 50-52 (L2) and 91-96 (L3) in the light chain variable domain and 26-32 (H1), 53-55 (H2) and 96-101 (H3) in the heavy chain variable domain;
Clothia and Lesk, J. Mol. Biol., 196:901-917 [1987]). "Framework" or "FR" residues are those variable domain residues other than the hypervariable region residues as herein defined.
"Humanized" forms of non-human (e.g., murine) antibodies are chimeric immunoglobulins, immunoglobulin chains or fragments thereof (such as Fv, Fab, Fab', F(ablz or other antigen-binding subsequences of antibodies) which contain minimal sequence derived from non-human immunoglobulin. For the most part, humanized antibodies are human imnnunoglobulins (recipient antibody) in which residues from a CDR of the recipient are replaced by residues from a CDR
of a non-human species (donor antibody) such as mouse, rat or rabbit having the desired specificity, affinity, and capacity.
In some instances, Fv FR residues of the human immunoglobulin are replaced by corresponding non-human residues.

Furthermore, humanized antibodies may comprise residues which are found neither in the recipient antibody nor in the imported CDR or framework sequences. These modifications are made to furthee refine and maximize antibody performance. In general, the humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the CDR regions correspond to those of a non-human immunoglobulin and all or substantially all of the FR regions are those of a human immunoglobulin sequence. The humanized antibody optimally also will comprise at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin.
For further details, see, Jones et al., Nature, 321:522-525 (1986); Reichmann et al., Nature, 332:323-329 [1988]; and Presta, Carr. Oe. Struct. Biol., 2:593-596 (1992). The humanized antibody includes a PRIMAT1ZEDT" antibody wherein the antigen-binding region of the antibody is derived from an antibody produced by immunizing macaque monkeys with the antigen of interest.
3 0 As used herein, the term "immunoadhesin" designates antibody-like molecules which combine the binding specificity of a heterologous protein (an "adhesin") with the effector functions of immunoglobulin constant domains.
Structurally, the immunoadhesins comprise a fusion of an amino acid sequence with the desired binding specificity which is other than the antigen recognition and binding site of an antibody (i.e., is "heterologous"), and an immunoglobulin constant domain sequence. The adhesin part of an immunoadhesin molecule typically is a contiguous amino acid sequence comprising at least the binding site of a receptor or a ligand. The immunoglobulin constant domain sequence in the immunoadhesin may be obtained from any immunoglobulin, such as IgG-1, IgG-2, IgG-3, or IgG-4 subtypes, IgA (including IgA-1 and IgA-2), IgE, Ig0 or IgM.
"Active" or "activity" for the purposes herein refers to form(s) of PRO10282 which retain a biological andlor an immunological activity of native or naturally-occurring PRO10282, wherein "biological" activity refers to a biological function (either inhibitory or stimulatory) caused by a native or naturally-occurring P11010282 other than the ability to induce the production of an antibody against an antigenic epitope possessed by a native or naturally-occurring PRO10282 and an "immunological" activity refers to the ability to induce the production of an antibody against an antigenic epitope possessed by a native or naturally-occurring PR010282. A preferred biological activity includes, for example, a role in growth, development or differentiation in response to retinoic acid. Based on close similarity with Stra6, a murine protein induced in response to retinoic acid, PR010282 polypeptide disclosed herein may play an important role, for example, in early dorsoventral limb patterning and later in the control of endochondral ossification. Another biological activity is involvement in tumor development and growth.
"Immunological activity" preferably is "immunological cross-reactivity," which is used to mean that the candidate polypeptide is capable of competitively inhibiting the qualitative biological activity of a P11010282 (Stra6) polypeptide having this activity with polyclonal antisera raised against the known active PRO10282 (Stra6) polypeptide. Such antisera are prepared in conventional fashion by injecting goats or rabbits, for example, subcutaneously with the known active analogue in complete Freund's adjuvant, followed by booster intraperitoneal or subcutaneous injection in incomplete Freund's. The immunological cross-reactivity is preferably "specific", which means that the binding affinity of the immunologically cross-reactive molecule (e.g. antibody) identified, to the corresponding Stra6 polypeptide is significantly higher (preferably at least about 2-times, more preferably at least about 4-times, even more preferably at least aout 8-times, most preferably at least about 10-times higher) than the binding affinity of that molecule to any other known native polypeptide.
The term "antagonist" is used in the broadest sense, and includes any molecule that partially or fully blocks, inhibits, or neutralizes a biological activity of a native PR010282 polypeptide disclosed herein. In a similar manner, the term "agonist" is used in the broadest sense and includes any molecule that mimics a biological activity of a native PR010282 polypeptide disclosed herein. Suitable agonist or antagonist molecules specifically include agonist or antagonist antibodies or antibody fragments, fragments or amino acid sequence variants of native PRO10282 polypeptides, peptides, small organic molecules, etc. Methods for identifying agonists or antagonists of a P11010282 polypeptide may comprise contacting a PRO10282 polypeptide with a candidate agonist o'r antagonist molecule and measuring a detectable change in one or more biological activities normally associated with the P11010282 polypeptide.
"Tumor", as used herein, refers to all neoplastic cell growth and proliferation, whether malignant or benign, and all pre-cancerous and cancerous cells and tissues.
The terms "cancer" and "cancerous" refer to or describe the physiological condition in mammals that is typically characterized by unregulated cell growth. Examples of cancer include but are not limited to, carcinoma, lymphoma, blastoma, sarcoma, and leukemia. More particular examples of such cancers include breast cancer, prostate cancer, colon cancer, squamous cell cancer, small-cell lung cancer, non-small cell lung cancer, gastrointestinal cancer, pancreatic cancer, glioblastoma, cervical cancer, ovarian cancer, liver cancer, bladder cancer, hepatoma, colorectal cancer, endometrial carcinoma, salivary gland carcinoma, kidney cancer, liver cancer, vulval cancer, thyroid cancer, hepatic carcinoma and various types of head and neck cancer.
"Treatment" refers to both therapeutic treatment and prophylactic or preventative measures, wherein the object is to prevent or slow down (lessen) the targeted pathologic condition or disorder. Those in need of treatment include those already with the disorder as well as those prone to have the disorder or those in whom the disorder is to be prevented. In tumor (e.g., cancer) treatment, a therapeutic agent may directly decrease the pathology of tumor cells, or render the tumor cells more susceptible to treatment by other therapeutic agents, e.g., radiation and/or chemotherapy.
The "pathology" of cancer includes all phenomena that compromise the well-being of the patient. This includes, without limitation, abnormal or uncontrollable cell growth, metastasis, interference with the normal functioning of neighboring cells, release of cytokines or other secretory products at abnormal levels, suppression or aggravation of inflammatory or immunological response, etc.
"Chronic" administration refers to administration of the agent(s) in a continuous mode as opposed to an acute mode, so as to maintain the initial therapeutic effect (activity) for an extended period of time. "Intermittent" administration is treatment that is not consecutively done without interruption, but rather is cyclic in nature.
"Mammal" for purposes of treatment refers to any animal classified as a mammal, including humans, domestic and farm animals, and zoo, sports, or pet animals, such as dogs, cats, cattle, horses, sheep, pigs, goats, rabbits, etc.
Preferably, the mammal is human.

Administration "in combination with" one or more further therapeutic agents includes simultaneous (concurrent) and consecutive administration in any order.
"Carriers" as used herein include pharmaceutically acceptable carriers, excipients, or stabilizers which are nontoxic to the cell or mammal being exposed thereto at the dosages and concentrations employed. Often the physiologically acceptable carrier is an aqueous pH buffered solution.
Examples of physiologically acceptable carriers include buffers such as phosphate, citrate, and other organic acids;
antioxidants including ascorbic acid; low molecular weight (less than about 10 residues) polypeptide; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, arginine or lysine;
monosaccharides, disaccharides, and other carbohydrates including glucose, rnannose, or dextrins; chelating agents such as EDTA; sugar alcohols such as mannitol or sorbitol; salt-forming counterions such as sodium; and/or nonionic surfactants 3 0 such as TWEEN', polyethylene glycol (PEG), and PLURONICP.
The word "label" when used herein refers to a detectable compound or composition which is conjugated directly or indirectly to the antibody so as to generate a "labeled" antibody. The label may be detectable by itself (e.g. radioisotope labels or fluorescent labels) or, in the case of an enzymatic label, may catalyze chemical alteration of a substrate compound or composition which is detectable.
3 5 By "solid phase" is meant a non-aqueous matrix to which the antibody of the present invention can adhere.

Examples of solid phases encompassed herein include those formed partially or entirely of glass (e.g., controlled pore glass), polysaccharides (e.g., agarose), polyacrylamides, polystyrene, polyvinyl alcohol and silicones. In certain embodiments, depending on the context, the solid phase can comprise the well of an assay plate; in others it is a purification column (e.g., an affinity chromatography column). This term also includes a discontinuous solid phase of discrete particles, such as those described in U.S. Patent No. 4,275,149.
A "liposome" is a small vesicle composed of various types of lipids, phospholipids andIor surfactant which is useful for delivery of a drug (such as a PRO10282 polypeptide or antibody thereto) to a mammal. The components of the liposome are commonly arranged in a bilayer formation, similar to the lipid arrangement of biological membranes.
A "small molecule" is defined herein to have a molecular weight below about 500 Da!tons.
The phrases "gene amplification" and "gene duplication" are used interchangeably and refer to a process by which multiple copies of a gene or gene fragment are formed in a particular cell or cell line. The duplicated region (a stretch of amplified DNA) is often referred to as "amplicon." Usually, the amount of the messenger RNA (mRNA) produced, i.e., the level of gene expression, also increases in the proportion of the number of copies made of the particular gene expressed.
The term "cytotoxic agent" as used herein refers to a substance that inhibits or prevents the function of cells andlor causes destruction of cells. The term is intended to include radioactive isotopes (e.g., l', 1, ro and Hen, chemotherapeutic agents, and toxins such as enzymatically active toxins of bacterial, fungal, plant or animal origin, or fragments thereof.
A "chemotherapeutic agent" is a chemical compound useful in the treatment of cancer. Examples of chemotherapeutic agents include adriamycin, doxorubicin, epirubicin, 5-fluorouracil, cytosine arabinoside ("Ara-C"), cyclophosphamide, thiotepa, busulfan, cytoxin, taxoids, e.g., paclitaxel (Taxol, Bristol-Myers Squibb Oncology, Princeton, NJ), and doxetaxel (Taxotere, Rhone-Poulenc Rorer, Antony, Rnace), toxotere, methotrexate, cisplatin, melphalan, vinblastine, bleomycin, etoposide, ifosfamide, mitomycin C, mitoxantrone, vincristine, vinorelbine, carboplatin, teniposide, daunornycin, carminomycin, aminopterin, dactinomycin, nnitomycins, esperamicins (see U.S. Pat. No. 4,675,187), 5-FU, 6-thioguanine, 6-mercaptopurine, actinomycin D, VP-16, chlorannbucil, melphalan, and other related nitrogen mustards. Also included in this definition are hormonal agents that act to regulate or inhibit hormone action on tumors such as tannoxifen and onapristone.
A "growth inhibitory agent" when used herein refers to a compound or composition which inhibits growth of a cell, especially cancer cell overexpressing any of the genes identified herein, either in vitro or in vivo. Thus, the growth inhibitory agent is one which significantly reduces the percentage of cells overexpressing such genes in S phase.
Examples of growth inhibitory agents include agents that block cell cycle progression (at a place other than S phase), such as agents that induce 131 arrest and M-phase arrest. Classical M-phase blockers include the vincas (vincristine and vinblastine), taxol, and topo II inhibitors such as doxorubicin, epirubicin, daunorubicin, etoposide, and bleomycin. Those agents that arrest 131 also spill over into S-phase arrest, for example, DNA
alkylating agents such as tamoxif en, prednisone, dacarbazine, mechlorethamine, cisplatin, methotrexate, 5-fluorouracil, and ara-C. Further information can be found in The Molecular Basis of Cancer, Mendelsohn and Israel, eds., Chapter 1, entitled "Cell cycle regulation, oncogens, and antineoplastic drugs" by Murakami et al., (WB Saunders: Philadelphia, 1995), especially p. 13.
"Doxorubicin" is an anthracycline antibiotic. The full chemical name of doxorubicin is (8S-cis)-10-113-amino-2,3,6-trideoxy- -L-Iyxo-hexapyranosyl)oxy1-7,8,9,10-tetrahydro-6,8,11-trihydroxy-8=(hydroxyacety1)-1-methoxy-5,12-naphthacenedione.
The term "cytokine" is a generic term for proteins released by one cell population which act on another cell as intercellular mediators. Examples of such cytokines are lymphokines, monokines, and traditional polypeptide hormones.
Included among the cytokines are growth hormone such as human growth hormone, N-methionyl human growth hormone, and bovine growth hormone; parathyroid hormone; thyroxine; insulin;
proinsulin; relaxin; prorelaxin; glycoprotein hormones such as follicle stimulating hormone (FSH), thyroid stimulating hormone (TSH), and luteinizing hormone (LH); hepatic growth factor; fibroblast growth factor; prolactin; placental lactogen; tumor necrosis factor- and -; mullerian-inhibiting substance;
mouse gonadotropin-associated peptide; inhibin; activin; vascular endothelial growth factor; integrin; thrombopoietin (TP0);
nerve growth factors such as NGF- ; platelet-growth factor; transforming growth factors (TGFs) such as TGF- and TGF- ;
insulin-like growth factor-I and -II; erythropoietin (EPO); osteoinductive factors; interferons such as interferon - , = , and - ;
colony stimulating factors (CSFs) such as macrophage-CSF (M-CSF); granulocyte-macrophage-CSF (GM-CSF); and granulocyte-CSF (G-CSF); interleukins (us) such as IL-1, IL- la, I1-2, I1-3, I1-4, I1-5, I1-6, I1-7, I1-8, I1-9, IL-11, 11-12; a tumor necrosis factor such as TNF- or INF-13; and other polypeptide factors including LIF and kit ligand (KL). As used herein, the term cytokine includes proteins from natural sources or from recombinant cell culture and biologically active equivalents of the native sequence cytokines.
The term "prodrug" as used in this application refers to a precursor or derivative form of a pharmaceutically active substance that is less cytotoxic to tumor cells compared to the parent drug and is capable of being enzymatically activated or converted into the more active parent form. See, e.g., Wilman, "Prodrugs in Cancer Chemotherapy", Biochemical Society Transactions, 14:375-382, 615th Meeting, Belfast (1986), and Stella et al., "Prodrugs: A Chemical Approach to Targeted Drug Delivery", Directed Drug Delivery, Borchardt et al., (ed.), pp. 147-267, Humana Press (1985).
The prodrugs of this invention include, but are not limited to, phosphate-containing prodrugs, thiophosphate-containing prodrugs, sulfate-containing prodrugs, peptide-containing prodrugs, 0-amino acid-modified prodrugs, glysocylated prodrugs, 6-lactam-containing prodrugs, optionally substituted phenoxyacetamide-containing prodrugs or optionally substituted phenylacetamide-containing prodrugs, 5-fluorocytosine and other 5-fluorouridine prodrugs which can be converted into the more active cytotoxic free drug. Examples of cytotoxic drugs that can be derivatized into a prodrugs form for use in this invention include, but are not limited to, those chemotherapeutic agents described above.
3 0 An "effective amount" of a polypeptide disclosed herein or an antagonist thereof, in reference to inhibition of neoplastic cell growth, tumor growth or cancer cell growth, is an amount capable of inhibiting, to some extent, the growth of target cells. The term includes an amount capable of invoking a growth inhibitory, cytostatic and/or cytotoxic effect and/or apoptosis of the target cells. An "effective amount" of a PRO10282 (including P11019578) polypeptide or an antagonist thereof for the purposes of inhibiting neoplastic cell growth, tumor cell or cancer cell growth, may be determined 3 5 empirically and in a routine manner.

A "therapeutically effective amount", in reference to the treatment of tumor, refers to an amount capable of invoking one or more of the following effects: (1) inhibition, to some extent, of tumor growth, including, slowing down and complete growth arrest; (2) reduction in the number of tumor cells; (3) reduction in tumor size; (4) inhibition (i.e., reduction, slowing down or complete stopping) of tumor cell infiltration into peripheral organs; (5) inhibition (i.e., reduction, slowing down or complete stopping) of metastasis; (6) enhancement of anti-tumor immune response, which may, but does not have to, result in the regression or rejection of the tumor; and/or (7) relief, to some extent, of one or more symptoms associated with the disorder. A "therapeutically efefctive amount" of a PRO10282 polypeptide antagonist for purpose of treatment of tumor may be determined empirically and in a routine manner.
A "growth inhibitory amount" of a PRO10282 antagonist is an amount capable of inhibiting the growth of a cell, especially tumor, e.g. cancer cell, eitther in vitro or in vivo. A "cytotoxic amount" of a PRO10282 antagonist for purposes of inhibiting neoplastic cell growth may be determined empirically and in a routine manner.
A "cytotoxic amount" or a PRO10282 antagonist is an amount capable of causing the destruction of a cell, especially tumor, e.g. cancer cell, either in vitro or in vivo.A "cytotoxic amount" of a PRO10282 antagonist for purposes of inhibiting neoplastic cell growth may be determined empirically and in a routine manner.
"Antisense oligodeoxynucleotides" or "antisense oligonucleotides" (which terms are used interchangeably) are defined as nucleic acid molecules that can inhibit the transcription and/or translation of target genes in a sequence-specific manner. The term "antisense" refers to the fact that the nucleic acid is complementary t the coding ("sense") genetic sequence of the target gene. Antisense oligonucleotides hybridize in an antiparallel orientation to nascent mRNA through Watson-Crick base-pairing. By binding the target mRNA template, antisense oligonucleotides block the successful 2 0 translation of the encoded protein. The term specifically includes antisense agents called "ribozomes" that have been designated to induce catalytic cleavage of a target RNA by addition of a sequence that has natural self-splicing activity (Warzocha and Wotowiec, "Antisense strategy" biological utility and prospects int he treatment of hematological malignancies." Leuk. Lymphoma 24:267-281 [1997]).
2 5 II. Compositions and Methods of the Invention A. Full-length native sequence human PR010282 and PRO

(Stra6) Polvpeptides The present invention provides newly identified and isolated nucleotide sequences encoding polypeptides referred to in the present application as native sequence human PR010282 (or also UNO3126) and PR019578, respectively. In 30 particular, cDNA encoding the native human PR010282 and PR019578 polypeptides has been identified and isolated, as disclosed in further detail in the Examples below. It is noted that proteins produced in separate expression rounds may be given different PRO numbers but the UNG number is unique for any given DNA
and the encoded protein, and will not be changed. However, for sake of simplicity, in the present specification the protein encoded by DNA148380-2827 as well as all further native homologues and variants included in the foregoing definition of PRO10282, will be referred to as 35 "PRO10282", regardless of their origin or mode of preparation. The polypeptide encoded by DNA148389-2827-1, which is a native variant of "P11010282," will be specifically referred to as "P11019578."
As disclosed in the Examples below, cDNA clones designated herein as DNA148380-2827 and DNA148389-2827-1 have been deposited with the ATCC. The actual nucleotide sequence of the clone can readily be determined by the skilled artisan by sequencing of the deposited clone using routine methods in the art. The predicted amino acid sequence can be determined from the nucleotide sequence using routine skill.
For the full-length native human PRO10282 and P11019578 polypeptides and encoding nucleic acid described herein, Applicants have identified what is believed to be the reading frame best identifiable with the sequence information available at the time.
Using the ALIGN-2 sequence alignment computer program referenced above, it has been found that the full-length native sequence PRO10282 (shown in Figure 2 and SEG ID N0:2) has certain amino acid sequence identity with Stra6, a murine protein induced in response to retinoic acid (AF062476). Accordingly, it is presently believed that the P11010282 polypeptide disclosed in the present application is a newly identified member of the Stra6 protein family and may possess one or more biological and/or immunological activities or properties typical of that protein family. PR019578 is currently believed to be an alternatively spliced variant of native, full-length human PR010282, which has 9 amino acids (SPVDFLAGD) deleted at positions 89-97 of the native human P11010282 amino acid sequence, and contains Ile (I) instead of Met (M) at position 518, which corresponds to position 527 of PR010282.
This indicates that Stra6 may be polymorphic.
B. PRO10282 (Stra6) Variants Two specific native sequence human Stra6 polypeptides (native sequence human P11010282 and P11019578) are disclosed herein. As noted above, PRO19578 is believed to be an alternatively spliced native variant of native sequence human P11010282 and is, therefore, a "P11010282 variant." In addition to the full-length native sequence P11010282 and PR019578 polypeptides described herein, it is contemplated that PR010282 and P11019578 variants can be prepared.
PR010282 and P11019578 variants can be prepared by introducing appropriate nucleotide changes into the PR010282 or P11019578 DNA, and/or by synthesis of the desired PR010282 variant polypeptide. Those skilled in the art will appreciate that amino acid changes may alter post-translational processes of the PRO10282 or PRO19578, such as changing the number or position of glycosylation sites or altering the membrane anchoring characteristics.
Variations in the native full-length sequence P11010282, PR019578 or in various domains of the P11010282 or P11019578 described herein, can be made, for example, using any of the techniques and guidelines for conservative and non-conservative mutations set forth, for instance, in U.S. Patent No.
5,364,934. Variations may be a substitution, deletion or insertion of one or more codons encoding the P11010282 or PR019578 that results in a change in the amino acid sequence of the PR010282 or P11019578 as compared with the native sequence PI1010282 or P11019578.
Optionally the variation is by substitution of at least one amino acid with any other amino acid in one or more of the domains of the PR010282 or P11019578. Guidance in determining which amino acid residue may be inserted, substituted or deleted without adversely affecting the desired activity may be found by comparing the sequence of the PRO10282 or P11019578 with that of homologous known protein molecules and minimizing the number of amino acid sequence changes made in regions of high homology. Amino acid substitutions can be the result of replacing one amino acid with another amino acid having similar structural and/or chemical properties, such as the replacement of a leucine with a serine, i.e., conservative amino acid replacements. Insertions or deletions may optionally be in the range of about 1 to 5 amino acids.
The variation allowed may be determined by systematically making insertions, deletions or substitutions of amino acids in the sequence and testing the resulting variants for activity exhibited by the full-length or mature native sequence.
P1I010282 polypeptide fragments are provided herein. Such fragments may be truncated at the N-terminus or C-terminus, or may lack internal residues, for example, when compared with a full-length native protein. Certain fragments lack amino acid residues that are not essential for a desired biological activity of the PRO10282 or P11019578 polypeptide.
PR010282 or PR019578 fragments may be prepared by any of a number of conventional techniques. Desired peptide fragments may be chemically synthesized. An alternative approach involves generating P11010282 or P11019578 fragments by enzymatic digestion, e.g., by treating the protein with an enzyme known to cleave proteins at sites defined by particular amino acid residues, or by digesting the DNA with suitable restriction enzymes and isolating the desired fragment. Yet another suitable technique involves isolating and amplifying a DNA fragment encoding a desired polypeptide fragment, by polymerase chain reaction (PCR). Oligonucleotides that define the desired termini of the DNA fragment are employed at the 5' and 3' primers in the PCR. Preferably, P11010282 or P11019578 polypeptide fragments share at least one biological and/or immunological activity with the native PR010282 or PR019578 polypeptide shown in Figure 2 (SEQ.
ID NO:2).
In particular embodiments, conservative substitutions of interest are shown in Table 1 under the heading of preferred substitutions. If such substitutions result in a change in biological activity, then more substantial changes, 2 0 denominated exemplary substitutions in Table 1, or as further described below in reference to amino acid classes, are introduced and the products screened.
Table 1 Original Exemplary Preferred Residue Substitutions Substitutions Ala (A) val; leu; ile val Arg (R) lys; gln; asn lys Asn (N) gln; his; lys; arg gln Asp (0) glu glu 3 5 Cys (C) ser ser Gin (G) asn asn Glu (E) asp asp Gly (G) pro; ala ala His (H) asn; gin; lys; arg arg 4 0 Ile (I) leu; val; met; ala; phe;
norleucine leu Leu (L) norleucine; ile; valmet; ale; phe lie Lys (K) arg; gin; asn arg Met (M) leu; phe; lie leu Phe (F) leu; vat; lie; ale; tyr leu Pro (P) ala ala Ser (S) thr thr Thr (T) ser ser Trp (W) tyr; phe tyr Tyr (Y) trp; phe; thr; ser phe Val (V) lie; leu; met; phe;a1a; norleucine leu Substantial modifications in function or immunological identity of the PR010282 polypeptide are accomplished by selecting substitutions that differ significantly in their effect on maintaining (a) the structure of the polypeptide backbone in the area of the substitution, for example, as a sheet or helical conformation, (b) the charge or hydrophobicity of the molecule at the target site, or (c) the bulk of the side chain.
Naturally occurring residues are divided into groups based on common side-chain properties:
(1) hydrophobic: norleucine, met, ala, val, leu, ile;
(2) neutral hydrophilic: cys, ser, thr;
(3) acidic: asp, glu;
2 0 (4) basic: asn, gin, his, lys, arg;
(5) residues that influence chain orientation: gly, pro; and (6) aromatic: trp, tyr, phe.
Non-conservative substitutions will entail exchanging a member of one of these classes for another class. Such 2 5 substituted residues also may be introduced into the conservative substitution sites or, more preferably, into the remaining (non-conserved) sites.
The variations can be made using methods known in the art such as oligonucleotide-mediated (site-directed) mutagenesis, alanine scanning, and PCR mutagenesis. Site-directed mutagenesis [Carter et al., Nucl. Acids Res., 13:4331 (1986); Zoller et al., Nucl. Acids Res., 10:6487 (1987)1, cassette mutagenesis [Wells et at., Gene, 34:315 (1985)1, 30 restriction selection mutagenesis [Wells et at., Philos. Trans. R. Soc.
London SerA, 317:415 (1986)1 or other known techniques can be performed on the cloned DNA to produce the P11010282 variant DNA.
Scanning amino acid analysis can also be employed to identify one or more amino acids along a contiguous sequence. Among the preferred scanning amino acids are relatively small, neutral amino acids. Such amino acids include alanine, glycine, serine, and cysteine. Alanine is typically a preferred scanning amino acid among this group because it 3 5 eliminates the side-chain beyond the beta-carbon and is less likely to alter the main-chain conformation of the variant [Cunningham and Wells, Science, 244: 1081-1085 (1989)]. Alanine is also typically preferred because it is the most common amino acid. Further, it is frequently found in both buried and exposed positions [Creighton, The Proteins, (W.-I.
Freeman & Co., N.Y.); Chothia, J. Mol. Biol., 150:1 (1976)1. If alanine substitution does not yield adequate amounts of variant, an isoteric amino acid can be used.
C. Modifications of native and variant PR010282 (Stra6) Covalent modifications of PR010282 and PR010282 variants, including PR019578, are included within the scope of this invention. One type of covalent modification includes reacting targeted amino acid residues of a PR010282.
polypeptide with an organic derivatizing agent that is capable of reacting with selected side chains or the N- or C- terminal residues of the PR010282. Derivatization with bifunctional agents is useful, for instance, for crosslinking PR010282 to a water-insoluble support matrix or surface for use in the method for purifying anti-PRO10282 antibodies, and vice-versa.
Commonly used crosslinking agents include, e.g., 1,1-bis(diazoacetyI)-2-phenylethane, glutaraldehyde, N-hydroxysuccinimide 1 0 esters, for example, esters with 4-azidosalicylic acid, homobifunctional innidoesters, including disuccinimidyl esters such as 3,3'-dithiobis(succinimidylpropionate), bifunctional maleimides such as bis-N-maleimido-1,8-octane and agents such as methyl-3-[(p-azidophenyl)dithio]propioimidate.
Other modifications include deamidation of glutaminyl and asparaginyl residues to the corresponding glutamyl and aspartyl residues, respectively, hydroxylation of proline and lysine, phosphorylation of hydroxyl groups of seryl or threonyl residues, methylation of the -amino groups of lysine, arginine, and histidine side chains [I.E. Creighton, Proteins:
Structure and Molecular Properties, W.H. Freeman & Co., San Francisco, pp. 79-86 (1983)], acetylation of the N-terminal amine, and amidation of any C-terminal carboxyl group.
Another type of covalent modification of the PRO10282 polypeptides included within the scope of this invention comprises altering the native glycosylation pattern of the polypeptide. Native sequence PR010282 and PR019578 polypeptides contain an N-linked glycosylation site at positions 8-12 of their amino acid sequence. "Altering the native glycosylation pattern" is intended for purposes herein to mean deleting one or more carbohydrate moieties found in a native sequence PR010282 or PR019578 (either by removing the underlying glycosylation site or by deleting the glycosylation by chemical and/or enzymatic means), and/or adding one or more glycosylation sites that are not present in the native sequence P11010282 or PRO19578. In addition, the phrase includes qualitative changes in the glycosylation of the native 2 5 proteins, involving a change in the nature and proportions of the various carbohydrate moieties present.
Addition of glycosylation sites to a native or variant PR010282 polypeptide may be accomplished by altering the amino acid sequence. The alteration may be made, for example, by the addition of, or substitution by, one or more serine or threonine residues to the native sequence PR010282 (for 0-linked glycosylation sites). The P11010282 amino acid sequence may optionally be altered through changes at the DNA level, particularly by mutating the DNA encoding the PRO10282 polypeptide at preselected bases such that codons are generated that will translate into the desired amino acids.
Another means of increasing the number of carbohydrate moieties on the PRO10282 polypeptide is by chemical or enzymatic coupling of glycosides to the polypeptide. Such methods are described in the art, e.g., in WO 87/05330 published 11 September 1987, and in Aplin and Wriston, CRC Crit. Rev.
Biochem., pp. 259-306 (1981).
3 5 Removal of carbohydrate moieties present on the PRO10282 polypeptide may be accomplished chemically or enzymatically or by mutational substitution of codons encoding for amino acid residues that serve as targets for glycosylation. Chemical deglycosylation techniques are known in the art and described, for instance, by Hakimuddin, et al., Arch. Biochem. Biophys., 259:52 (1987) and by Edge et al., Anal.
Biochem., 118:131 (1981). Enzymatic cleavage of carbohydrate moieties on polypeptides can be achieved by the use of a variety of endo- and exo-glycosidases as described by Thotakura et al., Meth. Enzvmol., 138:350 (1987).
Another type of covalent modification of PR010282 comprises linking the PRO10282 polypeptide to one of a variety of nonproteinaceous polymers, e.g., polyethylene glycol (PEG), polypropylene glycol, or polyoxyalkylenes, in the manner set forth in U.S. Patent Nos. 4,640,835; 4,496,689; 4,301,144;
4,670,417; 4,791,192 or 4,179,337.
The PR010282 polypeptides of the present invention may also be modified in a way to form a chimeric molecule comprising a PRO10282 fused to another, heterologous polypeptide or amino acid sequence.
In one embodiment, such a chimeric molecule comprises a fusion of the PR010282 with a tag polypeptide which provides an epitope to which an anti-tag antibody can selectively bind. The epitope tag is generally placed at the amino-or carboxyl- terminus of the PR010282. The presence of such epitope-tagged forms of the PR010282 can be detected using an antibody against the tag polypeptide. Also, provision of the epitope tag enables the PR010282 to be readily purified by affinity purification using an anti-tag antibody or another type of affinity matrix that binds to the epitope tag.
Various tag polypeptides and their respective antibodies are well known in the art. Examples include poly-histidine (poly-his) or poly-histidine-glycine (poly-his-gly) tags; the flu HA tag polypeptide and its antibody 12CA5 [Field et al., Mol. Cell. Biol., 8:2159-2165 (1988)1; the c-myc tag and the 8F9, 3C7, 6E10, 64, B7 and 9E10 antibodies thereto [Evan et al., Molecular and Cellular Biology, 5:3610.3616(1985)]; and the Herpes Simplex virus glycoprotein D (gD) tag and its antibody [Paborsky et al., Protein Engineering, 3(6):547-553 (1990)]. Other tag polypeptides include the Flag-peptide [Hopp et al., BioTechnology, 6:1204-1210 (1988)]; the KT3 epitope peptide [Martinet al., Science, 255:192-194 (1992)1; an -tubulin epitope peptide [Skinner et al., J. Biol. Chem., 266:15163-15166 (1991)]; and the T7 gene 10 protein peptide tag [Lutz-Freyermuth et al., Proc. Natl. Acad. Sci. USA, 87:6393-6397 (1990)].
In an alternative embodiment, the chimeric molecule may comprise a fusion of the PR010282 with an immunoglobulin or a particular region of an immunoglobulin. For a bivalent form of the chimeric molecule (also referred to as an "immonoadhesin"), such a fusion could be to the Fc region of an IgG
molecule. The Ig fusions preferably include the substitution of a soluble (transmembrane domain deleted or inactivated) form of a PRO10282 polypeptide in place of at least one variable region within an Ig molecule. In a particularly preferred embodiment, the immunoglobulin fusion includes the hinge, CH2 and CH3, or the hinge, CH1, CH2 and CH3 regions of an IgG1 molecule. For the production of immunoglobulin fusions see also US Patent No. 5,428,130 issued June 27, 1995.
D. Preparation of naitge or variant PR010282 (Stra6) polypeptide The description below relates primarily to production of native sequence and variant PR010282 (Stra6) polypeptides, specifically including PR019578, by culturing cells transformed or transfected with a vector containing PRO10282 nucleic acid. It is, of course, contemplated that alternative methods, which are well known in the art, may be employed to prepare PRO10282. For instance, the PRO10282 sequence, or portions thereof, may be produced by direct peptide synthesis using solid-phase techniques [see, e.g., Stewart et al., Solid-Phase Peptide Synthesis, W.H. Freeman Co., San Francisco, CA (1969); Merrifield, J. Am. Chem. Soc., 85:2149-2154 (1963)1.
In vitro protein synthesis may be performed using manual techniques or by automation. Automated synthesis may be accomplished, for instance, using an Applied Biosystems Peptide Synthesizer (Foster City, CA) using manufacturer's instructions. Various portions of the PR010282 may be chemically synthesized separately and combined using chemical or enzymatic methods to produce the full-length PRO10282.
1. Isolation of DNA Encoding PR010282 (Stra6) DNA encoding PR010282 may be obtained from a cDNA library prepared from tissue believed to possess the PR010282 nnRNA and to express it at a detectable level. Accordingly, human PR010282 DNA can be conveniently obtained from a cDNA library prepared from human tissue, such as described in the Examples. The PRO10282-encoding gene may also be obtained from a genomic library or by known synthetic procedures (e.g., automated nucleic acid synthesis).
Libraries can be screened with probes (such as antibodies to the PRO10282 or oligonucleotides of at least about 20-80 bases) designed to identify the gene of interest or the protein encoded by it. Screening the cDNA or genomic library with the selected probe may be conducted using standard procedures, such as described in Sambrook et al., Molecular Cloning: A Laboratory Manual (New York: Cold Spring Harbor Laboratory Press, 1989). An alternative means to isolate the gene encoding PR010282 is to use PCR methodology [Sambrook et al., supra;
Dieffenbach et al., PCR Primer: A
2 0 Laboratory Manual (Cold Spring Harbor Laboratory Press, 1995)].
The Examples below describe techniques for screening a cDNA library. The oligonucleotide sequences selected as probes should be of sufficient length and sufficiently unambiguous that false positives are minimized. The oligonucleotide is preferably labeled such that it can be detected upon hybridization to DNA in the library being screened.
Methods of labeling are well known in the art, and include the use of radiolabels like 32P-labeled ATP, biotinylation or enzyme labeling. Hybridization conditions, including moderate stringency and high stringency, are provided in Sambrook et al., supra.
Sequences identified in such library screening methods can be compared and aligned to other known sequences deposited and available in public databases such as GenBank or other private sequence databases. Sequence identity (at either the amino acid or nucleotide level) within defined regions of the molecule or across the full-length sequence can be 3 0 determined using methods known in the art and as described herein.
Nucleic acid having protein coding sequence may be obtained by screening selected cDNA or genomic libraries using the deduced amino acid sequence disclosed herein for the first time, and, if necessary, using conventional primer extension procedures as described in Sambrook et al., supra, to detect precursors and processing intermediates of mRNA
that may not have been reverse-transcribed into cDNA.

2. Selection and Transformation of Host Cells Host cells are transfected or transformed with expression or cloning vectors described herein for PR010282 production and cultured in conventional nutrient media modified as appropriate for inducing promoters, selecting transformants, or amplifying the genes encoding the desired sequences. The culture conditions, such as media, temperature, pH and the like, can be selected by the skilled artisan without undue experimentation. In general, principles, protocols, and practical techniques for maximizing the productivity of cell cultures can be found in Mammalian Cell Biotechnology: a Practical Approach, M. Butler, ed. (IRL Press, 1991) and Sambrook et al., supra.
Methods of eukaryotic cell transfection and prokaryotic cell transformation are known to the ordinarily skilled artisan, for example, CaCl2, CaPO4, liposome-mediated and electroporation.
Depending on the host cell used, transformation is performed using standard techniques appropriate to such cells. The calcium treatment employing calcium chloride, as described in Sambrook et al., supra, or electroporation is generally used for prokaryotes. Infection with Agrobacterium tumefaciens is used for transformation of certain plant cells, as described by Shaw et al., Gene, 23:315 (1983) and WO 89/05859 published 29 June 1989. For mammalian cells without such cell walls, the calcium phosphate precipitation method of Graham and van der Eb, Virology, 52:456-457 (1978) can be employed. General aspects of mammalian cell host system transfections have been described in U.S. Patent No. 4,399,216. Transformations into yeast are typically carried out according to the method of Van Solingen et al., J.
Bact., 130:946 (1977) and Hsiao et al., Proc.
Natl. Acad. Sci. (USA), 76:3829 (1979). However, other methods for introducing DNA into cells, such as by nuclear nnicroinjection, electroporation, bacterial protoplast fusion with intact cells, or polycations, e.g., polybrene, polyornithine, may also be used. For various techniques for transforming mammalian cells, see Keown et al., Methods in Enzymology, 185:527-537 (1990) and Mansour et al., Nature, 336:348-352 (1988).
Suitable host cells for cloning or expressing the DNA in the vectors herein include prokaryote, yeast, or higher eukaryote cells. Suitable prokaryotes include but are not limited to eubacteria, such as Gram-negative or Gram-positive organisms, for example, Enterobacteriaceae such as E. coli. Various E. coli strains are publicly available, such as E. coli K12 strain MM294 (ATCC 31,446); E. coli X1776 (ATCC 31,537); E. coli strain W3110 (ATCC 27,325) and K5 772 (ATCC
2 5 53,635). Other suitable prokaryotic host cells include Enterobacteriaceae such as Escherichia, e.g., E. coli, Enterobacter, Erwinia, Klebsiella, Proteus, Salmonella, e.g., Salmonella typhimurium, Serratia, e.g., Serratia marcescans, and Shigella, as well as Bacilli such as B. subtilis and B. licheniformis (e.g., B.
licheniformis 41P disclosed in DD 266,710 published 12 April 1989), Pseudomonas such as P. aeruginosa, and Streptomyces. These examples are illustrative rather than limiting.
Strain W3110 is one particularly preferred host or parent host because it is a common host strain for recombinant DNA
3 0 product fermentations. Preferably, the host cell secretes minimal amounts of proteolytic enzymes. For example, strain W3110 may be modified to effect a genetic mutation in the genes encoding proteins endogenous to the host, with examples of such hosts including E. coli W3110 strain 1A2, which has the complete genotype tonA; E. coli W3110 strain 9E4, which has the complete genotype tonA ptr3; E. coli W3110 strain 27C7 (ATCC 55,244), which has the complete genotype tonA
ptr3 phoA E15 (argFlac)169 degP ompT kanr; E. coli W3110 strain 3706, which has the complete genotype tonA ptr3 35 phoA E15 (argF-lac)169 degP ompT rbs7 ilvG kanr; E. coli W3110 strain 4064, which is strain 3706 with a non-kanamycin resistant degP deletion mutation; and an E. coli strain having mutant periplasmic protease disclosed in U.S.
Patent No. 4,946,783 issued 7 August 1990. Alternatively, in vitro methods of cloning, e.g., PCR or other nucleic acid polymerase reactions, are suitable.
In addition to prokaryotes, eukaryotic microbes such as filamentous fungi or yeast are suitable cloning or expression hosts for PR010282-encoding vectors. Saccharomyces cerevisiae is a commonly used lower eukaryotic host microorganism. Others include Schizosaccharomyces pombe (Beach and Nurse, Nature, 290: 140 [1981]; EP 139,383 published 2 May 1985); Kluyveromyces hosts (U.S. Patent No. 4,943,529; Fleer et al., BiolTechnoloey, 9:968-975 (1991)) such as, e.g., K. lactis (MW98-8C, CBS683, CBS4574; Louvencourt et al., J.
Bacterial., 154(2): 737-1742 [19831), K.
fragilis (ATCC 12,424), K. bulgaricus (ATCC 16,045), K. wickeramii (ATCC
24,178); K. waltii (ATCC 56,500), K.
drosophilarunn (ATCC 36,906; Van den Berg et al., Bio/Technology, 8:135 (1990)), K. thermotolerans, and K. marxianus;
yarrowia (EP 402,226); Pichia pastoris (EP 183,070; Sreekrishna et al., J.
Basic Microbial., 28:265-278 [19881); Candida;
Trichoderma reesia (EP 244,234); Neurospora crassa (Case et al., Proc. Natl.
Acad. Sci. USA, 76:5259-5263 [1979]);
Schwanniomyces such as Schwanniomyces occidentalis (EP 394,538 published 31 October 1990); and filamentous fungi such as, e.g., Neurospora; Penicilliunn, Tolypocladium (WO 91/00357 published 10 January 1991), and Aspergillus hosts such as A. nidulans (Ballance et al., Biochem. Biophys. Res. Commun., 112:284-289 [1983]; Tilburn et al., Gene, 26:205-221 [1983]; Yelton et al., Proc. Natl. Acad. Sci. USA, 81: 1470-1474 [19841) and A. niger (Kelly and Hynes, EMBO J., 4:475-479 [1985]). Methylotropic yeasts are suitable herein and include, but are not limited to, yeast capable of growth on methanol selected from the genera consisting of Hansenula, Candida, Kloeckera, Pichia, Saccharomyces, Torulopsis, and Rhodotorula. A list of specific species that are exemplary of this class of yeasts may be found in C. Anthony, The Biochemistry of Methylotrophs, 269 (1982).
Suitable host cells for the expression of glycosylated PR010282 are derived from multicellular organisms.
Examples of invertebrate cells include insect cells such as Drosophila S2 and Spodoptera Sf9, as well as plant cells.
Examples of useful mammalian host cell lines include Chinese hamster ovary (CHO) and COS cells. More specific examples include monkey kidney CV1 line transformed by SV40 (COS-7, ATCC CRL 1651);
human embryonic kidney line (293 or 293 cells subcloned for growth in suspension culture, Graham et al., J. Gen Viral., 36:59 (1977)); Chinese hamster ovary cells/-DHFR (CHO, Urlaub and Chasin, Proc. Natl. Acad. Sci. USA, 77:4216 (1980));
mouse sertoli cells (TM4, Mather, Biol.
Reprod., 23:243-251 (1980)); human lung cells (W138, ATCC CCL 75); human liver cells (Hep 02, HB 8065); and mouse mammary tumor (MMT 060562, ATCC CCL51). The selection of the appropriate host cell is deemed to be within the skill in the art.
3. Selection and Use of a Replicable Vector The nucleic acid (e.g., cDNA or genomic DNA) encoding a PR010282 may be inserted into a replicable vector for cloning (amplification of the DNA) or for expression. Various vectors are publicly available. The vector may, for example, be in the form of a plasmid, cosmid, viral particle, or phage. The appropriate nucleic acid sequence may be inserted into 3 5 the vector by a variety of procedures. In general, DNA is inserted into an appropriate restriction endonuclease site(s) using techniques known in the art. Vector components generally include, but are not limited to, one or more of a signal sequence, an origin of replication, one or more marker genes, an enhancer element, a promoter, and a transcription termination sequence. Construction of suitable vectors containing one or more of these components employs standard ligation techniques which are known to the skilled artisan.
The PR010282 may be produced recombinantly not only directly, but also as a fusion polypeptide with a heterologous polypeptide, which may be a signal sequence or other polypeptide having a specific cleavage site at the N-terminus of the mature protein or polypeptide. In general, the signal sequence may be a component of the vector, or it may be a part of the PRO10282-encoding DNA that is inserted into the vector. The signal sequence may be a prokaryotic signal sequence selected, for example, from the group of the alkaline phosphatase, penicillinase, Ipp, or heat-stable enterotoxin II leaders. For yeast secretion the signal sequence may be, e.g., the yeast invertase leader, alpha factor leader (including Saccharomyces and Kluyveromyces -factor leaders, the latter described in U.S.
Patent No. 5,010,182), or acid phosphatase leader, the C. albicans glucoamylase leader (EP 362,179 published 4 April 1990), or the signal described in WO 90113646 published 15 November 1990. In mammalian cell expression, mammalian signal sequences may be used to direct secretion of the protein, such as signal sequences from secreted polypeptides of the same or related species, as well as viral secretory leaders.
Both expression and cloning vectors contain a nucleic acid sequence that enables the vector to replicate in one or more selected host cells. Such sequences are well known for a variety of bacteria, yeast, and viruses. The origin of replication from the plasmid pBR322 is suitable for most Gram-negative bacteria, the 2 plasmid origin is suitable for yeast, and various viral origins (SV40, polyoma, adenovirus, VSV or BPV) are useful for cloning vectors in mammalian cells.
2 0 Expression and cloning vectors will typically contain a selection gene, also termed a selectable marker. Typical selection genes encode proteins that (a) confer resistance to antibiotics or other toxins, e.g., annpicillin, neomycin, methotrexate, or tetracycline, (b) complement auxotrophic deficiencies, or (c) supply critical nutrients not available from complex media, e.g., the gene encoding D-alanine racemase for Bacilli.
An example of suitable selectable markers for mammalian cells are those that enable the identification of cells competent to take up the PRO10282-encoding nucleic acid, such as DHFR or thymidine kinase. An appropriate host cell when wild-type DHFR is employed is the CHO cell line deficient in DHFR
activity, prepared and propagated as described by Urlaub et al., Proc. Natl. Acad. Sci. USA, 77:4216 (1980). A suitable selection gene for use in yeast is the trp1 gene present in the yeast plasmid YRp7 [Stinchcomb et al., Nature, 282:39 (1979);
Kingsman et al., Gene, 7:141 (1979);
Tschemper et al., Gene, 10:157 (1980)]. The trp1 gene provides a selection marker for a mutant strain of yeast lacking the ability to grow in tryptophan, for example, ATCC No. 44076 or PEP4-1 [Jones, Genetics, 85:12 (1977)1.
Expression and cloning vectors usually contain a promoter operably linked to the PRO10282-encoding nucleic acid sequence to direct mRNA synthesis. Promoters recognized by a variety of potential host cells are well known.
Promoters suitable for use with prokaryotic hosts include the -lactamase and lactose promoter systems [Chang et al., Nature, 275:615(1978); Goeddel et al., Nature, 281:544 (1979)], alkaline phosphatase, a tryptophan (trp) promoter system [Goeddel, Nucleic Acids Res., 8:4057 (1980); EP 36,7761, and hybrid promoters such as the tac promoter [deBoer et al., Proc. Natl. Acad. Sci. USA, 80:21-25 (1983)]. Promoters for use in bacterial systems also will contain a Shine-Dalgarno (S.D.) sequence operably linked to the DNA encoding PR010282.
Examples of suitable promoting sequences for use with yeast hosts include the promoters for 3-phosphoglycerato kinase [Hitzeman et al., J. Biol. Chem., 255:2073 (1980)1 or other glycolytic enzymes [Hess et al., J. Adv. Enzyme Reg., 7:149 (1968); Holland, Biochemistry, 17:4900 (1978)], such as enolase, glyceraldehyde-3-phosphate dehydrogenase, hexokinase, pyruvate decarboxylase, phosphofructokinase, glucose-6-phosphate isomerase, 3-phosphoglycerate mutase, pyruvate kinase, triosephosphate isomerase, phosphoglucose isomerase, and glucokinase.
Other yeast promoters, which are inducible promoters having the additional advantage of transcription controlled by growth conditions, are the promoter regions for alcohol dehydrogenase 2, isocytochrome C, acid phosphatase, 1 0 degradative enzymes associated with nitrogen metabolism, metallothionein, glyceraldehyde-3-phosphate dehydrogenase, and enzymes responsible for maltose and galactose utilization. Suitable vectors and promoters for use in yeast expression are further described in EP 73,657.
PRO10282 transcription from vectors in mammalian host cells is controlled, for example, by promoters obtained from the genomes of viruses such as polyoma virus, fowlpox virus (UK 2,211,504 published 5 July 1989), adenovirus (such as Adenovirus 2), bovine papillonna virus, avian sarcoma virus, cytomegalovirus, a retrovirus, hepatitis-B virus and Simian Virus 40 (SV40), from heterologous mammalian promoters, e.g., the actin promoter or an immunoglobulin promoter, and from heat-shock promoters, provided such promoters are compatible with the host cell systems.
Transcription of a DNA encoding the PR010282 by higher eukaryotes may be increased by inserting an enhancer sequence into the vector. Enhancers are cis-acting elements of DNA, usually about from 10 to 300 bp, that act on a 2 0 promoter to increase its transcription. Many enhancer sequences are now known from mammalian genes (globin, elastase, albumin, -fetoprotein, and insulin). Typically, however, one will use an enhancer from a eukaryotic cell virus. Examples include the SV40 enhancer on the late side of the replication origin (bp 100-270), the cytomogalovirus early promoter enhancer, the polyoma enhancer on the late side of the replication origin, and adenovirus enhancers. The enhancer may be spliced into the vector at a position 5' or 3' to the PRO10282 coding sequence, but is preferably located at a site 5' 2 5 from the promoter.
Expression vectors used in eukaryotic host cells (yeast, fungi, insect, plant, animal, human, or nucleated cells from other multicellular organisms) will also contain sequences necessary for the termination of transcription and for stabilizing the mRNA. Such sequences are commonly available from the 5' and, occasionally 3', untranslated regions of eukaryotic or viral DNAs or cDNAs. These regions contain nucleotide segments transcribed as polyadenylated fragments 30 in the untranslated portion of the mRNA encoding PR010282.
Still other methods, vectors, and host cells suitable for adaptation to the synthesis of PR010282 in recombinant vertebrate cell culture are described in Gething at at., Nature, 293:620-625 (1981); Mantei et al., Nature, 281:40-46 (1979); EP 117,060; and EP 117,058.
3 5 4. Detecting Gene Amplification/Expression =

Gene amplification and/or expression may be measured in a sample directly, for example, by conventional Southern blotting, Northern blotting to quantitate the transcription of mRNA
[Thomas, Proc. Natl. Acad. Sci. USA, 77:5201-5205 (1980)], dot blotting (DNA analysis), or in situ hybridization, using an appropriately labeled probe, based on the sequences provided herein. Alternatively, antibodies may be employed that can recognize specific duplexes, including DNA duplexes, RNA duplexes, and DNA-RNA hybrid duplexes or DNA-protein duplexes. The antibodies in turn may be labeled and the assay may be carried out where the duplex is bound to a surface, so that upon the formation of duplex on the surface, the presence of antibody bound to the duplex can be detected.
Gene expression, alternatively, may be measured by immunological methods, such as immunohistochemical staining of cells or tissue sections and assay of cell culture or body fluids, to quantitate directly the expression of gene product. Antibodies useful for immunohistochemical staining and/or assay of sample fluids may be either monoclonal or polyclonal, and may be prepared in any mammal. Conveniently, the antibodies may be prepared against a native sequence PR010282 polypeptide or against a synthetic peptide based on the DNA sequences provided herein or against exogenous sequence fused to PR010282 DNA and encoding a specific antibody epitope.
5. Purification of Polypeptide Forms of PRO10282 may be recovered from culture medium or from host cell lysates. If membrane-bound, it can be released from the membrane using a suitable detergent solution (e.g. Triton-X 100) or by enzymatic cleavage. Cells employed in expression of PRO10282 can be disrupted by various physical or chemical means, such as freeze-thaw cycling, sonication, mechanical disruption, or cell lysing agents.
2 0 It may be desired to purify PRO10282 from recombinant cell proteins or polypeptides. The following procedures are exemplary of suitable purification procedures: by fractionation on an ion-exchange column; ethanol precipitation; reverse phase HPLC; chromatography on silica or on a cation-exchange resin such as DEAE; chromatofocusing; SDS-PAGE;
ammonium sulfate precipitation; gel filtration using, for example, Sephadex G-75; protein A Sepharose columns to remove contaminants such as IgG; and metal chelating columns to bind epitope-tagged forms of the PRO10282. Various methods 2 5 of protein purification may be employed and such methods are known in the art and described for example in Deutscher, Methods in Enzymolouy, 182 (1990); Scopes, Protein Purification: Principles and Practice, Springer-Verlag, New York (1982). The purification step(s) selected will depend, for example, on the nature of the production process used and the particular PRO10282 produced.

E. Amplification of Genes Encoding the PR010282 (Stra6) Polypeptides in Tumor Tissues and Cell lines In one aspect, the present invention is based on the identification and characterization of genes that are amplified in certain cancer cells.
The genome of prokaryotic and eukaryotic organisms is subjected to two seemingly conflicting requirements.
One is the preservation and propagation of DNA as the genetic information in its original form, to guarantee stable inheritance through multiple generations. On the other hand, cells or organisms must be able to adapt to lasting environmental changes. The adaptive mechanisms can include qualitative or quantitative modifications of the genetic material. Qualitative modifications include DNA mutations, in which coding sequences are altered resulting in a structurally and/or functionally different protein. Gene amplification is a quantitative modification, whereby the actual number of complete coding sequence, i.e., a gene, increases, leading to an increased number of available templates for transcription, an increased number of translatable transcripts, and, ultimately, to an increased abundance of the protein encoded by the amplified gene.
The phenomenon of gene amplification and its underlying mechanisms have been investigated in vitro in several prokaryotic and eukaryotic culture systems. The best-characterized example of gene amplification involves the culture of eukaryotic cells in medium containing variable concentrations of the cytotoxic drug methotrexate (MTX). MTX is a folic acid analogue and interferes with DNA synthesis by blocking the enzyme dihydrofolate reductase (DHFR). During the initial exposure to low concentrations of MTX most cells ( > 99.9%) will die. A small number of cells survive, and are capable of growing in increasing concentrations of MTX by producing large amounts of DHFR-RNA and protein. The basis of this overproduction is the amplification of the single DHFR gene. The additional copies of the gene are found as extrachromosomal copies in the form of small, supernumerary chromosomes (double minutes) or as integrated chromosomal copies.
2 5 Gene amplification is most commonly encountered in the development of resistance to cytotoxic drugs (antibiotics for bacteria and chemotherapeutic agents for eukaryotic cells) and neoplastic transformation. Transformation of a eukaryotic cell as a spontaneous event or due to a viral or chemical/environmental insult is typically associated with changes in the genetic material of that cell. One of the most common genetic changes observed in human malignancies are mutations of the p53 protein. p53 controls the transition of cells from the stationary (G1) to the replicative (S) phase 3 0 and prevents this transition in the presence of DNA damage. In other words, one of the main consequences of disabling p53 mutations is the accumulation and propagation of DNA damage, i.e., genetic changes. Common types of genetic changes in neoplastic cells are, in addition to point mutations, amplifications and gross, structural alterations, such as translocations.
The amplification of DNA sequences may indicate a specific functional requirement as illustrated in the DHFR

experimental system. Therefore, the amplification of certain oncogenes in malignancies points toward a causative role of these genes in the process of malignant transformation and maintenance of the transformed phenotype. This hypothesis has gained support in recent studies. For example, the bc1-2 protein was found to be amplified in certain types of non-Hodgkin's lymphoma. This protein inhibits apoptosis and leads to the progressive accumulation of neoplastic cells.
Members of the gene family of growth factor receptors have been found to be amplified in various types of cancers suggesting that overexpression of these receptors may make neoplastic cells less susceptible to limiting amounts of available growth factor. Examples include the amplification of the androgen receptor in recurrent prostate cancer during androgen deprivation therapy and the amplification of the growth factor receptor homologue ERB2 in breast cancer. Lastly, genes involved in intracellular signaling and control of cell cycle progression can undergo amplification during malignant transformation. This is illustrated by the amplification of the bell and ras genes in various epithelial and lymphoid neoplasms.
These earlier studies illustrate the feasibility of identifying amplified DNA
sequences in neoplasms, because this approach can identify genes important for malignant transformation. The case of ERB2 also demonstrates the feasibility from a therapeutic standpoint, since transforming proteins may represent novel and specific targets for tumor therapy.
Several different techniques can be used to demonstrate amplified genomic sequences. Classical cytogenetic analysis of chromosome spreads prepared from cancer cells is adequate to identify gross structural alterations, such as translocations, deletions and inversions. Amplified genomic regions can only be visualized, if they involve large regions with high copy numbers or are present as extrachromosomal material. While cytogenetics was the first technique to demonstrate the consistent association of specific chromosomal changes with particular neoplasms, it is inadequate for the identification and isolation of manageable DNA sequences. The more recently developed technique of comparative 2 0 genomic hybridization (CGH) has illustrated the widespread phenomenon of genomic amplification in neoplasms. Tumor and normal DNA are hybridized simultaneously onto metaphases of normal cells and the entire genome can be screened by image analysis for DNA sequences that are present in the tumor at an increased frequency. (WO 93118,186; Gray et al., Radiation lies., 137:275-289 [1994]). As a screening method, this type of analysis has revealed a large number of recurring amplicons (a stretch of amplified DNA) in a variety of human neoplasms. Although CGH is more sensitive than 2 5 classical cytogenetic analysis in identifying amplified stretches of DNA, it does not allow a rapid identification and isolation of coding sequences within the amplicon by standard molecular genetic techniques.
The most sensitive methods to detect gene amplification are polymerase chain reaction (PCR)-based assays.
These assays utilize very small amount of tumor DNA as starting material, are exquisitely sensitive, provide DNA that is amenable to further analysis, such as sequencing and are suitable for high-volume throughput analysis.
3 0 The above-mentioned assays are not mutually exclusive, but are frequently used in combination to identify amplifications in neoplasms. While cytogenetic analysis and CGH represent screening methods to survey the entire genome for amplified regions, PCR-based assays are most suitable for the final identification of coding sequences, i.e., genes in amplified regions.
According to the present invention, such genes have been identified by quantitative PCR (S. Gelmini et al., Clin.
35 Chem. 43:752 [1997]), by comparing RNA from a variaty of primari tumors, including breast lung, colon, prostate, brain, liver, kidney, pancrease, spleen, thymus, testis, ovary, uterus, etc., tumor, or tumor cell lines, with pooled DNA from healthy donors. Quantitative PCR was performed using a TaqMan instrument (ABI). Gene-specific primers and fluorogenic probes were designed based upon the coding sequences of the DNAs.
Human lung carcinoma cell lines include A549 (3RCC768), Calu-1 (SRCC769), Calu-6 (8RCC770), H157 (SRCC771), H441 (SRCC772), H460 (8RCC773), SKMES-1 (SRCC774), SW900 (8RCC775), H522 (SRCC832),and H810 (SRCC833), all available from ATCC. Primary human lung tumor cells usually derive from adenocarcinomas, squamous cell carcinomas, large cell carcinomas, non-small cell carcinomas, small cell carcinomas, and broncho alveolar carcinomas, and include, for example, 8RCC724 (adenocarcinoma, abbreviated as "AdenoCa")(LT1), SRCC725 (squamous cell carcinoma, abbreviated as "SqCCa)(LT1a), SRCC726 (adenocarcinoma)(LT2), SRCC727 (adenocarcinoma)(1T3), 3RCC728 (adenocarcinoma)(L14), SRCC729 (squamous cell carcinoma)(LT6), SHCC730 (adenolsquamous cell carcinoma)(L17), SRCC731 (adenocarcinoma)(LT9), SRCC732 (squamous cell carcinoma)(LT10), SRCC733 (squamous cell carcinoma)(LT11), 8RCC734 (adenocarcinoma)(LT12), SRCC735 (adenolsquannous cell carcinoma)(LT13), 3RCC736 (squamous cell carcinoma)(LT15), SRCC737 (squamous cell carcinoma)(1T16), SRCC738 (squamous cell carcinoma)(LT17), SRCC739 (squamous cell carcinoma)(LT18), SRCC740 (squamous cell carcinoma)(1T19), SRCC741 (lung cell carcinoma, abbreviated as "LCCa")(LT21), SRCC811 (adenocarcinoma)(LT22), SRCC825 (adenocarcinoma)(LT8), SRCC886 (adenocarcinoma)(LT25), SRCC887 (squamous cell carcinoma) (LT26), SRCC888 (adeno-BAC carcinoma) (LT27), SRCC889 (squamous cell carcinoma) (LT28), SRCC890 (squamous cell carcinoma) (LT29), SRCC891 (adenocarcinoma) (1130), SRCC892 (squamous cell carcinoma) (1T31), SRCC894 (adenocarcinoma) (L133).
Also included are human lung tumors designated SRCC1125 [HF-000631], SRCC1127 [HF-000641], SRCC1129 [HF-000643], SRCC1133 [HF-0008401, SRCC1135 [HF-000842], SECC1227 [HF-001291], SRCC1229 [HF-001293], SRCC1230 [HF-001294], SRCC1231 [HF-001295], SRCC1232 [HF-001296], SRCC1233 [HF-001297], SRCC1235 [HF-001299], and SRCC1236 [HF-001300].
Colon cancer cell lines include, for example, ATCC cell lines SW480 (adenocarcinoma, SRCC776), SW620 (lymph node metastasis of colon adenocarcinoma, SRCC777), Colo320 (carcinoma, SRCC778), H129 (adenocarcinoma, SRCC779), HM7 (a high mucin producing variant of ATCC colon adenocarcinoma cell line, SRCC780, obtained from Dr.
Robert Warren, UCSF), CaWiDr (adenocarcinoma, SRCC781), HCT116 (carcinoma, SRCC782), SKCO1 (adenocarcinoma, SRCC783), SW403 (adenocarcinoma, SRCC784), [Si 741 (carcinoma, SRCC785), Co1o205 (carcinoma, SRCC828), HCT15 (carcinoma, SRCC829), HCC2998 (carcinoma, SRCC830), and KM12 (carcinoma, SRCC831). Primary colon tumors include colon adenocarcinomas designated CT2 (5RCC742), C13 (SRCC743), CT8 (SRCC744), CT10 (SRCC745), C112 (8RCC746), CT14 (3RCC747), C115 (SRCC748), C116 (SRCC749), CT17 (SRCC750), CT1 (SRCC751), CT4 (SRCC752), C15 (SRCC753), CT6 (SRCC754), C17 (SRCC755), CT9 (SRCC756), CT11 (SRCC757), CT18 (SRCC758), CT19 (adenocarcinoma, SRCC906), CT20 (adenocarcinoma, SFICC907), CT21 (adenocarcinoma, SHCC908), C122 (adenocarcinoma, SRCC909), C123 (adenocarcinoma, SRCC910), C124 (adenocarcinoma, SRCC911), CT25 (adenocarcinoma, SRCC912), CT26 (adenocarcinoma, SRCC913), CT27 (adenocarcinoma, SRCC914),CT28 (adenocarcinoma, SRCC915), C129 (adenocarcinoma, SRCC916), C130 (adenocarcinoma, SRCC917), CT31 (adenocarcinoma, SRCC918), C132 (adenocarcinoma, SRCC919), C133 (adenocarcinoma, SRCC920), C135 (adenocarcinoma, SRCC921), and CT36 (adenocarcinoma, SHCC922). Also included are human colon tumors designated SRCC1051 INF-000499], SRCC1052 [HF-000539], SRCC1053 [11F-000575], SRCC1054 [HF-0006981, SRCC1142 IFIF-000762], SRCC1144 [HF-000789], SRCC1146 [I-IF-000795] and SRCC1148[HF-000811].
Human breast carcinoma cell lines include, for example, HBL100 (SRCC759), MB435s (SRCC760), T47D
(SRCC761), MB468 (SRCC762), MB175 (SRCC763), MB361 (SRCC764), BT20 (SRCC765), MCF7 (SRCC766), and SKBR3 (SRCC767), and human breast tumor center designated SRCC1057 (HF-000545]. Also included are human breast tumors designated SRCC1094, SRCC1095, SRCC1096, SRCC1097, SRCC1098, SRCC1099, SRCC1100, SRCC1101, and human breast-met-lung-NS tumor designated SRCC893 [LT 32].
Human kidney tumor centers include SRCC989 [HF-000611] and SRCC1014 [HF-000613].
Human testis tumor center includes SBCC1001 [HF-000733] and testis tumor margin SRCC999 [1F-0007161.
Human parathyroid tumor includes SRCC1002 [HF-000831] and SRCC1003 [HF-000832].
F. Tissue Distribution The results of the gene amplification assays herein can be verified by further studies, such as, by determining mRNA expression in various human tissues.
As noted before, gene amplification and/or gene expression in various tissues may be measured by conventional Southern blotting, Northern blotting to quantitate the transcription of mRNA
(Thomas, Proc. Natl. Acad. Sci. USA, 77:5201-5205 [1980]), dot blotting (DNA analysis), or in situ hybridization, using an appropriately labeled probe, based on the sequences provided herein. Alternatively, antibodies may be employed that can recognize specific duplexes, including DNA duplexes, RNA duplexes, and DNA-RNA hybrid duplexes or DNA-protein duplexes.

Gene expression in various tissues, alternatively, may be measured by immunological methods, such as immunohistochemical staining of tissue sections and assay of cell culture or body fluids, to quantitate directly the expression of gene product. Antibodies useful for immunohistochemical staining and/or assay of sample fluids may be either monoclonal or polyclonal, and may be prepared in any mammal.
Conveniently, the antibodies may be prepared against a native sequence PRO10282 (Stra6) polypeptide or against a synthetic peptide based on the DNA and encoding 2 5 a specific antibody epitope. General techniques for generating antibodies, and special protocols for Northern blotting and in situ hybridization are provided hereinbelow.
G. Chromosome Mapping If the amplification of a given gene is functionally relevant, then that gene should be amplified more than neighboring genomic regions which are not important for tumor survival. To test this, the gene can be mapped to a particular chromosome, e.g., by radiation-hybrid analysis. The amplification level is then determined at the location identified, and at the neighboring genomic region. Selective or preferential amplification at the genomic region to which the gene has been mapped is consistent with the possibility that the gene amplification observed promotes tumor growth or survival. Chromosome mapping includes both framework and epicenter mapping.
For further details see, e.g., Stewart et al., Genonne Research, 7:422-433 (1997).

H. Antibody Binding Studies The results of the gene amplification study can be further verified by antibody binding studies, in which the ability of anti-PRO10282 (anti-Stra6) antibodies to inhibit the expression of Stra6 polypeptides on tumor (cancer) cells is tested. Exemplary antibodies include polyclonal, monoclonal, humanized, bispecific, and heteroconjugate antibodies, the preparation of which will be described hereinbelow.
Antibody binding studies may be carried out in any known assay method, such as competitive binding assays, direct and indirect sandwich assays, and immunoprecipitation assays. Zola, Monoclonal Antibodies: A Manual of Techniques, pp.147-158 (CRC Press, Inc., 1987).
Competitive binding assays rely on the ability of a labeled standard to compete with the test sample analyte for binding with a limited amount of antibody. The amount of target protein (encoded by a gene amplified in a tumor cell) in the test sample is inversely proportional to the amount of standard that becomes bound to the antibodies. To facilitate determining the amount of standard that becomes bound, the antibodies preferably are insolubilized before or after the competition, so that the standard and analyte that are bound to the antibodies may conveniently be separated from the standard and analyte which remain unbound.
Sandwich assays involve the use of two antibodies, each capable of binding to a different immunogenic portion, or epitope, of the protein to be detected. In a sandwich assay, the test sample analyte is bound by a first antibody which is immobilized on a solid support, and thereafter a second antibody binds to the analyte, thus forming an insoluble three-part complex. See, e.g., U.S. Patent No. 4,376,110. The second antibody may itself be labeled with a detectable moiety (direct sandwich assays) or may be measured using an anti-innmunoglobulin antibody that is labeled with a detectable 2 0 moiety (indirect sandwich assay). For example, one type of sandwich assay is an ELISA assay, in which case the detectable moiety is an enzyme.
For immunohistochemistry, the tumor sample may be fresh or frozen or may be embedded in paraffin and fixed with a preservative such as formalin, for example.
I. Cell-Based Tumor Assays Cell-based assays and animal models for tumors (e.g., cancers) can be used to verify the findings of the gene amplification assay, and further understand the relationship between the genes identified herein and the development and pathogenesis of neoplastic cell growth. The role of gene products identified herein in the development and pathology of tumor or cancer can be tested by using primary tumor cells or cells lines that have been identified to amplify the genes 3 0 herein. Such cells include, for example, the breast, colon and lung cancer cells and cell lines listed above.
In a different approach, cells of a cell type known to be involved in a particular tumor are transfected with the cDNAs herein, and the ability of these cDNAs to induce excessive growth is analyzed. Suitable cells include, for example, stable tumor cells lines such as, the B104-1-1 cell line (stable NIH-3T3 cell line transfected with the neu protooncogene) and ras-transfected NIH-3T3 cells, which can be transfected with the desired gene, and monitored for tumorogenic growth.
3 5 Such transfected cell lines can then be used to test the ability of poly- or monoclonal antibodies or antibody compositions to inhibit tumorogenic cell growth by exerting cytostatic or cytotoxic activity on the growth of the transformed cells, or by mediating antibody-dependent cellular cytotoxicity (ADCC). Cells transfected with the coding sequences of the genes identified herein can further be used to identify drug candidates for the treatment of cancer.
In addition, primary cultures derived from tumors in transgenic animals (as described below) can be used in the cell-based assays herein, although stable cell lines are preferred. Techniques to derive continuous cell lines from transgenic animals are well known in the art (see, e.g., Small et al., Mol. Cell. Biol., 5:642-648 [1985]).
J. Animal Models A variety of well known animal models can be used to further understand the role of the genes identified herein in the development and pathogenesis of tumors, and to test the efficacy of candidate therapeutic agents, including antibodies, and other antagonists of the native polypeptides, including small molecule antagonists. The in vivo nature of such models makes them particularly predictive of responses in human patients.
Animal models of tumors and cancers (e.g., breast cancer, colon cancer, prostate cancer, lung cancer, etc.) include both non-recombinant and recombinant (transgenic) animals. Non-recombinant animal models include, for example, rodent, e.g., nnurine models. Such models can be generated by introducing tumor cells into syngeneic mice using standard techniques, e.g., subcutaneous injection, tail vein injection, spleen implantation, intraperitoneal implantation, implantation under the renal capsule, or orthopin implantation, e.g., colon cancer cells implanted in colonic tissue. (See, e.g., PCT publication No. WO 97/33551, published September 18, 1997).
Probably the most often used animal species in oncological studies are immunodeficient mice and, in particular, 2 0 nude mice. The observation that the nude mouse with hypo/aplasia could successfully act as a host for human tumor xenografts has lead to its widespread use for this purpose. The autosomal recessive nu gene has been introduced into a very large number of distinct congenic strains of nude mouse, including, for example, ASW, AlHe, AKR, BALB/c, B10.LP, C17, C3H, C57B1, C57, CBA, DBA, ODD, list, NC, NFR, NFS, NFSIN, NZB, NZC, NZW, P, Rill and SJL. In addition, a wide variety of other animals with inherited immunological defects other than the nude mouse have been bred and used as recipients of tumor xenografts. For further details see, e.g., The Nude Mouse in Oncology Research, E. Boven and B.
Winograd, eds., CRC Press, Inc., 1991.
The cells introduced into such animals can be derived from known tumor/cancer cell lines, such as, any of the above-listed tumor cell lines, and, for example, the B104-1-1 cell line (stable NIH-3T3 cell line transfected with the neu protooncogene); ras-transfected NIH-3T3 cells; Caco-2 (ATCC HTB-37); a moderately well-differentiated grade ll human 3 0 colon adenocarcinoma cell line, HT-29 (ATCC HTB-38), or from tumors and cancers. Samples of tumor or cancer cells can be obtained from patients undergoing surgery, using standard conditions, involving freezing and storing in liquid nitrogen (Karmali et al., Br. J. Cancer, 48:689-696 [19831).
Tumor cells can be introduced into animals, such as nude mice, by a variety of procedures. The subcutaneous (s.c.) space in mice is very suitable for tumor implantation. Tumors can be transplanted s.c. as solid blocks, as needle 3 5 biopsies by use of a trochar, or as cell suspensions. For solid block or trochar implantation, tumor tissue fragments of suitable size are introduced into the s.c. space. Cell suspensions are freshly prepared from primary tumors or stable tumor cell lines, and injected subcutaneously. Tumor cells can also be injected as subdermal implants. In this location, the inoculum is deposited between the lower part of the dermal connective tissue and the s.c. tissue. Boven and Winograd (1991), supra.
Animal models of breast cancer can be generated, for example, by implanting rat neuroblastoma cells (from which the neu oncogen was initially isolated), or neu-transf armed NIH-3T3 cells into nude mice, essentially as described by Drebin et al., PNAS USA, 83:9129-9133 (1986).
Similarly, animal models of colon cancer can be generated by passaging colon cancer cells in animals, e.g., nude mice, leading to the appearance of tumors in these animals. An orthotopic transplant model of human colon cancer in nude mice has been described, for example, by Wang et al., Cancer Research, 54:4726-4728 (1994) and Too at al., Cancer Research, 55:681-684 (1995). This model is based on the so-called "METAMOUSE"
sold by AntiCancer, Inc., (San Diego, California).
Tumors that arise in animals can be removed and cultured in vitro. Cells from the in vitro cultures can then be passaged to animals. Such tumors can serve as targets for further testing or drug screening. Alternatively, the tumors resulting from the passage can be isolated and RNA from pre-passage cells and cells isolated after one or more rounds of passage analyzed for differential expression of genes of interest. Such passaging techniques can be performed with any known tumor or cancer cell lines.
For example, Meth A, CMS4, CMS5, CMS21, and WEHI-164 are chemically induced fibrosarcomas of BALBIc female mice (DeLeo et al., J. Exp. Med., 146:720 [1977]), which provide a highly controllable model system for studying 2 0 the anti-tumor activities of various agents (Palladino et al., J.
Immunol., 138:4023-4032 [1987]). Briefly, tumor cells are propagated in vitro in cell culture. Prior to injection into the animals, the cell lines are washed and suspended in buffer, at a cell density of about 10)(106 to 10x107 cells/mt. The animals are then infected subcutaneously with 10 to 100 pl of the cell suspension, allowing one to three weeks for a tumor to appear.
In addition, the Lewis lung (3LL) carcinoma of mice, which is one of the most thoroughly studied experimental 2 5 tumors, can be used as an investigational tumor model. Efficacy in this tumor model has been correlated with beneficial effects in the treatment of human patients diagnosed with small cell carcinoma of the lung (SCCL). This tumor can be introduced in normal mice upon injection of tumor fragments from an affected mouse or of cells maintained in culture (Zupi et al., Br. J. Cancer, 41:suppl. 4:309 119801 and evidence indicates that tumors can be started from injection of even a single cell and that a very high proportion of infected tumor cells survive.
For further information about this tumor model 30 see, Zacharski, Haemostasis, 16:300-320 [1986]).
One way of evaluating the efficacy of a test compound in an animal model on an implanted tumor is to measure the size of the tumor before and after treatment. Traditionally, the size of implanted tumors has been measured with a slide caliper in two or three dimensions. The measure limited to two dimensions does not accurately reflect the size of the tumor, therefore, it is usually converted into the corresponding volume by using a mathematical formula. However, the 3 5 measurement of tumor size is very inaccurate. The therapeutic effects of a drug candidate can be better described as treatment-induced growth delay and specific growth delay. Another important variable in the description of tumor growth is the tumor volume doubling time. Computer programs for the calculation and description of tumor growth are also available, such as the program reported by Rygaard and Spang-Thomsen, Proc.
6th Int. Workshop on Immune-Deficient Animals, Wu and Sheng eds., Basel, 1989, 301. It is noted, however, that necrosis and inflammatory responses following treatment may actually result in an increase in tumor size, at least initially. Therefore, these changes need to be carefully monitored, by a combination of a morphometric method and flow cytometric analysis.
Recombinant (transgenic) animal models can be engineered by introducing the coding portion of the genes identified herein into the genome of animals of interest, using standard techniques for producing transgenic animals.
Animals that can serve as a target for transgenic manipulation include, without limitation, mice, rats, rabbits, guinea pigs, sheep, goats, pigs, and non-human primates, e.g., baboons, chimpanzees and monkeys. Techniques known in the art to introduce a transgene into such animals include pronucleic microinjection (Hoppe and Wanger, U.S. Patent No. 4,873,191);
retrovirus-mediated gene transfer into germ lines (e.g., Van der Putten et al., Proc. Natl. Acad. Sci. USA, 82:6148-615 [1985]); gene targeting in embryonic stem cells (Thompson et al., Cell, 56:313-321 [1989]); electroporation of embryos (Lo, Mol. Cell Biol., 3:1803-1814 [1983]); sperm-mediated gene transfer (Lavitrano et al., Cell, 57:717-73 [1989]). For review, see, for example, US. Patent No. 4,736,866.
For the purpose of the present invention, transgenic animals include those that carry the transgene only in part of their cells ("mosaic animals"). The transgene can be integrated either as a single transgene, or in concatamers, e.g., head-to-head or head-to-tail tandems. Selective introduction of a transgene into a particular cell type is also possible by following, for example, the technique of Lasko et al., Proc. Natl. Acad. Sci.
USA, 89:6232-636 (1992).
2 0 The expression of the transgene in transgenic animals can be monitored by standard techniques. For example, Southern blot analysis or PCR amplification can be used to verify the integration of the transgene. The level of mRNA
expression can then be analyzed using techniques such as in situ hybridization, Northern blot analysis, PCR, or immunocytochemistry. The animals are further examined for signs of tumor or cancer development.
Alternatively, "knock out" animals can be constructed which have a defective or altered gene encoding a PRO10282 (Stra6) polypeptide identified herein, as a result of homologous recombination between the endogenous gene encoding the polypeptide and altered genomic DNA encoding the same polypeptide introduced into an embryonic cell of the animal. For example, cDNA encoding a P11010282 polypeptide can be used to clone genomic DNA encoding that polypeptide in accordance with established techniques. A portion of the genomic DNA encoding a particular P11010282 polypeptide can be deleted or replaced with another gene, such as a gene encoding a selectable marker which can be used 3 0 to monitor integration. Typically, several kilobases of unaltered flanking DNA (both at the 5' and 3' ends) are included in the vector [see, e.g., Thomas and Capecchi, Cell, 51:503 (1987) for a description of homologous recombination vectors].
The vector is introduced into an embryonic stem cell line (e.g., by electroporation) and cells in which the introduced DNA
has homologously recombined with the endogenous DNA are selected [see, e.g., Li et al., Cell, 69:915 (1992)]. The selected cells are then injected into a blastocyst of an animal (e.g., a mouse or rat) to form aggregation chimeras [see, e.g., Bradley, in Teratocarcinomas and Embryonic Stem Cells: A Practical Approach, E. J. Robertson, ed. (IR, Oxford, 1987), pp. 113-152J. A chimeric embryo can then be implanted into a suitable pseudopregnant female foster animal and the embryo brought to term to create a "knock out" animal. Progeny harboring the homologously recombined DNA in their germ cells can be identified by standard techniques and used to breed animals in which all cells of the animal contain the homologously recombined DNA. Knockout animals can be characterized for instance, by their ability to defend against certain pathological conditions and by their development of pathological conditions due to absence of the PR010282 polypeptide.
The efficacy of antibodies specifically binding the polypeptides identified herein and other drug candidates, can be tested also in the treatment of spontaneous animal tumors. A suitable target for such studies is the feline oral squamous cell carcinoma (SCC). Feline oral SCC is a highly invasive, malignant tumor that is the most common oral malignancy of cats, accounting for over 60% of the oral tumors reported in this species. It rarely metastasizes to distant sites, although this low incidence of metastasis may merely be a reflection of the short survival times for cats with this tumor. These tumors are usually not amenable to surgery, primarily because of the anatomy of the feline oral cavity. At present, there is no effective treatment for this tumor. Prior to entry into the study, each cat undergoes complete clinical examination, biopsy, and is scanned by computed tomography (CT). Cats diagnosed with sublingual oral squamous cell tumors are excluded from the study. The tongue can become paralyzed as a result of such tumor, and even if the treatment kills the tumor, the animals may not be able to feed themselves. Each cat is treated repeatedly, over a longer period of time. Photographs of the tumors will be taken daily during the treatment period, and at each subsequent recheck. After treatment, each cat undergoes another CT scan. CT scans and thoracic radiograms are evaluated every 8 weeks thereafter. The data are evaluated for differences in survival, response and toxicity as compared to control groups.
Positive response may require evidence of tumor regression, preferably with improvement of quality of life andlor increased life span.
In addition, other spontaneous animal tumors, such as fibrosarcoma, adenocarcinoma, lymphoma, chrondroma, leiomyosarcoma of dogs, cats, and baboons can also be tested. Of these mammary adenocarcinoma in dogs and cats is a preferred model as its appearance and behavior are very similar to those in humans. However, the use of this model is limited by the rare occurrence of this type of tumor in animals.
K. Screening Assays for Drug Candidates Screening assays for drug candidates are designed to identify compounds that bind or complex with the polypeptides encoded by the genes identified herein, or otherwise interfere with the interaction of the encoded polypeptides with other cellular proteins. Such screening assays will include assays amenable to high-throughput screening of chemical libraries, making them particularly suitable for identifying small molecule drug candidates. Small molecules contemplated include synthetic organic or inorganic compounds, including peptides, preferably soluble peptides, (poly)peptide-immunoglobulin fusions, and, in particular, antibodies including, without limitation, poly- and monoclonal antibodies and antibody fragments, single-chain antibodies, antildiotypic antibodies, and chimeric or humanized versions of such antibodies or fragments, as well as human antibodies and antibody fragments. The assays can be performed in a variety of formats, including protein-protein binding assays, biochemical screening assays, immunoassays and cell based assays, which are well characterized in the art.
All assays are common in that they call for contacting the drug candidate with a polypeptide encoded by a nucleic acid identified herein under conditions and for a time sufficient to allow these two components to interact.
In binding assays, the interaction is binding and the complex formed can be isolated or detected in the reaction mixture. In a particular embodiment, the polypeptide encoded by the gene identified herein or the drug candidate is immobilized on a solid phase, e.g., on a microtiter plate, by covalent or non-covalent attachments. Non-covalent attachment generally is accomplished by coating the solid surface with a solution of the polypeptide and drying. Alternatively, an immobilized antibody, e.g., a monoclonal antibody, specific for the polypeptide to be immobilized can be used to anchor it to a solid surface. The assay is performed by adding the non-immobilized component, which may be labeled by a detectable label, to the immobilized component, e.g., the coated surface containing the anchored component. When the reaction is complete, the non-reacted components are removed, e.g., by washing, and complexes anchored on the solid surface are detected. When the originally non-immobilized component carries a detectable label, the detection of label immobilized on the surface indicates that complexing occurred. Where the originally non-immobilized component does not carry a label, complexing can be detected, for example, by using a labeled antibody specifically binding the immobilized complex.
If the candidate compound interacts with but does not bind to a particular PR010282 polypeptide encoded by a gene identified herein, its interaction with that polypeptide can be assayed by methods well known for detecting protein-protein interactions. Such assays include traditional approaches, such as, cross-linking, co-immunoprecipitation, and co-purification through gradients or chromatographic columns. In addition, protein-protein interactions can be monitored by using a yeast-based genetic system described by Fields and co-workers [Fields and Song, Nature, 340:245-246 (1989);
Chien et al., Proc. Natl. Acad. Sci. USA, 88: 9578-9582 (1991)1 as disclosed by Chevray and Nathans, Proc. Natl. Acad.
Sci. USA, 89:5789-5793 (1991)1. Many transcriptional activators, such as yeast GAL4, consist of two physically discrete modular domains, one acting as the DNA-binding domain, while the other one functioning as the transcription activation domain. The yeast expression system described in the foregoing publications (generally referred to as the "two-hybrid system") takes advantage of this property, and employs two hybrid proteins, one in which the target protein is fused to the DNA-binding domain of GAL4, and another, in which candidate activating proteins are fused to the activation domain.
The expression of a GAL1-lacZ reporter gene under control of a GAL4-activated promoter depends on reconstitution of GAL4 activity via protein-protein interaction. Colonies containing interacting polypeptides are detected with a chromogenic substrate for -galactosidase. A complete kit (MATCHMAKER') for identifying protein-protein interactions between two specific proteins using the two-hybrid technique is commercially available from Clontech. This system can also be extended to map protein domains involved in specific protein interactions as well as to pinpoint amino acid residues that are crucial for these interactions.
Compounds that interfere with the interaction of a PRO10282-encoding gene identified herein and other intra-3 5 or extracellular components can be tested as follows: usually a reaction mixture is prepared containing the product of the amplified gene and the intra- or extracellular component under conditions and for a time allowing for the interaction and binding of the two products. To test the ability of a test compound to inhibit binding, the reaction is run in the absence and in the presence of the test compound. In addition, a placebo may be added to a third reaction mixture, to serve as positive control. The binding (complex formation) between the test compound and the intra- or extracellular component present in the mixture is monitored as described hereinabove. The formation of a complex in the control reaction(s) but not in the reaction mixture containing the test compound indicates that the test compound interferes with the interaction of the test compound and its reaction partner.
To assay for antagonists, the PR010282 polypeptide may be added to a cell along with the compound to be screened for a particular activity and the ability of the compound to inhibit the activity of interest in the presence of the PRO10282 polypeptide indicates that the compound is an antagonist to the PRO10282 polypeptide. Alternatively, antagonists may be detected by combining the PR010282 polypeptide and a potential antagonist with membrane-bound PR010282 polypeptide receptors or recombinant receptors under appropriate conditions for a competitive inhibition assay.
The PR010282 polypeptide can be labeled, such as by radioactivity, such that the number of PR010282 polypeptide molecules bound to the receptor can be used to determine the effectiveness of the potential antagonist. The gene encoding the receptor can be identified by numerous methods known to those of skill in the art, for example, ligand panning and FACS sorting. Coligan et al., Current Protocols in Immun., 1(2): Chapter 5 (1991). Preferably, expression cloning is employed wherein polyadenylated RNA is prepared from a cell responsive to the PRO10282 polypeptide and a cDNA library created from this RNA is divided into pools and used to transfect COS cells or other cells that are not responsive to the PR010282 polypeptide. Transfected cells that are grown on glass slides are exposed to labeled PRO10282 polypeptide.
The PRO10282 polypeptide can be labeled by a variety of means including iodination or inclusion of a recognition site for a site-specific protein kinase. Following fixation and incubation, the slides are subjected to autoradiographic analysis.
Positive pools are identified and sub-pools are prepared and re-transfected using an interactive sub-pooling and re-screening process, eventually yielding a single clone that encodes the putative receptor.
As an alternative approach for receptor identification, labeled PRO10282 polypeptide can be photoaffinity-linked 2 5 with cell membrane or extract preparations that express the receptor molecule.
Cross-linked material is resolved by PAGE
and exposed to X-ray film. The labeled complex containing the receptor can be excised, resolved into peptide fragments, and subjected to protein micro-sequencing. The amino acid sequence obtained from micro-sequencing would be used to design a set of degenerate oligonucleotide probes to screen a cDNA library to identify the gene encoding the putative receptor.
3 0 In another assay for antagonists, mammalian cells or a membrane preparation expressing the receptor would be incubated with labeled PR010282 polypeptide in the presence of the candidate compound. The ability of the compound to enhance or block this interaction could then be measured.
More specific examples of potential antagonists include an oligonucleotide that binds to the fusions of immunoglobulin with the PRO10282 polypeptide, and, in particular, antibodies including, without limitation, poly- and monoclonal antibodies and antibody fragments, single-chain antibodies, anti-idiotypic antibodies, and chimeric or humanized versions of such antibodies or fragments, as well as human antibodies and antibody fragments. Alternatively, a potential antagonist may be a closely related protein, for example, a mutated form of the PR010282 polypeptide that recognizes the receptor but imparts no effect, thereby competitively inhibiting the action of the PRO10282 polypeptide.
Another potential P11010282 polypeptide antagonist is an antisense RNA or DNA
construct prepared using antisense technology, where, e.g., an antisense RNA or DNA molecule acts to block directly the translation of mRNA by hybridizing to targeted mRNA and preventing protein translation. Antisense technology can be used to control gene expression through triple-helix formation or antisense DNA or RNA, both of which methods are based on binding of a polynucleotide to DNA or RNA. For example, the 5 coding portion of the polynucleotide sequence, which encodes the mature PR010282 polypeptide herein, is used to design an antisense RNA
oligonucleotide of from about 10 to 40 base pairs in length. A DNA oligonucleotide is designed to be complementary to a region of the gene involved in transcription (triple helix - see, Lee et al., Nucl. Acids Res., 6:3073 (1979); Cooney et al., Science, 241: 456 (1988); Dervan et al., Science, 251:1360 (1991)), thereby preventing transcription and the production of the P11010282 polypeptide. The antisense RNA oligonucleotide hybridizes to the mRNA in vivo and blocks translation of the mRNA molecule into the PR010282 polypeptide (antisense Okano, Neurochem., 56:560 (1991);
Oligodeoxynucleotides as Antisense Inhibitors of Gene Expression (CRC Press: Boca Raton, FL, 1988). The oligonucleotides described above can also be delivered to cells such that the antisense RNA or DNA may be expressed in vivo to inhibit production of the PRO10282 polypeptide. When antisense DNA is used, oligodeoxyribonucleotides derived from the translation-initiation site, e.g., between about -10 and + 10 positions of the target gene nucleotide sequence, are preferred.
Antisense RNA or DNA molecules are generally at least about 5 bases in length, about 10 bases in length, about 2 0 15 bases in length, about 20 bases in length, about 25 bases in length, about 30 bases in length, about 35 bases in length, about 40 bases in length, about 45 bases in length, about 50 bases in length, about 55 bases in length, about 60 bases in length, about 65 bases in length, about 70 bases in length, about 75 bases in length, about 80 bases in length, about 85 bases in length, about 90 bases in length, about 95 bases in length, about 100 bases in length, or more.
Potential antagonists include small molecules that bind to the active site, the receptor binding site, or growth factor or other relevant binding site of the PRO10282 polypeptide, thereby blocking the normal biological activity of the PR010282 polypeptide. Examples of small molecules include, but are not limited to, small peptides or peptide-like molecules, preferably soluble peptides, and synthetic non-peptidyl organic or inorganic compounds.
Ribozymes are enzymatic RNA molecules capable of catalyzing the specific cleavage of RNA. Ribozymes act by sequence-specific hybridization to the complementary target RNA, followed by endonucleolytic cleavage. Specific ribozyme 3 0 cleavage sites within a potential RNA target can be identified by known techniques. For further details see, e.g., Rossi, Current Biology, 4:469-471 (1994), and PCT publication No. WO 97/33551 (published September 18, 1997).
Nucleic acid molecules in triple-helix formation used to inhibit transcription should be single-stranded and composed of deoxynucleotides. The base composition of these oligonucleotides is designed such that it promotes triple-helix formation via Hoogsteen base-pairing rules, which generally require sizeable stretches of purines or pyrimidines on one strand of a duplex. For further details see, e.g., PCT publication No. WO
97/33551, supra.

These small molecules can be identified by any one or more of the screening assays discussed hereinabove andIor by any other screening techniques well known for those skilled in the art.
L. Uses for P11010282 (Stra6) Nucleotide sequences (or their complement) encoding PRO10282 (Stra6) polypeptides have various applications in the art of molecular biology, including uses as hybridization probes, in chromosome and gene mapping and in the generation of anti-sense RNA and DNA. P11010282 nucleic acid will also be useful for the preparation of PR010282 polypeptides by the recombinant techniques described herein.
The full-length native sequence P11010282 gene (SEG ID NO:1), or PR019578 gene (SEG ID NO: 4), or portions thereof, may be used as hybridization probes for a cDNA library to isolate the full-length PRO10282 or PRO19578 cDNA
or to isolate still other cDNAs (for instance, those encoding further naturally-occurring variants of PR010282, or P11010282 from other species) which have a desired sequence identity to the P11010282 coding sequence disclosed in Figure 1 (SEG ID N0:1) or the P11019578 coding sequence disclosed in Figure 6 (SEG ID NO: 4). Optionally, the length of the probes will be about 20 to about 50 bases. The hybridization probes may be derived from at least partially novel regions of the nucleotide sequence of SEG ID N0:1 wherein those regions may be determined without undue experimentation or from genomic sequences including promoters, enhancer elements and introns of the native PRO10282 or P11019578 gene. By way of example, a screening method will comprise isolating the coding region of the P11010282 or PRO19578 gene using the known DNA sequence to synthesize a selected probe of about 40 bases. Hybridization probes may be labeled by a variety of labels, including radionucleotides such as 3213 or 35, or enzymatic labels such as alkaline 2 0 phosphatase coupled to the probe via avidin/biotin coupling systems.
Labeled probes having a sequence complementary to that of the P11010282 or PI1019578 gene of the present invention can be used to screen libraries of human cDNA, genomic DNA or mRNA to determine which members of such libraries the probe hybridizes to. Hybridization techniques are described in further detail in the Examples below.
Any EST sequences disclosed in the present application may similarly be employed as probes, using the methods disclosed herein.
Other useful fragments of the P11010282 (Stra6) nucleic acids include antisense or sense oligonucleotides comprising a singe-stranded nucleic acid sequence (either RNA or DNA) capable of binding to target PR010282 (Stra6) mRNA (sense) or PRO10282 (Stra6) DNA (antisense) sequences. Antisense or sense oligonucleotides, according to the present invention, comprise a fragment of the coding region of PR010282 (Stra6) DNA. Such a fragment generally 3 0 comprises at least about 14 nucleotides, preferably from about 14 to 30 nucleotides. The ability to derive an antisense or a sense oligonucleotide, based upon a cDNA sequence encoding a given protein is described in, for example, Stein and Cohen (Cancer Res. 48:2659, 1988) and van der Krol at al. (BioTechnieues 6:958, 1988).
Binding of antisense or sense oligonucleotides to target nucleic acid sequences results in the formation of duplexes that block transcription or translation of the target sequence by one of several means, including enhanced 3 5 degradation of the duplexes, premature termination of transcription or translation, or by other means. The antisense oligonucleotides thus may be used to block expression of PRO10282 (Stra6) proteins. Antisense or sense oligonucleotides further comprise oligonucleotides having modified sugar-phosphodiester backbones (or other sugar linkages, such as those described in WO 91/06629) and wherein such sugar linkages are resistant to endogenous nucleases. Such oligonucleotides with resistant sugar linkages are stable in vivo (i.e., capable of resisting enzymatic degradation) but retain sequence specificity to be able to bind to target nucleotide sequences.
Other examples of sense or antisense oligonucleotides include those oligonucleotides which are covalently linked to organic moieties, such as those described in WO 90110048, and other moieties that increases affinity of the oligonucleotide for a target nucleic acid sequence, such as poly(L-lysine).
Further still, intercalating agents, such as ellipticine, and alkylating agents or metal complexes may be attached to sense or antisense oligonucleotides to modify 1 0 binding specificities of the antisense or sense oligonucleotide for the target nucleotide sequence.
Antisense or sense oligonucleotides may be introduced into a cell containing the target nucleic acid sequence by any gene transfer method, including, for example, CaPO4-mediated DNA
transfection, electroporation, or by using gene transfer vectors such as Epstein-Barr virus. In a preferred procedure, an antisense or sense oligonucleotide is inserted into 'a suitable retroviral vector. A cell containing the target nucleic acid sequence is contacted with the recombinant retroviral vector, either in vivo or ex vivo. Suitable retroviral vectors include, but are not limited to, those derived from the murine retrovirus M-MuLV, N2 (a retrovirus derived from M-MuLV), or the double copy vectors designated DCT5A, DCT5B and DCT5C (see WO 90/13641).
Sense or antisense oligonucleotides also may be introduced into a cell containing the target nucleotide sequence by formation of a conjugate with a ligand-binding molecule, as described in WO
91/04753. Suitable ligand binding 2 0 molecules include, but are not limited to, cell surface receptors, growth factors, other cytokines, or other ligands that bind to cell surface receptors. Preferably, conjugation of the ligand-binding molecule does not substantially interfere with the ability of the ligand-binding molecule to bind to its corresponding molecule or receptor, or block entry of the sense or antisense oligonucleotide or its conjugated version into the cell.
Alternatively, a sense or an antisense oligonucleotide may be introduced into a cell containing the target nucleic acid sequence by formation of an oligonucleotide-lipid complex, as described in WO 90/10448. The sense or antisense oligonucleotide-lipid complex is preferably dissociated within the cell by an endogenous lipase.
The probes may also be employed in PCR techniques to generate a pool of sequences for identification of closely related PRO10282 coding sequences.
Nucleotide sequences encoding a native sequence PRO10282 (Stra6) polypeptide can also be used to construct hybridization probes for mapping the gene, which encodes that native sequence PRO10282 (Stra6) and for the genetic analysis of individuals with genetic disorders. The nucleotide sequences provided herein may be mapped to a chromosome and specific regions of a chromosome using known techniques, such as in situ hybridization, linkage analysis against known chromosomal markers, and hybridization screening with libraries.
When the coding sequences for PRO10282 encode a protein which binds to another protein (for example, where the PRO10282 is a receptor), the PRO10282 can be used in assays to identify the other proteins or molecules involved in the binding interaction. By such methods, inhibitors of the receptorIligand binding interaction can be identified. Proteins involved in such binding interactions can also be used to screen for peptide or small molecule inhibitors or agonists of the binding interaction. Also, the receptor P11010282 (Stra6) can be used to isolate correlative ligand(s). Screening assays can be designed to find lead compounds that mimic the biological activity of a native P11010282 (Stra6) or a ntive polypeptide binding to P11010282 ("PR010282 binding protein"). Such screening assays will include assays amenable to high-throughput screening of chemical libraries, making them particularly suitable for identifying small molecule drug candidates. Small molecules contemplated include synthetic organic or inorganic compounds. The assays can be performed in a variety of formats, including protein-protein binding assays, biochemical screening assays, immunoassays and cell based assays, which are well characterized in the art.
Nucleic acids which encode P11010282 or its modified forms can also be used to generate either transgenic animals or "knock out" animals which, in turn, are useful in the development and screening of therapeutically useful reagents. A transgenic animal (e.g., a mouse or rat) is an animal having cells that contain a transgene, which transgene was introduced into the animal or an ancestor of the animal at a prenatal, e.g., an embryonic stage. A transgene is a DNA
which is integrated into the genome of a cell from which a transgenic animal develops. In one embodiment, cDNA encoding PR010282 can be used to clone genomic DNA encoding P11010282 in accordance with established techniques and the genomic sequences used to generate transgenic animals that contain cells which express DNA encoding PR010282.
Methods for generating transgenic animals, particularly animals such as mice or rats, have become conventional in the art and are described, for example, in U.S. Patent Nos. 4,736,866 and 4,870,009.
Typically, particular cells would be targeted for PR010282 transgene incorporation with tissue-specific enhancers.
Transgenic animals that include a copy of a transgene encoding PRO10282 introduced into the germ line of the animal at an embryonic stage can be used to examine the effect of increased expression of DNA encoding PRO10282. Such animals can be used as tester animals for reagents thought to confer protection from, for example, pathological conditions associated with its overexpression. In accordance with this facet of the invention, an animal is treated with the reagent and a reduced incidence of the pathological condition, compared to untreated animals bearing the transgene, would indicate a potential therapeutic intervention for the pathological condition.
Alternatively, non-human homologues of PRO10282 can be used to construct a PRO10282 "knock out" animal which has a defective or altered gene encoding PRO10282 as a result of homologous recombination between the endogenous gene encoding PR010282 and altered genomic DNA encoding P11010282 introduced into an embryonic stem cell of the animal. For example, cDNA encoding P11010282 can be used to clone genomic DNA encoding P11010282 in 3 0 accordance with established techniques. A portion of the genomic DNA
encoding PRO10282 can be deleted or replaced with another gene, such as a gene encoding a selectable marker which can be used to monitor integration. Typically, several kilobases of unaltered flanking DNA (both at the 5' and 3' ends) are included in the vector [see e.g., Thomas and Capecchi, Cell, 51:503 (1987) for a description of homologous recombination vectors]. The vector is introduced into an embryonic stem cell line (e.g., by electroporation) and cells in which the introduced DNA has homologously recombined with the endogenous DNA are selected [see e.g., Li et al., Cell, 69:915 (1992)].
The selected cells are then injected into a blastocyst of an animal (e.g., a mouse or rat) to form aggregation chimeras [see e.g., Bradley, in Teratocarcinomas and Embryonic Stem Cells: A Practical Approach, E. J. Robertson, ed. (IRL, Oxford, 1987), pp. 113-152]. A chimeric embryo can then be implanted into a suitable pseudopregnant female foster animal and the embryo brought to term to create a "knock out" animal. Progeny harboring the homologously recombined DNA in their germ cells can be identified by standard techniques and used to breed animals in which all cells of the animal contain the homologously recombined DNA. Knockout animals can be characterized for instance, for their ability to defend against certain pathological conditions and for their development of pathological conditions due to absence of the PRO10282 polypeptide.
Nucleic acid encoding the PR010282 polypeptides may also be used in gene therapy. In gene therapy applications, genes are introduced into cells in order to achieve in vivo synthesis of a therapeutically effective genetic product, for example for replacement of a defective gene. "Gene therapy"
includes both conventional gene therapy where a lasting effect is achieved by a single treatment, and the administration of gene therapeutic agents, which involves the one time or repeated administration of a therapeutically effective DNA or mRNA. Antisense RNAs and DNAs can be used as therapeutic agents for blocking the expression of certain genes in vivo. It has already been shown that short antisense oligonucleotides can be imported into cells where they act as inhibitors, despite their low intracellular concentrations caused by their restricted uptake by the cell membrane. (Zarnecnik et al., Proc. Natl. Acad. Sci. USA 83:4143-4146 [1986]). The oligonucleotides can be modified to enhance their uptake, e.g. by substituting their negatively charged phosphodiester groups by uncharged groups.
There are a variety of techniques available for introducing nucleic acids into viable cells. The techniques vary depending upon whether the nucleic acid is transferred into cultured cells in vitro, or in vivo in the cells of the intended host.
2 0 Techniques suitable for the transfer of nucleic acid into mammalian cells in vitro include the use of liposomes, electroporation, microinjection, cell fusion, DEAE-dextran, the calcium phosphate precipitation method, etc. The currently preferred in vivo gene transfer techniques include transfection with viral (typically retroviral) vectors and viral coat protein-liposome mediated transfection (Dzau et al., Trends in Biotechnology 11, 205-210 [1993]). In some situations it is desirable to provide the nucleic acid source with an agent that targets the target cells, such as an antibody specific for a cell surface membrane protein or the target cell, a ligand for a receptor on the target cell, etc. Where liposomes are employed, proteins which bind to a cell surface membrane protein associated with endocytosis may be used for targeting and/or to facilitate uptake, e.g. capsid proteins or fragments thereof tropic for a particular cell type, antibodies for proteins which undergo internalization in cycling, proteins that target intracellular localization and enhance intracellular half-life.
The technique of receptor-mediated endocytosis is described, for example, by Wu et al., J. Biol. Chem. 262, 4429-4432 (1987); and Wagner et al., Proc. Natl. Acad. Sci. USA 87, 3410-3414 (1990).
For review of gene marking and gene therapy protocols see Anderson et al., Science 256, 808-813 (1992).
The PRO10282 polypeptides described herein may also be employed as molecular weight markers for protein electrophoresis purposes.
The nucleic acid molecules encoding the PR010282 polypeptides or fragments thereof described herein are useful 3 5 for chromosome identification. In this regard, there exists an ongoing need to identify new chromosome markers, since relatively few chromosome marking reagents, based upon actual sequence data are presently available. Each PRO10282 nucleic acid molecule of the present invention can be used as a chromosome marker.
The PRO10282 polypeptides and nucleic acid molecules of the present invention may also be used for tissue typing, wherein the PRO10282 polypeptides of the present invention may be differentially expressed in one tissue as compared to another. PRO10282 nucleic acid molecules will find use for generating probes for PCR, Northern analysis, Southern analysis and Western analysis.
The PR010282 polypeptides described herein may also be employed as therapeutic agents. The PR010282 polypeptides of the present invention can be formulated according to known methods to prepare pharmaceutically useful compositions, whereby the PRO10282 product hereof is combined in admixture with a pharmaceutically acceptable carrier vehicle. Therapeutic formulations are prepared for storage by mixing the active ingredient having the desired degree of purity with optional physiologically acceptable carriers, excipients or stabilizers (Remington's Pharmaceutical Sciences 16th edition, Osol, A. Ed. (1980)), in the form of lyophilized formulations or aqueous solutions. Acceptable carriers, excipients or stabilizers are nontoxic to recipients at the dosages and concentrations employed, and include buffers such as phosphate, citrate and other organic acids; antioxidants including ascorbic acid; low molecular weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, gelatin or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone, amino acids such as glycine, glutamine, asparagine, arginine or lysine; monosaccharides, disaccharides and other carbohydrates including glucose, mannose, or dextrins; chelating agents such as EDTA; sugar alcohols such as mannitol or sorbitol; salt-forming counterions such as sodium; and/or nonionic surfactants such as TWEENTm, PLIJRONICSTM
or PEG.
2 0 The formulations to be used for in vivo administration must be sterile. This is readily accomplished by filtration through sterile filtration membranes, prior to or following lyophilization and reconstitution.
Therapeutic compositions herein generally are placed into a container having a sterile access port, for example, an intravenous solution bag or vial having a stopper pierceable by a hypodermic injection needle.
The route of administration is in accord with known methods, e.g. injection or infusion by intravenous, intraperitoneal, intracerebral, intramuscular, intraocular, intraarterial or intralesional routes, topical administration, or by sustained release systems.
Dosages and desired drug concentrations of pharmaceutical compositions of the present invention may vary depending on the particular use envisioned. The determination of the appropriate dosage or route of administration is well within the skill of an ordinary physician. Animal experiments provide reliable guidance for the determination of effective doses for human therapy. Interspecies scaling of effective doses can be performed following the principles laid down by Mordenti, J. and Chappell, W. "The use of interspecies scaling in toxicokinetics" In Toxicokinetics and New Drug Development, Yacobi et al., Eds., Pergamon Press, New York 1989, pp. 42-96.
When in vivo administration of a PR010282 polypeptide or agonist or antagonist thereof is employed, normal dosage amounts may vary from about 10 nglkg to up to 100 mglkg of mammal body weight or more per day, preferably about 1 glkgiday to 10 mg/kg/day, depending upon the route of administration.
Guidance as to particular dosages and methods of delivery is provided in the literature; see, for example, U.S. Pat.
Nos. 4,657,760; 5,206,344; or 5,225,212.
It is anticipated that different formulations will be effective for different treatment compounds and different disorders, that administration targeting one organ or tissue, for example, may necessitate delivery in a manner different from that to another organ or tissue.
Where sustained-release administration of a PR010282 polypeptide is desired in a formulation with release characteristics suitable for the treatment of any disease or disorder requiring administration of the PRO10282 polypeptide, microencapsulation of the PRO10282 polypeptide is contemplated.
Microencapsulation of recombinant proteins for sustained release has been successfully performed with human growth hormone (rhGH), interferon- (rhIF111- ), interleukin-2, and MN rgp120. Johnson et al., Nat. Med., 2:795-799 (1996); Yasuda, Biomed.
Ther., 27:1221-1223 (1993); Elora et al., BiolTechnology, 8:755-758 (1990); Cleland, "Design and Production of Single Immunization Vaccines Using Polylactide Polyglycolide Microsphere Systems," in Vaccine Design: The Subunit and Adjuvant Approach, Powell and Newman, eds, =
(Plenum Press: New York, 1995), pp. 439-462; WO 97/03692, WO 96/40072, WO
96/07399; and U.S. Pat. No.
5,654,010.
The sustained-release formulations of these proteins were developed using poly-lactic-coglycolic acid (PLGA) polymer due to its biocompatibility and wide range of biodegradable properties. The degradation products of PLGA, lactic and glycolic acids, can be cleared quickly within the human body. Moreover, the degradability of this polymer can be adjusted from months to years depending on its molecular weight and composition. Lewis, "Controlled release of bioactive agents from lactidelglycolide polymer," in: M. Chasin and R. Langer (Eds.), Biodegradable Polymers as Drug Delivery Systems, (Marcel Dekker: New York, 1990), pp. 1-41.
This invention encompasses methods of screening compounds to identify those that mimic the PR010282 polypeptide (agonists) or prevent the effect of the PRO10282 polypeptide (antagonists). Screening assays for antagonist drug candidates are designed to identify compounds that bind or complex with the PRO10282 polypeptides encoded by the genes identified herein, or otherwise interfere with the interaction of the encoded polypeptides with other cellular proteins. Such screening assays will include assays amenable to high-throughput screening of chemical libraries, making 2 5 them particularly suitable for identifying small molecule drug candidates.
The assays can be performed in a variety of formats, including protein-protein binding assays, biochemical screening assays, immunoassays, and cell-based assays, which are well characterized in the art.
All assays for antagonists are common in that they call for contacting the drug candidate with a PR010282 polypeptide encoded by a nucleic acid identified herein under conditions and for a time sufficient to allow these two 3 0 components to interact.
In binding assays, the interaction is binding and the complex formed can be isolated or detected in the reaction mixture. In a particular embodiment, the PR010282 polypeptide encoded by the gene identified herein or the drug candidate is immobilized on a solid phase, e.g., on a nnicrotiter plate, by covalent or non-covalent attachments. Non-covalent attachment generally is accomplished by coating the solid surface with a solution of the PRO10282 polypeptide 35 and drying. Alternatively, an immobilized antibody, e.g., a monoclonal antibody, specific for the PRO10282 polypeptide to be immobilized can be used to anchor it to a solid surface. The assay is performed by adding the non-immobilized component, which may be labeled by a detectable label, to the immobilized component, e.g., the coated surface containing the anchored component. When the reaction is complete, the non-reacted components are removed, e.g., by washing, and complexes anchored on the solid surface are detected. When the originally non-immobilized component carries a detectable label, the detection of label immobilized on the surface indicates that complexing occurred. Where the originally non-immobilized component does not carry a label, complexing can be detected, for example, by using a labeled antibody specifically binding the immobilized complex.
If the candidate compound interacts with but does not bind to a particular PR010282 polypeptide encoded by a gene identified herein, its interaction with that polypeptide can be assayed by methods well known for detecting protein-protein interactions. Such assays include traditional approaches, such as, e.g., cross-linking, co-immunoprecipitation, and co-purification through gradients or chromatographic columns. In addition, protein-protein interactions can be monitored by using a yeast-based genetic system described by Fields and co-workers (Fields and Song, Nature (London), 340:245-246 (1989); Chien et al., Proc. Natl. Acad. Sci. USA, 88:9578-9582 (1991)) as disclosed by Chevray and Nathans, Proc. Natl.
Acad. Sci. USA, 89: 5789-5793 (1991). Many transcriptional activators, such as yeast GAL4, consist of two physically discrete modular domains, one acting as the DNA-binding domain, the other one functioning as the transcription-activation domain. The yeast expression system described in the foregoing publications (generally referred to as the "two-hybrid system") takes advantage of this property, and employs two hybrid proteins, one in which the target protein is fused to the DNA-binding domain of GAL4, and another, in which candidate activating proteins are fused to the activation domain.
The expression of a GAL1-lacZ reporter gene under control of a GAL4-activated promoter depends on reconstitution of GAL4 activity via protein-protein interaction. Colonies containing interacting polypeptides are detected with a chromogenic substrate for -galactosidase. A complete kit (MATCHMAKER') for identifying protein-protein interactions between two specific proteins using the two-hybrid technique is commercially available from Clontech. This system can also be extended to map protein domains involved in specific protein interactions as well as to pinpoint amino acid residues that are crucial for these interactions.
Compounds that interfere with the interaction of a gene encoding a P11010282 polypeptide identified herein and other intra- or extracellular components can be tested as follows: usually a reaction mixture is prepared containing the product of the gene and the intra- or extracellular component under conditions and for a time allowing for the interaction and binding of the two products. To test the ability of a candidate compound to inhibit binding, the reaction is run in the absence and in the presence of the test compound. In addition, a placebo may be added to a third reaction mixture, to serve 3 0 as positive control. The binding (complex formation) between the test compound and the intra- or extracellular component present in the mixture is monitored as described hereinabove. The formation of a complex in the control reaction(s) but not in the reaction mixture containing the test compound indicates that the test compound interferes with the interaction of the test compound and its reaction partner.
To assay for antagonists, the PR010282 polypeptide may be added to a cell along with the compound to be screened for a particular activity and the ability of the compound to inhibit the activity of interest in the presence of the PR010282 polypeptide indicates that the compound is an antagonist to the PR010282 polypeptide. Alternatively, antagonists may be detected by combining the PRO10282 polypeptide and a potential antagonist with membrane-bound PRO10282 polypeptide receptors or recombinant receptors under appropriate conditions for a competitive inhibition assay.
The PR010282 polypeptide can be labeled, such as by radioactivity, such that the number of PR010282 polypeptide molecules bound to the receptor can be used to determine the effectiveness of the potential antagonist. The gene encoding the receptor can be identified by numerous methods known to those of skill in the art, for example, ligand panning and FACS sorting. Coligan et al., Current Protocols in lmmun., 1(2): Chapter 5 (1991). Preferably, expression cloning is employed wherein polyadenylated RNA is prepared from a cell responsive to the PRO10282 polypeptide and a cDNA library created from this RNA is divided into pools and used to transfect COS cells or other cells that are not responsive to the PRO10282 polypeptide. Transfected cells that are grown on glass slides are exposed to labeled PRO10282 polypeptide.
The PR010282 polypeptide can be labeled by a variety of means including iodination or inclusion of a recognition site for a site-specific protein kinase. Following fixation and incubation, the slides are subjected to autoradiographic analysis.
Positive pools are identified and sub-pools are prepared and re-transfected using an interactive sub-pooling and re-screening process, eventually yielding a single clone that encodes the putative receptor.
As an alternative approach for receptor identification, labeled PR010282 polypeptide can be photoaffinity-linked with cell membrane or extract preparations that express the receptor molecule.
Cross-linked material is resolved by PAGE
and exposed to X-ray film. The labeled complex containing the receptor can be excised, resolved into peptide fragments, and subjected to protein micro-sequencing. The amino acid sequence obtained from micro- sequencing would be used to design a set of degenerate oligonucleotide probes to screen a cDNA library to identify the gene encoding the putative receptor.
In another assay for antagonists, mammalian cells or a membrane preparation expressing the receptor would be incubated with labeled P11010282 polypeptide in the presence of the candidate compound. The ability of the compound to enhance or block this interaction could then be measured.
More specific examples of potential antagonists include an oligonucleotide that binds to the fusions of imnnunoglobulin with PR010282 polypeptide, and, in particular, antibodies including, without limitation, poly- and monoclonal antibodies and antibody fragments, single-chain antibodies, anti-idiotypic antibodies, and chimeric or humanized versions of such antibodies or fragments, as well as human antibodies and antibody fragments. Alternatively, a potential antagonist may be a closely related protein, for example, a mutated form of the PR010282 polypeptide that recognizes the receptor but imparts no effect, thereby competitively inhibiting the action of the PRO10282 polypeptide.
Another potential PR010282 polypeptide antagonist is an antisense RNA or DNA
construct prepared using antisense technology, where, e.g., an antisense RNA or DNA molecule acts to block directly the translation of mRNA by hybridizing to targeted mRNA and preventing protein translation. Antisense technology can be used to control gene expression through triple-helix formation or antisense DNA or RNA, both of which methods are based on binding of a polynucleotide to DNA or RNA. For example, the 5 coding portion of the polynucleotide sequence, which encodes the mature PRO10282 polypeptides herein, is used to design an antisense RNA
oligonucleotide of from about 10 to 40 base pairs in length. A DNA oligonucleotide is designed to be complementary to a region of the gene involved in transcription (triple helix - see Lee et al., Nucl. Acids Res., 6:3073 (1979); Cooney et al., Science, 241: 456 (1988); Dervan et al., Science, 251:1360 (1991)), thereby preventing transcription and the production of the PR010282 polypeptide. The antisense RNA oligonucleotide hybridizes to the mRNA in vivo and blocks translation of the mRNA molecule into the PRO10282 polypeptide (antisense- Okano, Neurochem., 56:560 (1991);
Oligodeoxynucleotides as Antisense Inhibitors of Gene Expression (CRC Press: Boca Raton, FL, 1988). The oligonucleotides described above can also be delivered to cells such that the antisense RNA or DNA may be expressed in vivo to inhibit production of the PR010282 polypeptide. When antisense DNA is used, oligodeoxyribonucleotides derived from the translation-initiation site, e.g., between about -10 and +10 positions of the target gene nucleotide sequence, are preferred.
Potential antagonists include small molecules that bind to the active site, the receptor binding site, or growth factor or other relevant binding site of the PR010282 polypeptide, thereby blocking the normal biological activity of the PR010282 polypeptide. Examples of small molecules include, but are not limited to, small peptides or peptide-like molecules, preferably soluble peptides, and synthetic non-peptidyl organic or inorganic compounds.
Ribozymes are enzymatic RNA molecules capable of catalyzing the specific cleavage of RNA. Ribozymes act by sequence-specific hybridization to the complementary target RNA, followed by endonucleolytic cleavage. Specific ribozyme cleavage sites within a potential RNA target can be identified by known techniques. For further details see, e.g., Rossi, Current Biology, 4:469-471 (1994), and PCT publication No. WO 97133551 (published September 18, 1997).
Nucleic acid molecules in triple-helix formation used to inhibit transcription should be single-stranded and composed of deoxynucleotides. The base composition of these oligonucleotides is designed such that it promotes triple-helix formation via Hoogsteen base-pairing rules, which generally require sizeable stretches of purines or pyrimidines on one strand of a duplex. For further details see, e.g., PCT publication No. WO
97133551, supra.
These small molecules can be identified by any one or more of the screening assays discussed hereinabove and/or by any other screening techniques well known for those skilled in the art.
Stra6, a murine retinoic acid inducible gene, has a spermatogenic cycle-dependent expression in Sertoli cells in testis. Based on sequence homology with Stra6, the PR010282 polypeptides disclosed herein may have an important role in spermatogenesis, and therefore has a potential use in fertility treatment.
It is currently believed that the expression of PR010282 polypeptide, just like that of its murine homolog Stra6, is induced in response to retinoic acid. Therefore, the expression of PR010282 at the level of mRNA, by hybridization, or protein, by immunological assays, can be used to determine whether a test compound has a potential for the treatment of a wide variety of retinoid responsive diseases.

Among the types of diseases contemplated as therapeutic targets include psoriasis, acne, dysplasias, cancers and autoimmune diseases. The category of contemplated dysplasias includes precancerous lesions of the epithelial tissues such as oral leukoplakias, dysplasia of the cervix, larynx and bronchi. The category of contemplated cancers includes carcinomas of colon (adenocarcinoma), lung (carcinoma and adenocarcinoma), skin, head and neck, cervix, uterus, breast and prostate.
The category of contemplated autoimmune diseases includes rheumatoid arthritis, osteoarthritis, systemic lupus erythematosus, pemphigus valgaris and pemphigus foliaceous. Potential therapeutics to treat such diseases and conditions are antibodies and antagonists (including small molecules) of PRO10282.
M. Compositions and Methods for the Treatment of Tumors The compositions useful in the treatment of tumors associated with the amplification of the genes identified herein include, without limitation, antibodies, small organic and inorganic molecules, peptides, phosphopeptides, antisense and ribozyme molecules, triple helix molecules, etc., that inhibit the expression and/or activity of the target gene product.
For example, antisense RNA and RNA molecules act to directly block the translation of mRNA by hybridizing to targeted mRNA and preventing protein translation. When antisense DNA is used, oligodeoxyribonucleotides derived from the translation initiation site, e.g., between about -10 and +10 positions of the target gene nucleotide sequence, are preferred.
Ribozymes are enzymatic RNA molecules capable of catalyzing the specific cleavage of RNA. Ribozymes act by sequence-specific hybridization to the complementary target RNA, followed by endonucleolytic cleavage. Specific ribozyme cleavage sites within a potential RNA target can be identified by known techniques. For further details see, e.g., Rossi, Current Biology, 4:469-471 (1994), and PCT publication No. WO 97133551 (published September 18, 1997).
Nucleic acid molecules in triple helix formation used to inhibit transcription should be single-stranded and composed of deoxynucleotides. The base composition of these oligonucleotides is designed such that it promotes triple helix formation via Hoogsteen base pairing rules, which generally require sizeable stretches of purines or pyrimidines on one strand of a duplex. For further details see, e.g., PCT publication No. WO
97/33551, supra.
These molecules can be identified by any or any combination of the screening assays discussed hereinabove and/or by any other screening techniques well known for those skilled in the art.
N. Anti-PR010282 (anti-Stra6) Antibodies The present invention further provides anti-PR010282 (anti-Stra6) antibodies.
Exemplary antibodies include polyclonal, monoclonal, humanized, bispecific, and heteroconjugate antibodies.
1. Polyclonal Antibodies The anti-PR010282 antibodies may comprise polyclonal antibodies. Methods of preparing polyclonal antibodies are known to the skilled artisan. Polyclonal antibodies can be raised in a mammal, for example, by one or more injections of an immunizing agent and, if desired, an adjuvant. Typically, the immunizing agent and/or adjuvant will be injected in the mammal by multiple subcutaneous or intraperitoneal injections. The immunizing agent may include the PRO10282 polypeptide or a fusion protein thereof. It may be useful to conjugate the immunizing agent to a protein known to be immunogenic in the mammal being immunized. Examples of such immunogenic proteins include but are not limited to keyhole limpet hemocyanin, serum albumin, bovine thyroglobulin, and soybean trypsin inhibitor. Examples of adjuvants which may be employed include Freund's complete adjuvant and MPL-TDM adjuvant (monophosphoryl Lipid A, synthetic trehalose dicorynomycolate). The immunization protocol may be selected by one skilled in the art without undue experimentation.
2. Monoclonal Antibodies The anti-PRO10282 antibodies may, alternatively, be monoclonal antibodies.
Monoclonal antibodies may be prepared using hybridoma methods, such as those described by Kohler and Milstein, Nature, 256:495 (1975). In a hybridoma method, a mouse, hamster, or other appropriate host animal, is typically immunized with an immunizing agent to elicit lymphocytes that produce or are capable of producing antibodies that will specifically bind to the immunizing agent.
Alternatively, the lymphocytes may be immunized in vitro.
The immunizing agent will typically include the PRO10282 polypeptide or a fusion protein thereof. Generally, 1 0 either peripheral blood lymphocytes ("PBLs") are used if cells of human origin are desired, or spleen cells or lymph node cells are used if non-human mammalian sources are desired. The lymphocytes are then fused with an immortalized cell line using a suitable fusing agent, such as polyethylene glycol, to form a hybridoma cell [Goding, Monoclonal Antibodies: Principles and Practice, Academic Press, (1986) pp. 59-103]. Immortalized cell lines are usually transformed mammalian cells, particularly myeloma cells of rodent, bovine and human origin. Usually, rat or mouse myeloma cell lines are employed. The hybridoma cells may be cultured in a suitable culture medium that preferably contains one or more substances that inhibit the growth or survival of the unfused, immortalized cells. For example, if the parental cells lack the enzyme hypoxanthine guanine phosphoribosyl transf erase (HGPRT or HPRT), the culture medium for the hybridomas typically will include hypoxanthine, aminopterin, and thymidine ("HAT medium"), which substances prevent the growth of HGPRT-deficient cells.
Preferred immortalized cell lines are those that fuse efficiently, support stable high level expression of antibody 2 0 by the selected antibody-producing cells, and are sensitive to a medium such as HAT medium. More preferred immortalized cell lines are murine myeloma lines, which can be obtained, for instance, from the Salk Institute Cell Distribution Center, San Diego, California and the American Type Culture Collection, Manassas, Virginia. Human myeloma and mouse-human heteromyeloma cell lines also have been described for the production of human monoclonal antibodies [Kozbor, J. Immunol., 133:3001 (1984); Brodeur et al., Monoclonal Antibody Production Techniques and Applications, Marcel Dekker, Inc., New York, (1987) pp. 51-63].
The culture medium in which the hybridoma cells are cultured can then be assayed for the presence of monoclonal antibodies directed against PRO10282. Preferably, the binding specificity of monoclonal antibodies produced by the hybridoma cells is determined by immunoprecipitation or by an in vitro binding assay, such as radioimmunoassay (RIA) or enzyme-linked immunoabsorbent assay (ELISA). Such techniques and assays are known in the art. The binding affinity 3 0 of the monoclonal antibody can, for example, be determined by the Scatchard analysis of Munson and Pollard, Anal.
Biochem., 107:220 (1980).
After the desired hybridoma cells are identified, the clones may be subcloned by limiting dilution procedures and grown by standard methods [Goding, supra]. Suitable culture media for this purpose include, for example, Dulbecco's Modified Eagle's Medium and RPMI-1640 medium. Alternatively, the hybridoma cells may be grown in vivo as ascites in a mammal.

The monoclonal antibodies secreted by the subclones may be isolated or purified from the culture medium or ascites fluid by conventional immunoglobulin purification procedures such as, for example, protein A-Sepharose, hydroxylapatite chromatography, gel electrophoresis, dialysis, or affinity chromatography.
The monoclonal antibodies may also be made by recombinant DNA methods, such as those described in U.S.
Patent No. 4,816,567. DNA encoding the monoclonal antibodies of the invention can be readily isolated and sequenced using conventional procedures (e.g., by using oligonucleotide probes that are capable of binding specifically to genes encoding the heavy and light chains of murine antibodies). The hybridoma cells of the invention serve as a preferred source of such DNA. Once isolated, the DNA may be placed into expression vectors, which are then transfected into host cells such as simian COS cells, Chinese hamster ovary (CHO) cells, or myeloma cells that do not otherwise produce 1 0 immunoglobulin protein, to obtain the synthesis of monoclonal antibodies in the recombinant host cells. The DNA also may be modified, for example, by substituting the coding sequence for human heavy and light chain constant domains in place of the homologous murine sequences [U.S. Patent No. 4,816,567; Morrison et al., supra] or by covalently joining to the immunoglobulin coding sequence all or part of the coding sequence for a non-immunoglobulin polypeptide. Such a non-immunoglobulin polypeptide can be substituted for the constant domains of an antibody of the invention, or can be substituted for the variable domains of one antigen-combining site of an antibody of the invention to create a chimeric bivalent antibody.
The antibodies may be monovalent antibodies. Methods for preparing monovalent antibodies are well known in the art. For example, one method involves recombinant expression of immunoglobulin light chain and modified heavy chain.
The heavy chain is truncated generally at any point in the Fc region so as to prevent heavy chain crosslinking.
2 0 Alternatively, the relevant cysteine residues are substituted with another amino acid residue or are deleted so as to prevent crosslinking.
In vitro methods are also suitable for preparing monovalent antibodies.
Digestion of antibodies to produce fragments thereof, particularly, Fab fragments, can be accomplished using routine techniques known in the art.
3. Human and Humanized Antibodies The anti-PR010282 antibodies of the invention may further comprise humanized antibodies or human antibodies.
Humanized forms of non-human (e.g., murine) antibodies are chimeric immunoglobulins, immunoglobulin chains or fragments thereof (such as Fv, Fab, Fab', Flab% or other antigen-binding subsequences of antibodies) which contain minimal sequence derived from non-human immunoglobulin. Humanized antibodies include human immunoglobulins (recipient antibody) in 3 0 which residues from a complementary determining region (CDR) of the recipient are replaced by residues from a CDR of a non-human species (donor antibody) such as mouse, rat or rabbit having the desired specificity, affinity and capacity. In some instances, Fv framework residues of the human immunoglobulin are replaced by corresponding non-human residues.
Humanized antibodies may also comprise residues which are found neither in the recipient antibody nor in the imported CDR
or framework sequences. In general, the humanized antibody will comprise substantially all of at least one, and typically 3 5 two, variable domains, in which all or substantially all of the CDR
regions correspond to those of a non-human immunoglobulin and all or substantially all of the FR regions are those of a human immunoglobulin consensus sequence.
The humanized antibody optimally also will comprise at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin [Jones et al., Nature, 321:522-525 (1986);
Riechmann et al., Nature, 332:323-329 (1988); and Presta, Curr. Op. Struct. Biol., 2:593-596 (1992)].
Methods for humanizing non-human antibodies are well known in the art.
Generally, a humanized antibody has one or more amino acid residues introduced into it from a source which is non-human. These non-human amino acid residues are often referred to as "import" residues, which are typically taken from an "import" variable domain.
Humanization can be essentially performed following the method of Winter and co-workers [Jones et al., Nature, 321:522-525 (1986); Riechmann et al., Nature, 332:323-327 (1988); Verhoeyen et al., Science, 239:1534-1536 (1988)], by substituting rodent CDRs or CDR sequences for the corresponding sequences of a human antibody. Accordingly, such "humanized" antibodies are chimeric antibodies (U.S. Patent No. 4,816,567), wherein substantially less than an intact human variable domain has been substituted by the corresponding sequence from a non-human species. In practice, humanized antibodies are typically human antibodies in which some CDR residues and possibly some FR residues are substituted by residues from analogous sites in rodent antibodies.
Human antibodies can also be produced using various techniques known in the art, including phage display libraries [Hoogenboonn and Winter, J. Mol. Biol., 227:381 (1991); Marks et al., J. Mol. Biol., 222:581 (1991)]. The techniques of Cole et al. and Boerner et al. are also available for the preparation of human monoclonal antibodies (Cole et al., Monoclonal Antibodies and Cancer Therapy, Alan R. Liss, p. 77 (1985) and Boerner et al., J. Immunol., 147(1):86-95 (1991)]. Similarly, human antibodies can be made by introducing of human immunoglobulin loci into transgenic animals, e.g., mice in which the endogenous immunoglobulin genes have been partially or completely inactivated. Upon challenge, human antibody production is observed, which closely resembles that seen in humans in all respects, including gene rearrangement, assembly, and antibody repertoire. This approach is described, for example, in U.S. Patent Nos. 5,545,807;
5,545,806; 5,569,825; 5,625,126; 5,633,425; 5,661,016, and in the following scientific publications: Marks et al., Biorfechnology 10, 779-783 (1992); Lonberg et al., Nature 368 856-859 (1994);
Morrison, Nature 368, 812-13 (1994);
Fishwild et al., Nature Biotechnology 14, 845-51 (1996); Neuberger, Nature Biotechnology 14, 826 (1996); Lonberg and Huszar, Intern. Rev. Immunol. 13 65-93 (1995).
4. Bispecific Antibodies Bispecific antibodies are monoclonal, preferably human or humanized, antibodies that have binding specificities for at least two different antigens. In the present case, one of the binding specificities is for the PRO10282, the other one is for any other antigen, and preferably for a cell-surface protein or receptor or receptor subunit.
Methods for making bispecific antibodies are known in the art. Traditionally, the recombinant production of bispecific antibodies is based on the co-expression of two immunoglobulin heavy-chain/light-chain pairs, where the two heavy chains have different specificities [Milstein and Cuello, Nature, 305:537-539 (1983)1. Because of the random assortment of immunoglobulin heavy and light chains, these hybridomas (quadromas) produce a potential mixture of ten different antibody molecules, of which only one has the correct bispecific structure. The purification of the correct molecule is usually accomplished by affinity chromatography steps. Similar procedures are disclosed in WO 93108829, published 13 May 1993, and in Traunecker et al., EMBO J., 10:3655-3659 (1991).
Antibody variable domains with the desired binding specificities (antibody-antigen combining sites) can be fused to immunoglobulin constant domain sequences. The fusion preferably is with an immunoglobulin heavy-chain constant domain, comprising at least part of the hinge, CH2, and CH3 regions. It is preferred to have the first heavy-chain constant region (CH1) containing the site necessary for light-chain binding present in at least one of the fusions. DNAs encoding the immunoglobulin heavy-chain fusions and, if desired, the immunoglobulin light chain, are inserted into separate expression vectors, and are co-transfected into a suitable host organism. For further details of generating bispecific antibodies see, for example, Suresh et at., Methods in Enzymology, 121:210 (1986).
According to another approach described in WO 96127011, the interface between a pair of antibody molecules can be engineered to maximize the percentage of heterodimers which are recovered from recombinant cell culture. The preferred interface comprises at least a part of the CH3 region of an antibody constant domain. In this method, one or more small amino acid side chains from the interface of the first antibody molecule are replaced with larger side chains (e.g. tyrosine or tryptophan). Compensatory "cavities" of identical or similar size to the large side chain(s) are created on the interface of the second antibody molecule by replacing large amino acid side chains with smaller ones (e.g. alanine or threonine). This provides a mechanism for increasing the yield of the heterodimer over other unwanted end-products such as homodimers.
Bispecific antibodies can be prepared as full length antibodies or antibody fragments (e.g. F(abl2 bispecific 2 0 antibodies). Techniques for generating bispecific antibodies from antibody fragments have been described in the literature.
For example, bispecific antibodies can be prepared can be prepared using chemical linkage. Brennan et al., Science 229:81 (1985) describe a procedure wherein intact antibodies are proteolytically cleaved to generate F(alf)2 fragments. These fragments are reduced in the presence of the dithiol complexing agent sodium arsenite to stabilize vicinal dithiols and prevent intermolecular disulfide formation. The Fab' fragments generated are then converted to thionitrobenzoate (TNB) 2 5 derivatives. One of the Fab'-TNB derivatives is then reconverted to the Fab'-thiol by reduction with nnercaptoethylamine and is mixed with an equimolar amount of the other Fab'-TNB derivative to form the bispecific antibody. The bispecific antibodies produced can be used as agents for the selective immobilization of enzymes.
Fab' fragments may be directly recovered from E. coli and chemically coupled to form bispecific antibodies.
Shalaby et at., J. Exp. Med. 175:217-225 (1992) describe the production of a fully humanized bispecific antibody F(abl2 3 0 molecule. Each Fab' fragment was separately secreted from E. coli and subjected to directed chemical coupling in vitro to form the bispecific antibody. The bispecific antibody thus formed was able to bind to cells overexpressing the ErbB2 receptor and normal human T cells, as well as trigger the lytic activity of human cytotoxic lymphocytes against human breast tumor targets.
Various technique for making and isolating bispecific antibody fragments directly from recombinant cell culture 3 5 have also been described. For example, bispecific antibodies have been produced using leucine zippers. Kostelny et at., J. Immunol. 148(51:1547-1553 (1992). The leucine zipper peptides from the Fos and Jun proteins were linked to the Fab' portions of two different antibodies by gene fusion. The antibody homodimers were reduced at the hinge region to form monomers and then re-oxidized to form the antibody heteradimers. This method can also be utilized for the production of antibody homodimers. The "diabody" technology described by Hollinger et al., Proc. Natl. Acad. Sci. USA 90:6444-6448 (1993) has provided an alternative mechanism for making bispecific antibody fragments. The fragments comprise a heavy-chain variable domain (VH) connected to a light-chain variable domain (1/1) by a linker which is too short to allow pairing between the two domains on the same chain. Accordingly, the VH and V1 domains of one fragment are forced to pair with the complementary VI. and VH domains of another fragment, thereby forming two antigen-binding sites. Another strategy for making bispecific antibody fragments by the use of single-chain Fv (sFv) dinners has also been reported. See, Gruber et al., J. Immunol. 152:5368 (1994). Antibodies with more than two valencies are contemplated. For example, trispecific antibodies can be prepared. Tutt et al., J. Immunol. 147:60 (1991).
Exemplary bispecific antibodies may bind to two different epitopes on a given PRO10282 polypeptide herein.
Alternatively, an anti-PRO10282 polypeptide arm may be combined with an arm which binds to a triggering molecule on a leukocyte such as a 1-cell receptor molecule (e.g. CD2, CD3, CO28, or B7), or Fc receptors for IgG (Fc R), such as Fc RI
(CD64), Fc RII (CD32) and Fc Rill (CD16) so as to focus cellular defense mechanisms to the cell expressing the particular PR010282 polypeptide. Bispecyic antibodies may also be used to localize cytotoxic agents to cells which express a particular PRO10282 polypeptide. These antibodies possess a PRO10282-binding arm and an arm which binds a cytotoxic agent or a radionuclide chelator, such as EOTUBE, DPTA, DOTA, or TETA. Another bispecific antibody of interest binds the PRO10282 polypeptide and further binds tissue factor (IF).
5. Heteroconiugate Antibodies Heteroconjugate antibodies are also within the scope of the present invention.
Heteroconjugate antibodies are composed of two covalently joined antibodies. Such antibodies have, for example, been proposed to target immune system cells to unwanted cells [U.S. Patent No. 4,676,980], and for treatment of HIV
infection [WO 91100360; WO 921200373;
2 5 EP 03089]. It is contemplated that the antibodies may be prepared in vitro using known methods in synthetic protein chemistry, including those involving crosslinking agents. For example, immunotoxins may be constructed using a disulfide exchange reaction or by forming a thioether bond. Examples of suitable reagents for this purpose include iminothiolate and methyl-4-nnercaptobutyrimidate and those disclosed, for example, in U.S.
Patent No. 4,676,980.
3 0 6. Effector Function Engineering It may be desirable to modify the antibody of the invention with respect to effector function, so as to enhance, e.g., the effectiveness of the antibody in treating cancer. For example, cysteine residue(s) may be introduced into the Fc region, thereby allowing interchain disulfide bond formation in this region.
The homodimeric antibody thus generated may have improved internalization capability and/or increased complement-mediated cell killing and antibody-dependent cellular 35 cytotoxicity (ADCC). See Caron et al., J. Exp Med., 176: 1191-1195 (1992) and Shopes, J. Immunol., 148: 2918-2922 (1992). Homodimeric antibodies with enhanced anti-tumor activity may also be prepared using heterobifunctional cross-linkers as described in Wolff et al. Cancer Research, 53: 2560-2565 (1993).
Alternatively, an antibody can be engineered that has dual Fc regions and may thereby have enhanced complement lysis and ADCC capabilities. See Stevenson et al., Anti-Cancer Druo Design, 3: 219-230 (1989).
7. Immunoconjunates The invention also pertains to immunoconjugates comprising an antibody conjugated to a cytotoxic agent such as a chemotherapeutic agent, toxin (e.g., an enzymatically active toxin of bacterial, fungal, plant, or animal origin, or fragments thereof), or a radioactive isotope (i.e., a radioconjugate).
Chemotherapeutic agents useful in the generation of such immunoconjugates have been described above.
Enzymatically active toxins and fragments thereof that can be used include diphtheria A chain, nonbinding active fragments of diphtheria toxin, exotoxin A chain (from Pseudomonas aeruginosa), ricin A
chain, abrin A chain, modeccin A chain, alpha-sarcin, Aleurites fordii proteins, dianthin proteins, Phytolaca americana proteins (PAPI, PAPII, and PAP-S), momordica charantia inhibitor, curcin, crotin, sapaonaria officinalis inhibitor, gelonin, mitogellin, restrictocin, phenomycin, enomycin, and the tricothecenes. A variety of radionuclides are available for the production of radioconjugated antibodies. Examples include 212Bi, 1311, 1311n, "Y, and 186Re.
Conjugates of the antibody and cytotoxic agent are made using a variety of bifunctional protein-coupling agents such as N-succinimidy1-3-(2-pyridyldithiol) propionate (SPDP), iminothiolane (IT), bifunctional derivatives of imidoesters (such as dimethyl adipimidate HCL), active esters (such as disuccinimidyl suberate), aldehydes (such as glutareldehyde), 2 0 bis-azido compounds (such as his (p-azidobenzoyl) hexanediamine), bis-diazonium derivatives (such as bis-(p-diazoniumbenzoy1)-ethylenediamine), diisocyanates (such as tolyene 2,6-diisocyanate), and bis-active fluorine compounds (such as 1,5-difluoro-2,4-dinitrobenzene). For example, a ricin immunotoxin can be prepared as described in Vitetta et al., Science, 238: 1098 (1987). Carbon-14-labeled 1-isothiocyanatobenzy1-3-methyldiethylene triaminepentaacetic acid (MX-DTPA) is an exemplary chelating agent for conjugation of radionucleotide to the antibody. See W094111026.
2 5 In another embodiment, the antibody may be conjugated to a "receptor" (such streptavidin) for utilization in tumor pretargeting wherein the antibody-receptor conjugate is administered to the patient, followed by removal of unbound conjugate from the circulation using a clearing agent and then administration of a "ligand" (e.g., avidin) that is conjugated to a cytotoxic agent (e.g., a radionucleotide).
3 0 8. Immunoliposomes The antibodies disclosed herein may also be formulated as immunoliposomes.
Liposomes containing the antibody are prepared by methods known in the art, such as described in Epstein et al., Proc. Natl. Acad. Sci. USA, 82: 3688 (1985);
Hwang et al., Proc. Natl Acad. Sci. USA, 77: 4030 (1980); and U.S. Pat. Nos.
4,485,045 and 4,544,545. Liposomes with enhanced circulation time are disclosed in U.S. Patent No. 5,013,556.

Particularly useful liposomes can be generated by the reverse-phase evaporation method with a lipid composition comprising phosphatidylcholine, cholesterol, and PEG-derivatized phosphatidylethanolamine (PEG-PE). Liposomes are extruded through filters of defined pore size to yield liposomes with the desired diameter. Fab fragments of the antibody of the present invention can be conjugated to the liposomes as described in Martin et at ., J. Biol. Chem., 257: 286-288 (1982) via a disulfide-interchange reaction. A chemotherapeutic agent (such as Doxorubicin) is optionally contained within the liposome. See Gabizon et al., J. National Cancer Inst., 81(19): 1484 (1989).
8. Antibody-Dependent Enzyme Mediated Prodrup Therapy (ADEPT) The antibodies of the present invention may also be used in ADEPT by conjugating the antibody to a prodrug-activating enzyme which converts a prodrug (e.g., a peptidyl chemotherapeutic agent, see WO 81101145) to an active anti-cancer drug. See, for example, WO 88/07378 and U. S. Patent No. 4,975,278.
The enzyme component of the innmunoconjugate useful for ADEPT includes any enzyme capable of acting on a prodrug in such as way so as to convert it into its more active, cytotoxic form.
Enzymes that are useful in the method of this invention include, but are not limited to, glycosidase, glucose oxidase, human lysosyme, human glucuronidase, alkaline phosphatase useful for converting phosphate-containing prodrugs into free drugs; arylsulfatase useful for converting sulfate-containing prodrugs into free drugs; cytosine deaminase useful for converting non-toxic 5-fluorocytosine into the anti-cancer drug 5-fluorouracil; proteases, such as serratia protease, thernnolysin, subtilisin, carboxypeptidases (e.g., carboxypeptidase 02 and carboxypeptidase A) and cathepsins (such as cathepsins B and L), that are useful for converting peptide-containing prodrugs into free drugs; D-alanylcarboxypeptidases, useful for converting prodrugs that contain D-amino acid substituents;
carbohydrate-cleaving enzymes such as -2 0 galactosidase and neuraminidase useful for converting glycosylated prodrugs into free drugs; -lactamase useful for converting drugs derivatized with -lactanns into free drugs; and penicillin amidases, such as penicillin Vamidase or penicillin amidase, useful for converting drugs derivatized at their amine nitrogens with phenoxyacetyl or phenylacetyl groups, respectively, into free drugs. Alternatively, antibodies with enzymatic activity, also known in the art as "abzymes" can be used to convert the prodrugs of the invention into free active drugs (see, e.g., Massey, Nature, 328:457-458 (1987)).
2 5 Antibody-abzyme conjugates can be prepared as described herein for delivery of the abzyme to a tumor cell population.
The enzymes of this invention can be covalently bound to the anti-PR010282 (anti-Stra6) antibodies by techniques well known in the art such as the use of the heterobifunctional cross-linking agents discussed above.
Alternatively, fusion proteins comprising at least the antigen binding region of the antibody of the invention linked to at least a functionally active portion of an enzyme of the invention can be constructed using recombinant DNA techniques 3 0 well known in the art (see, e.g., Neuberger et at., Nature, 312:604-608 (1984)).
10. Pharmaceutical Compositions of Antibodies Antibodies specifically binding a PRO10282 polypeptide identified herein, as well as other molecules identified by the screening assays disclosed hereinbef ore, can be administered for the treatment of various disorders in the form of 35 pharmaceutical compositions.

If the PRO10282 polypeptide is intracellular and whole antibodies are used as inhibitors, internalizing antibodies are preferred. However, lipofections or liposomes can also be used to deliver the antibody, or an antibody fragment, into cells. Where antibody fragments are used, the smallest inhibitory fragment that specifically binds to the binding domain of the target protein is preferred. For example, based upon the variable-region sequences of an antibody, peptide molecules can be designed that retain the ability to bind the target protein sequence.
Such peptidestan be synthesized chemically and/or produced by recombinant DNA technology. See, e.g., Marasco et al., Proc. Natl. Acad. Sci. USA, 90: 7889-7893 (1993). The formulation herein may also contain more than one active compound as necessary for the particular indication being treated, preferably those with complementary activities that do not adversely affect each other. Alternatively, or in addition, the composition may comprise an agent that enhances its function, such as, for example, a cytotoxic agent, cytokine, chemotherapeutic agent, or growth-inhibitory agent. Such molecules are suitably present in combination in amounts that are effective for the purpose intended. .
The active ingredients may also be entrapped in microcapsules prepared, for example, by coacervation techniques or by interfacial polymerization, for example, hydroxymethylcellulose or gelatin-microcapsules and poly-(methylmethacylate) microcapsules, respectively, in colloidal drug delivery systems (for example, liposonnes, albumin nnicrospheres, microemulsions, nano-particles, and nanocapsules) or in macroemulsions. Such techniques are disclosed in Rennington's Pharmaceutical Sciences, supra.
The formulations to be used for in vivo administration must be sterile. This is readily accomplished by filtration through sterile filtration membranes.
Sustained-release preparations may be prepared. Suitable examples of sustained-release preparations include 2 0 semipermeable matrices of solid hydrophobic polymers containing the antibody, which matrices areln the form of shaped articles, e.g., films, or microcapsules. Examples of sustained-release matrices include polyesters, hydrogels (for example, poly(2-hydroxyethyl-methacrylate), or poly(vinylalcohol)), polylactides (U.S.
Pat. No. 3,773,919), copolymers of L-glutamic acid and ethyl-L-glutamate, non-degradable ethylene-vinyl acetate, degradable lactic acid-glycolic acid copolymers such as the LUPRON DEPOT TM (injectable microspheres composed of lactic acid-glycolic acid copolymer and leuprolide acetate), 2 5 and poly-D-(-)-3-hydroxybutyric acid. While polymers such as ethylene-vinyl acetate and lactic acid-glycolic acid enable release of molecules for over 100 days, certain hydrogels release proteins for shorter time periods. When encapsulated antibodies remain in the body for a long time, they may denature or aggregate as a result of exposure to moisture at 37 C, resulting in a loss of biological activity and possible changes in immunogenicity. Rational strategies can be devised for stabilization depending on the mechanism involved. For example, if the aggregation mechanism is discovered to be 3 0 intermolecular S-S bond formation through thio-disulfide interchange, stabilization may be achieved by modifying sulfhydryl residues, lyophilizing from acidic solutions, controlling moisture content, using appropriate additives, and developing specific polymer matrix compositions.
0. Uses for anti-PRO10282 (anti-Stra6) Antibodies 3 5 The anti-PRO10282 antibodies of the invention have various utilities.
For example, anti-PRO10282 antibodies may be used in diagnostic assays for PR010282, e.g., detecting its expression in specific cells, tissues, or serum. Various diagnostic assay techniques known in the art may be used, such as competitive binding assays, direct or indirect sandwich assays and immunoprecipitation assays conducted in either heterogeneous or homogeneous phases [Zola, Monoclonal Antibodies: A Manual of Techniques, CRC Press, Inc. (1987) pp. 147-1581. The antibodies used in the diagnostic assays can be labeled with a detectable moiety. The detectable moiety should be capable of producing, either directly or indirectly, a detectable signal. For example, the detectable moiety may be a radioisotope, such as 3F1, 14C, 32p, 35^, or 1251, a fluorescent or chemiluminescent compound, such as fluorescein isothiocyanate, rhodamine, or luciferin, or an enzyme, such as alkaline phosphatase, beta-galactosidase or horseradish peroxidase. Any method known in the art for conjugating the antibody to the detectable moiety may be employed, including those methods described by Hunter et al., Nature, 144:945 (1962); David et al., Biochemistry, 13:1014 (1974); Pain et al., J. Immunol.
Meth., 40:219 (1981); and Nygren, J.
Histochem. and Cytochem., 30:407 (1982).
Anti-PR010282 antibodies also are useful for the affinity purification of PR010282 from recombinant cell culture or natural sources. In this process, the antibodies against PRO10282 are immobilized on a suitable support, such a Sephadex resin or filter paper, using methods well known in the art. The immobilized antibody then is contacted with a sample containing the PR010282 to be purified, and thereafter the support is washed with a suitable solvent that will remove substantially all the material in the sample except the PRO10282, which is bound to the immobilized antibody.
Finally, the support is washed with another suitable solvent that will release the PRO10282 from the antibody.
Anti-PR010282 antibodies can be used to monitor the expression of PR010282 polypeptide which has a potential use as a sensitive method to screen for therapeutically useful compounds against a wide variety of retinoid 2 0 responsive diseases. Among the types of diseases contemplated as therapeutic targets include psoriasis, acne, dysplasias, cancers and autoimmune diseases. The category of contemplated dysplasias includes precancerous lesions of the epithelial tissues such as oral leukoplakias, dysplasia of the cervix, larynx and bronchi. The category of contemplated cancers includes carcinomas of colon (adenocarcinoma), lung (adenocarcinoma and carcinoma), skin, head and neck, cervix, uterus, breast and prostate. The category of contemplated autoimmune diseases includes rheumatoid arthritis, osteoarthritis, 2 5 systemic lupus erythematosus, pemphigus valgaris and pemphigus foliaceous. Anti-PRO10282 antibodies have potential therapeutic use in the treatment of such diseases and conditions.
P. Methods of Treatment Using anti-PR010282 (Stra6) antibodies and other Stra6 antagonists It is contemplated that the antibodies and other anti-tumor compounds of the present invention may be used to 3 0 treat various conditions, including those characterized by overexpression and/or activation of the amplified genes identified herein. Exemplary conditions or disorders to be treated with such antibodies and other compounds, including, but not limited to, small organic and inorganic molecules, peptides, antisense molecules, etc., include benign or malignant tumors (e.g., renal, liver, kidney, bladder, breast, gastric, ovarian, colorectal, prostate, pancreatic, lung, vulval, thyroid, hepatic carcinomas; sarcomas; glioblastomas; and various head and neck tumors);
leukemias and lymphoid malignancies; other 35 disorders such as neuronal, glial, astrocytal, hypothalamic and other glandular, macrophagal, epithelial, stromal and blastocoelic disorders; and inflammatory, angiogenic and immunologic disorders. Particularly preferred targets for treatment with anti-Stra6 antibodies and other antagonists of the present invention are tumors that harbor genetic defects in the Wnt-1 pathway (e.g. leading to abnormal activation of 13-catenin signaling) and/or overexpress Stra6. It has been found that human cancers harboring genetic defects in the VI/tit-1 pathway typically also exhibit overexpression of Stra6, but not all Stra6 overexpressing tumors have been known to be associated with mutations in the Wnt-1 pathway. The Stra6 antagonists of the present invention have great potential in the treatment of Stra6 overexpressing tumors. A
preferred group of such tumors includes colorectal tumors, ovary, endometrium, and Wilm's kidney tumors, melanoma, and pheochromocytome (a tumor derived from the adrenal medulla).
The anti-tumor agents of the present invention, e.g., antibodies, are administered to a mammal, preferably a 1 0 human, in accord with known methods, such as intravenous administration as a bolus or by continuous infusion over a period of time, by intramuscular, intraperitoneal, intracerobrospinal, subcutaneous, intra-articular, intrasynovial, intrathecal, oral, topical, or inhalation routes. Intravenous administration of the antibody is preferred.
Other therapeutic regimens may be combined with the administration of the anti-cancer agents, e.g., antibodies of the instant invention. For example, the patient to be treated with such anti-cancer agents may also receive radiation therapy. Alternatively, or in addition, a chemotherapeutic agent may be administered to the patient. Preparation and ' dosing schedules for such chemotherapeutic agents may be used according to manufacturers instructions or as determined empirically by the skilled practitioner. Preparation and dosing schedules for such chemotherapy are also described in Chemotherapy Service Ed., M.G. Perry, Williams & Wilkins, Baltimore, MD
(1992). The chemotherapeutic agent may precede, or follow administration of the anti-tumor agent, e.g., antibody, or may be given simultaneously therewith. The 2 0 antibody may be combined with an anti-oestrogen compound such as tamoxifen or an anti-progesterone such as onapristone (see, EP 616812) in dosages known for such molecules.
It may be desirable to also administer antibodies against other tumor associated antigens, such as antibodies which bind to the ErbB2, EGFR, ErbB3, ErbB4, or vascular endothelial factor (VEGF). Alternatively, or in addition, two or more antibodies binding the same or two or more different antigens disclosed herein may be co-administered to the patient.
2 5 Sometimes, it may be beneficial to also administer one or more cytokines to the patient. In a preferred embodiment, the antibodies herein are co-administered with a growth inhibitory agent. For example, the growth inhibitory agent may be administered first, followed by an antibody of the present invention. However, simultaneous administration or administration of the antibody of the present invention first is also contemplated. Suitable dosages for the growth inhibitory agent are those presently used and may be lowered due to the combined action (synergy) of the growth inhibitory 3 0 agent and the antibody herein.
For the prevention or treatment of disease, the appropriate dosage of an anti-tumor agent, e.g., an antibody herein will depend on the type of disease to be treated, as defined above, the severity and course of the disease, whether the agent is administered for preventive or therapeutic purposes, previous therapy, the patient's clinical history and response to the agent, and the discretion of the attending physician. The agent is suitably administered to the patient at 3 5 one time or over a series of treatments.

For example, depending on the type and severity of the disease, about 1 glkg to 15 mg/kg (e.g., 0.1-20 mg/kg) of antibody is an initial candidate dosage for administration to the patient, whether, for example, by one or more separate administrations, or by continuous infusion. A typical daily dosage might range from about 1 glkg to 100 mg/kg or more, depending on the factors mentioned above. For repeated administrations over several days or longer, depending on the condition, the treatment is sustained until a desired suppression of disease symptoms occurs. However, other dosage regimens may be useful. The progress of this therapy is easily monitored by conventional techniques and assays.
Ii Articles of Manufacture In another embodiment of the invention, an article of manufacture containing materials useful for the diagnosis or treatment of the disorders described above is provided. The article of manufacture comprises a container and a label.
Suitable containers include, for example, bottles, vials, syringes, and test tubes. The containers may be formed from a variety of materials such as glass or plastic. The container holds a composition which is effective for diagnosing or treating the condition and may have a sterile access port (for example the container may be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle). The active agent in the composition is usually an anti-tumor agent capable of interfering with the activity of a gene product identified herein, e.g., an antibody. The label on, or associated with, the container indicates that the composition is used for diagnosing or treating the condition of choice. The article of manufacture may further comprise a second container comprising a pharmaceutically-acceptable buffer, such as phosphate-buffered saline, Ringers solution and dextrose solution. It may further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, needles, syringes, and package 2 0 inserts with instructions for use.
R. Diagnosis and Prognosis of Tumors While cell surface proteins, such as growth receptors overexpressed in certain tumors are excellent targets for drug candidates or tumor (e.g., cancer) treatment, the same proteins along with secreted proteins encoded by the genes amplified in tumor cells find additional use in the diagnosis and prognosis of tumors. For example, antibodies directed against the protein products of genes amplified in tumor cells can be used as tumor diagnostics or prognostics.
For example, antibodies, including antibody fragments, can be used to qualitatively or quantitatively detect the expression of proteins encoded by the amplified genes ("marker gene products"). The antibody preferably is equipped with a detectable, e.g., fluorescent label, and binding can be monitored by light microscopy, flow cytometry, fluorimetry, or other 3 0 techniques known in the art. These techniques are particularly suitable, if the amplified gene encodes a cell surface protein, e.g., a growth factor. Such binding assays are performed essentially as described in section 5 above.
In situ detection of antibody binding to the marker gene products can be performed, for example, by immunofluorescence or immunoelectron microscopy. For this purpose, a histological specimen is removed from the patient, and a labeled antibody is applied to it, preferably by overlaying the antibody on a biological sample. This procedure also 3 5 allows for determining the distribution of the marker gene product in the tissue examined. It will be apparent for those skilled in the art that a wide variety of histological methods are readily available for in situ detection.
The following examples are offered for illustrative purposes only, and are not intended to limit the scope of the present invention in any way.
EXAMPLES
Commercially available reagents referred to in the examples were used according to manufacturer's instructions unless otherwise indicated. The source of those cells identified in the following examples, and throughout the specification, by ATCC accession numbers is the American Type Culture Collection, Manassas, VA. In the following Examples, unless otherwise specified, "Sra6" will refer to native sequence P8010282 polypeptide.

Isolation of cDNA Clones Encoding a Human PR010282 and P8019578 nolvpeptides cDNA clones (DNA148380-2827 and DNA148389-2827-1) encoding native human P8010282 and P8019578 polypeptides were identified using a yeast screen, in a human fetal brain cDNA
library that preferentially represents the 5' ends of the primary cDNA clones.
Clone DNA148380-2827 contains a single open reading frame with an apparent translational initiation site at nucleotide positions 49-51 and ending at the stop codon at nucleotide positions 2050-2052 (Figure 1). The predicted polypeptide precursor is 667 amino acids long (Figure 2). The full-length P8010282 protein shown in Figure 2 has an estimated molecular weight of about 73502 daltons and a pl of about 9.26.
Analysis of the full-length P8010282 sequence shown in Figure 2 (SE0 ID N0:2) evidences the presence of a variety of important polypeptide domains as shown in Figure 2, wherein the locations given for those important polypeptide domains are approximate as described above. Clone DNA148380-2827 has been deposited with ATCC on January 11, 2000 and is assigned ATCC Deposit No. PTA-1181.
Clone 0NA148389-2827-1 contains a single open reading frame with an apparent translational initiation bite at nucleotide positions 186-188 and ending at the stop codon at nucleotide positions 2160-2162 (Figure 6, SEQ ID NO: 4).
The predicted polypeptide precursor is 658 amino acids long (Figure 7, SEQ ID
NO: 5). The full-length P8019578 protein shown in Figure 7 has an estimated molecular weight of about 72583 daltons and a pl of about 9.36. Analysis of the full-length P8019578 sequence shown in Figure 7 (SE(1 ID NO: 5) evidences the presence of a variety of important polypeptide domains as shown in Figure 7, wherein the locations given for those important polypeptide domains are approximate as described above. Noteworthy is the presence of nine potential transmembrane domains and fourteen cysteine residues conserved between the human and the corresponding mouse sequence. While mouse Stra6 has three potential N-linked glycosylation sites, the human P8019578 (native human Stra6) polypeptide has one. Clone 148389-2827-1 has been =

deposited with ATCC on February 23, 2000, and is assigned ATCC Deposit No. PTA-1402.
An analysis of the Dayhoff database (version 35.45 SwissProt 35), using the ALIGN-2 sequence alignment analysis of the full-length sequence shown in Figure 2 (SEQ ID NO:2), and of the full-length sequence shown in Figure 7 (SEQ ID NO: 5) evidenced sequence identity between the PR010282 amino acid sequence and the following Dayhoff sequences: AF062476, P_W88559 and HGS_RE259.
As shown in Figure 8, comparison of the full-length human PRO10282 and PRO19578 polypeptides shows that PR019578 contains a deletion of nine amino acids (SPVDFLAGD; SEQ ID NO: 13) at positions 89-97 of the PRO10282 amino acid sequence. In addition, PRO19578 contains an isoleucine (I) at amino acid position 518 in place of methionine (M) at the corresponding position (position 527) of PR010282, which results from a GIA polymorphism at this position.
Both the PR010282 and the native sequence PR019578 polypeptides are believed to be the human homologues of the mouse Stra6 polypeptide, and are, therefore, also referred to as "Stra6."
Mouse Stra6 and the native sequence human full-length PRO10282 polypeptide encoded by DNA148340-2827 show about 74%
amino acid sequence identity.
Figure 9 shows the hydrophobicity plot of the native sequence human full-length PRO10282 polypeptide encoded by DNA148380-2827, briefly referred to as "human Stra6." As shown in Figure 9, about 50% of the amino acid residues in this 667 amino acids long polypeptide are hydrophobic.
The human Stra6 gene was localized to chromosome 15q23 as determined by UNIGENE. Preliminary fine mapping indicates that Stra6 is located in the STS interval D15S124-D15S160 and the GeneMap'98 position corresponds to 244.52 on the 03 map.

Use of PR010282 and PRO19578 as a hybridization probe The following method describes use of a nucleotide sequence encoding PR010282 and PR019578 as a hybridization probe.
DNA comprising the coding sequence of full-length or mature PRO10282 or PR019578 is employed as a probe to screen for homologous DNAs (such as those encoding naturally-occurring variants of PRO10282 or PR019578) in human tissue cDNA libraries or human tissue genomic libraries.
Hybridization and washing of filters containing either library DNAs is performed under the following high stringency conditions. Hybridization of radiolabeled PRO10282-derived probe to the filters is performed in a solution of 50% formamide, 5x SSC, 0.1% SDS, 0.1% sodium pyrophosphate, 50 rnM sodium phosphate, pH 6.8, 2x Denhardt's solution, and 10% dextran sulfate at 42 C for 20 hours. Washing of the filters is performed in an aqueous solution of 0.1x SSC and 0.1% SOS at 42 C.
DNAs having a desired sequence identity with the DNA encoding full-length native sequence PRO10282 or PR019578 can then be identified using standard techniques known in the art.

Expression of PRO10282 and PRO19578 in E. coli This example illustrates preparation of an unglycosylated form of PR010282 or PR019578 by recombinant expression in E. coli.
The DNA sequence encoding PR010282 or PR019578 is initially amplified using selected PCR primers. The primers should contain restriction enzyme sites which correspond to the restriction enzyme sites on the selected expression vector. A variety of expression vectors may be employed. An example of a suitable vector is pBR322 (derived from E. coil;
see Bolivar et al., Gene, 2:95 (1977)) which contains genes for ampicillin and tetracycline resistance. The vector is digested with restriction enzyme and dephosphorylated. The PCR amplified sequences are then ligated into the vector.
The vector will preferably include sequences which encode for an antibiotic resistance gene, a trp promoter, a polyhis leader (including the first six STII codons, polyhis sequence, and enterokinase cleavage site), the PRO10282 or PRO19578 coding region, lambda transcriptional terminator, and an argU gene.
The ligation mixture is then used to transform a selected E. coli strain using the methods described in Sambrook at al., supra. Transformants are identified by their ability to grow on LB
plates and antibiotic resistant colonies are then selected. Plasmid DNA can be isolated and confirmed by restriction analysis and DNA sequencing.
Selected clones can be grown overnight in liquid culture medium such as LB
broth supplemented with antibiotics.
The overnight culture may subsequently be used to inoculate a larger scale culture. The cells are then grown to a desired optical density, during which the expression promoter is turned on.
After culturing the cells for several more hours, the cells can be harvested by centrifugation. The cell pellet obtained by the centrifugation can be solubilized using various agents known in the art, and the solubilized PR010282 protein can then be purified using a metal chelating column under conditions that allow tight binding of the protein.
PR010282 or PR019578 may be expressed in E. call in a poly-His tagged form, using the following procedure.
The DNA encoding PR010282 or PR019578 is initially amplified using selected PCR primers. The primers will contain restriction enzyme sites which correspond to the restriction enzyme sites on the selected expression vector, and other useful sequences providing for efficient and reliable translation initiation, rapid purification on a metal chelation column, and proteolytic removal with enterokinase. The PCR-amplified, poly-His tagged sequences are then ligated into an expression vector, which is used to transform an E. coli host based on strain 52 (W3110 fuhA(tonA) Ion galE rpoHts(htpRts) cIpP(laclq). Transformants are first grown in LB containing 50 mgIml carbenicillin at 30 C with shaking until an 0Ø600 of 3-5 is reached. Cultures are then diluted 50-100 fold into CRAP media (prepared by mixing 3.57 g (NH4)2304, 0.71 g 3 0 sodium citrate 2H20, 1.07 g KCI, 5.36 g Difco yeast extract, 5.36 g Sheffield hycase SF in 500 mL water, as well as 110 mM MPOS, pH 7.3, 0.55% (wIv) glucose and 7 mM MgSO4) and grown for approximately 20-30 hours at 30 C with shaking. Samples are removed to verify expression by SOS-PAGE analysis, and the bulk culture is centrifuged to pellet the cells. Cell pellets are frozen until purification and refolding.
E. coli paste from 0.5 to 1 L fermentations (6-10 g pellets) is resuspended in 10 volumes (wlv) in 7 M guanidine, 3 5 20 mM Tris, pH 8 buffer. Solid sodium sulfite and sodium tetrathionate is added to make final concentrations of 0.1M

and 0.02 M, respectively, and the solution is stirred overnight at 4 C. This step results in a denatured protein with all cysteine residues blocked by sulfitolization. The solution is centrifuged at 40,000 rpm in a Beckman Ultracentrifuge for 30 min. The supernatant is diluted with 3-5 volumes of metal chelate column buffer (6 M guanidine, 20 mM Iris, pH 7.4) and filtered through 0.22 micron filters to clarify. The clarified extract is loaded onto a 5 ml Qiagen Ni-NTA metal chelate column equilibrated in the metal chelate column buffer. The column is washed with additional buffer containing 50 mM
imidazole (Calbiochem, Utrol grade), pH 7.4. The protein is eluted with buffer containing 250 mM imidazole. Fractions containing the desired protein are pooled and stored at 4 C. Protein concentration is estimated by its absorbance at 280 nm using the calculated extinction coefficient based on its amino acid sequence.
The proteins are refolded by diluting the sample slowly into freshly prepared refolding buffer consisting of: 20 mM Tris, pH 8.6, 0.3 M NaCI, 2.5 M urea, 5 mM cysteine, 20 mM glycine and 1 mM
EDTA. Refolding volumes are chosen so that the final protein concentration is between 50 to 100 micrograms/ml.
The refolding solution is stirred gently at 4 C
for 12-36 hours. The refolding reaction is quenched by the addition of TFA to a final concentration of 0.4% (pH of approximately 3). Before further purification of the protein, the solution is filtered through a 0.22 micron filter and acetonitrile is added to 2-10% final concentration. The refolded protein is chromatographed on a Poros R1111 reversed phase column using a mobile buffer of 0.1% TFA with elution with a gradient of acetonitrile from 10 to 80%. Aliquots of fractions with A280 absorbance are analyzed on SOS polyacrylamide gels and fractions containing homogeneous refolded protein are pooled. Generally, the properly refolded species of most proteins are eluted at the lowest concentrations of acetonitrile since those species are the most compact with their hydrophobic interiors shielded from interaction with the reversed phase resin. Aggregated species are usually eluted at higher acetonitrile concentrations. In addition to resolving 2 0 misfolded forms of proteins from the desired form, the reversed phase step also removes endotoxin from the samples.
Fractions containing the desired folded PR010282 or PR019578 polypeptide are pooled and the acetonitrile removed using a gentle stream of nitrogen directed at the solution. Proteins are formulated into 20 mM Hepes, pH 6.8 with 0.14 M sodium chloride and 4% mannitol by dialysis or by gel filtration using 625 Superfine (Pharmacia) resins equilibrated in the formulation buffer and sterile filtered.
Specifically, two extracellular domains (ECD) of the native human Stra6 protein PRO10282, Pepetide A (amino acids 229-295) and Pepetide B (amino acids 532-667) were expressed separately as peptides in the E. coli cytoplasm with an N-terminal polyhistidine leader having the amino acid sequence MKHOHQHQHQHQHQMHQ (SEQ ID NO: 12). This leader provides for optimal translation initiation, purification on a nickel chelation column, and efficient removal if desired with the TAGZyme system (Unizyme Laboratories). Transcription was controlled by the E. coli alkaline phosphatase promoter - 30 (Kikuchi et al., Nucleic Acids Res. 9:5671-5678 [19811) and the trp operon ribosome binding site (Yanofsky et al, Nucleic Acids Res. 9:6647-6668 [1981]) provided for translation. Downstream pf the translation termination codon, is the Xto transcriptional terminator (Scholtissek and Grosse, Nucleic Acids Res. 15:3185 [19871) followed by the rare codon tRNA
genes pro2, argU, and glyT (Komine at al., J. Mol. Biol. 212:579-598 [1990];
Fournier and Ozeki, Microbiol. Rev. 49:379-397 [1985]).
The two Stra6 ECD coding sequence DNA fragments were prepared by PCR from a full length cDNA clone, and inserted into the expression vector described above, which was designated as pST239. After DNA sequence verification, the new Stra6 expression plasmids, designated PE148380A and PE1483806, were transformed into the E. coli strain 58F3 ((fhuAA(tonAA) lonA galE rpoHts (htpRts) AcIpP laclq AompTA (nmpc-fepE) AslyD). Luria Broth cultures of these transformants were first grown overnight at 30 C, and then diluted 100 fold into a phosphate limiting media to induce the alkaline phosphatase promoter. After 24 hours at 30 C with shaking, the cultures were centrifuged, and the cell pastes frozen until the start of peptide purification.
For purification, E. coil pastes (6-10 gm pellets) were resuspended in 10 volumes (wlv) of 7 M guanidine HCI, 20 mM Iris, pH 8, buffer. Solid sodium sulfite and sodium tetrathionate were added to make final concentrations of 0.1 M and 0.02 M, respectively, and the solution was stirred overnight at 4 C. The solution was clarified by centrifugation and loaded onto a Qiagen Ni-NTA metal chelate column equilibrated in 6 M guanidine, HCI, 20 mM Iris, pH 7.4. The column was washed with additional buffer containing 50 mM
imidazole (Calbiochem, Utrol grade). The protein was eluted with buffer containing 250 mM imidazole. Fractions containing the desired protein were pooled, dialyzed against 1 mM HCI and stored at 4 C.

Expression of PR010282 and PR019578 in mammalian cells This example illustrates preparation of a potentially glycosylated form of PR010282 and PR019578 by recombinant expression in mammalian cells.
The vector, pRK5 (see EP 307,247, published March 15, 1989), is employed as the expression vector.
Optionally, the PR010282 or PR019578 DNA is ligated into pRK5 with selected restriction enzymes to allow insertion of the PR010282 DNA using ligation methods such as described in Sambrook et al., supra. The resulting vector is called pRK5-PR010282 and pRK5-PR019578, respectively.
In one embodiment, the selected host cells may be 293 cells. Human 293 cells (ATCC CCL 1573) are grown to confluence in tissue culture plates in medium such as DMEM supplemented with fetal calf serum and optionally, nutrient components andlor antibiotics. About 10 g pRK5-PR010282 or pRK5-PR019578 DNA
is mixed with about 1 g DNA
encoding the VA RNA gene rThimmappaya et al., Cell, 31:543 (1982)] and dissolved in 500 I of 1 mM Tris-HCI, 0.1 mM
EDTA, 0.227 M CaCl2. To this mixture is added, dropwise, 500 I of 50 mM HEPES
(pH 7.35), 280 mM NaCI, 1.5 mM
NaPO4, and a precipitate is allowed to form for 10 minutes at 25 C. The precipitate is suspended and added to the 293 cells and allowed to settle for about four hours at 37 C. The culture medium is aspirated off and 2 ml of 20% glycerol 3 0 in PBS is added for 30 seconds. The 293 cells are then washed with serum free medium, fresh medium is added and the cells are incubated for about 5 days.
Approximately 24 hours after the transfections, the culture medium is removed and replaced with culture medium (alone) or culture medium containing 200 Cilml 35S-cysteine and 200 Cilml 35S-methionine. After a 12 hour incubation, the conditioned medium is collected, concentrated on a spin filter, and loaded onto a 15% SDS gel. The processed gel may 3 5 be dried and exposed to film for a selected period of time to reveal the presence of PRO10282 polypeptide. The cultures containing transfected cells may undergo further incubation (in serum free medium) and the medium is tested in selected bioassays.
In an alternative technique, PR010282 or PR019578 may be introduced into 293 cells transiently using the dextran sulfate method described by Somparyrac et al., Proc. Natl. Acad. Sc., 12:7575 (1981). 293 cells are grown to maximal density in a spinner flask and 700 g pRK5-PR010282 or pRK5-PR019578 DNA is added. The cells are first concentrated from the spinner flask by centrifugation and washed with PBS. The DNA-dextran precipitate is incubated on the cell pellet for four hours. The cells are treated with 20% glycerol for 90 seconds, washed with tissue culture medium, and re-introduced into the spinner flask containing tissue culture medium, 5 glml bovine insulin and 0.1 glml bovine transferrin. After about four days, the conditioned media is centrifuged and filtered to remove cells and debris. The sample containing expressed PR010282 or PR019578 can then be concentrated and purified by any selected method, such as dialysis and/or column chromatography.
In another embodiment, PR010282 or PRO19578 can be expressed in CHO cells. The pRK5-PRO10282 or pRK5-PR019578 can be transfected into CHO cells using known reagents such as CaPO4 or DEAE-dextran. As described above, the cell cultures can be incubated, and the medium replaced with culture medium (alone) or medium containing a radiolabel such as 355-methionine. After determining the presence of PRO10282 or PRO19578 polypeptide, the culture medium may be replaced with serum free medium. Preferably, the cultures are incubated for about 6 days, and then the conditioned medium is harvested. The medium containing the expressed PR010282 or PR019578 can then be concentrated and purified by any selected method.
Epitope-tagged PR010282 or pR019578 may also be expressed in host CHO cells.
The PR010282 or PRO19578 may be subcloned out of the pRK5 vector. The subclone insert can undergo PCR to fuse in frame with a selected epitope tag such as a poly-his tag into a Baculovirus expression vector. The poly-his tagged PRO10282 or PR019578 insert can then be subcloned into a SV40 driven vector containing a selection marker such as DHFR for selection of stable clones. Finally, the CHO cells can be transfected (as described above) with the 3V40 driven vector.
Labeling may be performed, as described above, to verify expression. The culture medium containing the expressed poly-His tagged PR010282 or PR019578 can then be concentrated and purified by any selected method, such as by Ni2+-chelate affinity chromatography.
PRO10282 or PR019578 may also be expressed in CHO and/or COS cells by a transient expression procedure or in CHO cells by another stable expression procedure.
Stable expression in CHO cells is performed using the following procedure. The proteins are expressed as an IgG
3 0 construct (immunoadhesin), in which the coding sequences for the soluble forms (e.g. extracellular domains) of the respective proteins are fused to an IgG1 constant region sequence containing the hinge, CH2 and CH2 domains and/or is a poly-His tagged form.
Following PCR amplification, the respective DNAs are subcloned in a CHO
expression vector using standard techniques as described in Ausubel et al., Current Protocols of Molecular Biology, Unit 3.16, John Wiley and Sons (1997).
3 5 CHO expression vectors are constructed to have compatible restriction sites 5' and 3' of the DNA of interest to allow the convenient shuttling of cDNA's. The vector used expression in CHO cells is as described in Lucas et al., Nucl. Acids Res.
24:9 (1774-1779 (1996), and uses the SV40 early promoter/enhancer to drive expression of the cDNA of interest and dihydrofolate reductase (DHFR). DHFR expression permits selection for stable maintenance of the plasmid following transfection.
Twelve micrograms of the desired plasmid DNA is introduced into approximately 10 million CHO cells using commercially available transfection reagents Superfect (Ouiagen), Dosper or Fugene (Boehringer Mannheim). The cells are grown as described in Lucas et al., supra. Approximately 3 x la cells are frozen in an ampule for further growth and production as described below.
The ampules containing the plasmid DNA are thawed by placement into water bath and mixed by vortexing. The contents are pipetted into a centrifuge tube containing 10 mLs of media and centrifuged at 1000 rpm for 5 minutes. The supernatant is aspirated and the cells are resuspended in 10 mL of selective media (0.2 jam filtered P520 with 5% 0.2 jam diafiltered fetal bovine serum). The cells are then aliquoted into a 100 mL
spinner containing 90 mL of selective media.
After 1-2 days, the cells are transferred into a 250 mL spinner filled with 150 mL selective growth medium and incubated at 37 C. After another 2-3 days, 250 mL, 500 mL and 2000 mL spinners are seeded with 3 x 106 cells/mL. The cell media is exchanged with fresh media by centrifugation and resuspension in production medium. Although any suitable CHO media may be employed, a production medium described in U.S. Patent No. 5,122,469, issued June 16, 1992 may actually be used. A 3L production spinner is seeded at 1.2 x 106 cells/mL. On day 0, the cell number pH is determined. On day 1, the spinner is sampled and sparging with filtered air is commenced. On day 2, the spinner is sampled, the temperature shifted to 33 C, and 30 mL of 500 gIL glucose and 0.6 mL of 10% antifoam (e.g., 35%
polydimethylsiloxane emulsion, Dow 2 0 Corning 365 Medical Grade Emulsion) taken. Throughout the production, the pH is adjusted as necessary to keep it at around 7.2. After 10 days, or until the viability dropped below 70%, the cell culture is harvested by centrifugation and filtering through a 0.22 jam filter. The filtrate was either stored at 4 C or immediately loaded onto columns for purification.
For the poly-His tagged constructs, the proteins are purified using a Ni-NTA
column ((Hagen). Before purification, 2 5 imidazole is added to the conditioned media to a concentration of 5 mM.
The conditioned media is pumped onto a 6 ml Ni-NTA column equilibrated in 20 mM Hopes, pH 7.4, buffer containing 0.3 M NaCl and 5 mM imidazole at a flow rate of 4-5 ml/min. at 4 C. After loading, the column is washed with additional equilibration buffer and the protein eluted with equilibration buffer containing 0.25 M imidazole. The highly purified protein is subsequently desalted into a storage buffer containing 10 mM Hepes, 0.14 M NaCI and 4% mannitol, pH 6.8, with a 25 ml G25 Superfine (Pharmacia) column and 30 stored at -80 C.
Immunoadhesin (Fc-containing) constructs are purified from the conditioned media as follows. The conditioned medium is pumped onto a 5 ml Protein A column (Pharmacia) which had been equilibrated in 20 mM Na phosphate buffer, pH 6.8. After loading, the column is washed extensively with equilibration buffer before elution with 100 mM citric acid, pH 3.5. The eluted protein is immediately neutralized by collecting 1 ml fractions into tubes containing 275 jaL of 1 M Tris 3 5 buffer, pH 9. The highly purified protein is subsequently desalted into storage buffer as described above for the poly-His tagged proteins. The homogeneity is assessed by SOS polyacrylamide gels and by N-terminal amino acid sequencing by Ednnan degradation.

Expression of PR010282 and PR019578 in Yeast The following method describes recombinant expression of PR010282 and PR019578 in yeast.
First, yeast expression vectors are constructed for intracellular production or secretion of PR010282 polypeptides from the ADH2IGAPDH promoter. DNA encoding a PR010282 polyepeptide (including PR019578) and the promoter is inserted into suitable restriction enzyme sites in the selected plasmid to direct intracellular expression of PRO10282. For secretion, DNA encoding PRO10282 can be cloned into the selected plasmid, together with DNA encoding the ADH2IGAPDH promoter, a native PR010282 signal peptide or other mammalian signal peptide, or, for example, a yeast alpha-factor or invertase secretory signallleader sequence, and linker sequences (if needed) for expression of PRO10282.
Yeast cells, such as yeast strain AB110, can then be transformed with the expression plasmids described above and cultured in selected fermentation media. The transformed yeast supernatants can be analyzed by precipitation with 10% trichloroacetic acid and separation by SDS-PAGE, followed by staining of the gels with Coomassie Blue stain.
Recombinant PRO10282 (including PRO19578) can subsequently be isolated and purified by removing the yeast cells from the fermentation medium by centrifugation and then concentrating the medium using selected cartridge filters.
The concentrate containing PR010282 (e.g. PR019578) may further be purified using selected column chromatography resins.

Expression of PRO10282 and PR019578 in Baculovirus-Infected Insect Cells The following method describes recombinant expression of PR010282 and PR019578 in Baculovirus-infected insect cells.
The sequence coding for PR010282 or PR019578 is fused upstream of an epitope tag contained within a baculovirus expression vector. Such epitope tags include poly-his tags and innmunoglobulin tags (like Fc regions of IgG).
A variety of plasmids may be employed, including plasmids derived from commercially available plasmids such as pVL1393 (Novagen). Briefly, the sequence encoding PR010282 or PR019578 or the desired portion of the coding sequence of PR010282 or PR019578, such as the sequence encoding the extracellular domain of a transmembrane protein or the 3 0 sequence encoding the mature protein if the protein is extracellular, is amplified by PCR with primers complementary to the 5' and 3' regions. The 5' primer may incorporate flanking (selected) restriction enzyme sites. The product is then digested with those selected restriction enzymes and subcloned into the expression vector.
Recombinant baculovirus is generated by co-transfecting the above plasmid and BaculoGoldTM virus DNA
(Pharmingen) into Spodoptera frugiperda ("Sf9") cells (ATCC CRL 1711) using lipofectin (commercially available from 3 5 GIBCO-BRL). After 4- 5 days of incubation at 28 C, the released viruses are harvested and used for further amplifications.

Viral infection and protein expression are performed as described by O'Reilley et al., Baculovirus expression vectors: A
Laboratory Manual, Oxford: Oxford University Press (1994).
Expressed poly-his tagged PR010282 or PRO19578 can then be purified, for example, by Ni'-chelate affinity chromatography as follows. Extracts are prepared from recombinant virus-infected Sf9 cells as described by Rupert et al., Nature, 362:175-179 (1993). Briefly, Sf9 cells are washed, resuspended in sonication buffer (25 mL Hepes, pH 7.9;
12.5 mM MgCl2; 0.1 mM EDTA; 10% glycerol; 0.1% NP-40; 0.4 M KCI), and sonicated twice for 20 seconds on ice. The sonicates are cleared by centrifugation, and the supernatant is diluted 50-fold in loading buffer (50 mM phosphate, 300 mM NaCI, 10% glycerol, pH 7.8) and filtered through a 0.45 gm filter. A Ni2+-NTA agarose column (commercially available from (Hagen) is prepared with a bed volume of 5 mL, washed with 25 mL of water and equilibrated with 25 mL of loading buffer. The filtered cell extract is loaded onto the column at 0.5 mL per minute. The column is washed to baseline A280 with loading buffer, at which point fraction collection is started. Next, the column is washed with a secondary wash buffer (50 mM phosphate; 300 mM NaCI, 10% glycerol, pH 6.0), which elutes nonspecifically bound protein. After reaching A280 baseline again, the column is developed with a 0 to 500 mM Imidazole gradient in the secondary wash buffer. One mL
fractions are collected and analyzed by SDS-PAGE and silver staining or Western blot with Ni2+-NTA-conjugated to alkaline phosphatase ((Hagen). Fractions containing the eluted Hisio-tagged PR010282 or PR019578 are pooled and dialyzed against loading buffer.
Alternatively, purification of the IgG tagged (or Fc tagged) PR010282 or PR019578 can be performed using known chromatography techniques, including for instance, Protein A or protein G column chromatography.

Preparation of Antibodies that Bind PR010282 or PRO19578 This example illustrates preparation of monoclonal antibodies which can specifically bind PR010282 or PRO19578.
Techniques for producing the monoclonal antibodies are known in the art and are described, for instance, in Goding, supra. lmmunogens that may be employed include purified PRO10282 and PRO19578, fusion proteins containing PR010282 or PRO19578, and cells expressing recombinant PR010282 or PR019578 on the cell surface. Selection of the immunogen can be made by the skilled artisan without undue experimentation.
Mice, such as BalbIc, are immunized with the PR010282 or PRO19578 immunogen emulsified in complete Freund's adjuvant and injected subcutaneously or intraperitoneally in an amount from 1-100 micrograms. Alternatively, 3 0 the immunogen is emulsified in MPL-TDM adjuvant (Ribi Immunochemical Research, Hamilton, MT) and injected into the animal's hind foot pads. The immunized mice are then boosted 10 to 12 days later with additional immunogen emulsified in the selected adjuvant. Thereafter, for several weeks, the mice may also be boosted with additional immunization injections. Serum samples may be periodically obtained from the mice by retro-orbital bleeding for testing in ELISA assays to detect anti-PR010282 antibodies or anti-PRO19578 antibodies.

After a suitable antibody titer has been detected, the animals "positive" for antibodies can be injected with a final intravenous injection of PR010282 or PR019578. Three to four days later, the mice are sacrificed and the spleen cells are harvested. The spleen cells are then fused (using 35% polyethylene glycol) to a selected murine myeloma cell line such as P3X63AgU.1, available from ATCC, No. CRL 1597. The fusions generate hybridoma cells which can then be plated in 96 well tissue culture plates containing HAT (hypoxanthine, aminopterin, and thymidine) medium to inhibit proliferation of non-fused cells, myeloma hybrids, and spleen cell hybrids.
The hybridoma cells will be screened in an ELISA for reactivity against PR010282 or PR019578. Determination of "positive" hybridoma cells secreting the desired monoclonal antibodies against PRO10282 or PRO19578 is within the skill in the art.
The positive hybridoma cells can be injected intraperitoneally into syngeneic Ball* mice to produce ascites containing the anti-PR010282 or anti-PR019578 monoclonal antibodies.
Alternatively, the hybridoma cells can be grown in tissue culture flasks or roller bottles. Purification of the monoclonal antibodies produced in the ascites can be accomplished using ammonium sulfate precipitation, followed by gel exclusion chromatography. Alternatively, affinity chromatography based upon binding of antibody to protein A or protein G can be employed.
Specifically, five BaIblc mice (Charles River Laboratories, Wilmington, DE) were hyper-immunized with purified Unizyme-conjugated amino acid peptide, corresponding to amino acids 532-667 of human Stra6, in Ribi adjuvant (Ribi lmnnunochem Research, Inc., Hamilton, MO). B-cells from popliteal lymph nodes were fused with mouse myeloma cells (X63.Ag8.653; American Type Culture Collection, Rockville, MD) as previously described (Hongo et al., Hybridoma 14:253-260 [1995]). After 10-14 days, supernatants were harvested and screened for antibody production by direct enzyme-linked immunosorbant assay (ELISA). Eight positive clones, showing the highest immunobinding by 2 0 direct ELISA and immunohistochemistry after two rounds of subcloning by limiting dilution, were injected into Pristine-primed mice for in vivo production of mAb (Freud and Blair, J. Immunol.
129:2826-2830 (19821 The ascites fluids were pooled and purified by Protein A affinity chromatography (Pharmacia fast protein liquid chromatography (FPLC);
Pharmacia, Uppsala, Sweden) as previously described (Hongo et al., supra). The purified antibody preparations were sterile filtered (0.2-jam pore size; Nalgene, Rochester, NY) and stored at 4 C in phosphate buffered saline (PBS).

Purification of PR010282 and PR019578 Polypeptides Using Specific Antibodies Native or recombinant PR010282 and PR019578 polypeptides may be purified by a variety of standard techniques in the art of protein purification. For example, pro-PR010282 or pro-PR019578 polypeptide, mature PR010282 or PRO19578 polypeptide, or pre-PRO10282 or pre-PR019578 polypeptide is purified by immunoaffinity chromatography using antibodies specific for the PRO10282 polypeptide of interest. In general, an immunoaffinity column is constructed by covalently coupling the anti-PR010282 or anti-PR019578 polypeptide antibody to an activated chromatographic resin.
Polyclonal immunoglobulins are prepared from immune sera either by precipitation with ammonium sulfate or by purification on immobilized Protein A (Pharmacia LKB Biotechnology, Piscataway, N.J.). Likewise, monoclonal antibodies 3 5 are prepared from mouse ascites fluid by ammonium sulfate precipitation or chromatography on immobilized Protein A.

Partially purified immunoglobulin is covalently attached to a chromatographic resin such as CnBractivated SEPHAROSE' (Pharmacia LKB Biotechnology). The antibody is coupled to the resin, the resin is blocked, and the derivative resin is washed according to the manufacturer's instructions.
Such an immunoaffinity column is utilized in the purification of PR010282 or PR019578 polypeptide by preparing a fraction from cells containing P11010282 or P11019578 polypeptide in a soluble form. This preparation is derived by solubilization of the whole cell or of a subcellular fraction obtained via differential centrifugation by the addition of detergent or by other methods well known in the art. Alternatively, soluble P11010282 or P11019578 polypeptide containing a signal sequence may be secreted in useful quantity into the medium in which the cells are grown.
A soluble PRO10282 or PRO19578 polypeptide-containing preparation is passed over the immunoaffinity column, and the column is washed under conditions that allow the preferential absorbance of PR010282 or P11019578 polypeptide (e.g., high ionic strength buffers in the presence of detergent). Then, the column is eluted under conditions that disrupt antibody/PRO10282 or PR019578 polypeptide binding (e.g., a low pH buffer such as approximately pH 2-3, or a high concentration of a chaotrope such as urea or thiocyanate ion), and P11010282 or p11019578 polypeptide is collected.

Drug Screening This invention is particularly useful for screening compounds by using PR010282 (Stra6) polypeptides (including P11019578) or binding fragment thereof in any of a variety of drug screening techniques. The P11010282 polypeptide or fragment employed in such a test may either be free in solution, affixed to a solid support, borne on a cell surface, or 2 0 located intracellularly. One method of drug screening utilizes eukaryotic or prokaryotic host cells which are stably transformed with recombinant nucleic acids expressing the PI1010282 polypeptide or fragment. Drugs are screened against such transformed cells in competitive binding assays. Such cells, either in viable or fixed form, can be used for standard binding assays. One may measure, for example, the formation of complexes between P11010282 polypeptide or a fragment and the agent being tested. Alternatively, one can examine the diminution in complex formation between the PRO10282 2 5 polypeptide and its target cell or target receptors caused by the agent being tested.
Thus, the present invention provides methods of screening for drugs or any other agents which can affect a PR010282 (Stra6) polypeptide-associated disease or disorder. These methods comprise contacting such an agent with a Stra6 polypeptide or fragment thereof and assaying (i) for the presence of a complex between the agent and the Stra6 polypeptide or fragment, or (ii) for the presence of a complex between the Stra6 polypeptide or fragment and the cell, by 3 0 methods well known in the art. In such competitive binding assays, the Stra6 polypeptide or fragment is typically labeled.
After suitable incubation, free Stra6 polypeptide or fragment is separated from that present in bound form, and the amount of free or uncomplexed label is a measure of the ability of the particular agent to bind to Stra6 polypeptide or to interfere with the Stra6 polypeptidelcell complex.
Another technique for drug screening provides high throughput screening for compounds having suitable binding 3 5 affinity to a polypeptide and is described in detail in WO 84103564, published on September 13, 1984. Briefly stated, large numbers of different small peptide test compounds are synthesized on a solid substrate, such as plastic pins or some other surface. As applied to a PR010282 (Stra6) polypeptide, the peptide test compounds are reacted with Stra6 polypeptide and washed. Bound Stra6 polypeptide is detected by methods well known in the art. Purified Stra6 polypeptide can also be coated directly onto plates for use in the aforementioned drug screening techniques. In addition, non-neutralizing antibodies can be used to capture the peptide and immobilize it on the solid support.
This invention also contemplates the use of competitive drug screening assays in which neutralizing antibodies capable of binding a Stra6 polypeptide specifically compete with a test compound for binding to a Stra6 polypeptide or fragments thereof. In this manner, the antibodies can be used to detect the presence of any peptide which shares one or more antigenic determinants with the Stra6 polypeptide.

Rational Druq Design The goal of rational drug design is to produce structural analogs of biologically active polypeptide of interest (i.e., a PR010282 (Stra6) polypeptide) or of small molecules with which they interact, e.g., agonists, antagonists, or inhibitors.
Any of these examples can be used to fashion drugs which are more active or stable forms of the native sequence PRO10282 polypeptide or PRO19578 polypeptide, or which enhance or interfere with the function of the native sequence P11010282 or PR019578 polypeptide in vivo (c.f., Hodgson, BiolTechnoloqy, 9:
19-21 (1991)).
In one approach, the three-dimensional structure of the native sequence PR010282 or PR019578 polypeptide, or of a PR010282 or PR019578 polypeptide-inhibitor complex, is determined by x-ray crystallography, by computer modeling or, most typically, by a combination of the two approaches. Both the shape and charges of the native P11010282 or PR019578 polypeptide must be ascertained to elucidate the structure and to determine active site(s) of the molecule.
Less often, useful information regarding the structure of the PR010282 or PR019578 polypeptide may be gained by modeling based on the structure of homologous proteins. In both cases, relevant structural information is used to design 2 5 analogous PRO10282 polypeptide-like molecules or to identify efficient inhibitors. Useful examples of rational drug design may include molecules which have improved activity or stability as shown by Braxton and Wells, Biochemistry, 31:7796-7801 (1992) or which act as inhibitors, agonists, or antagonists of native peptides as shown by Athauda et al., J. Biochem., 113:742-746 (1993).
It is also possible to isolate a target-specific antibody, selected by functional assay, as described above, and then 3 0 to solve its crystal structure. This approach, in principle, yields a pharmacore upon which subsequent drug design can be based. It is possible to bypass protein crystallography altogether by generating anti-idiotypic antibodies (anti-ids) to a functional, pharmacologically active antibody. As a mirror image of a mirror image, the binding site of the anti-ids would be expected to be an analog of the original receptor. The anti-id could then be used to identify and isolate peptides from banks of chemically or biologically produced peptides. The isolated peptides would then act as the pharmacore.
35 By virtue of the present invention, sufficient amounts of the PR010282 polypeptides (including PR019578) may be made available to perform such analytical studies as X-ray crystallography.
In addition, knowledge of the P11010282 polypeptide amino acid sequences provided herein will provide guidance to those employing computer modeling techniques in place of or in addition to x-ray crystallography.

Tissue Expression Distribution Oligonucleotide probes were constructed from the PRO10282 polypeptide-encoding nucleotide sequence shown in the accompanying figures for use in quantitative PCR amplification reactions. The oligonucleotide probes were chosen so as to give an approximately 200-600 base pair amplified fragment from the 3' end of its associated template in a 1 0 standard PCR reaction. The oligonucleotide probes were employed in standard quantitative PCR amplification reactions with Clontech RNA isolated from different adult human tissue sources and analyzed by agarose gel electrophoresis so as to obtain a quantitative determination of the level of expression of the PR010282 polypeptide-encoding nucleic acid in the various tissues tested. Knowledge of the expression pattern or the differential expression of the PR010282 polypeptide-encoding nucleic acid in various different human tissue types provides a diagnostic marker useful for tissue typing, with or without other tissue-specific markers, for determining the primary tissue source of a metastatic tumor, and the like. The results of these assays (shown in Figure 10) demonstrated that the DNA148380-2827 molecule is highly expressed in the adult kidney, testis and uterus; significantly expressed in breast, prostate and trachea; weakly expressed in brain, heart, lung and thymus; and not expressed in liver, bone marrow, colon, skeletal muscle, small intestine, spleen and stomach.
Total RNA was purchased from Clontech (Palo Alto, California) and analyzed using the following primer/probe 2 0 set for PCR amplification:
h.Stra6.tmf3: 5' CACACTCGAGAGCCAGATATTTT (SEG ID NO: 6) h.Stra5.tmr4: 5' AACAAGTTTATTGCAGGGAACAC (SEG ID NO: 7) h.Stra6.tmp4: 5' TGTAGTTTTTATGCCTTTGGCTATTATGAAAGAGGT
(SEG ID NO: 8) tmf = forward primer tmr = reverse primer tnnp = probe In situ hybridization, performed as described in Example 12 below, confirmed Stra6 expression in kidney tubular epithelial cells, myometrium, and stromal cells surrounding breast ducts and lobules, whereas little or no expression was detected in sections of brain, liver, spleen, pancreas, heart, lung, stomach, small intestine, colon, prostate, spleen, and adrenal cortex (data not shown). Of the normal tissues examined by in situ hybridization, highest expression levels were seen in placenta and adrenal medulla, which were not included in the PCR
analysis.

Over-expression of native human PR010282 (Stra6) transcript in Human Tumors This example shows that the gene encoding native human full-length PRO10282 (Stra6) is significantly over-expressed in certain human colon tumors and also in cell lines derived from various human tumors such as colon, lung, kidney and breast.
The starting material for the screen was total RNA isolated from human colon tumors, or various human colon, kidney, breast, or lung tumor cell lines. In colon tumor tissue, Stra6 RNA
expression was determined relative to RNA from normal colon tissue (mucosa) from the same patient. Stra6 RNA expression in various tumor cell lines was determined in comparison with various normal cell lines (i.e., normal colon, kidney and lung cell lines).
Real-time quantitative PCR (RT-PCR, for example, TAGMAN ABI PRIZM 7700 Sequence Detection System' [Perkin Elmer, Applied Biosystems Division, Foster City, California]), was used to monitor quantitative differences in the level of expression of the PRO10292 (Stra6) encoding gene (corresponding to DNA148380-2827) in normal cells and cells derived from certain cancers or cancer cell lines, using Taqman assay reagents. 50 pf RT-PCR reactions consisted of 5 ill 10x Taqman Buffer A, 300 iuM of each dNTF, 5 mM MgCl2, 10 units of RNAse inhibitor, 12.5 units ofMuLV Reverse Transcriptase, 1.25 units of AmpliTaq Gold DNA Polymerase, 200 nM probe, 500 nM primers and 100 ng RNA. Reaction conditions consisted of reverse transcription at 48 C for 30 minutes, denaturation at 95 C for 25 seconds and 65 C
for one minute. Reaction products were analyzed on 4-20% polyacrylamide gels (Novex).
Standard curves were used to determine relative levels of expression for each gene of interest as well as the glyceraldehyde-3-phosphate dehydrogenase (GAPDH) housekeeping gene for each sample analyzed. Relative normalized units were obtained by dividing the gene of interest mRNA level by the GAPDH
mRNA level. Relative normalized units were 2 0 compared between experimental sample and control to determine fold induction.
The results were used to determine whether the mRNA encoding PRO10282 is over-expressed in any of the primary colon cancers or colon, kidney, breast, or lung cancer cell lines that were screened. The histology of some matched human normal and colon tumor samples used for the analysis (see Figures 11 and 12) is shown below:
Number Histoloov 850 Invasive adenocarcinoma; no necrosis; good preservation.
851 Invasive adenocarcinoma; minimal necrosis; good condition.
892 Invasive adenocarcinoma; minimal necrosis; good condition.
869 Invasive adenocarcinoma; minimal necrosis; good condition.
3 0 893 Normal mucosa - dysplasia - invasive adenocarcinoma;
minor necrosis; good condition.
870 Adenocarcinoma - severe dysplasia; minimal necrosis; good condition.
871 Adenocarcinoma dysplasia - normal mucosa; no necrosis;
good condition.

848 Adenocarcinoma - appears to be arising in vinous adenoma;
normal mucosalsubmucosa; no necrosis, good condition.
872 Invasive adenocarcinoma; about 70% of tumor is necrotic;
overall good preservation.
778 Adenocarcinoma - unusually papillary morphology;
normal/hyperplastic mucosa; minimal necrosis; acceptable preservation.
17 Moderately well-differantiated adenocarcinoma.
18 Well-differentiated adenocarcinoma.
64 Moderately well-differentiated adenocarcinoma.
76 Moderately well-differentiated adenocarcinoma.
Human lung canines include the normal human lung fibroblast canines MRC5 (CCL-171) and IMR90 (CCL-186), the human lung carcinoma epithelial canine A549 (SRCC768, CCL-185), the human epidermoid lung carcinoma cell line Calu-1 (SRCC769; HTB-54), the human anaplastic carcinoma cell line Calu-6 (SRCC770, HTB-56 - probably lung), the human epithelial cell line NCI-H441 (SRCC772; HTB-174) which was derived from pericardial fluid of a patient with papillary adenocarcinoma of the lung, and the human lung squamous cell carcinoma canine SW900 (SRCC775; HTB-59), all available from ATCC.
Colon cell lines include, for example, the normal colon flroblast cell line CC0112Co (CRL-1541), the human colorectal adenocarcinoma cell line CaCo-2 (HTB-37), the human colorectal adenocarcinoma cell line WiOr (CCL-218), the human colorectal carcinoma cell line HCT116 (CCL-247), the human colorectal adenocarcinoma cell line SK-Col (HTB-39) the human colorectal adenocarcinoma cell line C010320 (SRCC778; CCL-220), the human colorectal adenocarcinoma cell line HT29 (SRCC779; HTB-38), the human colorectal adenocarcinoma canine SW403 (CCL-230), the human colon cancer cell line NCl/HCC2998, and the human colorectal adenocarcinoma cell line Colo3200M (CCL-220), all available from ATCC or other public sources.
Human breast carcinoma cell lines include the human breast adenocarcinoma cell line MCF7 (SRCC766; HTB-22), and the human breast cancer cell line NCl/AIM-RES, both of which are publicly available.
Kidney lines include the 293 cell line (CRL-1573) which is transformed with adenovirus 5 DNA. Two Wilm's tumor cell lines were also incuded in the analysis, G401 (CRL-1441) and SK-NEP-1 (CR1-1573).
RNA preparation:
RNA was prepared from the foregoing cultured cell lines. The isolation was performed using purification kit, buffer set and protease from Qiagen, according to the manufacturer's instructions and the description below. More specifically, total RNA from cells in culture was isolated using Qiagen RNeas;midrcolumns. Total RNA from tissue samples was isolated using RNA Stat-60 (Tel-Test). RNA prepared from tumor was isolated by cesium chloride density *Trade¨mark =

gradient centrifugation.
Solid human tumor sample preparation and lysis:
Tumor samples were weighed and placed into 50 ml conical tubes and held on ice. Processing was limited to no more than 250 mg tissue per preparation (1 tip/preparation). The protease solution was freshly prepared by diluting into 6.25 ml cold ddH20 to a final concentration of 20 lig/mland stored at 4 C. G2 buffer (20 ml) was prepared by diluting DNAse A to a final concentration of 200 ilg/m1 (from 100 mglml stock). The tumor tissue was homogenized in 10 ml G2 buffer for 60 seconds using the large tip of the polytron in a laminar-flow IC
hood in order to avoid inhalation of aerosols, and held at room temperature. Between samples, the polytron was cleaned by spinning at 2 x 30 seconds each in 2L, ddH20, followed by G2 buffer (50 ml). If tissue was still present on the generator tip, the apparatus was disassembled and cleaned.
Giagen protease (prepared as indicated above, 1.0 ml) was added, followed by vortexing and incubation at 50 C for 3 hours. The incubation and centrifugation were prepeated until the lysates were clear (e.g., incubating additional 30-60 minutes, pelleting at 3000 x g for 10 minutes, 4 C).
Ouantitation The results obtained from the real-time PCR analysis of RNA were initially expressed as delta CT units. One unit corresponds to one PCR cycle or approximately a 2-fold amplification relative to normal, two units correspond to 4-fold, 3 units to 8-fold amplificaiton and so on. The data is converted to fold difference and presented as such. Initially, reverse transcriptase was used to synthesize cDNA from 100 ng total RNA or polyA+ RNA
using oligo(dT) as a primer. The resultant cDNA was then used as a template for PCR. Gantitation was obtained using primers derived from the 3'-' untranslated regions of the PRO10282 encoding sequence and a TAGMAN
fluorescent probe corresponding to the respective intervening sequences. Using the 3' region tends to avoid crossing intron-exon boundaries in the genomic DNA, an essential requirement for accurate assessment of RNA expression using this method. The sequences for the primers and probes (forward, reverse, and probe) using for the PRO10282 encoding gene amplification were as follows:
One set included the forward and reverse primers and probe described in Example 11 above as SEO ID Nos: 6, 7 and 8, respectively.
Another set included:
hStra6.tmf11: 5' AGACCAGGTCCCACACTGA (SEG ID NO: 9) hStra6.tmr1: 5' TTCATAATAGCCAAAGGCATAAAA (SEG ID NO: 10), and h.Stra6.tmp1: 5' CTGCCCACACTCGAGAGCCAGAT 3' (SEG ID NO: 11) Human GAPDH:
forward primer: 5'-GAAGATGGTGATGGGATTTC-3' (SEG ID NO: 14) reverse primer; 5'-GAAGGTGAAGGTCGGAGTC-3' (SEG ID NO: 15) probe: 5'-CAAGCTTCCCGTTCTCAGCC-3' (SEG ID NO: 16) The 5' nuclease assay reaction is a fluorescent PCR-based technique which makes use of the 5' exonuclease activity of Taq DNA polymerase enzyme to monitor amplification in real time.
Two oligonucleotide primers are used to generate an amplicon typical of a PCR reaction. A third oligonucleotide, or probe, is designed to detect nucleotide sequence located between the two PCR primers. The probe is non-extendible by Taq DNA
polymerase enzyme, and is labeled with a reporter fluorescent dye and a quencher fluorescent dye. Any laser-induced emission from the reporter dye is quenched by the quenching dye when the two dyes are located close together as they are on the probe. During the amplification reaction, the Taq DNA polymerase enzyme cleaves the probe in a template-dependent manner. The resultant probe 1 0 fragments disassociate in solution, and signal from the released reporter dye is free from the quenching effect of the second fluorophore. One molecule of reporter dye is liberated for each new molecule synthesized, and detection of the unquenched reporter dye provides the basis for quantitative interpretation of the data.
As noted above, the 5' nuclease procedure is run on a real-time quantitative PCR device such as the ABI PRIZM
7700TM Sequence Detection SystemTM. The system consists of a thermocyler, laser, charge-coupled device (CCD), camera and computer. The system amplifies samples in a 96-well format on a thermocycler. During amplification, laser-induced fluorescent signal is collected in real-time through fiber optics cables for all 96 wells, and detected at the CCD. The system includes software for running the instrument and for analyzing the data.
5'-Nuclease assay data are initially expressed as Ct, or the threshold cycle.
This is defined as the cycle at which the reporter signal accumulates above the background level of fluorescence.
The Ct values are used as quantitative measurement of the relative number of starting copies of a particular target sequence in a nucleic acid sample when comparing the expression of RNA in a cancer cell with that from a normal cell.
Results The human Stra6 RNA (corresponding to DNA148380-2827) shows strong over-expression in human colon tumor tissues, when compared with corresponding normal human colon tissues. As shown in Figure 11, human Stra6 RNA
(corresponding to DNA148380-2827) was found to be over-expressed in all 14 human tumor colon tissues examined, relative to RNA from matched normal colorectal mucosa from the same patient.
The over-expression varied between two-and 170-fold, and in 7 out of 14 tumor tissue samples was at least about 10-fold. The cycle threshold values obtained by quantitative PCR indicated that Stra6 mRNA levels were extremely low or possibly absent in many of the nomal mucosa samples.
As a second method of detection, the products obtained after completion of the quantitative PCR reactions (40 cycles each) were subjected to electrophoresis in polyacrylamide gels and visualized by ethidiunn bromide staining. As shown in Figure 12A, using expression of a housekeeping gene, glyceraldehyde 3-phosphate dehydrogenase (GAPDH) as the standard, substantially greater amounts of PCR products were generated off Stra6 mRNA in tumor samples compared to their normal counterparts. By contrast, a comparable level of product from the internal control GAPDH mRNA was generated from all samples.
Stra6 expression in colon adenocarcinomas was localized to the epithelial tumor cells by in situ hybridization (ISH) performed as follows. 93-labeled sense and antisense riboprobes were transcribed from an 874 bp PCR product corresponding to nucleotides 432-1247 of the coding sequence of human Stra6 polypeptide encoded by DNA148380-2827.
Formalin-fixed, paraffin-embedded tissue sections were processes as described previously (Pennica et al., Proc. Natl. Acad.
Sci USA 95:14717-22 [1998]). The results are shown in Figure 12B.
As shown in Figure 13, the human Stra6 RNA (corresponding to DNA148380-2827) is also significantly over-expressed in various breast, kidney, colon and lung tumor cell lines.
"Relative RNA Expression" means that the RNA
expression in normal and tumor tissues is shown relative to expression in an arbitrarily chosen stadard cell line SW480.
In situ hybridization results obtained in various tumor sections are also shown in Figure 16. Several tumor types other than colon adenocarcinomas also showed high levels of Stra6 expression.
These included 3 of 3 melanomas (Figures 16A and B), 3 of 4 endometrial adenocarcinomas (Figures 16C and D), 2 of 3 ovarian adenocarcinomas, and a Wilm's tumor of the kidney (Figures 16E and F). The Stra6 in situ hybridization signal in these various tumors was considerably greater than in colon adenocarcinomas consistent with data showing relatively high expression levels in normal kidney and uterus and low levels in normal colon. Since Stra6 was detected in normal adrenal medulla, we also examined two pheochronnocytomas, which are tumors derived from this tissue. In these tumors, Stra6 expression exceeded that of any other tumor or tissue examined in this study (Figures 16G and H). Although Stra6 was detected in normal kidney and was strongly expressed in Wilm's tumor, it was not detected in renal cell carcinomas. In kidney transitional cell carcinomas, tumor-associated stromal cells rather than tumor epithelial cells expressed Stra6 (data not shown).
Because mRNA DNA148380-2827 encoding PRO10282 is overexpressed in various tumors as well as in a number of tumor derived cell lines, it is likely associated with tumor formation and/or growth. As a result, antagonists (e.g., antibodies, organic and inorganic small molecules, peptides and polypeptides, such as Stra6 variants, antisense oligonucleotides) directed against the protein encoded by DNA148380-2827 (PR010282) or other naturally occurring variants of this protein, such as PR019578 encoded by DNA148389-2827-1, are expected to be useful in diagnosis, 2 5 prevention and/or treatment of cancer particularly, without limitation, colong, lung, breast and/or kidney cancer.
The efficacy of antagonists such as therapeutic antibodies directed against the protein encoded by DNA148380-2827 (PR010282) could be enhanced by agents that stimulate the expression of the gene encoding PR010282. For example, treatment of human colorectal cancer cell lines with 9-cis-retinoic acid or all-trans retinoic acid resulted in a dramatic enhancement of the expression of the Stra6 mRNA (Figure 15). Thus, the treatment of cancer patients with therapeutic antibodies directed against PR010282 in combination with the appropriate retinoids would be expected to enhance tumor killing by the antibodies. The same will be true to antibodies directed against other native Stra6 polypepides over-expressed in various tumors, such as the human splice variant encoded by DNA148389-2827-1.
EXAMPLE' 13 3 5 Synergistic Induction of Stra6 by Wnt-1 and Retinoids Materials and Methods Cell Culture C57MG and C57MG/Wnt-1 cells were grown in Dulbecco's Modified Eagle medium supplemented with 10%
fetal bovine serum, 2 mM L-glutarnine, and 2.5 pg1m1puromycin (Edge Biosystems). C57MG cells with tetracycline-repressible Wnt-1 expression were grown in complete medium without puromycin, supplemented with 400 pglml G418 (Gibco BRL), 100 pglml hygromycin B (Gibco BRL), and 50 nglml tetracycline (Korinek et al., Mol. Cell Biol. 18:1248-56 [1998]). For Wnt-1 induction studies, cells were washed with phosphate buffered saline, cultured in tetracycline-free media for 10, 24, 48, 72, and 96 hours and then harvested. A 0-hour control dish was maintained entirely in media containing tetracycline. All dishes were simultaneously harvested and total RNA was extracted for each time point. RT-PCR was carried out with Wnt-1, Stra6, and CAPON specific primers and probes on 100 ng total RNA from each sample.
Human colon adenocarcinoma cell lines HCT116 and With cells were obtained from the American Type Culture Collection. HCT116 cells were maintained in McCoy's 5A media supplemented with 10% fetal bovine serum (FBS). WiDr cells were maintained in Dulbecco's minimal essential media (OMEM) supplemented with 10% FBS. For retinoic acid induction studies, cells were plated at 105 cellsI60 mm dish containing 2.5 ml of medium and allowed to grow for 24 hours. Cells were treated with Vitamin 03, all-trans-RA (Spectrum Laboratory Products), or 9-cis-RA
(Toronto Research Chemicals Inc.) (1pM final concentration in DMSO) for the indicated times. Control cells were treated with an equal volume of DMSO.
For the treatment of C57MGIParent cells with Wnt-3a conditioned media, cells were incubated in regular media or conditioned media from L-cells or L-W3A cells in the presence or absence of 1 pM 9-cis-RA. Conditioned media was prepared as previously described (VVillert et al.,Genes Dev. 13:1768-73 [19991 At 48 hours, cells were harvested by scraping into PBS containing sodium fluoride and vanadate and quick frozen in liquid nitrogen. RNA was prepared using the RNAeas;kit (Wagon), including the additional DNasel treatment on the columns according to manufacturer's instructions.
Western Blotting For the C57MGIWnt-1 TET asinine, Wnt-1 expression was induced by culturing cells in the absence of tetracycline for 0, 24.48, or 72 hours. Cells were lysed in Triton X-100 lysis buffer [20 mM tris-HC1 (pH 8.0), 137 mM NaCl, 1% Triton X-100, 1 mM EGTA, 10% glycerol, 1.5 mM MgC12, 1 mM
dithiothreitol, 1 mM sodium vanadate, 50 mM sodium fluoride, and complete protease inhibitor cocktail (Boehringer Mannheim) and protein-equivalents were subjected to SOS-PAGE and imtnunoblotting. Blots were incubated with either 0.2 pgImlaffinity purified rabbit polyclonal antibody against 13-catenin (Rubinfeld at al., Science 262:1731-173411993D, 0.1 pg/mlanti-ERK2 monoclonal antibody (Transduction Laboratories), or 1:2000 rabbit polyclonal antisera against RAlirl (Affinity Bioreagents). For the With cell line, cells were treated with 1 pM all-trans-RA for 48 hours and then lysed in Triton X.
100 lysis buffer and processed as indicated above. Blots were incubated with 1:50 anti-Stra6 peptide B monoclonal hybridoma culture supernatant (clone 12F4.2H9.105).
*Trademark Immunohistochemistrv WiDr cells were treated with 9-cis-retinoic acid or DMSO, then detached and pelleted by low-speed centrifugation. Cell pellets were fixed overnight in 10% neutral buffered formalin, dehydrated, and embedded in paraffin. lmmunohistochemistry was performed using anti-Stra6 peptide B
monoclonal hybridoma culture supernatant (clone 12F4.2H9.105) or nonspecific mouse isotype IgG2A as primary antibodies, followed by detection using avidin-biotin complex method with diaminobenzidine as chromogen (Vectastain Elite Kit, Vector Laboratories) as described previously (Eberhard et al., Am. J. Pathol. 145:640-9 [1994]). Sections were counterstained with hematoxylin.
Wnt-1 Transoenic Mice Transgenic mice that express the Wnt-1 proto-oncogene in the mammary gland typically exhibit hyperplastic lesions and develop neoplasms in this tissue (Tsukannoto et al., Cell 55:619-625 [1988]). Such mice were used in the following experiments.
Results Easwaran et al. previously reported enhanced activation of a synthetic retinoic acid responsive reporter gene when MCF-7 cells were co-transfected with mutant P-catenin and treated with retinoids (Easwaran at al., Curr. Biol.
9:1415-1418 [1999]). Considering this, together with the original identification of Stra6 as a retinoic acid inducible gene (Bouillet et al., Mech Dev. 63:173-186 [1997]), we asked whether retinoic acid could synergize with Wnt-1 to increase the level of Stra6 in the C57M6 cell line. Treatment of parental C57M0 cells with either 9-cis-RA or all-trans-RA (ATRA) for 48 hours significantly increased the level of Stra6 mRNA
while DMSO and vitamin D3 had no effect (Figure 17A). As expected, the C57MG/Wnt-1 cells treated with either DMSO or vitamin 03 exhibited enhanced levels of Stra6 mRNA relative to the parent cell line. The level of Stra6 induction by Wnt-1 was comparable to that observed on stimulation of the parental C57MG cells with 9-cis-RA. However, 9-cis-RA treatment of the C57MGIWnt-1 cell line induced a further 10-fold increase in Stra6 mRNA relative to either untreated C57MGIWnt-1 or 9-cis-RA
treated C57MG parent cells. Similar results were obtained with all-trans-RA.
It was possible that potential clonal variations in the C57MG/Wnt-1 cell line, that were unrelated to Wnt-1 expression, accounted for their differential response to retinoic acid relative to parental control cells. To address this, we tested the response of the parental C57MG cells to stimulation by soluble Wnt-3a in the presence or absence of 9-cis-RA. Wnt-3a is a Wnt-1 homolog that exhibits transforming properties similar to those of Wnt-1 and can be produced as a soluble ligand in mouse L-cells. Conditioned media from cultured L-cells expressing Wnt-3a, but not from control L-cells, induced the expression of Stra6 in the C57MG cells (Figure 17B). A slightly higher level of induction was observed on treatment of C57M6 cells with 9-cis-RA. However, the combination of 9-cis-RA and Wnt-3a resulted in levels of Stra6 transcript vastly exceeding that seen with either agent alone.
If the induction of Stra6 in the retinoic acid treated C57MG IWnt-1cells was potentiated by increased f3-catenin levels, then one might expect a similar induction of Stra6 in response to retinoic acid in human colon carcinoma 3 5 cells containing mutations in either 13-catenin or APC. To determine whether this occurs, Stra6 mRNA levels were measured before and after retinoic acid treatment in HCT116 cells, which carry an activating mutation in f3-catenin, and in WiDr cells, which have lost both copies of wild-type APC. In both cell lines, a significant increase in Stra6 mRNA levels was seen following treatment with either ATRA or 9-cis-RA compared to DMSO or vitamin D3 (Figure 17C). The activation of the Stra6 gene by ATRA in the HCT116 cell line was confirmed by in situ hybridization (Figure 17D). Induction of the predicted 73kDa Stra6 protein band in WiDr cells treated with ATRA was detected by Western blot analysis with a Stra6 specific monoclonal antibody (Figure 17E).
Immunohistochemical analysis of the WiDr cells revealed that the increase in Stra6 protein in response to retinoic acid was localized to the plasma membrane (Fig.
17F).
The RARy gene has been proposed as a target for Wnt signaling in Xenopus embryos, and the induction of Stra6 by retinoids was shown to be dependent upon the presence of this retinoic acid receptor subtype (McGrew et al., Mech. Dev. 87:21-32 [1999]; Taneja et al., Proc. Natl. Acad. Sci. USA 92:7854-8 [1995]). Together, these observations suggest that the synergistic activation of Stra6 by Wnt and retinoids might be due to the activation of RARy expression by Wnt signaling. To determine whether Wnt-1 signaling had any influence over the levels of RARy in mammalian cells, we performed Western blots for the receptor on lysates prepared from C57MG cells that conditionally express Wnt-1. Upon Wnt-1 expression, a protein reactive with antibody specific to RARy-1, and migrating with an apparent molecular mass of 64 kDa, was induced at 24 hours and its expression was increased at 48 hours (Figure 18A). We also analyzed hyperplastic mammary glands and mammary gland tumors obtained from Wnt-1 transgenic mice and detected elevated levels of RARy mRNA in these tissues relative to normal mammary gland (Figure 183). The level of RARy transcript present in equivalent amounts of RNA isolated from 19 adenocarcinomas = 20 was assessed by quantitative PCR. Notably, RARy mRNA expression was increased approximately 2-4 fold in 14 of the 19 (74%) of human colon tumors examined compared to normal human colon tissue (Figure 18C). These results demonstrate that Wnt-1 signaling promotes the expression of RARy that the receptors are elevated in mouse and human tumors that are driven by the Wnt-1 pathway.
Discussion of Experimental Findings Gene expression profiling approaches are based on unbiased detection of mRNA
transcripts and can therefore lead to unexpected insights into the mechanisms by which gene activation occurs. Here it has been shown that Wnt-1 promoted the induction of the retinoic acid responsive gene Stra6, suggesting a connection between signaling pathways elicited by Wnt and retinoic acid. This connection was further supported by demonstrating a synergistic 3 0 induction of Stra6 by a combination of Wnt and retinoic acid. There are at least three alternative explanations that could account for this synergy: i) transcription factors directly responsive to Wnt such as the TCFILEFs might bind to and activate promoter elements in the Stra6 gene; ii) signaling components in the Wnt pathway might directly interact with the appropriate retinoic acid receptor (RAR) and potentiate gene activation mediated by the RAR; or iii) signaling by Wnt-1 could activate expression of the appropriate RAR, which could then sensitize cells to retinoic acid. Although this final proposal is favored because the data presented herein show that Wnt-1 signaling rapidly induces expression of the RARy receptor, the present invention is not limited by any particular theory or mechanism of action.
Previous work has shown that specific disruption of the RARy gene greatly reduced induction of the Stra6 gene by retinoids, which was then rescued on re-expression of this receptor (Taneja et at., supra). However, it remains possible that mechanisms in addition to the Wnt-1-induced expression of RARy may also contribute to the observed synergy. The proposal that 13-catenin binds to retinoic acid receptors is attractive, but we have not observed any specific association of endogenous RARy with 13-catenin in our cell lines.
However, the binding of 13-catenin to RARy, as was shown in vitro (Easwaran et al., 1999, supra), might relate to the activation of Stra6 by Wnt-1. The expression pattern of Stra6 in transgenic animals null for RARy was more widespread than that observed in wild-type littermates, and induction of Stra6 by retinoic acid was enhanced in cells from the null animals compared to controls (Bouillet et at., 1997, supra; Taneja et at., 1995, supra). Thus RARy might be inhibitory to Stra6 expression, and activation of I3-catenin by Wnt-1 could potentially relieve this inhibition if p-catenin inhibited RARy function.
The synergistic induction of a retinoic acid responsive gene by Wnt-1 signaling implies that human cancers that harbor genetic defects in the Wnt-1 pathway would exhibit overexpression of Stra6. The vast majority of colorectal tumors contain mutations in the genes coding for either the APC
tumor suppressor or P-catenin (Polakis, Curr. Opin. Genet. Dev. 9:15-21 [1999]) and, accordingly, we have detected overexpression of the Stra6 transcript in 14 of 14 colorectal tumors relative to matched normal tissue (see Example 12).
Activating mutations in f3-catenin have also been identified in cancers of the ovary and endometrium, Wilm's kidney tumors and melanomas, demonstrating that defects in Wnt-1 signaling contribute to the progression of these cancers (Kobayashi et al.Jpn. J.
2 0 Cancer Res. 90:55-9 [1999]; Koesters et al., Cencer Res. 59:3880-2 [1999]; Palacios and Hamallo, Cancer Res.
58:1344-7 [1998]; Rimm et at. Am. J. Pathol.154:325-9 [1999]; Rubinfeld et al.Science 262:1731-1734 [1993];
Wright et al. Int. J. Cancer 82:625-9 [1999]). All four of these human cancers displayed overexpression of Stra6 mRNA as determined by in situ hybridization (see Example 12).
Pheochromocytoma, a tumor derived from the adrenal medulla, exhibited extremely high levels of Stra6 mRNA, however, the status of these tumors with respect to mutations in the Wnt-1 signaling pathway has not been reported. It is intriguing that the cell types that give rise to melanomas and pheochromocytomas share in common an embryological derivation from the neural crest. Notably, Wilms' tumor of the kidney, pheochromocytoma, and endometrial carcinomas all arise in organs in which Stra6 is normally expressed. This suggests that turnorigenic signaling, such as that driven by the Wnt-1 pathway, might interact with signals responsible for differentiation, thereby hyper-activating the expression of Stra6.
3 0 The human Stra6 protein resides at the cell surface, as determined by the staining of colorectal cancer cells with monoclonal antibodies specific to human Stra6 protein. Moreover, the staining intensity observed at the cell membrane was increased after treatment of cells with retinoic acid. Thus, Stra6 likely encodes a multi-pass transmembrane protein that is localized to the cell surface.

Deposit of Material The following materials have been deposited with the Amedcan Type Culture Collection, 10801 University Blvd., Manassas, VA 20110-2209, USA (ATCC):
Material ATCC Dan. No.spggp_j_p_t ate 0NA148380-2827 PTA-1181 January 11, 2000 DNA148389-2827-1 PTA-1405 February 23,2000 These deposits were made under the provisions of the Budapest Treaty on the International Recognition of the Deposit of Microorganisms for the Purpose of Patent Procedure and the Regulations thereunder (Budapest Treaty). This assures inaintenance of a viable reefcultu the deposit far 30 years from the date of deposit The deposit will be nmde available by ATCC under the terms of the Budapest Treaty, and subject to an agreement between Genentech, Inc. and ATCC, which assures immanent and unrestricted avallabity of the progeny of the culture of the deposit to the public upon issuance of the pertinent U.S. patent or upon laying open to the public of any U.S. or foreign patent appicatioe, whichever comes first, and assures availability of the. progeny to one detennined by the U.S. Commissioner of Patents and Trademarks to be entitled thereto acceding to 35 U.S.C. 122 and the Commissioner's rules pursuant thereto (including 37 C.F.R. 1.14 with particular reference to 88600 838).
The assignee of the present appication has agreed that if a came of the materials on deposit should die or be lost or destroyed when cultivated under suitable conditions, the materials will be promptly replaced on notification with another of the same. Avadablity of the deposited material is not to be construed as a loose to practice the invention in contravention of the rights granted under the authority of any government in accordance with it: patent laws.
The foregoing written specification is considered to be sufficient to enable one skilled in the art to practice the invention. The present invention is not tube kited in scope by the construct deposited, since the deposited embedment is intended as a single illustration of certain aspects of the invention and any constructs that are functionally equivalent are within the scope of this hwention. The deposit of material heroin does not constitute an admission that the written _ description herein contained is inadequate to enable the practice of any aspect of the invention, including the best mode thereof, nor is it to be construed as kiting the scope of the claims to the specific illustrations that it represents. Indeed, various modifications of the invention in addition to those shown and deserted herein will become apparent to those skilled In the art from the foregoing description and fall within the scope of the appended daims.

SEQUENCE LISTING
<110> Pennica, Diane Smith, Victoria Wood, William I.
<120> Novel STRA6 Polypeptides <130> GENENT.2827VPC
<150> 60/197089 <151> 2000-04-14 <150> 60/175849 <151> 2000-01-13 <150> 60/228914 <151> 2000-08-29 <160> 5 <170> FastSEQ for Windows Version 4.0 <210> 1 <211> 2732 <212> DNA
<213> Homo Sapiens <220>
<221> CDS
<222> (49)...(2052) <400> 1 agtcccagac gggcttttcc cagagagcta aaagagaagg gccagaga atg tcg tcc 57 Met Ser Ser cag cca gca ggg aac cag acc tcc ccc ggg gcc aca gag gac tac tcc 105 Gin Pro Ala Gly Asn Gin Thr Ser Pro Gly Ala Thr Glu Asp Tyr Ser tat ggc agc tgg tac atc gat gag ccc cag ggg ggc gag gag ctc cag 153 Tyr Gly Ser Trp Tyr Ile Asp Glu Pro Gin Gly Gly Glu Glu Leu Gin cca gag ggg gaa gtg ccc tcc tgc cac acc agc ata cca ccc ggc ctg 201 Pro Glu Gly Glu Val Pro Ser Cys His Thr Ser Ile Pro Pro Gly Leu tac cac gcc tgc ctg gcc tcg ctg tca atc ctt gtg ctg ctg ctc ctg 249 Tyr His Ala Cys Leu Ala Ser Leu Ser Ile Leu Val Leu Leu Leu Leu gcc atg ctg gtg agg cgc cgc cag ctc tgg cot gac tgt gtg cgt ggc 297 Ala Met Leu Val Arg Arg Arg Gin Leu Trp Pro Asp Cys Val Arg Gly agg ccc ggc ctg ccc agc cct gtg gat ttc ttg gct ggg gac agg ccc 345 Arg Pro Gly Leu Pro Ser Pro Val Asp Phe Leu Ala Gly Asp Arg Pro cgg gca gtg cot got got gtt ttc atg gtc ctc ctg ago too ctg tgt 393 Arg Ala Val Pro Ala Ala Val Phe Met Val Leu Leu Ser Ser Leu Cys ttg ctg ctc ccc gac gag gac gca ttg ccc ttc ctg act ctc goo tca 441 Leu Leu Leu Pro Asp Glu Asp Ala Leu Pro Phe Leu Thr Leu Ala Ser gca ccc ago caa gat ggg aaa act gag got cca aga ggg gcc tgg aag 489 Ala Pro Ser Gin Asp Gly Lys Thr Glu Ala Pro Arg Gly Al-Trp Lys 135 140 145' ata ctg gga ctg ttc tat tat got gcc ctc tac tac cot ctg got goo 537 Ile Leu Gly Leu Phe Tyr Tyr Ala Ala Leu Tyr Tyr Pro Leu Ala Ala tgt gcc acg got ggc cac aca got gca cac ctg ctc ggc ago acg ctg 585 Cys Ala Thr Ala Gly His Thr Ala Ala His Leu Leu Gly Ser Thr Leu too tgg goo cac ctt ggg gtc cag gtc tgg cag agg gca gag tgt ccc 633 Ser Trp Ala His Leu Gly Val Gin Val Trp Gin Arg Ala Glu Cys Pro cag gtg ccc aag atc tac aag tac tac tdc ctg ctg gcc too ctg cct 681 Gln Val Pro Lys Ile Tyr Lys Tyr Tyr Ser Leu Leu Ala Ser Leu Pro ctc ctg ctg ggc ctc gga ttc ctg ago ctt tgg tac cot gtg cag ctg 729 Leu Leu Leu Gly Leu Gly Phe Leu Ser Leu Trp Tyr Pro Val Gin Leu gtg aga ago ttc ago cgt agg aca gga gca ggc tcc aag ggg ctg cag 777 Val Arg Ser Phe Ser Arg Arg Thr Gly Ala Gly Ser Lys Gly Leu Gin ago ago tac tot gag gaa tat ctg agg aac ctc ctt tgc agg aag aag 825 Ser Ser Tyr Ser Glu Glu Tyr Leu Arg Asn Leu Leu Cys Arg Lys Lys ctg gga ago ago tac cac acc too aag cat ggc ttc ctg tcc tgg gcc 873 Leu Gly Ser Ser Tyr His Thr Ser Lys His Gly Phe Leu Ser Trp Ala 260 265 '270 275 cgc gtc tgc ttg aga cac tgc atc tac act cca cag cca gga ttc cat 921 Arg Val Cys Leu Arg His Cys Ile Tyr Thr Pro Gin Pro Gly Phe His ctc ccg ctg aag ctg gtg ctt tca got aca ctg aca ggg acg gcc att 969 Leu Pro Leu Lys Leu Val Leu Ser Ala Thr Leu Thr Gly Thr Ala Ile tac cag gtg gcc ctg ctg ctg ctg gtg ggc gtg gta ccc act atc cag 1017 Tyr Gin Val Ala Leu Leu Leu Leu Val Gly Val Val Pro Thr Ile Gin aag gtg agg gca ggg gtc acc acg gat gtc tcc tac ctg ctg gcc ggc 1065 Lys Val Arg Ala Gly Val Thr Thr Asp Val Ser Tyr Leu Leu Ala Gly ttt gga atc gtg ctc tcc gag gac aag cag gag gtg gtg gag ctg gtg 1113 Phe Gly Ile Val Leu Ser Glu Asp Lys Gin Glu Val Val Glu Leu Val aag cac cat ctg tgg gct ctg gaa gtg tgc tac atc tca gcc ttg gtc 1161 Lys His His Leu Trp Ala Leu Glu Val Cys Tyr Ile Ser Ala Leu Val ttg tcc tgc tta ctc acc ttc ctg gtc ctg atg cgc tca ctg gtg aca 1209 Leu Ser Cys Leu Leu Thr Phe Leu Val Leu Met Arg Ser Leu Val Thr cac agg acc aac ctt cga gct ctg cac cga gga gct gcc ctg gac ttg 1257 His Arg Thr Asn Leu Arg Ala Leu His Arg Gly Ala Ala Leu Asp Leu agt ccc ttg cat cgg agt ccc cat ccc tcc cgc caa gcc ata ttc tgt 1305 Ser Pro Leu His Arg Ser Pro His Pro Ser Arg Gin Ala Ile She Cys tgg atg agc ttc agt gcc tac cag aca gcc ttt atc tgc ctt ggg ctc 1353 Trp Met Ser Phe Ser Ala Tyr Gin Thr Ala She Ile Cys Leu Gly Leu ctg gtg cag cag atc atc ttc ttc ctg gga acc acg gcc ctg gcc ttc 1401 Leu Val Gln Gin Ile Ile Phe She Leu Gly Thr Thr Ala Leu Ala She ctg gtg ctc atg cct gtg ctc cat ggc agg aac ctc ctg ctc ttc cgt 1449 Leu Val Leu Met Pro Val Leu His Gly Arg Asn Leu Leu Leu Phe Arg 455 460 . 465 tcc ctg gag tcc tcg tgg ccc ttc tgg ctg act ttg gcc ctg gct gtg 1497 Ser Leu Glu Ser Ser Trp Pro Phe Trp Leu Thr Leu Ala Leu Ala Val atc ctg cag aac atg gca gcc cat tgg gtc ttc ctg gag act cat gat 1545 Ile Leu Gin Asn Met Ala Ala His Trp Val She Leu Glu Thr His Asp gga cac cca cag ctg acc aac cgg cga gtg ctc tat gca gcc acc ttt 1593 Gly His Pro Gin Leu Thr Asn Arg Arg Val Leu Tyr Ala Ala Thr She ctt ctc ttc ccc ctc aat gtg ctg gtg ggt gcc atg gtg gcc acc tgg 1641 Leu Leu She Pro Leu Asn Val Leu Val Gly Ala Met Val Ala Thr Trp cga gtg ctc ctc tct gcc ctc tac aac gcc atc cac ctt ggc cag atg 1689 Arg Val Leu Leu Ser Ala Leu Tyr Asn Ala Ile His Leu Gly Gin Met gac ctc agc ctg ctg cca ccg aga gcc gcc act ctc gac ccc ggc tac 1737 Asp Leu Ser Leu Leu Pro Pro Arg Ala Ala Thr Leu Asp Pro Gly Tyr tac acg tac cga aac ttc ttg aag att gaa gtc agc cag tcg cat cca 1785 Tyr Thr Tyr Arg Asn Phe Leu Lys Ile Glu Val Ser Gin Ser His Pro gcc atg aca gcc ttc tgc tcc ctg ctc ctg caa gcg cag agc ctc cta 1833 Ala Met Thr Ala Phe Cys Ser Leu Leu Leu Gin Ala Gin Ser Leu Leu ccc agg acc atg gca gcc ccc cag gac agc ctc aga cca ggg gag gaa 1881 Pro Arg Thr Met Ala Ala Pro Gin Asp Ser Leu Arg Pro Gly Glu Glu gac gaa ggg atg cag ctg cta cag aca aag gac tcc atg gcc aag gga 1929 Asp Glu Gly Met Gin Leu Leu Gin Thr Lys Asp Ser Met Ala Lys Gly get agg ccc ggg gcc agc cgc ggc agg get cgc tgg ggt ctg gcc tac 1977 Ala Arg Pro Gly Ala Ser Arg Gly Arg Ala Arg Trp Gly Leu Ala Tyr acg ctg ctg cac aac cca ace ctg cag gtc ttc cgc aag acg gcc ctg 2025 Thr Leu Leu His Asn Pro Thr Leu Gin Val Phe Arg Lys Thr Ala Leu ttg ggt gcc aat ggt gcc cag ccc tga gggcagggaa ggtcaaccca 2072 Leu Gly Ala Asn Gly Ala Gin Pro *

cctgcccatc tgtgctgagg catgttcctg cctaccatcc tcctccctcc ccggctctcc 2132 tcccagcatc acaccagcca tgcagccagc aggtcctccg gatcactgtg gttgggtgga 2192 ggtctgtctg cactgggagc ctcaggaggg ctctgctcca cccacttggc tatgggagag 2252 ccagcagggg ttctggagaa aaaaactggt gggttagggc cttggtccag gagccagttg 2312 agccagggca gccacatcca ggcgtctccc taccctggct ctgccatcag ccttgaaggg 2372 cctcgatgaa gccttctctg gaaccactcc agcccagctc cacctcagcc ttggccttca 2432 cgctgtggaa gcagccaagg cacttcctca ccccctcagc gccacggacc tctctgggga 2492 gtggccggaa agctcccggt cctctggcct gcagggcagc ccaagtcatg actcagacca 2552 ggtcccacac tgagctgccc acactcgaga gccagatatt tttgtagttt ttatgccttt 2612 ggctattatg aaagaggtta gtgtgttccc tgcaataaac ttgttcctga gaaaaaaaaa 2672 aaaaaaaaaa aaaaaaaaaa aaaaaaaaaa aaaaaaaaaa aaaaaaaaaa aaaaaaaaaa 2732 <210> 2 <211> 667 <212> PRT
<213> Homo Sapiens <400> 2 Met Ser Ser Gin Pro Ala Gly Asn Gin Thr Ser Pro Gly Ala Thr Glu VIM) 01/51635 Asp Tyr Ser Tyr Gly Ser Trp Tyr Ile Asp Glu Pro Gin Gly Gly Glu Glu Leu Gin Pro Glu Gly Glu Val Pro Ser Cys His Thr Ser Ile Pro Pro Gly Leu Tyr His Ala Cys Leu Ala Ser Leu Ser Ile Leu Val Leu Leu Leu Leu Ala Met Leu Val Arg Arg Arg Gin Leu Trp Pro Asp Cys Val Arg Gly Arg Pro Gly Leu Pro Ser Pro Val Asp Phe Leu Ala Gly Asp Arg Pro Arg Ala Val Pro Ala Ala Val Phe Met Val Leu Leu Ser Ser Leu Cys Leu Leu Leu Pro Asp Glu Asp Ala Leu Pro Phe Leu Thr Leu Ala Ser Ala Pro Ser Gin Asp Gly Lys Thr Glu Ala Pro Arg Gly Ala Trp Lys Ile Leu Gly Leu She Tyr Tyr Ala Ala Leu Tyr Tyr Pro Leu Ala Ala Cys Ala Thr Ala Gly His Thr Ala Ala His Leu Leu Gly Ser Thr Leu Ser Trp Ala His Leu Gly Val Gin Val Trp Gin Arg Ala Glu Cys Pro Gin Val Pro Lys Ile Tyr Lys Tyr Tyr Ser Leu Leu Ala Ser Leu Pro Leu Leu Leu Gly Leu Gly Phe Leu Ser Leu Trp Tyr Pro Val Gin Leu Val Arg Ser She Ser Arg Arg Thr Gly Ala Gly Ser Lys Gly Leu Gln Ser Ser Tyr Ser Glu Glu Tyr Leu Arg Asn Leu Leu Cys Arg Lys Lys Leu Gly Ser Ser Tyr His Thr Ser Lys His Gly Phe Leu Ser Trp Ala Arg Val Cys Leu Arg His Cys Ile Tyr Thr Pro Gin Pro Gly Phe His Leu Pro Leu Lys Leu Val Leu Ser Ala Thr Leu Thr Gly Thr Ala Ile Tyr Gin Val Ala Leu Leu Leu Leu Val Gly Val Val Pro Thr Ile Gin Lys Val Arg Ala Gly Val Thr Thr Asp Val Ser Tyr Leu Leu Ala Gly She Gly Ile Val Leu Ser Glu Asp Lys Gin Glu Val Val Glu Leu Val Lys His His Leu Trp Ala Leu Glu Val Cys Tyr Ile Ser Ala Leu Val Leu Ser Cys Leu Leu Thr Phe Leu Val Leu Met Arg Ser Leu Val Thr His Arg Thr Asn Leu Arg Ala Leu His Arg Gly Ala Ala Leu Asp Leu Ser Pro Leu His Arg Ser Pro His Pro Ser Arg Gin Ala Ile Phe Cys Trp Met Ser She Ser Ala Tyr Gin Thr Ala Phe Ile Cys Leu Gly Leu Leu Val Gin in Ile Ile She Phe Leu Gly Thr Thr Ala Leu Ala Phe Leu Val Leu Met Pro Val Leu His Gly Arg Asn Leu Leu Leu Phe Arg Ser Leu Glu Ser Ser Trp Pro Phe Trp Leu Thr Leu Ala Leu Ala Val Ile Leu Gln Asn Met Ala Ala His Trp Val Phe Leu Glu Thr His Asp Gly His Pro Gln Leu Thr Asn Arg Arg Val Leu Tyr Ala Ala Thr Phe Leu Leu Phe Pro Leu Asn Val Leu Val Gly Ala Met Val Ala Thr Trp Arg Val Leu Leu Ser Ala Leu Tyr Asn Ala Ile His Leu Gly Gln Met Asp Leu Ser Leu Leu Pro Pro Arg Ala Ala Thr Leu Asp Pro Gly Tyr Tyr Thr Tyr Arg Asn Phe Leu Lys Ile Glu Val Her Gln Ser His Pro Ala Met Thr Ala Phe Cys Ser Leu Leu Leu Gln Ala Gln Ser Leu Leu Pro Arg Thr Met Ala Ala Pro Gln Asp Ser Leu Arg Pro Gly Glu Glu Asp Glu Gly Met Gln Leu Leu Gln Thr Lys Asp Ser Met Ala Lys Gly Ala Arg Pro Gly Ala Ser Arg Gly Arg Ala Arg Trp Gly Leu Ala Tyr Thr Leu Leu His Asn Pro Thr Leu Gln Val Phe Arg Lys Thr Ala Leu Leu Gly Ala Asn Gly Ala Gln Pro <210> 3 <211> 676 <212> DNA
<213> Homo sapiens <220>
<221> misc_feature <222> (26)...(26) <223> n ¨ A, T, C or G
<400> 3 gtgctctccg aggacaagca ggaggnggtg gagctggtga agcaccatct gtgggctctg 60 gaagtgtgct acatctcagc cttggtcttg tcctgattac tcaccttcct ggtcctgatg 120 cgctcactgg tgacacacag gaccaacctt cgagctctgc accgaggagc tgccctggac 180 ttgagtccct tgcatcggag tccccatccc toccgccaag ccatattctg ttggatgagc 240 ttcagtgcct accagacagc ctttatctgc cttgggctcc tggtgcagca gatcatcttc 300 ttcctgggaa ccacggccct ggccttcctg gtgctcatgc ctgtgctcca tggcaggaac 360 ctcctgctct tccgttccct ggagtcctcg tggcccttct ggctgacttt ggccctggct 420 gtgatcctgc agaacatggc agcccattgg gtcttcctgg agactcatga tggacaccca 480 cagctgacca accggcgagt gctctatgca gccacctttc ttctcttccc cctcaatgtg 540 ctggtgggtg ccatggtggc cacctggcga gtgctcctct ctgccctcta caacgccatc 600 caccttggcc agatggacct cagcctgctg ccaccgagag ccgccactct cgaccccggc 660 tactacacgt accgaa 676 <210> 4 <211> 2777 <212> DNA
<213> Homo sapiens <220>
<221> CDS
<222> (186)...(2160) <400> 4 cacaaccagc cacccctcta ggatcccagc ccagctggtg ctgggctcag aggagaaggc 60 cccgtgttgg gagcaccctg cttgcctgga gggacaagtt tccgggagag atcaataaag 120 gaaaggaaag agacaaggaa gggagaggtc aggagagcgc ttgattggag gagaagggcc 180 agaga atg tcg tcc cag cca gca ggg aac cag acc tcc ccc ggg gcc aca 230 Met Ser Ser Gin Pro Ala Gly Asn Gin Thr Ser Pro Gly Ala Thr gag gac tac tcc tat ggc agc tgg tac atc gat gag ccc cag ggg ggc 278 Glu Asp Tyr Ser Tyr Gly Ser Trp Tyr Ile Asp Glu Pro Gin Gly Gly gag gag ctc cag cca gag ggg gaa gtg ccc tcc tgc cac acc agc ata 326 Glu Glu Leu Gin Pro Glu Gly Glu Val Pro Ser Cys His Thr Ser Ile cca ccc ggc ctg tac cac gcc tgc ctg gcc tcg ctg tca atc ctt gtg 374 Pro Pro Gly Leu Tyr His Ala Cys Leu Ala Ser Leu Ser Ile Leu Val ctg ctg ctc ctg gcc atg ctg gtg agg cgc cgc cag ctc tgg cct gac 422 Leu Leu Leu Leu Ala Met Leu Val Arg Arg Arg Gin Leu Trp Pro Asp tgt gtg cgt ggc agg ccc ggc ctg ccc agg ccc cgg gca gtg cct got 470 Cys Val Arg Gly Arg Pro Gly Leu Pro Arg Pro Arg Ala Val Pro Ala got gtt ttc atg gtc ctc ctg agc tcc ctg tgt ttg ctg ctc ccc gac 518 Ala Val Phe Met Val Leu Leu Ser Ser Leu Cys Leu Leu Leu Pro Asp gag gac gca ttg ccc ttc ctg act ctc gcc tca gca ccc agc caa gat 566 Glu Asp Ala Leu Pro Phe Leu Thr Leu Ala Ser Ala Pro Ser Gin Asp ggg aaa act gag got cca aga ggg gcc tgg aag ata ctg gga ctg ttc 614 Gly Lys Thr Glu Ala Pro Arg Gly Ala Trp Lys Ile Leu Gly Leu Phe tat tat got gcc ctc tac tac cct ctg got gcc tgt gcc acg got ggc 662 Tyr Tyr Ala Ala Leu Tyr Tyr Pro Leu Ala Ala Cys Ala Thr Ala Gly cac aca got gca cac ctg ctc ggc agc acg ctg tcc tgg gcc cac ctt 710 His Thr Ala Ala His Leu Leu Gly Ser Thr Leu Ser Trp Ala His Leu ggg gtc cag gtc tgg cag agg gca gag tgt ccc cag gtg ccc aag atc 758 Gly Val Gin Val Trp Gin Arg Ala Glu Cys Pro Gln Val Pro Lys Ile tac aag tac tac tcc ctg ctg gcc tcc ctg cct ctc ctg ctg ggc ctc 806 Tyr Lys Tyr Tyr Ser Leu Leu Ala Ser Leu Pro Leu Leu Lau Gly Leu gga ttc ctg ago ctt tgg tac cot gtg cag ctg gtg aga ago ttc ago 854 Gly Phe Leu Ser Leu Trp Tyr Pro Val Gin Leu Val Arg Ser Phe Ser cgt agg aca gga gca ggc too aag ggg ctg cag ago agc tac tot gag 902 Arg Arg Thr Gly Ala Gly Ser Lys Gly Leu Gin Ser Ser Tyr Ser Glu gaa tat ctg agg aac ctc ctt tgc agg aag aag ctg gga ago ago tac 950 Glu Tyr Leu Arg Asn Leu Leu Cys Arg Lys Lys Leu Gly Ser Ser Tyr cac acc too aag cat ggc ttc ctg too tgg gcc cgc gtc tgc ttg aga 998 His Thr Ser Lys His Gly Phe Leu Ser Trp Ala Arg Val Cys Leu Arg cac tgc atc tac act cca cag cca gga ttc cat ctc cog ctg aag ctg 1046 His Cys Ile Tyr Thr Pro Gin Pro Gly Phe His Leu Pro Leu Lys Leu gtg ctt tca got aca ctg aca ggg acg gcc att tac cag gtg gcc ctg 1094 Val Leu Ser Ala Thr Leu Thr Gly Thr Ala Ile Tyr Gin Val Ala Leu ctg ctg ctg gtg ggc gtg gta ccc act atc cag aag gtg agg gca ggg 1142 Leu Leu Leu Val Gly Val Val Pro Thr Ile Gin Lys Val Arg Ala Gly gtc acc acg gat gtc too tac ctg ctg gcc ggc ttt gga atc gtg ctc 1190 Val Thr Thr Asp Val Ser Tyr Leu Leu Ala Gly Phe Gly Ile Val Leu too gag gac aag cag gag gtg gtg gag ctg gtg aag cac cat ctg tgg 1238 Ser Glu Asp Lys Gin Glu Val Val Glu Leu Val Lys His His Leu Trp got ctg gaa gtg tgc tac atc tca gcc ttg gtc ttg too tgc tta ctc 1286 Ala Leu Glu Val Cys Tyr Ile Ser Ala Leu Val Leu Ser Cys Leu Leu acc ttc ctg gtc ctg atg cgc tca ctg gtg aca cac agg acc aac ctt 1334 Thr Phe Leu Val Leu Met Arg Ser Leu Val Thr His Arg Thr Asn Leu cga got ctg cac cga gga got gcc ctg gac ttg agt ccc ttg cat cgg 1382 Arg Ala Leu His Arg Gly Ala Ala Leu Asp Leu Ser Pro Leu His Arg agt ccc cat ccc too cgc caa gcc ata ttc tgt tgg atg ago ttc agt 1430 Ser Pro His Pro Ser Arg Gin Ala Ile Phe Cys Trp Met Ser Phe Ser gcc tac cag aca gcc ttt atc tgc ctt ggg ctc ctg gtg cag cag atc 1478 Ala Tyr Gin Thr Ala Phe Ile Cys Leu Gly Leu Leu Val Gin Gin Ile atc ttc ttc ctg gga acc acg gcc ctg gcc ttc ctg gtg ctc atg cct 1526 Ile Phe Phe Leu Gly Thr Thr Ala Leu Ala Phe Leu Val Leu Met Pro gtg ctc cat ggc agg aac ctc ctg ctc ttc cgt tcc ctg gag tcc tog 1574 Val Leu His Gly Arg Asn Leu Leu Leu Phe Arg Ser Leu Glu Ser Ser tgg ccc ttc tgg ctg act ttg gcc ctg got gtg atc ctg cag aac atg 1622 Trp Pro Phe Trp Leu Thr Leu Ala Leu Ala Val Ile Leu Gin Asn Met gca gcc cat tgg gtc ttc ctg gag act cat gat gga cac cca cag ctg 1670 Ala Ala His Trp Val Phe Leu Glu Thr His Asp Gly His Pro Gin Leu acc aac cgg cga gtg ctc tat gca gcc acc ttt ctt ctc ttc ccc ctc 1718 Thr Asn Arg Arg Val Leu Tyr Ala Ala Thr Phe Leu Leu Phe Pro Leu aat gtg ctg gtg ggt gcc ata gtg gcc acc tgg cga gtg ctc ctc tct 1766 Asn Val Leu Val Gly Ala Ile Val Ala Thr Trp Arg Val Leu Leu Ser gcc ctc tac aac gcc atc cac ctt ggc cag atg gac ctc ago ctg ctg 1814 Ala Leu Tyr Asn Ala Ile His Leu Gly Gin Met Asp Leu Ser Leu Leu cca cog aga gcc gcc act ctc gac ccc ggc tac tac acg tac cga aac 1862 Pro Pro Arg Ala Ala Thr Leu Asp Pro Gly Tyr Tyr Thr Tyr Arg Asn ttc ttg aag att gaa gtc ago cag tog cat cca gcc atg aca gcc ttc 1910 Phe Leu Lys Ile Glu Val Ser Gin Ser His Pro Ala Met Thr Ala Phe tgc tcc ctg ctc ctg caa gcg cag agc ctc cta ccc agg acc atg gca 1958 Cys Ser Leu Leu Leu Gin Ala Gin Ser Leu Leu Pro Arg Thr Met Ala gcc ccc cag gac ago ctc aga cca ggg gag gaa gac gaa ggg atg cag 2006 Ala Pro Gin Asp Ser Leu Arg Pro Gly Glu Glu Asp Glu Gly Met Gin ctg cta cag aca aag gac tcc atg gcc aag gga got agg ccc ggg gcc 2054 Leu Leu Gin Thr Lys Asp Ser Met Ala Lys Gly Ala Arg Pro Gly Ala ago cgc ggc agg got cgc tgg ggt ctg gcc tac acg ctg ctg cac aac 2102 Ser Arg Gly Arg Ala Arg Trp Gly Leu Ala Tyr Thr Leu Leu His Asn cca acc ctg cag gtc ttc cgc aag acg gcc ctg ttg ggt gcc aat ggt 2150 Pro Thr Leu Gin Val Phe Arg Lys Thr Ala Leu Leu Gly Ala Asn Gly gcc cag ccc t gagggcaggg aaggtcaacc cacctgccca tctgtgctga 2200 Ala Gln Pro ggcatgttcc tgcctaccac ctcctccctc cccggctctc ctcccagcat cacaccagcc 2260 atgcagccag caggtcctcc ggatcactgt ggttgggtgg aggtctgtct gcactgggag 2320 cctcaggagg gctctgctcc acccacttgg ctatgggaga gccagcaggg gttctggaga 2380 aagaaactgg tgggttaggg ccttggtcca ggagccagtt gagccagggc agccacatcc 2440 aggcgtctcc ctaccctggc tctgccatca gccttgaagg gcctcgatga agccttctct 2500 ggaaccactc cagcccagct ccacctcagc cttggccttc acgctgtgga agcagccaag 2560 gcacttcctc accccctcag cgccacggac ctctctgggg agtggccgga aagctcccgg 2620 gcctctggcc tgcagggcag cccaagtcat gactcagacc aggtcccaca ctgagctgcc 2680 cacactcgag agccagatat ttttgtagtt tttatgcctt tggctattat gaaagaggtt 2740 agtgtgttcc ctgcaataaa cttgttcctg agaaaaa 2777 <210> 5 <211> 658 <212> PRT
<213> Homo sapiens <400> 5 Met Ser Ser Gin Pro Ala Gly Asn Gln Thr Ser Pro Gly Ala Thr Glu Asp Tyr Ser Tyr Gly Ser Trp Tyr Ile Asp Glu Pro Gln Gly Gly Glu Glu Leu Gln Pro Glu Gly Glu Val Pro Ser Cys His Thr Ser Ile Pro Pro Gly Leu Tyr His Ala Cys Leu Ala Ser Leu Ser Ile Leu Val Leu Leu Leu Leu Ala Met Leu Val Arg Arg Arg Gln Leu Trp Pro Asp Cys Val Arg Gly Arg Pro Gly Leu Pro Arg Pro Arg Ala Val Pro Ala Ala Val Phe Met Val Leu Leu Ser Ser Leu Cys Leu Leu Leu Pro Asp Glu Asp Ala Leu Pro Phe Leu Thr Leu Ala Ser Ala Pro Ser Gln Asp Gly Lys Thr Glu Ala Pro Arg Gly Ala Trp Lys Ile Leu Gly Leu Phe Tyr Tyr Ala Ala Leu Tyr Tyr Pro Leu Ala Ala Cys Ala Thr Ala Gly His Thr Ala Ala His Leu Leu Gly Ser Thr Leu Ser Trp Ala His Leu Gly Val Gln Val Trp Gln Arg Ala Glu Cys Pro Gln Val Pro Lys Ile Tyr Lys Tyr Tyr Ser Leu Leu Ala Ser Leu Pro Leu Leu Leta Gly Leu Gly Phe Leu Ser Leu Trp Tyr Pro Val Gln Leu Val Arg Ser Phe Ser Arg Arg Thr Gly Ala Gly Ser Lys Gly Leu Gln Ser Ser Tyr Ser Glu Glu =

Tyr Leu Arg Asn Leu Leu Cys Arg Lys Lys Leu Gly Ser Ser Tyr His Thr Ser Lys His Gly Phe Leu Ser Trp Ala Arg Val Cys Leu Arg His Cys Ile Tyr Thr Pro Gin Pro Gly Phe His Leu Pro Leu Lys Leu Val Leu Ser Ala Thr Leu Thr Gly Thr Ala Ile Tyr Gln Val Ala Leu Leu Leu Leu Val Gly Val Val Pro Thr Ile Gln Lys Val Arg Ala Gly Val Thr Thr Asp Val Ser Tyr Leu Leu Ala Gly Phe Gly Ile Val Leu Ser Glu Asp Lys Gln Glu Val Val Glu Leu Val Lys His His Leu Trp Ala Leu Glu Val Cys Tyr Ile Ser Ala Leu Val Leu Ser Cys Leu Leu Thr Phe Leu Val Leu Met Arg Ser Leu Val Thr His Arg Thr Asn Leu Arg Ala Leu His Arg Gly Ala Ala Leu Asp Leu Ser Pro Leu His Arg Ser Pro His Pro Ser Arg Gln Ala Ile Phe Cys Trp Met Ser Phe Ser Ala Tyr Gln Thr Ala Phe Ile Cys Leu Gly Leu Leu Val Gln Gln Ile Ile Phe Phe Leu Gly Thr Thr Ala Leu Ala Phe Leu Val Leu Met Pro Val Leu His Gly Arg Asn Leu Leu Leu Phe Arg Ser Leu Glu Ser Ser Trp Pro She Trp Leu Thr Leu Ala Leu Ala Val Ile Leu Gln Asn Met Ala Ala His Trp Val Phe Leu Glu Thr His Asp Gly His Pro Gln Leu Thr Asn Arg Arg Val Leu Tyr Ala Ala Thr Phe Leu Leu Phe Pro Leu Asn Val Leu Val Gly Ala Ile Val Ala Thr Trp Arg Val Leu Leu Ser Ala Leu Tyr Asn Ala Ile His Leu Gly Gln Met Asp Leu Ser Leu Leu Pro Pro Arg Ala Ala Thr Leu Asp Pro Gly' Tyr Tyr Thr Tyr Arg Asn She Leu Lys Ile Glu Val Ser Gln Ser His Pro Ala Met Thr Ala She Cys Ser Leu Leu Leu Gln Ala Gln Ser Leu Leu Pro Arg Thr Met Ala Ala Pro Gln Asp Ser Leu Arg Pro Gly Glu Glu Asp Glu Gly Met Gln Leu Leu Gln Thr Lys Asp Ser Met Ala Lys Gly Ala Arg Pro Gly Ala Ser Arg Gly Arg Ala Arg Trp Gly Leu Ala Tyr Thr Leu Leu His Asn Pro Thr Leu Gln Val Phe Arg Lys Thr Ala Leu Leu Gly Ala Asn Gly Ala Gln Pro

Claims (25)

CLAIMS:
1. An isolated nucleic acid molecule which comprises (a) a DNA molecule encoding Stra6 polypeptide having the sequence of amino acid residues from about 1 to about 658 of SEQ ID NO:5, or (b) the complement of the DNA molecule of (a).
2. The isolated nucleic acid molecule of claim 1 comprising the sequence of (a) nucleotide positions from about 186 to about 2159 of SEQ ID NO:4, or (b) the complement of the nucleotide sequence of (a).
3. The isolated nucleic acid molecule of claim 1 comprising the nucleotide sequence of SEQ ID NO:4.
4. An isolated nucleic acid molecule encoding a Stra6 polypeptide comprising DNA that hybridizes under stringent hybridization conditions to the complement of the nucleic acid sequence that encodes amino acids 1 to about 658 of SEQ ID NO:5, wherein the hybridization is conducted under the following conditions:
(a) 50% (v/v) formamide with 0.1% bovine serum albumin/0.1% Fico11/0.1% polyvinylpyrrolidone/50 mM sodium phosphate buffer at pH 6.5 with 750 mM sodium chloride, 75 mM sodium citrate at 42°C;
(b) 50% formamide, 5 x SSC (0.75 M NaCI, 0.075 M sodium citrate), 0.1% sodium dodecyl phosphate (SDS), 0.1% sodium pyrophosphate, 50 mM sodium phosphate, pH 6.8, 2 x Denhardt's solution, and 10% dextran sulfate at 42°C; or (c) 50% formamide, 5 x SSC, 50 mM sodium phosphate (pH 6.8), 0.1%
sodium pyrophosphate, 5 x Denhardt's solution, sonicated salmon sperm DNA
(50 g/ml), 0.1% SDS, and 10% dextran sulfate at 42°C;
and washing is conducted under the following conditions:
(d) 0.1 x SSC and 0.1% SDS at 42°C;

(e) 0.015 M sodium chloride/0.0015 M sodium citrate/0.1% SDS
at 50°C;
(f) 0.2 x SSC at 42°C;
(g) 50% formamide at 55°C; or (h) 0.1 x SSC containing EDTA at 55°C.
5. The isolated nucleic acid molecule of claim 4, wherein the nucleic acid that encodes amino acids 1 to about 658 of SEQ ID NO:5 comprises nucleotides to about 2159 of SEQ ID NO:4.
6. A vector comprising the nucleic acid molecule of any one of claims 1 to 5.
7. The vector of claim 6, wherein said nucleic acid molecule is operably linked to control sequences recognized by a host cell transformed with the vector.
8. A host cell comprising the vector of claim 6 or claim 7.
9. The host cell of claim 8, wherein said cell is a CHO cell.
10. The host cell of claim 8, wherein said cell is an E. coli.
11. The host cell of claim 8, wherein said cell is a yeast cell.
12. A process for producing a Stra6 polypeptide comprising culturing the host cell of any one of claims 8-11 under conditions suitable for expression of said Stra6 polypeptide and recovering said Stra6 polypeptide from the cell culture.
13. An isolated Stra6 polypeptide comprising amino acid residues 1 to 658 of SEQ ID NO:5.
14. A chimeric molecule comprising a Stra6 polypeptide according to claim 13 fused to a heterologous amino acid sequence.
15. The chimeric molecule of claim 14, wherein said heterologous amino acid sequence is an epitope tag sequence.
16. The chimeric molecule of claim 14, wherein said heterologous amino acid sequence is a Fc region of an immunoglobulin.
17. An antibody which specifically binds to a Stra6 polypeptide according to claim 13.
18. The antibody of claim 17 which is a monoclonal antibody.
19. The antibody of claim 17 which is labeled.
20. The antibody of any one of claims 17-19 wherein the antibody is conjugated to a cytotoxic agent or prodrug activating agent.
21. The antibody of any one of claims 17-19, wherein the antibody is a heteroconjugate antibody.
22. A composition which comprises the antibody of any one of claims 17-19 in admixture with a pharmaceutical acceptable carrier.
23. The composition of matter of claim 22, further comprising a retinoid.
24. The composition of claim 23, wherein the retinoid comprises retinoic acid or an analogue thereof.
25. The composition of matter of claim 22, which further comprises a cytotoxic agent or prodrug activating agent.
CA2397207A 2000-01-13 2001-01-11 Novel stra6 polypeptides Expired - Fee Related CA2397207C (en)

Applications Claiming Priority (7)

Application Number Priority Date Filing Date Title
US17584900P 2000-01-13 2000-01-13
US60/175,849 2000-01-13
US19708900P 2000-04-14 2000-04-14
US60/197,089 2000-04-14
US22891400P 2000-08-29 2000-08-29
US60/228,914 2000-08-29
PCT/US2001/000847 WO2001051635A2 (en) 2000-01-13 2001-01-11 Novel stra6 polypeptides

Publications (2)

Publication Number Publication Date
CA2397207A1 CA2397207A1 (en) 2001-07-19
CA2397207C true CA2397207C (en) 2013-12-03

Family

ID=27390600

Family Applications (1)

Application Number Title Priority Date Filing Date
CA2397207A Expired - Fee Related CA2397207C (en) 2000-01-13 2001-01-11 Novel stra6 polypeptides

Country Status (15)

Country Link
US (7) US7173115B2 (en)
EP (1) EP1246917B1 (en)
JP (2) JP2003519491A (en)
KR (2) KR20080006002A (en)
CN (2) CN101643733A (en)
AT (1) ATE424457T1 (en)
AU (2) AU785055B2 (en)
CA (1) CA2397207C (en)
DE (1) DE60137829D1 (en)
DK (1) DK1246917T3 (en)
ES (1) ES2323220T3 (en)
IL (2) IL150676A0 (en)
NZ (1) NZ520095A (en)
PT (1) PT1246917E (en)
WO (1) WO2001051635A2 (en)

Families Citing this family (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ATE424457T1 (en) * 2000-01-13 2009-03-15 Genentech Inc HUMAN STRA6 POLYPEPTIDES
EP1334126A1 (en) * 2000-03-23 2003-08-13 Curagen Corporation Human stra6-like protein and nucleic acids encoding the same
CN1339489A (en) * 2000-08-23 2002-03-13 上海博德基因开发有限公司 New polypeptide-human retinoic acid modulator protein 53.57 and polynucleotide for encoding such polypeptide
DE60140966D1 (en) * 2000-08-29 2010-02-11 Genentech Inc Method for improving cancer therapy
US7682607B2 (en) * 2001-05-01 2010-03-23 The Regents Of The University Of California Wnt and frizzled receptors as targets for immunotherapy in head and neck squamous cell carcinomas
EP2799555B1 (en) * 2002-03-13 2017-02-22 Genomic Health, Inc. Gene expression profiling in biopsied tumor tissues
US7666627B2 (en) * 2002-08-08 2010-02-23 Targetex Kft. Folded recombinant catalytic fragments of multidomain serine proteases, preparation and uses thereof
AU2003282306A1 (en) * 2002-10-29 2004-05-25 Engene, Inc. Compositions for cancer treatment
CA2524172A1 (en) * 2003-04-30 2004-11-18 Centocor, Inc. Cngh0010 specific polynucleotides, polypeptides, antibodies, compositions, methods and uses
US7763067B2 (en) * 2004-09-01 2010-07-27 C. R. Bard, Inc. Stent and method for manufacturing the stent
WO2008089438A2 (en) * 2007-01-18 2008-07-24 The Regents Of The University Of California A membrane receptor for retinol binding protein mediates cellular uptake of vitamin a, methods of use and compositions
US20110159017A1 (en) * 2008-04-11 2011-06-30 Ludwig Institute For Cancer Research Ltd. Trytophan catabolism in cancer treatment and diagnosis
EP2445539A4 (en) * 2009-06-26 2015-05-20 Five Prime Therapeutics Inc Therapeutic antibody target validation and screening in vivo
JP6286124B2 (en) * 2010-01-11 2018-02-28 ジェノミック ヘルス, インコーポレイテッド A method to determine the likelihood of clinical outcome of renal cancer using gene expression
SG10201912361VA (en) * 2010-07-27 2020-02-27 Genomic Health Inc Method for using gene expression to determine prognosis of prostate cancer
SG10201605210PA (en) 2012-01-31 2016-08-30 Genomic Health Inc Gene expression profile algorithm and test for determining prognosis of prostate cancer
EP2812350B1 (en) 2012-02-11 2019-04-03 F.Hoffmann-La Roche Ag R-spondin translocations and methods using the same
DK3004392T3 (en) 2013-05-30 2020-10-26 Genomic Health Inc GENE EXPRESSION PROFILE ALGORM FOR CALCULATING A RECURRENCY SCORE FOR A PATIENT WITH KIDNEY CANCER
EP3636661A1 (en) * 2018-10-12 2020-04-15 Bioc3 Phosphate translocator

Family Cites Families (81)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3773919A (en) 1969-10-23 1973-11-20 Du Pont Polylactide-drug mixtures
US4179337A (en) 1973-07-20 1979-12-18 Davis Frank F Non-immunogenic polypeptides
FR2413974A1 (en) 1978-01-06 1979-08-03 David Bernard DRYER FOR SCREEN-PRINTED SHEETS
US4275149A (en) 1978-11-24 1981-06-23 Syva Company Macromolecular environment control in specific receptor assays
US4657760A (en) 1979-03-20 1987-04-14 Ortho Pharmaceutical Corporation Methods and compositions using monoclonal antibody to human T cells
WO1981001145A1 (en) 1979-10-18 1981-04-30 Univ Illinois Hydrolytic enzyme-activatible pro-drugs
US4399216A (en) 1980-02-25 1983-08-16 The Trustees Of Columbia University Processes for inserting DNA into eucaryotic cells and for producing proteinaceous materials
ZA811368B (en) 1980-03-24 1982-04-28 Genentech Inc Bacterial polypedtide expression employing tryptophan promoter-operator
US4873191A (en) 1981-06-12 1989-10-10 Ohio University Genetic transformation of zygotes
US4485045A (en) 1981-07-06 1984-11-27 Research Corporation Synthetic phosphatidyl cholines useful in forming liposomes
NZ201705A (en) 1981-08-31 1986-03-14 Genentech Inc Recombinant dna method for production of hepatitis b surface antigen in yeast
US4640835A (en) 1981-10-30 1987-02-03 Nippon Chemiphar Company, Ltd. Plasminogen activator derivatives
US4444545A (en) * 1982-04-08 1984-04-24 Sanders David F Pump control system
US4943529A (en) 1982-05-19 1990-07-24 Gist-Brocades Nv Kluyveromyces as a host strain
US4485054A (en) * 1982-10-04 1984-11-27 Lipoderm Pharmaceuticals Limited Method of encapsulating biologically active materials in multilamellar lipid vesicles (MLV)
US4870009A (en) 1982-11-22 1989-09-26 The Salk Institute For Biological Studies Method of obtaining gene product through the generation of transgenic animals
US4713339A (en) 1983-01-19 1987-12-15 Genentech, Inc. Polycistronic expression vector construction
AU2353384A (en) 1983-01-19 1984-07-26 Genentech Inc. Amplification in eukaryotic host cells
NZ207394A (en) 1983-03-08 1987-03-06 Commw Serum Lab Commission Detecting or determining sequence of amino acids
US4816567A (en) * 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US4544545A (en) 1983-06-20 1985-10-01 Trustees University Of Massachusetts Liposomes containing modified cholesterol for organ targeting
AU3145184A (en) 1983-08-16 1985-02-21 Zymogenetics Inc. High expression of foreign genes in schizosaccharomyces pombe
US4496689A (en) 1983-12-27 1985-01-29 Miles Laboratories, Inc. Covalently attached complex of alpha-1-proteinase inhibitor with a water soluble polymer
US4736866B1 (en) 1984-06-22 1988-04-12 Transgenic non-human mammals
EP0206448B1 (en) 1985-06-19 1990-11-14 Ajinomoto Co., Inc. Hemoglobin combined with a poly(alkylene oxide)
US5206344A (en) 1985-06-26 1993-04-27 Cetus Oncology Corporation Interleukin-2 muteins and polymer conjugation thereof
US4676980A (en) 1985-09-23 1987-06-30 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Target specific cross-linked heteroantibodies
AU597574B2 (en) 1986-03-07 1990-06-07 Massachusetts Institute Of Technology Method for enhancing glycoprotein stability
US4676417A (en) * 1986-04-14 1987-06-30 Richard Hirschkoff Ski carrier
US4791192A (en) 1986-06-26 1988-12-13 Takeda Chemical Industries, Ltd. Chemically modified protein with polyethyleneglycol
GB8705477D0 (en) 1987-03-09 1987-04-15 Carlton Med Prod Drug delivery systems
US5010182A (en) 1987-07-28 1991-04-23 Chiron Corporation DNA constructs containing a Kluyveromyces alpha factor leader sequence for directing secretion of heterologous polypeptides
US4975278A (en) 1988-02-26 1990-12-04 Bristol-Myers Company Antibody-enzyme conjugates in combination with prodrugs for the delivery of cytotoxic agents to tumor cells
US4997852A (en) 1987-08-26 1991-03-05 Ohio State University Research Foundation Method and composition for achieving cancer chemopreventive and chemotherapeutic activity
IL87737A (en) 1987-09-11 1993-08-18 Genentech Inc Method for culturing polypeptide factor dependent vertebrate recombinant cells
DE3889546T2 (en) 1987-12-21 1994-09-08 Univ Toledo TRANSFORMATION OF Germinating PLANT SEEDS WITH THE HELP OF AGROBACTERIUM.
SE8802525L (en) 1988-07-06 1990-01-07 Fmg Filipstad Ab CHAIR
AU4005289A (en) 1988-08-25 1990-03-01 Smithkline Beecham Corporation Recombinant saccharomyces
GB8823869D0 (en) 1988-10-12 1988-11-16 Medical Res Council Production of antibodies
US5530101A (en) * 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
US5225538A (en) 1989-02-23 1993-07-06 Genentech, Inc. Lymphocyte homing receptor/immunoglobulin fusion proteins
US5009772A (en) 1989-02-27 1991-04-23 Kerr-Mcgee Corporation Solvent extraction process
ATE144281T1 (en) 1989-04-28 1996-11-15 Rhein Biotech Proz & Prod Gmbh YEAST CELLS OF THE GENUS SCHWANNIOMYCES
FR2646437B1 (en) 1989-04-28 1991-08-30 Transgene Sa NOVEL DNA SEQUENCES, THEIR APPLICATION AS A SEQUENCE ENCODING A SIGNAL PEPTIDE FOR THE SECRETION OF MATURE PROTEINS BY RECOMBINANT YEASTS, EXPRESSION CASSETTES, PROCESSED YEASTS AND PROCESS FOR PREPARING THE SAME
WO1990013641A1 (en) 1989-05-10 1990-11-15 Sloan-Kettering Institute For Cancer Research Stably transformed eucaryotic cells comprising a foreign transcribable dna under the control of a pol iii promoter
DE3920358A1 (en) 1989-06-22 1991-01-17 Behringwerke Ag BISPECIFIC AND OLIGO-SPECIFIC, MONO- AND OLIGOVALENT ANTI-BODY CONSTRUCTS, THEIR PRODUCTION AND USE
ATE144793T1 (en) 1989-06-29 1996-11-15 Medarex Inc BISPECIFIC REAGENTS FOR AIDS THERAPY
FR2649120B1 (en) 1989-06-30 1994-01-28 Cayla NOVEL STRAIN AND ITS MUTANTS OF FILAMENTOUS MUSHROOMS, PROCESS FOR PRODUCING RECOMBINANT PROTEINS USING SAID STRAIN, AND STRAINS AND PROTEINS OBTAINED BY SAID METHOD
WO1991004753A1 (en) 1989-10-02 1991-04-18 Cetus Corporation Conjugates of antisense oligonucleotides and therapeutic uses thereof
US5225212A (en) 1989-10-20 1993-07-06 Liposome Technology, Inc. Microreservoir liposome composition and method
US5013556A (en) 1989-10-20 1991-05-07 Liposome Technology, Inc. Liposomes with enhanced circulation time
US5625126A (en) 1990-08-29 1997-04-29 Genpharm International, Inc. Transgenic non-human animals for producing heterologous antibodies
KR100272077B1 (en) 1990-08-29 2000-11-15 젠팜인터내셔날,인코포레이티드 Transgenic non-human animals capable of producing heterologous antibodies
US5545806A (en) 1990-08-29 1996-08-13 Genpharm International, Inc. Ransgenic non-human animals for producing heterologous antibodies
US5661016A (en) 1990-08-29 1997-08-26 Genpharm International Inc. Transgenic non-human animals capable of producing heterologous antibodies of various isotypes
US5633425A (en) 1990-08-29 1997-05-27 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
US5122469A (en) 1990-10-03 1992-06-16 Genentech, Inc. Method for culturing Chinese hamster ovary cells to improve production of recombinant proteins
JPH07501451A (en) 1991-11-25 1995-02-16 エンゾン・インコーポレイテッド Multivalent antigen binding protein
US5731167A (en) 1992-01-17 1998-03-24 The United States Of America As Represented By The Department Of Health And Human Services Autotaxin: motility stimulating protein useful in cancer diagnosis and therapy
US5756448A (en) 1992-02-26 1998-05-26 The General Hospital Corporation Constitute activator of retinoid (CAR) receptor polypeptides
WO1993018186A1 (en) 1992-03-04 1993-09-16 The Regents Of The University Of California Comparative genomic hybridization (cgh)
DE122004000036I1 (en) 1992-11-13 2005-07-07 Biogen Idec Inc Therapeutic use of chimeric and labeled antibodies to human B lymphocyte limited differentiation antigen for the treatment of B-cell lymphoma.
EP0674506B1 (en) 1992-12-02 2000-08-23 Alkermes Controlled Therapeutics, Inc. Controlled release growth hormone containing microspheres
US5547947A (en) * 1993-03-11 1996-08-20 Hoffmann-La Roche Inc. Methods of treatment
PT616812E (en) 1993-03-24 2000-04-28 Berlex Biosciences COMBINATION OF ANTI-HORMONAL COMPOUNDS AND LIGACATION MOLECULES IN CANCER TREATMENT
WO1996007399A1 (en) 1994-09-09 1996-03-14 Takeda Chemical Industries, Ltd. Sustained release preparation containing metal salt of a peptide
US5681860A (en) 1993-09-21 1997-10-28 The Trustees Of Columbia University In The City Of New York Method of increasing expression of HLA, cell surface and TAA antigens of cells using 3-(N-acetylamino)-5-(N-decyl-N-methylamino)-benzyl alcohol
US5731168A (en) 1995-03-01 1998-03-24 Genentech, Inc. Method for making heteromultimeric polypeptides
US6316007B1 (en) 1995-04-04 2001-11-13 Wound Healing Of Oklahoma Combined physical and immunotherapy for cancer
SK281571B6 (en) 1995-06-07 2001-05-10 Alkermes Controlled Therapeutics, Inc. Composition for sustained release of human growth hormone
ZA965368B (en) 1995-07-14 1997-01-14 Novo Nordisk As A pharmaceutical formulation
US5871697A (en) * 1995-10-24 1999-02-16 Curagen Corporation Method and apparatus for identifying, classifying, or quantifying DNA sequences in a sample without sequencing
US6458939B1 (en) 1996-03-15 2002-10-01 Millennium Pharmaceuticals, Inc. Compositions and methods for the diagnosis, prevention, and treatment of neoplastic cell growth and proliferation
EP1039801A4 (en) 1997-06-06 2003-03-26 Human Genome Sciences Inc 207 human secreted proteins
US6525174B1 (en) * 1997-06-06 2003-02-25 Human Genome Sciences, Inc. Precerebellin-like protein
WO2001012660A2 (en) * 1999-08-17 2001-02-22 Sagami Chemical Research Center HUMAN PROTEINS HAVING HYDROPHOBIC DOMAINS AND DNAs ENCODING THESE PROTEINS
ATE424457T1 (en) * 2000-01-13 2009-03-15 Genentech Inc HUMAN STRA6 POLYPEPTIDES
EP1334126A1 (en) 2000-03-23 2003-08-13 Curagen Corporation Human stra6-like protein and nucleic acids encoding the same
US20030100710A1 (en) 2000-06-05 2003-05-29 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
US20030187201A1 (en) 2000-09-15 2003-10-02 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
US20030120040A1 (en) 2001-06-29 2003-06-26 Genentech, Inc. Secreted and Transmembrane polypeptides and nucleic acids encoding the same

Also Published As

Publication number Publication date
CA2397207A1 (en) 2001-07-19
ATE424457T1 (en) 2009-03-15
US20020156252A1 (en) 2002-10-24
EP1246917A2 (en) 2002-10-09
AU3276801A (en) 2001-07-24
AU2006241374A1 (en) 2006-12-14
US7939650B2 (en) 2011-05-10
JP5701624B2 (en) 2015-04-15
US20080299547A1 (en) 2008-12-04
JP2011103891A (en) 2011-06-02
AU2006241374B2 (en) 2009-10-22
CN1425066B (en) 2010-05-26
US20070025998A1 (en) 2007-02-01
US20070128188A1 (en) 2007-06-07
DK1246917T3 (en) 2009-06-08
WO2001051635A2 (en) 2001-07-19
PT1246917E (en) 2009-06-02
KR20020073168A (en) 2002-09-19
IL150676A0 (en) 2003-02-12
IL150676A (en) 2010-05-17
WO2001051635A3 (en) 2002-05-02
KR20080006002A (en) 2008-01-15
EP1246917B1 (en) 2009-03-04
US20090010936A1 (en) 2009-01-08
US7855278B2 (en) 2010-12-21
US20110171238A1 (en) 2011-07-14
US20020173461A1 (en) 2002-11-21
CN101643733A (en) 2010-02-10
US7741439B2 (en) 2010-06-22
CN1425066A (en) 2003-06-18
AU785055B2 (en) 2006-09-07
KR100929057B1 (en) 2009-11-30
DE60137829D1 (en) 2009-04-16
ES2323220T3 (en) 2009-07-09
US7173115B2 (en) 2007-02-06
NZ520095A (en) 2004-05-28
JP2003519491A (en) 2003-06-24

Similar Documents

Publication Publication Date Title
US7741439B2 (en) Isolated stra6 polypeptides
JP4260778B2 (en) Compositions and methods for the treatment of tumors
EP1607402B1 (en) Method for the diagnosis of tumour
US20050176104A1 (en) Compositions and methods for the treatment of tumor
US20080175842A1 (en) Compositions and methods for treatment
JP2003519491A5 (en)
EP1642968B1 (en) Composition and methods for the diagnosis of tumours
CA2420867C (en) Methods for enhancing the efficacy of cancer therapy
AU2007203291B2 (en) Methods for enhancing the efficacy of cancer therapy
ES2261559T3 (en) SECRETED AND TRANSMENBRANE POLYPEPTIDES AS WELL AS THE NUCLEIC ACIDS THAT CODE THEM.

Legal Events

Date Code Title Description
EEER Examination request
MKLA Lapsed

Effective date: 20180111